Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
PLoS Biol ; 21(12): e3002424, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38048351

RESUMEN

Metazoan chromosomes are organized into discrete spatial domains (TADs), believed to contribute to the regulation of transcriptional programs. Despite extensive correlation between domain organization and gene activity, a direct mechanistic link is unclear, with perturbation studies often showing little effect. To follow chromatin architecture changes during development, we used Capture Hi-C to interrogate the domains around key differentially expressed genes during mouse thymocyte maturation, uncovering specific remodeling events. Notably, one TAD boundary was broadened to accommodate RNA polymerase elongation past the border, and subdomains were formed around some activated genes without changes in CTCF binding. The ectopic induction of some genes was sufficient to recapitulate domain formation in embryonic stem cells, providing strong evidence that transcription can directly remodel chromatin structure. These results suggest that transcriptional processes drive complex chromosome folding patterns that can be important in certain genomic contexts.


Asunto(s)
Cromatina , Cromosomas , Animales , Ratones , Cromatina/genética , Cromosomas/metabolismo , Transcripción Genética , Diferenciación Celular/genética , Genoma , Ensamble y Desensamble de Cromatina , Factor de Unión a CCCTC/genética
2.
Cell ; 146(1): 67-79, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21722948

RESUMEN

DNA methylation is a major epigenetic mechanism for gene silencing. Whereas methyltransferases mediate cytosine methylation, it is less clear how unmethylated regions in mammalian genomes are protected from de novo methylation and whether an active demethylating activity is involved. Here, we show that either knockout or catalytic inactivation of the DNA repair enzyme thymine DNA glycosylase (TDG) leads to embryonic lethality in mice. TDG is necessary for recruiting p300 to retinoic acid (RA)-regulated promoters, protection of CpG islands from hypermethylation, and active demethylation of tissue-specific developmentally and hormonally regulated promoters and enhancers. TDG interacts with the deaminase AID and the damage response protein GADD45a. These findings highlight a dual role for TDG in promoting proper epigenetic states during development and suggest a two-step mechanism for DNA demethylation in mammals, whereby 5-methylcytosine and 5-hydroxymethylcytosine are first deaminated by AID to thymine and 5-hydroxymethyluracil, respectively, followed by TDG-mediated thymine and 5-hydroxymethyluracil excision repair.


Asunto(s)
Metilación de ADN , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Timina ADN Glicosilasa/metabolismo , 5-Metilcitosina/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , Citidina Desaminasa/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Femenino , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Timina ADN Glicosilasa/genética , Transcripción Genética
3.
J Cell Sci ; 128(13): 2303-18, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25991548

RESUMEN

Mouse embryonic stem cells (ESCs) treated with all-trans retinoic acid differentiate into a homogenous population of glutamatergic neurons. Although differentiation is initiated through activation of target genes by the retinoic acid receptors, the downstream transcription factors specifying neuronal fate are less well characterised. Here, we show that the transcription factor Brn2 (also known as Pou3f2) is essential for the neuronal differentiation programme. By integrating results from RNA-seq following Brn2 silencing with results from Brn2 ChIP-seq, we identify a set of Brn2 target genes required for the neurogenic programme. Further integration of Brn2 ChIP-seq data from retinoic-acid-treated ESCs and P19 cells with data from ESCs differentiated into neuronal precursors by Fgf2 treatment and that from fibroblasts trans-differentiated into neurons by ectopic Brn2 expression showed that Brn2 occupied a distinct but overlapping set of genomic loci in these differing conditions. However, a set of common binding sites and target genes defined the core of the Brn2-regulated neuronal programme, among which was that encoding the transcription factor Zic1. Small hairpin RNA (shRNA)-mediated silencing of Zic1 prevented ESCs from differentiating into neuronal precursors, thus defining a hierarchical Brn2-Zic1 axis that is essential to specify neural fate in retinoic-acid-treated ESCs.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Células Madre Embrionarias de Ratones/citología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/citología , Factores del Dominio POU/metabolismo , Factores de Transcripción/metabolismo , Tretinoina/farmacología , Animales , Secuencia de Bases , Diferenciación Celular/efectos de los fármacos , Cuerpos Embrioides/citología , Cuerpos Embrioides/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Genoma , Células HEK293 , Humanos , Ratones , Datos de Secuencia Molecular , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo
4.
Nucleic Acids Res ; 40(4): 1446-59, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22013162

RESUMEN

The TATA binding protein (TBP) plays a pivotal role in RNA polymerase II (Pol II) transcription through incorporation into the TFIID and B-TFIID complexes. The role of mammalian B-TFIID composed of TBP and B-TAF1 is poorly understood. Using a complementation system in genetically modified mouse cells where endogenous TBP can be conditionally inactivated and replaced by exogenous mutant TBP coupled to tandem affinity purification and mass spectrometry, we identify two TBP mutations, R188E and K243E, that disrupt the TBP-BTAF1 interaction and B-TFIID complex formation. Transcriptome and ChIP-seq analyses show that loss of B-TFIID does not generally alter gene expression or genomic distribution of TBP, but positively or negatively affects TBP and/or Pol II recruitment to a subset of promoters. We identify promoters where wild-type TBP assembles a partial inactive preinitiation complex comprising B-TFIID, TFIIB and Mediator complex, but lacking TFIID, TFIIE and Pol II. Exchange of B-TFIID in wild-type cells for TFIID in R188E and K243E mutant cells at these primed promoters completes preinitiation complex formation and recruits Pol II to activate their expression. We propose a novel regulatory mechanism involving formation of a partial preinitiation complex comprising B-TFIID that primes the promoter for productive preinitiation complex formation in mammalian cells.


Asunto(s)
Factores Asociados con la Proteína de Unión a TATA/metabolismo , Proteína de Unión a TATA-Box/metabolismo , Transcripción Genética , Animales , Regulación de la Expresión Génica , Prueba de Complementación Genética , Genoma , Ratones , Mutación , Regiones Promotoras Genéticas , ARN Polimerasa II/metabolismo , Factores Asociados con la Proteína de Unión a TATA/química , Proteína de Unión a TATA-Box/química , Proteína de Unión a TATA-Box/genética , Factor de Transcripción TFIID/metabolismo , Transcriptoma
5.
Nat Commun ; 12(1): 6184, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702821

RESUMEN

The spatiotemporal organization of chromatin influences many nuclear processes: from chromosome segregation to transcriptional regulation. To get a deeper understanding of these processes, it is essential to go beyond static viewpoints of chromosome structures, to accurately characterize chromatin's diffusion properties. We present GP-FBM: a computational framework based on Gaussian processes and fractional Brownian motion to extract diffusion properties from stochastic trajectories of labeled chromatin loci. GP-FBM uses higher-order temporal correlations present in the data, therefore, outperforming existing methods. Furthermore, GP-FBM allows to interpolate incomplete trajectories and account for substrate movement when two or more particles are present. Using our method, we show that average chromatin diffusion properties are surprisingly similar in interphase and mitosis in mouse embryonic stem cells. We observe surprising heterogeneity in local chromatin dynamics, correlating with potential regulatory activity. We also present GP-Tool, a user-friendly graphical interface to facilitate usage of GP-FBM by the research community.


Asunto(s)
Cromatina/fisiología , Modelos Biológicos , Animales , Ensamble y Desensamble de Cromatina , Biología Computacional , Proteínas de Homeodominio/genética , Interfase , Ratones , Mitosis , Movimiento (Física) , Células Madre Embrionarias de Ratones , Distribución Normal
6.
Pigment Cell Melanoma Res ; 23(3): 404-18, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20337985

RESUMEN

POU3F2 is a POU-Homeodomain transcription factor expressed in neurons and melanoma cells. In melanoma lesions, cells expressing high levels of POU3F2 show enhanced invasive and metastatic capacity that can in part be explained by repression of Micropthalmia-associated Transcription Factor (MITF) expression via POU3F2 binding to its promoter. To identify other POU3F2 target genes that may be involved in modulating the properties of melanoma cells, we performed ChIP-chip experiments in 501Mel melanoma cells. 2108 binding loci located in the regulatory regions of 1700 potential target genes were identified. Bioinformatic and experimental assays showed the presence of known POU3F2-binding motifs, but also many AT-rich sequences with only partial similarity to the known motifs at the occupied loci. Functional analysis indicates that POU3F2 regulates the stem cell factor (Kit ligand, Kitl) promoter via a cluster of four closely spaced binding sites located in the proximal promoter. Our results suggest that POU3F2 may regulate the properties of melanoma cells via autocrine KIT ligand signalling.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Proteínas de Homeodominio/genética , Melanoma/genética , Factores del Dominio POU/genética , Regiones Promotoras Genéticas , Factor de Células Madre/genética , Secuencias de Aminoácidos , Secuencia de Bases , Sitios de Unión , Antígenos CD36/genética , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Genes Relacionados con las Neoplasias/genética , Sitios Genéticos/genética , Proteínas de Homeodominio/química , Humanos , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/genética , Datos de Secuencia Molecular , Factores del Dominio POU/química , Unión Proteica , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Activación Transcripcional/genética , Proteínas Wnt/genética
7.
Cancer Res ; 68(19): 7788-94, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18829533

RESUMEN

The origin of tumor heterogeneity is poorly understood, yet it represents a major barrier to effective therapy. In melanoma and in melanocyte development, the microphthalmia-associated transcription factor (Mitf) controls survival, differentiation, proliferation, and migration/metastasis. The Brn-2 (N-Oct-3, POU3F2) transcription factor also regulates melanoma proliferation and is up-regulated by BRAF and beta-catenin, two key melanoma-associated signaling molecules. Here, we show that Brn-2 also regulates invasiveness and directly represses Mitf expression. Remarkably, in melanoma biopsies, Mitf and Brn-2 each mark a distinct subpopulation of melanoma cells, providing a striking illustration of melanoma tumor heterogeneity with implications for melanoma therapy.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas de Homeodominio/fisiología , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factores del Dominio POU/fisiología , Animales , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factor de Transcripción Asociado a Microftalmía/metabolismo , Datos de Secuencia Molecular , Invasividad Neoplásica , Factores del Dominio POU/genética , Factores del Dominio POU/metabolismo , Unión Proteica , Trasplante Heterólogo , Células Tumorales Cultivadas
8.
Development ; 134(16): 2947-58, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17626060

RESUMEN

The TAF4 subunit of transcription factor TFIID was inactivated in the basal keratinocytes of foetal and adult mouse epidermis. Loss of TAF4 in the foetal epidermis results in reduced expression of the genes required for skin barrier function, leading to early neonatal death. By contrast, TAF4 inactivation in adult epidermis leads to extensive fur loss and an aberrant hair cycle characterised by a defective anagen phase. Although the mutant epidermis contains few normal anagen-phase hair follicles, many genes expressed at this stage are strongly upregulated indicating desynchronized and inappropriate gene expression. The TAF4 mutant adult epidermis also displays interfollicular hyperplasia associated with a potent upregulation of several members of the EGF family of mitogens. Moreover, loss of TAF4 leads to malignant transformation of chemically induced papillomas and the appearance of invasive melanocytic tumours. Together, our results show that TAF4 is an important regulator of keratinocyte proliferation and has cell-autonomous and non-cell-autonomous tumour suppressor activity.


Asunto(s)
Proliferación Celular , Epidermis/metabolismo , Queratinocitos/citología , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factores Asociados con la Proteína de Unión a TATA/fisiología , Factor de Transcripción TFIID/metabolismo , Factor de Transcripción TFIID/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular/genética , Epidermis/embriología , Epidermis/patología , Femenino , Predisposición Genética a la Enfermedad , Cabello/citología , Cabello/embriología , Hiperplasia/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Nevo Pigmentado/inducido químicamente , Nevo Pigmentado/genética , Subunidades de Proteína/fisiología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética , Tretinoina/efectos adversos , Proteínas Supresoras de Tumor/fisiología
9.
Cell Cycle ; 4(11): 1486-90, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16205117

RESUMEN

RNA polymerase II general transcription factor TFIID is a macromolecular complex comprising the TATA-binding protein, TBP and 13-14 evolutionary conserved TBP-associated factors, TAFs. Although genetic experiments have shown that TAFs are essential for cell cycle progression in yeast and in rapidly proliferating vertebrate cells in vitro, new experiments indicate they may be dispensible in specific developmental and physiological processes. Moreover, the TAF4 subunit of TFIID negatively regulates proliferation by inhibiting activation of the TGFbeta signalling pathway by its paralogue TAF4b. TAF4 is however essential in the retinoic acid and cAMP signalling pathways acting as a cofactor for CREB and the retinoic acid receptor, but is a negative regulator of the ATF7 transcription factor.


Asunto(s)
Ciclo Celular/fisiología , Transducción de Señal/fisiología , Factores Asociados con la Proteína de Unión a TATA/fisiología , Animales , Ciclo Celular/genética , Humanos , Transducción de Señal/genética
10.
EMBO J ; 24(15): 2753-67, 2005 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-16015375

RESUMEN

We have inactivated transcription factor TFIID subunit TBP-associated factor 4 (TAF4) in mouse embryonic fibroblasts. Mutant taf4(-/-) cells are viable and contain intact TFIID comprising the related TAF4b showing that TAF4 is not an essential protein. TAF4 inactivation deregulates more than 1000 genes indicating that TFIID complexes containing TAF4 and TAF4b have distinct target gene specificities. However, taf4(-/-) cell lines have altered morphology and exhibit serum-independent autocrine growth correlated with the induced expression of several secreted mitotic factors and activators of the transforming growth factor beta signalling pathway. In addition to TAF4 inactivation, many of these genes can also be induced by overexpression of TAF4b. A competitive equilibrium between TAF4 and TAF4b therefore regulates expression of genes controlling cell proliferation. We have further identified a set of genes that are regulated both by TAF4 and upon adaptation to serum starvation and which may be important downstream mediators of serum-independent growth. Our study also shows that TAF4 is an essential cofactor for activation by the retinoic acid receptor and CREB, but not for Sp1 and the vitamin D3 receptor.


Asunto(s)
Comunicación Autocrina/fisiología , Fibroblastos/metabolismo , Transducción de Señal/fisiología , Factores Asociados con la Proteína de Unión a TATA/antagonistas & inhibidores , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factor de Transcripción TFIID/antagonistas & inhibidores , Factor de Transcripción TFIID/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Supervivencia Celular/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Regulación de la Expresión Génica , Immunoblotting , Ratones , Estructura Terciaria de Proteína , Receptores de Ácido Retinoico/metabolismo , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética
11.
FEMS Yeast Res ; 5(3): 213-30, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15556083

RESUMEN

The first protein map of an ale-fermenting yeast is presented in this paper: 205 spots corresponding to 133 different proteins were identified. Comparison of the proteome of this ale strain with a lager brewing yeast and the Saccharomyces cerevisiae strain S288c confirmed that this ale strain is much closer to S288c than the lager strain at the proteome level. The dynamics of the ale-brewing yeast proteome during production-scale fermentation was analysed at the beginning and end of the first and the third usage of the yeast (called generation in the brewing industry). During the first generation, most changes were related to the switch from aerobic propagation to anaerobic fermentation. Fewer changes were observed during the third generation but certain stress-response proteins such as Hsp26p, Ssa4p and Pnc1p exhibited constitutive expression in subsequent generations. The ale brewing yeast strain appears to be quite well adapted to fermentation conditions and stresses.


Asunto(s)
Proteínas Fúngicas/metabolismo , Proteoma/metabolismo , Levaduras/metabolismo , Bebidas Alcohólicas , Electroforesis en Gel Bidimensional , Fermentación , Proteínas Fúngicas/análisis , Microbiología Industrial/métodos , Proteoma/análisis , Saccharomyces cerevisiae/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Levaduras/crecimiento & desarrollo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA