Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 42(4): 438-50, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21596310

RESUMO

We have identified human MBT domain-containing protein L3MBTL2 as an integral component of a protein complex that we termed Polycomb repressive complex 1 (PRC1)-like 4 (PRC1L4), given the copresence of PcG proteins RING1, RING2, and PCGF6/MBLR. PRC1L4 also contained E2F6 and CBX3/HP1γ, known to function in transcriptional repression. PRC1L4-mediated repression necessitated L3MBTL2 that compacted chromatin in a histone modification-independent manner. Genome-wide location analyses identified several hundred genes simultaneously bound by L3MBTL2 and E2F6, preferentially around transcriptional start sites that exhibited little overlap with those targeted by other E2Fs or by L3MBTL1, another MBT domain-containing protein that interacts with RB1. L3MBTL2-specific RNAi resulted in increased expression of target genes that exhibited a significant reduction in H2A lysine 119 monoubiquitination. Our findings highlight a PcG/MBT collaboration that attains repressive chromatin without entailing histone lysine methylation marks.


Assuntos
Cromatina/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Ubiquitinação , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Células HEK293 , Histonas/genética , Humanos , Proteínas Nucleares/genética , Proteínas do Grupo Polycomb , Proteínas Repressoras/genética , Fatores de Transcrição/genética
2.
Development ; 140(1): 167-75, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23154409

RESUMO

Transforming growth factor ß (TGFß) is widely recognised as an important factor that regulates many steps of normal mammary gland (MG) development, including branching morphogenesis, functional differentiation and involution. Tif1γ has previously been reported to temporally and spatially control TGFß signalling during early vertebrate development by exerting negative effects over SMAD4 availability. To evaluate the contribution of Tif1 γ to MG development, we developed a Cre/LoxP system to specifically invalidate the Tif1g gene in mammary epithelial cells in vivo. Tif1g-null mammary gland development appeared to be normal and no defects were observed during the lifespan of virgin mice. However, a lactation defect was observed in mammary glands of Tif1g-null mice. We demonstrate that Tif1 γ is essential for the terminal differentiation of alveolar epithelial cells at the end of pregnancy and to ensure lactation. Tif1 γ appears to play a crucial role in the crosstalk between TGFß and prolactin pathways by negatively regulating both PRL receptor expression and STAT5 phosphorylation, thereby impairing the subsequent transactivation of PRL target genes. Using HC11 cells as a model, we demonstrate that the effects of Tif1g knockdown on lactation depend on both SMAD4 and TGFß. Interestingly, we found that the Tif1γ expression pattern in mammary epithelial cells is almost symmetrically opposite to that described for TGFß. We propose that Tif1γ contributes to the repression of TGFß activity during late pregnancy and prevents lactation by inhibiting SMAD4.


Assuntos
Diferenciação Celular/genética , Células Epiteliais/citologia , Lactação/genética , Glândulas Mamárias Animais/citologia , Proteína Smad4/antagonistas & inibidores , Fatores de Transcrição/fisiologia , Animais , Células Epiteliais/fisiologia , Feminino , Masculino , Glândulas Mamárias Animais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Gravidez , Transdução de Sinais/genética , Proteína Smad4/fisiologia
3.
Blood ; 122(23): 3798-807, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24092935

RESUMO

In previous mass spectrometry and coimmune precipitation studies, we identified tripartite motif-containing 28 (TRIM28; also known as transcriptional intermediary factor1ß and Krüppel-associated box-associated protein-1) as a cofactor that specifically copurified with an NR2C1/NR2C2 (TR2/TR4) orphan nuclear receptor heterodimer that previous studies had implicated as an embryonic/fetal ß-type globin gene repressor. TRIM28 has been characterized as a transcriptional corepressor that can associate with many different transcription factors and can play functional roles in multiple tissues and cell types. Here, we tested the contribution of TRIM28 to globin gene regulation and erythropoiesis using a conditional loss-of-function in vivo model. We discovered that Trim28 genetic loss in the adult mouse leads to defective immature erythropoiesis in the bone marrow and consequently to anemia. We further found that TRIM28 controls erythropoiesis in a cell-autonomous manner by inducibly deleting Trim28 exclusively in hematopoietic cells. Finally, in the absence of TRIM28, we observed increased apoptosis as well as diminished expression of multiple erythroid transcription factors and heme biosynthetic enzymes in immature erythroid cells. Thus, TRIM28 is essential for the cell-autonomous development of immature erythroblasts in the bone marrow.


Assuntos
Eritroblastos/citologia , Eritroblastos/metabolismo , Proteínas Nucleares/fisiologia , Proteínas Repressoras/fisiologia , Anemia/genética , Anemia/patologia , Animais , Apoptose/genética , Apoptose/fisiologia , Medula Óssea/metabolismo , Medula Óssea/patologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Eritropoese/genética , Eritropoese/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Fatores de Transcrição/metabolismo , Proteína 28 com Motivo Tripartido , Globinas beta/genética
4.
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
5.
Development ; 137(15): 2571-8, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20573697

RESUMO

The definition of embryonic potency and induction of specific cell fates are intimately linked to the tight control over TGFbeta signaling. Although extracellular regulation of ligand availability has received considerable attention in recent years, surprisingly little is known about the intracellular factors that negatively control Smad activity in mammalian tissues. By means of genetic ablation, we show that the Smad4 inhibitor ectodermin (Ecto, also known as Trim33 or Tif1gamma) is required to limit Nodal responsiveness in vivo. New phenotypes, which are linked to excessive Nodal activity, emerge from such a modified landscape of Smad responsiveness in both embryonic and extra-embryonic territories. In extra-embryonic endoderm, Ecto is required to confine expression of Nodal antagonists to the anterior visceral endoderm. In trophoblast cells, Ecto precisely doses Nodal activity, balancing stem cell self-renewal and differentiation. Epiblast-specific Ecto deficiency shifts mesoderm fates towards node/organizer fates, revealing the requirement of Smad inhibition for the precise allocation of cells along the primitive streak. This study unveils that intracellular negative control of Smad function by ectodermin/Tif1gamma is a crucial element in the cellular response to TGFbeta signals in mammalian tissues.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Smad/metabolismo , Fatores de Transcrição/metabolismo , Alelos , Animais , Padronização Corporal , Diferenciação Celular , Cruzamentos Genéticos , Ectoderma/metabolismo , Mesoderma/metabolismo , Camundongos , Modelos Biológicos , Fenótipo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
6.
Am J Pathol ; 180(6): 2214-21, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22469842

RESUMO

Transcriptional intermediary factor 1γ (TIF1γ; alias, TRIM33/RFG7/PTC7/ectodermin) belongs to an evolutionarily conserved family of nuclear factors that have been implicated in stem cell pluripotency, embryonic development, and tumor suppression. TIF1γ expression is markedly down-regulated in human pancreatic tumors, and Pdx1-driven Tif1γ inactivation cooperates with the Kras(G12D) oncogene in the mouse pancreas to induce intraductal papillary mucinous neoplasms. In this study, we report that aged Pdx1-Cre; LSL-Kras(G12D); Tif1γ(lox/lox) mice develop pancreatic ductal adenocarcinomas (PDACs), an aggressive and always fatal neoplasm, demonstrating a Tif1γ tumor-suppressive function in the development of pancreatic carcinogenesis. Deletion of SMAD4/DPC4 (deleted in pancreatic carcinoma locus 4) occurs in approximately 50% of human cases of PDAC. We, therefore, assessed the genetic relationship between Tif1γ and Smad4 signaling in pancreatic tumors and found that Pdx1-Cre; LSL-Kras(G12D); Smad4(lox/lox); Tif1γ(lox/lox) (alias, KSSTT) mutant mice exhibit accelerated tumor progression. Consequently, Tif1γ tumor-suppressor effects during progression from a premalignant to a malignant state in our mouse model of pancreatic cancer are independent of Smad4. These findings establish, for the first time to our knowledge, that Tif1γ and Smad4 both regulate an intraductal papillary mucinous neoplasm-to-PDAC sequence through distinct tumor-suppressor programs.


Assuntos
Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Proteína Smad4/genética , Fatores de Transcrição/genética , Animais , Carcinoma Ductal Pancreático/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Progressão da Doença , Deleção de Genes , Genes Supressores de Tumor , Predisposição Genética para Doença , Imageamento por Ressonância Magnética , Camundongos , Camundongos Mutantes , Neoplasias Pancreáticas/patologia , Lesões Pré-Cancerosas/genética , Transdução de Sinais/genética , Proteína Smad4/fisiologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/fisiologia
7.
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
8.
J Biol Chem ; 286(38): 33369-79, 2011 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-21768647

RESUMO

Recent genetic studies in mice have established that the nuclear receptor coregulator Trim24/Tif1α suppresses hepatocarcinogenesis by inhibiting retinoic acid receptor α (Rara)-dependent transcription and cell proliferation. However, Rara targets regulated by Trim24 remain unknown. We report that the loss of Trim24 resulted in interferon (IFN)/STAT pathway overactivation soon after birth (week 5). Despite a transient attenuation of this pathway by the induction of several IFN/STAT pathway repressors later in the disease, this phenomenon became more pronounced in tumors. Remarkably, Rara haplodeficiency, which suppresses tumorigenesis in Trim24(-/-) mice, prevented IFN/STAT overactivation. Moreover, together with Rara, Trim24 bound to the retinoic acid-responsive element of the Stat1 promoter and repressed its retinoic acid-induced transcription. Altogether, these results identify Trim24 as a novel negative regulator of the IFN/STAT pathway and suggest that this repression through Rara inhibition may prevent liver cancer.


Assuntos
Interferons/metabolismo , Proteínas Nucleares/metabolismo , Receptores do Ácido Retinoico/antagonistas & inibidores , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Carcinoma Hepatocelular/genética , Análise por Conglomerados , Dosagem de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Neoplasias Hepáticas/genética , Camundongos , Modelos Biológicos , Proteínas Nucleares/deficiência , Receptores do Ácido Retinoico/metabolismo , Reprodutibilidade dos Testes , Receptor alfa de Ácido Retinoico , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Fatores de Transcrição/deficiência , Transcriptoma , Tretinoína/farmacologia
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.
Proc Natl Acad Sci U S A ; 106(51): 21830-5, 2009 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-20018718

RESUMO

Sotos syndrome is an autosomal dominant condition characterized by overgrowth resulting in tall stature and macrocephaly, together with an increased risk of tumorigenesis. The disease is caused by loss-of-function mutations and deletions of the nuclear receptor SET domain containing protein-1 (NSD1) gene, which encodes a histone methyltransferase involved in chromatin regulation. However, despite its causal role in Sotos syndrome and the typical accelerated growth of these patients, little is known about the putative contribution of NSD1 to human sporadic malignancies. Here, we report that NSD1 function is abrogated in human neuroblastoma and glioma cells by transcriptional silencing associated with CpG island-promoter hypermethylation. We also demonstrate that the epigenetic inactivation of NSD1 in transformed cells leads to the specifically diminished methylation of the histone lysine residues H4-K20 and H3-K36. The described phenotype is also observed in Sotos syndrome patients with NSD1 genetic disruption. Expression microarray data from NSD1-depleted cells, followed by ChIP analysis, revealed that the oncogene MEIS1 is one of the main NSD1 targets in neuroblastoma. Furthermore, we show that the restoration of NSD1 expression induces tumor suppressor-like features, such as reduced colony formation density and inhibition of cellular growth. Screening a large collection of different tumor types revealed that NSD1 CpG island hypermethylation was a common event in neuroblastomas and gliomas. Most importantly, NSD1 hypermethylation was a predictor of poor outcome in high-risk neuroblastoma. These findings highlight the importance of NSD1 epigenetic inactivation in neuroblastoma and glioma that leads to a disrupted histone methylation landscape and might have a translational value as a prognostic marker.


Assuntos
Anormalidades Múltiplas/genética , Epigênese Genética , Glioma/enzimologia , Transtornos do Crescimento/genética , Histona-Lisina N-Metiltransferase/genética , Neuroblastoma/enzimologia , Imunoprecipitação da Cromatina , Ilhas de CpG , Metilação de DNA , Glioma/genética , Histona Metiltransferases , Humanos , Neuroblastoma/genética , Regiões Promotoras Genéticas , Síndrome
11.
PLoS Genet ; 5(7): e1000575, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19629168

RESUMO

Inactivation of the Transforming Growth Factor Beta (TGFbeta) tumor suppressor pathway contributes to the progression of Pancreatic Ductal AdenoCarcinoma (PDAC) since it is inactivated in virtually all cases of this malignancy. Genetic lesions inactivating this pathway contribute to pancreatic tumor progression in mouse models. Transcriptional Intermediary Factor 1 gamma (TIF1gamma) has recently been proposed to be involved in TGFbeta signaling, functioning as either a positive or negative regulator of the pathway. Here, we addressed the role of TIF1gamma in pancreatic carcinogenesis. Using conditional Tif1gamma knockout mice (Tif1gamma(lox/lox)), we selectively abrogated Tif1gamma expression in the pancreas of Pdx1-Cre;Tif1gamma(lox/lox) mice. We also generated Pdx1-Cre;LSL-Kras(G12D);Tif1gamma(lox/lox) mice to address the effect of Tif1gamma loss-of-function in precancerous lesions induced by oncogenic Kras(G12D). Finally, we analyzed TIF1gamma expression in human pancreatic tumors. In our mouse model, we showed that Tif1gamma was dispensable for normal pancreatic development but cooperated with Kras activation to induce pancreatic tumors reminiscent of human Intraductal Papillary Mucinous Neoplasms (IPMNs). Interestingly, these cystic lesions resemble those observed in Pdx1-Cre;LSL-Kras(G12D);Smad4(lox/lox) mice described by others. However, distinctive characteristics, such as the systematic presence of endocrine pseudo-islets within the papillary projections, suggest that SMAD4 and TIF1gamma don't have strictly redundant functions. Finally, we report that TIF1gamma expression is markedly down-regulated in human pancreatic tumors by quantitative RT-PCR and immunohistochemistry supporting the relevance of these findings to human malignancy. This study suggests that TIF1gamma is critical for tumor suppression in the pancreas, brings new insight into the genetics of pancreatic cancer, and constitutes a promising model to decipher the respective roles of SMAD4 and TIF1gamma in the multifaceted functions of TGFbeta in carcinogenesis and development.


Assuntos
Carcinoma Ductal Pancreático/metabolismo , Genes Supressores de Tumor , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Transcrição/metabolismo , Animais , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Humanos , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Fatores de Transcrição/genética
12.
Biochim Biophys Acta ; 1799(5-6): 463-8, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20176155

RESUMO

The NSD1 histone methyltransferase is involved in the outgrowth disorders Sotos and Weaver syndromes and childhood acute myeloid leukemia. NSD1 is a bona fida transcriptional co-repressor for Nizp1 which is a protein including SCAN, KRAB, C2HR and zinc-finger domains. In this study the Nizp1 KRAB-domain was identified to possess an intrinsic transcriptional activation capacity suppressed in cis by the presence of the C2HR domain. Oppositely, the KRAB-domain supported C2HR domain mediated transcriptional repression. The presence of the KRAB-domain resulted in increased NSD1 co-repressor association with the C2HR domain. This study shows a new function of the KRAB-domain, C2HR-domain, and the associated factors to confer Nizp1 mediated transcriptional regulation.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Transativadores/química , Transativadores/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Sítios de Ligação/genética , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Técnicas In Vitro , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Domínios e Motivos de Interação entre Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homologia de Sequência de Aminoácidos , Transativadores/genética , Transfecção , Dedos de Zinco/genética
13.
Biochim Biophys Acta ; 1799(8): 539-45, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20538088

RESUMO

The NSD1 histone methyl transferase is involved in childhood acute myeloid leukemia and the outgrowth disorders Sotos and Weaver syndromes. NSD1 is a transcriptional co-repressor for the zinc finger protein Nizp1 (also abbreviated Zfp496 and Zkscan17). Nizp1 includes a SCAN-domain, a KRAB-domain, four C2H2 Krüppel related zinc fingers, and a C2HR transcriptional repression and protein interaction domain required for NSD1 interaction. In this study we have identified alternative splicing of the Nizp1 gene resulting in transcripts encoding Nizp1 protein isoforms with a short N-terminal deletion or a SCAN-domain deletion. The alternative Nizp1 transcripts are expressed in lower levels relative to the canonical Nizp1 transcript. The Nizp1 SCAN-domain mediates Nizp1 self-association but lacks intrinsic transcriptional activating or repressing capacity and has no influence on the transcriptional repression activity of Nizp1 in reporter assays. Sub-cellular localization analysis showed that a fraction of Nizp1 localizes to CBP nuclear bodies and that the SCAN-domain is required for the localization to nuclear bodies. The presented results show that alternative splicing is a functional mechanism to generate Nizp1 protein isoforms with different SCAN-domain compositions and accordingly different sub-cellular localizations.


Assuntos
Processamento Alternativo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Domínios e Motivos de Interação entre Proteínas/genética , Dedos de Zinco , Animais , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Imunofluorescência , Células Hep G2 , Humanos , Rim/citologia , Rim/metabolismo , Camundongos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Transporte Proteico , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saccharomyces cerevisiae , Transcrição Gênica
14.
Proc Natl Acad Sci U S A ; 105(7): 2598-603, 2008 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-18287084

RESUMO

Calcification of arteries is a major risk factor for cardiovascular mortality in humans. Using genetic approaches, we demonstrate here that the transcriptional intermediary factor 1alpha (TIF1alpha), recently shown to function as a tumor suppressor in murine hepatocytes, also participates in a molecular cascade that prevents calcifications in arterioles and medium-sized arteries. We further provide genetic evidence that this function of TIF1alpha is not exerted in hepatocytes. The sites of ectopic calcifications in mutant mice lacking TIF1alpha resemble those seen in mice carrying an activating mutation of the calcium sensor receptor (Casr) gene and, in TIF1alpha-deficient kidneys, Casr expression is increased together with that of many other vitamin D receptor (VDR) direct target genes, namely Car2, Cyp24a1, Trpv5, Trpv6, Calb1, S100g, Pthlh, and Spp1. Thus, our data indicate that TIF1alpha represses the VDR pathway in kidney and suggest that an up-regulation of Casr expression in this organ could account for ectopic calcifications generated upon TIF1alpha deficiency. Interestingly, the calcifying arteriopathy of TIF1alpha-null mutant mice shares features with the human age-related Mönckeberg's disease and, overall, the TIF1alpha-null mutant pathological phenotype supports the hypothesis that aging is promoted by increased activity of the vitamin D signaling pathway.


Assuntos
Artérias/metabolismo , Calcinose/metabolismo , Proteínas Nucleares/deficiência , Proteínas Nucleares/metabolismo , Receptores de Calcitriol/metabolismo , Fatores de Transcrição/deficiência , Fatores de Transcrição/metabolismo , Envelhecimento/fisiologia , Animais , Calcinose/genética , Cálcio/metabolismo , Células Endoteliais/metabolismo , Regulação da Expressão Gênica , Hepatócitos/metabolismo , Homeostase , Rim/metabolismo , Pulmão/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Vibrissas/metabolismo
15.
Nat Commun ; 11(1): 2807, 2020 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-32533074

RESUMO

The nuclear receptor binding SET domain protein 1 (NSD1) is recurrently mutated in human cancers including acute leukemia. We show that NSD1 knockdown alters erythroid clonogenic growth of human CD34+ hematopoietic cells. Ablation of Nsd1 in the hematopoietic system of mice induces a transplantable erythroleukemia. In vitro differentiation of Nsd1-/- erythroblasts is majorly impaired despite abundant expression of GATA1, the transcriptional master regulator of erythropoiesis, and associated with an impaired activation of GATA1-induced targets. Retroviral expression of wildtype NSD1, but not a catalytically-inactive NSD1N1918Q SET-domain mutant induces terminal maturation of Nsd1-/- erythroblasts. Despite similar GATA1 protein levels, exogenous NSD1 but not NSDN1918Q significantly increases the occupancy of GATA1 at target genes and their expression. Notably, exogenous NSD1 reduces the association of GATA1 with the co-repressor SKI, and knockdown of SKI induces differentiation of Nsd1-/- erythroblasts. Collectively, we identify the NSD1 methyltransferase as a regulator of GATA1-controlled erythroid differentiation and leukemogenesis.


Assuntos
Diferenciação Celular , Células Eritroides/metabolismo , Células Eritroides/patologia , Fator de Transcrição GATA1/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patologia , Adulto , Animais , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Linhagem Celular Tumoral , Linhagem da Célula , Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Eritroblastos/metabolismo , Fator de Transcrição GATA1/genética , Regulação Leucêmica da Expressão Gênica , Técnicas de Silenciamento de Genes , Hematopoese , Histona-Lisina N-Metiltransferase/genética , Humanos , Estimativa de Kaplan-Meier , Leucemia Eritroblástica Aguda/genética , Masculino , Camundongos , Ligação Proteica , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores da Transferrina/metabolismo
16.
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
17.
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
18.
Mol Cell Biol ; 24(12): 5184-96, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15169884

RESUMO

Haploinsufficiency of the NSD1 gene is a hallmark of Sotos syndrome, and rearrangements of this gene by translocation can cause acute myeloid leukemia. The NSD1 gene product is a SET-domain histone lysine methyltransferase that has previously been shown to interact with nuclear receptors. We describe here a novel NSD1-interacting protein, Nizp1, that contains a SCAN box, a KRAB-A domain, and four consensus C2H2-type zinc fingers preceded by a unique finger derivative, referred to herein as the C2HR motif. The C2HR motif functions to mediate protein-protein interaction with the cysteine-rich (C5HCH) domain of NSD1 in a Zn(II)-dependent fashion, and when tethered to RNA polymerase II promoters, represses transcription in an NSD1-dependent manner. Mutations of the cysteine or histidine residues in the C2HR motif abolish the interaction of Nizp1 with NSD1 and compromise the ability of Nizp1 to repress transcription. Interestingly, converting the C2HR motif into a canonical C2H2 zinc finger has a similar effect. Thus, Nizp1 contains a novel type of zinc finger motif that functions as a docking site for NSD1 and is more than just a degenerate evolutionary remnant of a C2H2 motif.


Assuntos
Proteínas de Transporte/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas Nucleares/metabolismo , Dedos de Zinco/fisiologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Proteínas de Transporte/genética , Linhagem Celular , Sequência Conservada , DNA Complementar/genética , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/genética , Humanos , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular , Mutação , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras/química , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Homologia de Sequência de Aminoácidos , Síndrome , Técnicas do Sistema de Duplo-Híbrido , Dedos de Zinco/genética
19.
Mol Endocrinol ; 20(6): 1276-86, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16322096

RESUMO

In previous studies transcriptional intermediary factor 1alpha (TIF1alpha) was identified as a direct binding partner and potential transcriptional coactivator for nuclear receptors (NRs) but its overexpression inhibited, rather than enhanced, transcriptional activation by NRs. Here we show that TIF1alpha bound to and enhanced the function of the C-terminal activation domain (AD) of coactivator associated arginine methyltransferase 1 (CARM1) and the N-terminal AD of glucocorticoid receptor-interacting protein 1 (GRIP1). Furthermore, although TIF1alpha had little or no NR coactivator activity by itself, it cooperated synergistically with GRIP1 and CARM1 to enhance NR-mediated transcription. Inhibition of endogenous TIF1alpha expression reduced transcriptional activation by the GRIP1 N-terminal domain but not by the CARM1 C-terminal domain, suggesting that TIF1alpha may be more important for mediating the activity of the former than the latter. Reduction of endogenous TIF1alpha levels also compromised the androgen-dependent induction of an endogenous target gene of the androgen receptor. Finally, TIF1alpha formed a ternary complex with the GRIP1 N-terminal and CARM1 C-terminal domains. Thus, we conclude that TIF1alpha cooperates with NR coactivators GRIP1 and CARM1 by forming a stable ternary complex with them and enhancing the AD function of one or both of them.


Assuntos
Proteínas Nucleares/metabolismo , Coativador 2 de Receptor Nuclear/metabolismo , Proteína-Arginina N-Metiltransferases/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Humanos , Técnicas In Vitro , Modelos Biológicos , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Coativador 2 de Receptor Nuclear/química , Coativador 2 de Receptor Nuclear/genética , Ligação Proteica , Estrutura Terciária de Proteína , Proteína-Arginina N-Metiltransferases/química , Proteína-Arginina N-Metiltransferases/genética , RNA Interferente Pequeno/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Ativação Transcricional , Técnicas do Sistema de Duplo-Híbrido
20.
Nucleic Acids Res ; 33(19): 6393-404, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16282588

RESUMO

Gene activation involves protein complexes with diverse enzymatic activities, some of which are involved in chromatin modification. We have shown previously that the base excision repair enzyme thymine DNA glycosylase (TDG) acts as a potent coactivator for estrogen receptor-alpha. To further understand how TDG acts in this context, we studied its interaction with known coactivators of nuclear receptors. We find that TDG interacts in vitro and in vivo with the p160 coactivator SRC1, with the interaction being mediated by a previously undescribed motif encoding four equally spaced tyrosine residues in TDG, each tyrosine being separated by three amino acids. This is found to interact with two motifs in SRC1 also containing tyrosine residues separated by three amino acids. Site-directed mutagenesis shows that the tyrosines encoded in these motifs are critical for the interaction. The related p160 protein TIF2 does not interact with TDG and has the altered sequence, F-X-X-X-Y, at the equivalent positions relative to SRC1. Substitution of the phenylalanines to tyrosines is sufficient to bring about interaction of TIF2 with TDG. These findings highlight a new protein-protein interaction motif based on Y-X-X-X-Y and provide new insight into the interaction of diverse proteins in coactivator complexes.


Assuntos
Timina DNA Glicosilase/química , Timina DNA Glicosilase/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Histona Acetiltransferases , Humanos , Dados de Sequência Molecular , Coativador 1 de Receptor Nuclear , Sequências Repetitivas de Aminoácidos , Transativadores/química , Fatores de Transcrição/química , Tirosina/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA