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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 42(4): 438-50, 2011 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-21596310

RESUMEN

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.


Asunto(s)
Cromatina/metabolismo , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitinación , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células HEK293 , Histonas/genética , Humanos , Proteínas Nucleares/genética , Proteínas del Grupo Polycomb , Proteínas Represoras/genética , Factores de Transcripción/genética
2.
Development ; 140(1): 167-75, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23154409

RESUMEN

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.


Asunto(s)
Diferenciación Celular/genética , Células Epiteliales/citología , Lactancia/genética , Glándulas Mamarias Animales/citología , Proteína Smad4/antagonistas & inhibidores , Factores de Transcripción/fisiología , Animales , Células Epiteliales/fisiología , Femenino , Masculino , Glándulas Mamarias Animales/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Embarazo , Transducción de Señal/genética , Proteína Smad4/fisiología
3.
Blood ; 122(23): 3798-807, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-24092935

RESUMEN

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.


Asunto(s)
Eritroblastos/citología , Eritroblastos/metabolismo , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Anemia/genética , Anemia/patología , Animales , Apoptosis/genética , Apoptosis/fisiología , Médula Ósea/metabolismo , Médula Ósea/patología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Eritropoyesis/genética , Eritropoyesis/fisiología , Regulación del Desarrollo de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito , Globinas beta/genética
4.
Proc Natl Acad Sci U S A ; 108(20): 8212-7, 2011 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-21531907

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/etiología , Neoplasias Hepáticas/etiología , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Hepatocelular/patología , Transformación Celular Neoplásica/patología , Hepatocitos/patología , Neoplasias Hepáticas/patología , Ratones , Complejos Multiproteicos/aislamiento & purificación , Complejos Multiproteicos/fisiología , Unión Proteica , Receptores de Ácido Retinoico , Proteína 28 que Contiene Motivos Tripartito
5.
Development ; 137(15): 2571-8, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20573697

RESUMEN

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.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Smad/metabolismo , Factores de Transcripción/metabolismo , Alelos , Animales , Tipificación del Cuerpo , Diferenciación Celular , Cruzamientos Genéticos , Ectodermo/metabolismo , Mesodermo/metabolismo , Ratones , Modelos Biológicos , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
6.
Am J Pathol ; 180(6): 2214-21, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22469842

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Proteína Smad4/genética , Factores de Transcripción/genética , Animales , Carcinoma Ductal Pancreático/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Progresión de la Enfermedad , Eliminación de Gen , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Imagen por Resonancia Magnética , Ratones , Ratones Mutantes , Neoplasias Pancreáticas/patología , Lesiones Precancerosas/genética , Transducción de Señal/genética , Proteína Smad4/fisiología , Factores de Transcripción/deficiencia , Factores de Transcripción/fisiología
7.
Dev Biol ; 350(2): 548-58, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21163256

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/fisiología , Gastrulación , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Células de Sertoli/fisiología , Espermatogénesis , Animales , Ciclo Celular , Homólogo de la Proteína Chromobox 5 , Desarrollo Embrionario , Femenino , Proteínas de Homeodominio/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Homeótica Nanog , Proteínas Nucleares/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Proteínas Represoras/genética , Proteína 28 que Contiene Motivos Tripartito
8.
J Biol Chem ; 286(38): 33369-79, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21768647

RESUMEN

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.


Asunto(s)
Interferones/metabolismo , Proteínas Nucleares/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Factores de Transcripción STAT/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Animales , Carcinoma Hepatocelular/genética , Análisis por Conglomerados , Dosificación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/metabolismo , Neoplasias Hepáticas/genética , Ratones , Modelos Biológicos , Proteínas Nucleares/deficiencia , Receptores de Ácido Retinoico/metabolismo , Reproducibilidad de los Resultados , Receptor alfa de Ácido Retinoico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factores de Transcripción/deficiencia , Transcriptoma , Tretinoina/farmacología
9.
Adv Exp Med Biol ; 770: 59-76, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23631000

RESUMEN

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.


Asunto(s)
Secuencias de Aminoácidos , Regulación de la Expresión Génica , Proteínas/química , Proteínas/metabolismo , Transcripción Genética , Animales , Humanos
10.
Proc Natl Acad Sci U S A ; 106(51): 21830-5, 2009 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-20018718

RESUMEN

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.


Asunto(s)
Anomalías Múltiples/genética , Epigénesis Genética , Glioma/enzimología , Trastornos del Crecimiento/genética , N-Metiltransferasa de Histona-Lisina/genética , Neuroblastoma/enzimología , Inmunoprecipitación de Cromatina , Islas de CpG , Metilación de ADN , Glioma/genética , Histona Metiltransferasas , Humanos , Neuroblastoma/genética , Regiones Promotoras Genéticas , Síndrome
11.
PLoS Genet ; 5(7): e1000575, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19629168

RESUMEN

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.


Asunto(s)
Carcinoma Ductal Pancreático/metabolismo , Genes Supresores de Tumor , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patología , Humanos , Ratones , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Factores de Transcripción/genética
12.
Biochim Biophys Acta ; 1799(5-6): 463-8, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20176155

RESUMEN

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.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/química , Transactivadores/metabolismo , Transcripción Genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Homología de Secuencia de Aminoácido , Transactivadores/genética , Transfección , Dedos de Zinc/genética
13.
Biochim Biophys Acta ; 1799(8): 539-45, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20538088

RESUMEN

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.


Asunto(s)
Empalme Alternativo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Dominios y Motivos de Interacción de Proteínas/genética , Dedos de Zinc , Animales , Western Blotting , Núcleo Celular/metabolismo , Células Cultivadas , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Humanos , Riñón/citología , Riñón/metabolismo , Ratones , Neuroblastoma/metabolismo , Neuroblastoma/patología , Transporte de Proteínas , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Saccharomyces cerevisiae , Transcripción Genética
14.
Proc Natl Acad Sci U S A ; 105(7): 2598-603, 2008 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-18287084

RESUMEN

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.


Asunto(s)
Arterias/metabolismo , Calcinosis/metabolismo , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Receptores de Calcitriol/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/metabolismo , Envejecimiento/fisiología , Animales , Calcinosis/genética , Calcio/metabolismo , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Homeostasis , Riñón/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Proteínas Nucleares/genética , Factores de Transcripción/genética , Vibrisas/metabolismo
15.
Nat Commun ; 11(1): 2807, 2020 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-32533074

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Células Eritroides/metabolismo , Células Eritroides/patología , Factor de Transcripción GATA1/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/patología , Adulto , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Línea Celular Tumoral , Linaje de la Célula , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Eritroblastos/metabolismo , Factor de Transcripción GATA1/genética , Regulación Leucémica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hematopoyesis , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Estimación de Kaplan-Meier , Leucemia Eritroblástica Aguda/genética , Masculino , Ratones , Unión Proteica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Transferrina/metabolismo
16.
J Virol ; 82(9): 4675-9, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18287239

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Virus de la Leucemia Murina/fisiología , Proteínas Represoras/metabolismo , Replicación Viral , Animales , Sitios de Unión , Línea Celular , Homólogo de la Proteína Chromobox 5 , Células Madre de Carcinoma Embrionario/virología , Células Madre Embrionarias/virología , Ratones , Proteínas Nucleares/metabolismo , Mutación Puntual , Unión Proteica/genética , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito
17.
FASEB J ; 22(11): 3853-65, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18676401

RESUMEN

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.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Endodermo/enzimología , Heterocromatina/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Homólogo de la Proteína Chromobox 5 , Endodermo/citología , Humanos , Poli(ADP-Ribosa) Polimerasa-1 , Proteína 28 que Contiene Motivos Tripartito
18.
Mol Cell Biol ; 24(12): 5184-96, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15169884

RESUMEN

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.


Asunto(s)
Proteínas Portadoras/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas Nucleares/metabolismo , Dedos de Zinc/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Sitios de Unión , Proteínas Portadoras/genética , Línea Celular , Secuencia Conservada , ADN Complementario/genética , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Técnicas In Vitro , Ratones , Datos de Secuencia Molecular , Mutación , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/química , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Homología de Secuencia de Aminoácido , Síndrome , Técnicas del Sistema de Dos Híbridos , Dedos de Zinc/genética
19.
Mol Endocrinol ; 20(6): 1276-86, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16322096

RESUMEN

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.


Asunto(s)
Proteínas Nucleares/metabolismo , Coactivador 2 del Receptor Nuclear/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Factores de Transcripción/metabolismo , Animales , Secuencia de Bases , Proteínas Portadoras/química , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Humanos , Técnicas In Vitro , Modelos Biológicos , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Coactivador 2 del Receptor Nuclear/química , Coactivador 2 del Receptor Nuclear/genética , Unión Proteica , Estructura Terciaria de Proteína , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Activación Transcripcional , Técnicas del Sistema de Dos Híbridos
20.
Nucleic Acids Res ; 33(19): 6393-404, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16282588

RESUMEN

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.


Asunto(s)
Timina ADN Glicosilasa/química , Timina ADN Glicosilasa/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Histona Acetiltransferasas , Humanos , Datos de Secuencia Molecular , Coactivador 1 de Receptor Nuclear , Secuencias Repetitivas de Aminoácido , Transactivadores/química , Factores de Transcripción/química , Tirosina/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA