Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(51): 25839-25849, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31776254

RESUMO

Naive CD4+ T lymphocytes differentiate into different effector types, including helper and regulatory cells (Th and Treg, respectively). Heritable gene expression programs that define these effector types are established during differentiation, but little is known about the epigenetic mechanisms that install and maintain these programs. Here, we use mice defective for different components of heterochromatin-dependent gene silencing to investigate the epigenetic control of CD4+ T cell plasticity. We show that, upon T cell receptor (TCR) engagement, naive and regulatory T cells defective for TRIM28 (an epigenetic adaptor for histone binding modules) or for heterochromatin protein 1 ß and γ isoforms (HP1ß/γ, 2 histone-binding factors involved in gene silencing) fail to effectively signal through the PI3K-AKT-mTOR axis and switch to glycolysis. While differentiation of naive TRIM28-/- T cells into cytokine-producing effector T cells is impaired, resulting in reduced induction of autoimmune colitis, TRIM28-/- regulatory T cells also fail to expand in vivo and to suppress autoimmunity effectively. Using a combination of transcriptome and chromatin immunoprecipitation-sequencing (ChIP-seq) analyses for H3K9me3, H3K9Ac, and RNA polymerase II, we show that reduced effector differentiation correlates with impaired transcriptional silencing at distal regulatory regions of a defined set of Treg-associated genes, including, for example, NRP1 or Snai3. We conclude that TRIM28 and HP1ß/γ control metabolic reprograming through epigenetic silencing of a defined set of Treg-characteristic genes, thus allowing effective T cell expansion and differentiation into helper and regulatory phenotypes.


Assuntos
Diferenciação Celular/fisiologia , Reprogramação Celular/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Epigênese Genética/fisiologia , Linfócitos T/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Animais , Autoimunidade/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/genética , Plasticidade Celular/fisiologia , Reprogramação Celular/genética , Homólogo 5 da Proteína Cromobox , Colo/patologia , Citocinas/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Inativação Gênica , Histonas/metabolismo , Camundongos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Transcriptoma , Proteína 28 com Motivo Tripartido/genética
2.
Biochem J ; 477(5): 1033-1047, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-32091571

RESUMO

Hepatocellular carcinoma (HCC) is the most frequent primary liver cancer in adults. Among the altered pathways leading to HCC, an increasing role is attributed to abnormal epigenetic regulation. Members of the Heterochromatin Protein (HP1) 1 family are key players in chromatin organisation, acting as docking sites for chromatin modifiers. Here, we inactivated HP1α in HepG2 human liver carcinoma cells and showed that HP1α participated in cell proliferation. HP1α-depleted cells have a global decrease in DNA methylation and consequently a perturbed chromatin organisation, as exemplified by the reactivation of transcription at centromeric and pericentromeric regions, eventhough the protein levels of chromatin writers depositing methylation marks, such as EZH2, SETDB1, SUV39H1, G9A and DNMT3A remained unaltered. This decrease was attributed mainly to a low S-Adenosyl Methionine (SAM) level, a cofactor involved in methylation processes. Furthermore, we showed that this decrease was due to a modification in the Methionine adenosyl transferase 2A RNA (MAT2A) level, which modifies the ratio of MAT1A/MAT2A, two enzymes that generate SAM. Importantly, HP1α reintroduction into HP1α-depleted cells restored the MAT2A protein to its initial level. Finally, we demonstrated that this transcriptional deregulation of MAT2A in HP1α-depleted cells relied on a lack of recruitment of HP1ß and HP1γ to MAT2A promoter where an improper non-CpG methylation site was promoted in the vicinity of the transcription start site where HP1ß and HP1γ bound. Altogether, these results highlight an unanticipated link between HP1 and the SAM synthesis pathway, and emphasise emerging functions of HP1s as sensors of some aspects of liver cell metabolism.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas Cromossômicas não Histona/deficiência , Neoplasias Hepáticas/metabolismo , S-Adenosilmetionina/metabolismo , Vias Biossintéticas/fisiologia , Homólogo 5 da Proteína Cromobox , Células HEK293 , Células Hep G2 , Humanos
3.
Nucleic Acids Res ; 45(12): 7191-7211, 2017 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-28472341

RESUMO

In mammalian embryonic gonads, SOX9 is required for the determination of Sertoli cells that orchestrate testis morphogenesis. To identify genetic networks directly regulated by SOX9, we combined analysis of SOX9-bound chromatin regions from murine and bovine foetal testes with sequencing of RNA samples from mouse testes lacking Sox9. We found that SOX9 controls a conserved genetic programme that involves most of the sex-determining genes. In foetal testes, SOX9 modulates both transcription and directly or indirectly sex-specific differential splicing of its target genes through binding to genomic regions with sequence motifs that are conserved among mammals and that we called 'Sertoli Cell Signature' (SCS). The SCS is characterized by a precise organization of binding motifs for the Sertoli cell reprogramming factors SOX9, GATA4 and DMRT1. As SOX9 biological role in mammalian gonads is to determine Sertoli cells, we correlated this genomic signature with the presence of SOX9 on chromatin in foetal testes, therefore equating this signature to a genomic bar code of the fate of foetal Sertoli cells. Starting from the hypothesis that nuclear factors that bind to genomic regions with SCS could functionally interact with SOX9, we identified TRIM28 as a new SOX9 partner in foetal testes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Fatores de Transcrição SOX9/genética , Células de Sertoli/metabolismo , Transcriptoma , Animais , Bovinos , Cromatina/química , Cromatina/metabolismo , Embrião de Mamíferos , Feminino , Feto , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Redes Reguladoras de Genes , Masculino , Camundongos , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOX9/metabolismo , Análise de Sequência de RNA , Células de Sertoli/citologia , Processos de Determinação Sexual , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido
4.
EMBO J ; 33(22): 2606-22, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25216677

RESUMO

HP1 proteins are transcriptional regulators that, like histones, are targets for post-translational modifications defining an HP1-mediated subcode. HP1γ has multiple phosphorylation sites, including serine 83 (S83) that marks it to sites of active transcription. In a guinea pig model for Shigella enterocolitis, we observed that the defective type III secretion mxiD Shigella flexneri strain caused more HP1γ phosphorylation in the colon than the wild-type strain. Shigella interferes with HP1 phosphorylation by injecting the phospholyase OspF. This effector interacts with HP1γ and alters its phosphorylation at S83 by inactivating ERK and consequently MSK1, a downstream kinase. MSK1 that here arises as a novel HP1γ kinase, phosphorylates HP1γ at S83 in the context of an MSK1-HP1γ complex, and thereby favors its accumulation on its target genes. Genome-wide transcriptome analysis reveals that this mechanism is linked to up-regulation of proliferative gene and fine-tuning of immune gene expression. Thus, in addition to histones, bacteria control host transcription by modulating the activity of HP1 proteins, with potential implications in transcriptional reprogramming at the mucosal barrier.


Assuntos
Proteínas da Membrana Bacteriana Externa/metabolismo , Carbono-Oxigênio Liases/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Disenteria Bacilar/metabolismo , Enterocolite/metabolismo , Shigella flexneri/metabolismo , Transcriptoma , Animais , Proteínas da Membrana Bacteriana Externa/genética , Carbono-Oxigênio Liases/genética , Células Cultivadas , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Modelos Animais de Doenças , Disenteria Bacilar/genética , Disenteria Bacilar/patologia , Enterocolite/genética , Enterocolite/patologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Estudo de Associação Genômica Ampla , Cobaias , Camundongos , Camundongos Mutantes , Fosforilação , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Shigella flexneri/genética
5.
Nature ; 487(7406): 249-53, 2012 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-22763435

RESUMO

During immune responses, naive CD4+ T cells differentiate into several T helper (TH) cell subsets under the control of lineage-specifying genes. These subsets (TH1, TH2 and TH17 cells and regulatory T cells) secrete distinct cytokines and are involved in protection against different types of infection. Epigenetic mechanisms are involved in the regulation of these developmental programs, and correlations have been drawn between the levels of particular epigenetic marks and the activity or silencing of specifying genes during differentiation. Nevertheless, the functional relevance of the epigenetic pathways involved in TH cell subset differentiation and commitment is still unclear. Here we explore the role of the SUV39H1­H3K9me3­HP1α silencing pathway in the control of TH2 lineage stability. This pathway involves the histone methylase SUV39H1, which participates in the trimethylation of histone H3 on lysine 9 (H3K9me3), a modification that provides binding sites for heterochromatin protein 1α (HP1α) and promotes transcriptional silencing. This pathway was initially associated with heterochromatin formation and maintenance but can also contribute to the regulation of euchromatic genes. We now propose that the SUV39H1­H3K9me3­HP1α pathway participates in maintaining the silencing of TH1 loci, ensuring TH2 lineage stability. In TH2 cells that are deficient in SUV39H1, the ratio between trimethylated and acetylated H3K9 is impaired, and the binding of HP1α at the promoters of silenced TH1 genes is reduced. Despite showing normal differentiation, both SUV39H1-deficient TH2 cells and HP1α-deficient TH2 cells, in contrast to wild-type cells, expressed TH1 genes when recultured under conditions that drive differentiation into TH1 cells. In a mouse model of TH2-driven allergic asthma, the chemical inhibition or loss of SUV39H1 skewed T-cell responses towards TH1 responses and decreased the lung pathology. These results establish a link between the SUV39H1­H3K9me3­HP1α pathway and the stability of TH2 cells, and they identify potential targets for therapeutic intervention in TH2-cell-mediated inflammatory diseases.


Assuntos
Epigênese Genética , Células Th2/citologia , Células Th2/imunologia , Animais , Asma/enzimologia , Asma/imunologia , Asma/patologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Inativação Gênica , Histonas/metabolismo , Masculino , Metiltransferases/deficiência , Metiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Proteínas Repressoras/deficiência , Proteínas Repressoras/metabolismo , Células Th1/metabolismo , Células Th2/enzimologia
6.
Proc Natl Acad Sci U S A ; 109(49): 20083-8, 2012 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-23169648

RESUMO

T-cell receptor-α (TCRα) rearrangement in CD4(+)CD8(+) double-positive immature thymocytes is a prerequisite for production of αß T cells and invariant natural killer T cells. This developmental event is regulated by the TCRα enhancer (Eα), which induces chromatin modification and recruitment of the recombination-activating proteins Rag1 and Rag2. However, the molecular mechanism underlying the activation and long-range action of Eα remains incompletely understood. We show here that the chromatin-modifying factor TRIM28 is highly expressed in double-positive thymocytes and persistently phosphorylated at serine 473. TRIM28 binds to Eα and induces histone 3 lysine 4 trimethylation in the Eα and distant regions of the TCRα locus, coupled with recruitment of Rag proteins. T-cell-conditional ablation of TRIM28 impaired TCRα gene rearrangement and compromised the development of αß T cells and invariant natural killer T cells. These findings establish TRIM28 as a unique regulator of thymocyte development and highlight an epigenetic mechanism involving TRIM28-mediated active chromatin modification in the TCRα locus.


Assuntos
Diferenciação Celular/imunologia , Montagem e Desmontagem da Cromatina/fisiologia , Elementos Facilitadores Genéticos/genética , Células T Matadoras Naturais/citologia , Proteínas Nucleares/metabolismo , Receptores de Antígenos de Linfócitos T alfa-beta/metabolismo , Proteínas Repressoras/metabolismo , Subpopulações de Linfócitos T/citologia , Animais , Imunoprecipitação da Cromatina , Metilação de DNA , Citometria de Fluxo , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Immunoblotting , Camundongos , Camundongos Transgênicos , Células T Matadoras Naturais/imunologia , Fosforilação , Subpopulações de Linfócitos T/imunologia , Timócitos/metabolismo , Proteína 28 com Motivo Tripartido
7.
Proc Natl Acad Sci U S A ; 108(20): 8212-7, 2011 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-21531907

RESUMO

TRIM24 (TIF1α), TRIM28 (TIF1ß), and TRIM33 (TIF1γ) are three related cofactors belonging to the tripartite motif superfamily that interact with distinct transcription factors. TRIM24 interacts with the liganded retinoic acid (RA) receptor to repress its transcriptional activity. Germ line inactivation of TRIM24 in mice deregulates RA-signaling in hepatocytes leading to the development of hepatocellular carcinoma (HCC). Here we show that TRIM24 can be purified as at least two macromolecular complexes comprising either TRIM33 or TRIM33 and TRIM28. Somatic hepatocyte-specific inactivation of TRIM24, TRIM28, or TRIM33 all promote HCC in a cell-autonomous manner in mice. Moreover, HCC formation upon TRIM24 inactivation is strongly potentiated by further loss of TRIM33. These results demonstrate that the TIF1-related subfamily of TRIM proteins interact both physically and functionally to modulate HCC formation in mice.


Assuntos
Carcinoma Hepatocelular/etiologia , Neoplasias Hepáticas/etiologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Transformação Celular Neoplásica/patologia , Hepatócitos/patologia , Neoplasias Hepáticas/patologia , Camundongos , Complexos Multiproteicos/isolamento & purificação , Complexos Multiproteicos/fisiologia , Ligação Proteica , Receptores do Ácido Retinoico , Proteína 28 com Motivo Tripartido
8.
Dev Biol ; 350(2): 548-58, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21163256

RESUMO

TIF1ß is an essential mammalian transcriptional corepressor. It interacts with the heterochromatin proteins HP1 through a highly conserved motif, the HP1box, and we have previously shown that this interaction is essential for the differentiation of F9 cells to occur. Here we address the in vivo functions of the TIF1ß-HP1 interaction, by generating mice in which the TIF1ß HP1box is mutated, leading to the loss of TIF1ß interaction with HP1. The effects of the mutation were monitored in two instances, where TIF1ß is known to play key roles: early embryonic development and spermatogenesis. We find that mutating the HP1box of TIF1ß disrupts embryonic development soon after gastrulation. This effect is likely caused by the misexpression of TIF1ß targets that regulate mitotic progression and pluripotency. In contrast, in Sertoli cells, we found that the absence of TIF1ß but not its mutation in the HP1box leads to a clear defect of spermatogenesis characterized by a failure of spermatid release and a testicular degeneration. These data show that the interaction between TIF1ß and HP1 is essential for some but not all TIF1ß functions in vivo. Furthermore, we observed that TIF1ß is dispersed through the nucleoplasm of E7.0 embryos, whereas it is mainly associated with pericentromeric heterochromatin of E8.5 embryos and of Sertoli cells, an association that is lost upon TIF1ß HP1box mutation. Altogether, these data provide strong evidence that nuclear organization plays key roles during early embryonic development.


Assuntos
Proteínas Cromossômicas não Histona/fisiologia , Gastrulação , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Células de Sertoli/fisiologia , Espermatogênese , Animais , Ciclo Celular , Homólogo 5 da Proteína Cromobox , Desenvolvimento Embrionário , Feminino , Proteínas de Homeodomínio/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Homeobox Nanog , Proteínas Nucleares/genética , Fator 3 de Transcrição de Octâmero/genética , Proteínas Repressoras/genética , Proteína 28 com Motivo Tripartido
9.
Adv Exp Med Biol ; 770: 59-76, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23631000

RESUMO

Members of the tripartite motif (TRIM) protein family are found in all multicellular eukaryotes and function in a wide range of cellular processes such as cell cycle regulation, differentiation, development, oncogenesis and viral response. Over the past few years, several TRIM proteins have been reported to control gene expression through regulation of the transcriptional activity of numerous sequence-specific transcription factors. These proteins include the transcriptional intermediary factor 1 (TIF1) regulators, the promyelocytic leukemia tumor suppressor PML and the RET finger protein (RFP). In this chapter, we will consider the molecular interactions made by these TRIM proteins and will attempt to clarify some of the molecular mechanisms underlying their regulatory effect on transcription.


Assuntos
Motivos de Aminoácidos , Regulação da Expressão Gênica , Proteínas/química , Proteínas/metabolismo , Transcrição Gênica , Animais , Humanos
10.
Cells ; 11(15)2022 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-35954237

RESUMO

About half of the mammalian genome is constituted of repeated elements, among which endogenous retroviruses (ERVs) are known to influence gene expression and cancer development. The HP1 (Heterochromatin Protein 1) proteins are known to be essential for heterochromatin establishment and function and its loss in hepatocytes leads to the reactivation of specific ERVs and to liver tumorigenesis. Here, by studying two ERVs located upstream of genes upregulated upon loss of HP1, Mbd1 and Trim24, we show that these HP1-dependent ERVs behave as either alternative promoters or as putative enhancers forming a loop with promoters of endogenous genes depending on the genomic context and HP1 expression level. These ERVs are characterised by a specific HP1-independent enrichment in heterochromatin-associated marks H3K9me3 and H4K20me3 as well as in the enhancer-specific mark H3K4me1, a combination that might represent a bookmark of putative ERV-derived enhancers. These ERVs are further enriched in a HP1-dependent manner in H3K27me3, suggesting a critical role of this mark together with HP1 in the silencing of the ERVs, as well as for the repression of the associated genes. Altogether, these results lead to the identification of a new regulatory hub involving the HP1-dependent formation of a physical loop between specific ERVs and endogenous genes.


Assuntos
Retrovirus Endógenos , Animais , Cromatina/genética , Homólogo 5 da Proteína Cromobox , Retrovirus Endógenos/genética , Expressão Gênica , Heterocromatina , Mamíferos/genética
11.
Nat Commun ; 13(1): 4412, 2022 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-35906245

RESUMO

Gonadal sexual fate in mammals is determined during embryonic development and must be actively maintained in adulthood. In the mouse ovary, oestrogen receptors and FOXL2 protect ovarian granulosa cells from transdifferentiation into Sertoli cells, their testicular counterpart. However, the mechanism underlying their protective effect is unknown. Here, we show that TRIM28 is required to prevent female-to-male sex reversal of the mouse ovary after birth. We found that upon loss of Trim28, ovarian granulosa cells transdifferentiate to Sertoli cells through an intermediate cell type, different from gonadal embryonic progenitors. TRIM28 is recruited on chromatin in the proximity of FOXL2 to maintain the ovarian pathway and to repress testicular-specific genes. The role of TRIM28 in ovarian maintenance depends on its E3-SUMO ligase activity that regulates the sex-specific SUMOylation profile of ovarian-specific genes. Our study identifies TRIM28 as a key factor in protecting the adult ovary from the testicular pathway.


Assuntos
Ovário , Sumoilação , Animais , Feminino , Masculino , Mamíferos/metabolismo , Camundongos , Ovário/metabolismo , Células de Sertoli/metabolismo , Testículo/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido/genética , Proteína 28 com Motivo Tripartido/metabolismo
12.
Oncogene ; 39(13): 2676-2691, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32020053

RESUMO

Chromatin organization is essential for appropriate interpretation of the genetic information. Here, we demonstrated that the chromatin-associated proteins HP1 are dispensable for hepatocytes survival but are essential within hepatocytes to prevent liver tumor development in mice with HP1ß being pivotal in these functions. Yet, we found that the loss of HP1 per se is not sufficient to induce cell transformation but renders cells more resistant to specific stress such as the expression of oncogenes and thus in fine, more prone to cell transformation. Molecular characterization of HP1-Triple KO premalignant livers and BMEL cells revealed that HP1 are essential for the maintenance of heterochromatin organization and for the regulation of specific genes with most of them having well characterized functions in liver functions and homeostasis. We further showed that some specific retrotransposons get reactivated upon loss of HP1, correlating with overexpression of genes in their neighborhood. Interestingly, we found that, although HP1-dependent genes are characterized by enrichment H3K9me3, this mark does not require HP1 for its maintenance and is not sufficient to maintain gene repression in absence of HP1. Finally, we demonstrated that the loss of TRIM28 association with HP1 recapitulated several phenotypes induced by the loss of HP1 including the reactivation of some retrotransposons and the increased incidence of liver cancer development. Altogether, our findings indicate that HP1 proteins act as guardians of liver homeostasis to prevent tumor development by modulating multiple chromatin-associated events within both the heterochromatic and euchromatic compartments, partly through regulation of the corepressor TRIM28 activity.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas Cromossômicas não Histona/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Animais , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Modelos Animais de Doenças , Feminino , Hepatócitos , Heterocromatina/metabolismo , Humanos , Fígado/citologia , Fígado/patologia , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Camundongos Knockout , Ligação Proteica/genética , RNA-Seq , Retroelementos/genética , Proteína 28 com Motivo Tripartido/metabolismo
13.
J Virol ; 82(9): 4675-9, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18287239

RESUMO

TRIM28 is a transcriptional corepressor which is required for primer binding site (PBS)-dependent restriction of murine leukemia virus (MLV) replication in embryonic stem and embryonic carcinoma (EC) cells. PBS-dependent restriction of MLV leads to transcriptional silencing of the integrated provirus and has been shown to correlate with TRIM28-mediated recruitment of HP1 to the silenced loci. Here we show, using a cell line with a point mutation in the HP1 binding domain of TRIM28, that interaction with HP1 is absolutely required for the PBS-dependent restriction of MLV in the F9 EC cell line.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Vírus da Leucemia Murina/fisiologia , Proteínas Repressoras/metabolismo , Replicação Viral , Animais , Sítios de Ligação , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Células-Tronco de Carcinoma Embrionário/virologia , Células-Tronco Embrionárias/virologia , Camundongos , Proteínas Nucleares/metabolismo , Mutação Puntual , Ligação Proteica/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido
14.
FASEB J ; 22(11): 3853-65, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18676401

RESUMO

Recent advances reveal emerging unique functions of poly(ADP-ribose) polymerase-1 (Parp-1) and Parp-2 in heterochromatin integrity and cell differentiation. However, the chromatin-mediated molecular and cellular events involved remain elusive. Here we describe specific physical and functional interactions of Parp-1 and Parp-2 with the transcriptional intermediary factor (TIF1beta) and the heterochromatin proteins (HP1) that affect endodermal differentiation. We show that Parp-2 binds to TIF1beta with high affinity both directly and through HP1alpha. Both partners colocalize at pericentric heterochromatin in primitive endoderm-like cells. Parp-2 also binds to HP1beta but not to HP1gamma. In contrast Parp-1 binds weakly to TIF1beta and HP1beta only. Both Parps selectively poly(ADP-ribosyl)ate HP1alpha. Using shRNA approaches, we provide evidence for distinct participation of both Parps in endodermal differentiation. Whereas Parp-2 and its activity are required for the relocation of TIF1beta to heterochromatic foci during primitive endodermal differentiation, Parp-1 and its activity modulate TIF1beta-HP1alpha association with consequences on parietal endodermal differentiation. Both Parps control TIF1beta transcriptional activity. In addition, this work identifies both Parps as new modulators of the HP1-mediated subcode histone.-Quénet, D., Gasser, V., Fouillen, L., Cammas, F., Sanglier-Cianferani, S., Losson, R., Dantzer, F. The histone subcode: poly(ADP-ribose) polymerase-1 (Parp-1) and Parp-2 control cell differentiation by regulating the transcriptional intermediary factor TIF1beta and the heterochromatin protein HP1alpha.


Assuntos
Diferenciação Celular/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Proteínas de Ligação a DNA/metabolismo , Endoderma/enzimologia , Heterocromatina/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Repressoras/metabolismo , Linhagem Celular , Homólogo 5 da Proteína Cromobox , Endoderma/citologia , Humanos , Poli(ADP-Ribose) Polimerase-1 , Proteína 28 com Motivo Tripartido
16.
Cell Rep ; 10(1): 20-8, 2015 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-25543143

RESUMO

TRIM28 is a corepressor that mediates transcriptional silencing by establishing local heterochromatin. Here, we show that deletion of TRIM28 in neural progenitor cells (NPCs) results in high-level expression of two groups of endogenous retroviruses (ERVs): IAP1 and MMERVK10C. We find that NPCs use TRIM28-mediated histone modifications to dynamically regulate transcription and silencing of ERVs, which is in contrast to other somatic cell types using DNA methylation. We also show that derepression of ERVs influences transcriptional dynamics in NPCs through the activation of nearby genes and the expression of long noncoding RNAs. These findings demonstrate a unique dynamic transcriptional regulation of ERVs in NPCs. Our results warrant future studies on the role of ERVs in the healthy and diseased brain.


Assuntos
Retrovirus Endógenos/genética , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Transcrição Gênica , Animais , Metilação de DNA/genética , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/patogenicidade , Regulação da Expressão Gênica no Desenvolvimento , Heterocromatina/genética , Histonas/metabolismo , Humanos , Camundongos , Neurônios/virologia , Proteínas Nucleares/biossíntese , Proteínas Repressoras/biossíntese , Células-Tronco/metabolismo , Células-Tronco/virologia , Proteína 28 com Motivo Tripartido
17.
Nat Struct Mol Biol ; 20(3): 339-46, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23377542

RESUMO

Trim24 (Tif1α) and Trim33 (Tif1γ) interact to form a co-repressor complex that suppresses murine hepatocellular carcinoma. Here we show that Trim24 and Trim33 cooperatively repress retinoic acid receptor-dependent activity of VL30-class endogenous retroviruses (ERVs) in liver. In Trim24-knockout hepatocytes, VL30 derepression leads to accumulation of reverse-transcribed VL30 cDNA in the cytoplasm that correlates with activation of the viral-defense interferon responses mimicking the preneoplastic inflammatory state seen in human liver following exogenous viral infection. Furthermore, upon derepression, VL30 long terminal repeats (LTRs) act as promoter and enhancer elements deregulating expression of neighboring genes and generating enhancer RNAs that are required for LTR enhancer activity in hepatocytes in vivo. These data reinforce the role of the TRIM family of proteins in retroviral restriction and antiviral defense and provide an example of an ERV-derived oncogenic regulatory network.


Assuntos
Proteínas Nucleares/metabolismo , RNA não Traduzido , Retroelementos/genética , Retroviridae/genética , Fatores de Transcrição/metabolismo , Animais , Carcinoma Hepatocelular/genética , Proteínas Correpressoras/genética , Proteínas Correpressoras/metabolismo , Citoplasma/genética , DNA Complementar , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Hepatócitos/citologia , Hepatócitos/fisiologia , Neoplasias Hepáticas/genética , Camundongos , Camundongos Mutantes , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Sequências Repetidas Terminais , Fatores de Transcrição/genética
18.
J Exp Med ; 208(8): 1649-60, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21746811

RESUMO

Immunoglobulin class switch recombination (CSR) is initiated by double-stranded DNA breaks (DSBs) in switch regions triggered by activation-induced cytidine deaminase (AID). Although CSR correlates with epigenetic modifications at the IgH locus, the relationship between these modifications and AID remains unknown. In this study, we show that during CSR, AID forms a complex with KAP1 (KRAB domain-associated protein 1) and HP1 (heterochromatin protein 1) that is tethered to the donor switch region (Sµ) bearing H3K9me3 (trimethylated histone H3 at lysine 9) in vivo. Furthermore, in vivo disruption of this complex results in impaired AID recruitment to Sµ, inefficient DSB formation, and a concomitant defect in CSR but not in somatic hypermutation. We propose that KAP1 and HP1 tether AID to H3K9me3 residues at the donor switch region, thus providing a mechanism linking AID to epigenetic modifications during CSR.


Assuntos
Linfócitos B/citologia , Citidina Desaminase/imunologia , Epigênese Genética/imunologia , Switching de Imunoglobulina/imunologia , Região de Troca de Imunoglobulinas/imunologia , Proteínas Nucleares/imunologia , Proteínas Repressoras/imunologia , Animais , Linfócitos B/imunologia , Western Blotting , Imunoprecipitação da Cromatina , Cromatografia em Gel , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/metabolismo , Análise Mutacional de DNA , Primers do DNA/genética , Citometria de Fluxo , Histonas/metabolismo , Hibridização in Situ Fluorescente , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína 28 com Motivo Tripartido
19.
Mol Biol Cell ; 20(1): 296-305, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18923144

RESUMO

Here, we identified the imprinted mesoderm-specific transcript (MEST) gene as an endogenous TIF1beta primary target gene and demonstrated that transcriptional intermediary factor (TIF) 1beta, through its interaction with heterochromatin protein (HP) 1, is essential in establishing and maintaining a local heterochromatin-like structure on MEST promoter region characterized by H3K9 trimethylation and hypoacetylation, H4K20 trimethylation, DNA hypermethylation, and enrichment in HP1 that correlates with preferential association to foci of pericentromeric heterochromatin and transcriptional repression. On disruption of the interaction between TIF1beta and HP1, TIF1beta is released from the promoter region, and there is a switch from DNA hypermethylation and histone H3K9 trimethylation to DNA hypomethylation and histone H3K27 trimethylation correlating with rapid reactivation of MEST expression. Interestingly, we provide evidence that the imprinted MEST allele DNA methylation is insensitive to TIF1beta loss of function, whereas the nonimprinted allele is regulated through a distinct TIF1beta-DNA methylation mechanism.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Metilação de DNA , Impressão Genômica , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas/genética , Fatores de Transcrição/metabolismo , Animais , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Heterocromatina/metabolismo , Histonas/genética , Humanos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Isoformas de Proteínas/genética , Proteínas/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica
20.
Neuron ; 60(5): 818-31, 2008 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-19081377

RESUMO

KAP1 is an essential cofactor of KRAB-zinc finger proteins, a family of vertebrate-specific epigenetic repressors of largely unknown functions encoded in the hundreds by the mouse and human genomes. Here, we report that KAP1 is expressed at high levels and necessary for KRAB-mediated repression in mature neurons of the mouse brain. Mice deleted for KAP1 in the adult forebrain exhibit heightened levels of anxiety-like and exploratory activity and stress-induced alterations in spatial learning and memory. In the hippocampus, a small number of genes are dysregulated, including some imprinted genes. Chromatin analyses of the promoters of two genes markedly upregulated in knockout mice reveal decreased histone 3 K9-trimethylation and increased histone 3 and histone 4 acetylation. We propose a model in which the tethering of KAP1-associated chromatin remodeling factors via KRAB-ZFPs epigenetically controls gene expression in the hippocampus, thereby conditioning responses to behavioral stress.


Assuntos
Comportamento Animal/fisiologia , Proteínas Nucleares/fisiologia , Prosencéfalo/metabolismo , Proteínas Repressoras/fisiologia , Estresse Psicológico/patologia , Estresse Psicológico/fisiopatologia , Regulação para Cima/genética , Acetilação , Análise de Variância , Animais , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Imunoprecipitação da Cromatina/métodos , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Feminino , Hipocampo/metabolismo , Histonas/metabolismo , Locomoção/genética , Masculino , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Camundongos , Camundongos Transgênicos , Análise em Microsséries/métodos , Modelos Biológicos , Mutação , Estresse Psicológico/genética , Proteína 28 com Motivo Tripartido
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA