Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Life Sci Alliance ; 6(10)2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37487640

RESUMEN

Polycomb repressive complex 1 (PRC1) strongly influences 3D genome organization, mediating local chromatin compaction and clustering of target loci. Several PRC1 subunits have the capacity to form biomolecular condensates through liquid-liquid phase separation in vitro and when tagged and over-expressed in cells. Here, we use 1,6-hexanediol, which can disrupt liquid-like condensates, to examine the role of endogenous PRC1 biomolecular condensates on local and chromosome-wide clustering of PRC1-bound loci. Using imaging and chromatin immunoprecipitation, we show that PRC1-mediated chromatin compaction and clustering of targeted genomic loci-at different length scales-can be reversibly disrupted by the addition and subsequent removal of 1,6-hexanediol to mouse embryonic stem cells. Decompaction and dispersal of polycomb domains and clusters cannot be solely attributable to reduced PRC1 occupancy detected by chromatin immunoprecipitation following 1,6-hexanediol treatment as the addition of 2,5-hexanediol has similar effects on binding despite this alcohol not perturbing PRC1-mediated 3D clustering, at least at the sub-megabase and megabase scales. These results suggest that weak hydrophobic interactions between PRC1 molecules may have a role in polycomb-mediated genome organization.


Asunto(s)
Cromatina , Proteínas de Drosophila , Animales , Ratones , Complejo Represivo Polycomb 1 , Núcleo Celular , Proteínas del Grupo Polycomb
3.
PLoS One ; 17(11): e0268149, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36413568

RESUMEN

Classical aniridia is a congenital and progressive panocular disorder almost exclusively caused by heterozygous loss-of-function variants at the PAX6 locus. We report nine individuals from five families with severe aniridia and/or microphthalmia (with no detectable PAX6 mutation) with ultrarare monoallelic missense variants altering the Arg51 codon of MAB21L1. These mutations occurred de novo in 3/5 families, with the remaining families being compatible with autosomal dominant inheritance. Mice engineered to carry the p.Arg51Leu change showed a highly-penetrant optic disc anomaly in heterozygous animals with severe microphthalmia in homozygotes. Substitutions of the same codon (Arg51) in MAB21L2, a close homolog of MAB21L1, cause severe ocular and skeletal malformations in humans and mice. The predicted nucleotidyltransferase function of MAB21L1 could not be demonstrated using purified protein with a variety of nucleotide substrates and oligonucleotide activators. Induced expression of GFP-tagged wildtype and mutant MAB21L1 in human cells caused only modest transcriptional changes. Mass spectrometry of immunoprecipitated protein revealed that both mutant and wildtype MAB21L1 associate with transcription factors that are known regulators of PAX6 (MEIS1, MEIS2 and PBX1) and with poly(A) RNA binding proteins. Arg51 substitutions reduce the association of wild-type MAB21L1 with TBL1XR1, a component of the NCoR complex. We found limited evidence for mutation-specific interactions with MSI2/Musashi-2, an RNA-binding proteins with effects on many different developmental pathways. Given that biallelic loss-of-function variants in MAB21L1 result in a milder eye phenotype we suggest that Arg51-altering monoallelic variants most plausibly perturb eye development via a gain-of-function mechanism.


Asunto(s)
Aniridia , Microftalmía , Humanos , Animales , Ratones , Microftalmía/genética , Factor de Transcripción PAX6/genética , Aniridia/genética , Mutación Missense , Heterocigoto , Factores de Transcripción/genética , Proteínas de Homeodominio/genética , Proteínas de Unión al ARN/genética , Proteínas del Ojo/genética , Péptidos y Proteínas de Señalización Intracelular/genética
4.
Nat Commun ; 13(1): 5609, 2022 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-36153345

RESUMEN

Human centromeres appear as constrictions on mitotic chromosomes and form a platform for kinetochore assembly in mitosis. Biophysical experiments led to a suggestion that repetitive DNA at centromeric regions form a compact scaffold necessary for function, but this was revised when neocentromeres were discovered on non-repetitive DNA. To test whether centromeres have a special chromatin structure we have analysed the architecture of a neocentromere. Centromere repositioning is accompanied by RNA polymerase II recruitment and active transcription to form a decompacted, negatively supercoiled domain enriched in 'open' chromatin fibres. In contrast, centromerisation causes a spreading of repressive epigenetic marks to surrounding regions, delimited by H3K27me3 polycomb boundaries and divergent genes. This flanking domain is transcriptionally silent and partially remodelled to form 'compact' chromatin, similar to satellite-containing DNA sequences, and exhibits genomic instability. We suggest transcription disrupts chromatin to provide a foundation for kinetochore formation whilst compact pericentromeric heterochromatin generates mechanical rigidity.


Asunto(s)
Heterocromatina , Histonas , Centrómero/genética , Cromatina/genética , ADN/genética , ADN Satélite , Heterocromatina/genética , Histonas/genética , Humanos , ARN Polimerasa II/genética
5.
Nat Commun ; 13(1): 3053, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35650196

RESUMEN

In addition to central functions in cell adhesion signalling, integrin-associated proteins have wider roles at sites distal to adhesion receptors. In experimentally defined adhesomes, we noticed that there is clear enrichment of proteins that localise to the nucleus, and conversely, we now report that nuclear proteomes contain a class of adhesome components that localise to the nucleus. We here define a nucleo-adhesome, providing experimental evidence for a remarkable scale of nuclear localisation of adhesion proteins, establishing a framework for interrogating nuclear adhesion protein functions. Adding to nuclear FAK's known roles in regulating transcription, we now show that nuclear FAK regulates expression of many adhesion-related proteins that localise to the nucleus and that nuclear FAK binds to the adhesome component and nuclear protein Hic-5. FAK and Hic-5 work together in the nucleus, co-regulating a subset of genes transcriptionally. We demonstrate the principle that there are subcomplexes of nuclear adhesion proteins that cooperate to control transcription.


Asunto(s)
Núcleo Celular , Proteoma , Adhesión Celular , Núcleo Celular/metabolismo , Proteoma/metabolismo , Transducción de Señal
6.
Cancer Res ; 82(8): 1548-1559, 2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35074757

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is an aggressive malignancy of the bile ducts within the liver characterized by high levels of genetic heterogeneity. In the context of such genetic variability, determining which oncogenic mutations drive ICC growth has been difficult, and developing modes of patient stratification and targeted therapies remains challenging. Here we model the interactions between rare mutations with more common driver genes and combine in silico analysis of patient data with highly multiplexed in vivo CRISPR-spCas9 screens to perform a functional in vivo study into the role genetic heterogeneity plays in driving ICC. Novel tumor suppressors were uncovered, which, when lost, cooperate with the RAS oncoprotein to drive ICC growth. Focusing on a set of driver mutations that interact with KRAS to initiate aggressive, sarcomatoid-type ICC revealed that tumor growth relies on Wnt and PI3K signaling. Pharmacologic coinhibition of Wnt and PI3K in vivo impeded ICC growth regardless of mutational profile. Therefore, Wnt and PI3K activity should be considered as a signature by which patients can be stratified for treatment independent of tumor genotype, and inhibitors of these pathways should be levied to treat ICC. SIGNIFICANCE: This work shows that, despite significant genetic heterogeneity, intrahepatic cholangiocarcinoma relies on a limited number of signaling pathways to grow, suggesting common therapeutic vulnerabilities across patients.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Colangiocarcinoma/patología , Heterogeneidad Genética , Humanos , Fosfatidilinositol 3-Quinasas/genética
7.
Nat Commun ; 12(1): 3127, 2021 05 25.
Artículo en Inglés | MEDLINE | ID: mdl-34035299

RESUMEN

Cornelia de Lange syndrome is a multisystem developmental disorder typically caused by mutations in the gene encoding the cohesin loader NIPBL. The associated phenotype is generally assumed to be the consequence of aberrant transcriptional regulation. Recently, we identified a missense mutation in BRD4 associated with a Cornelia de Lange-like syndrome that reduces BRD4 binding to acetylated histones. Here we show that, although this mutation reduces BRD4-occupancy at enhancers it does not affect transcription of the pluripotency network in mouse embryonic stem cells. Rather, it delays the cell cycle, increases DNA damage signalling, and perturbs regulation of DNA repair in mutant cells. This uncovers a role for BRD4 in DNA repair pathway choice. Furthermore, we find evidence of a similar increase in DNA damage signalling in cells derived from NIPBL-deficient individuals, suggesting that defective DNA damage signalling and repair is also a feature of typical Cornelia de Lange syndrome.


Asunto(s)
Daño del ADN , Reparación del ADN , Síndrome de Cornelia de Lange/genética , Mutación , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Predisposición Genética a la Enfermedad/genética , Humanos , Ratones , RNA-Seq/métodos , Transducción de Señal/genética , Factores de Transcripción/genética
8.
Clin Cancer Res ; 27(11): 3201-3214, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33741650

RESUMEN

PURPOSE: The abundance and effects of structural variation at BRCA1/2 in tumors are not well understood. In particular, the impact of these events on homologous recombination repair deficiency (HRD) has yet to be demonstrated. EXPERIMENTAL DESIGN: Exploiting a large collection of whole-genome sequencing data from high-grade serous ovarian carcinoma (N = 205) together with matched RNA sequencing for the majority of tumors (N = 150), we have comprehensively characterized mutation and expression at BRCA1/2. RESULTS: In addition to the known spectrum of short somatic mutations (SSM), we discovered that multi-megabase structural variants (SV) were a frequent, unappreciated source of BRCA1/2 disruption in these tumors, and we found a genome-wide enrichment for large deletions at the BRCA1/2 loci across the cohort. These SVs independently affected a substantial proportion of patients (16%) in addition to those affected by SSMs (24%), conferring HRD and impacting patient survival. We also detail compound deficiencies involving SSMs and SVs at both loci, demonstrating that the strongest risk of HRD emerges from combined SVs at both BRCA1 and BRCA2 in the absence of SSMs. Furthermore, these SVs are abundant and disruptive in other cancer types. CONCLUSIONS: These results extend our understanding of the mutational landscape underlying HRD, increase the number of patients predicted to benefit from therapies exploiting HRD, and suggest there is currently untapped potential in SV detection for patient stratification.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patología , Recombinación Homóloga/genética , Mutación/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Reparación del ADN por Recombinación/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Femenino , Expresión Génica , Humanos , Secuenciación Completa del Genoma
9.
Sci Rep ; 11(1): 229, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33420223

RESUMEN

Focal adhesion kinase (FAK) localizes to focal adhesions and is overexpressed in many cancers. FAK can also translocate to the nucleus, where it binds to, and regulates, several transcription factors, including MBD2, p53 and IL-33, to control gene expression by unknown mechanisms. We have used ATAC-seq to reveal that FAK controls chromatin accessibility at a subset of regulated genes. Integration of ATAC-seq and RNA-seq data showed that FAK-dependent chromatin accessibility is linked to differential gene expression, including of the FAK-regulated cytokine and transcriptional regulator interleukin-33 (Il33), which controls anti-tumor immunity. Analysis of the accessibility peaks on the Il33 gene promoter/enhancer regions revealed sequences for several transcription factors, including ETS and AP-1 motifs, and we show that c-Jun, a component of AP-1, regulates Il33 gene expression by binding to its enhancer in a FAK kinase-dependent manner. This work provides the first demonstration that FAK controls transcription via chromatin accessibility, identifying a novel mechanism by which nuclear FAK regulates biologically important gene expression.


Asunto(s)
Cromatina/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Regulación de la Expresión Génica , Interleucina-33/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Secuencias de Aminoácidos , Comunicación Celular , Núcleo Celular/metabolismo , Humanos , Unión Proteica
10.
Cell Rep ; 29(7): 1974-1985.e6, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31722211

RESUMEN

The DNA hypomethylation that occurs when embryonic stem cells (ESCs) are directed to the ground state of naive pluripotency by culturing in two small molecule inhibitors (2i) results in redistribution of polycomb (H3K27me3) away from its target loci. Here, we demonstrate that 3D genome organization is also altered in 2i, with chromatin decompaction at polycomb target loci and a loss of long-range polycomb interactions. By preventing DNA hypomethylation during the transition to the ground state, we are able to restore to ESC in 2i the H3K27me3 distribution, as well as polycomb-mediated 3D genome organization that is characteristic of primed ESCs grown in serum. However, these cells retain the functional characteristics of 2i ground-state ESCs. Our findings demonstrate the central role of DNA methylation in shaping major aspects of 3D genome organization but caution against assuming causal roles for the epigenome and 3D genome in gene regulation and function in ESCs.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Metilación de ADN , Epigenoma , Células Madre Embrionarias de Ratones/metabolismo , Animales , Cromatina/genética , Masculino , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones/citología
11.
Mol Cell ; 76(3): 473-484.e7, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31494034

RESUMEN

Enhancers can regulate the promoters of their target genes over very large genomic distances. It is widely assumed that mechanisms of enhancer action involve the reorganization of three-dimensional chromatin architecture, but this is poorly understood. The predominant model involves physical enhancer-promoter interaction by looping out the intervening chromatin. However, studying the enhancer-driven activation of the Sonic hedgehog gene (Shh), we have identified a change in chromosome conformation that is incompatible with this simple looping model. Using super-resolution 3D-FISH and chromosome conformation capture, we observe a decreased spatial proximity between Shh and its enhancers during the differentiation of embryonic stem cells to neural progenitors. We show that this can be recapitulated by synthetic enhancer activation, is impeded by chromatin-bound proteins located between the enhancer and the promoter, and appears to involve the catalytic activity of poly (ADP-ribose) polymerase. Our data suggest that models of enhancer-promoter communication need to encompass chromatin conformations other than looping.


Asunto(s)
Ensamble y Desensamble de Cromatina , Elementos de Facilitación Genéticos , Proteínas Hedgehog/metabolismo , Células Madre Embrionarias de Ratones/metabolismo , Neurogénesis , Neuronas/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Animales , Línea Celular , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/genética , Ratones , Modelos Genéticos , Neurogénesis/genética , Conformación de Ácido Nucleico , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo
13.
Nat Genet ; 50(5): 767, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29440723

RESUMEN

In the version of this article initially published, Wendy Bickmore and Madapura Pradeepa were incorrectly not indicated as corresponding authors. The error has been corrected in the HTML and PDF versions of the paper.

14.
Nat Genet ; 50(3): 329-332, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29379197

RESUMEN

We found that the clinical phenotype associated with BRD4 haploinsufficiency overlapped with that of Cornelia de Lange syndrome (CdLS), which is most often caused by mutation of NIPBL. More typical CdLS was observed with a de novo BRD4 missense variant, which retained the ability to coimmunoprecipitate with NIPBL, but bound poorly to acetylated histones. BRD4 and NIPBL displayed correlated binding at super-enhancers and appeared to co-regulate developmental gene expression.


Asunto(s)
Síndrome de Cornelia de Lange/genética , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Animales , Sitios de Unión/genética , Proteínas de Ciclo Celular , Células Cultivadas , Niño , Preescolar , Elementos de Facilitación Genéticos , Femenino , Regulación del Desarrollo de la Expresión Génica , Haploinsuficiencia , Humanos , Masculino , Ratones , Ratones Transgénicos , Linaje , Fenotipo , Unión Proteica
15.
PLoS Genet ; 13(4): e1006677, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28384324

RESUMEN

Long noncoding RNAs (lncRNAs) have been implicated in various biological functions including the regulation of gene expression, however, the functionality of lncRNAs is not clearly understood and conflicting conclusions have often been reached when comparing different methods to investigate them. Moreover, little is known about the upstream regulation of lncRNAs. Here we show that the short isoform (p52) of a transcriptional co-activator-PC4 and SF2 interacting protein (Psip1), which is known to be involved in linking transcription to RNA processing, specifically regulates the expression of the lncRNA Hottip-located at the 5' end of the Hoxa locus. Using both knockdown and knockout approaches we show that Hottip expression is required for activation of the 5' Hoxa genes (Hoxa13 and Hoxa10/11) and for retaining Mll1 at the 5' end of Hoxa. Moreover, we demonstrate that artificially inducing Hottip expression is sufficient to activate the 5' Hoxa genes and that Hottip RNA binds to the 5' end of Hoxa. By engineering premature transcription termination, we show that it is the Hottip lncRNA molecule itself, not just Hottip transcription that is required to maintains active expression of posterior Hox genes. Our data show a direct role for a lncRNA molecule in regulating the expression of developmentally-regulated mRNA genes in cis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas de Homeodominio/genética , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Transcripción Genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proliferación Celular/genética , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas Homeobox A10 , Humanos , Procesamiento Postranscripcional del ARN/genética , ARN Largo no Codificante/biosíntesis , Factores de Transcripción/biosíntesis
16.
Nat Genet ; 48(6): 681-6, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27089178

RESUMEN

Histone acetylation is generally associated with active chromatin, but most studies have focused on the acetylation of histone tails. Various histone H3 and H4 tail acetylations mark the promoters of active genes. These modifications include acetylation of histone H3 at lysine 27 (H3K27ac), which blocks Polycomb-mediated trimethylation of H3K27 (H3K27me3). H3K27ac is also widely used to identify active enhancers, and the assumption has been that profiling H3K27ac is a comprehensive way of cataloguing the set of active enhancers in mammalian cell types. Here we show that acetylation of lysine residues in the globular domain of histone H3 (lysine 64 (H3K64ac) and lysine 122 (H3K122ac)) marks active gene promoters and also a subset of active enhancers. Moreover, we find a new class of active functional enhancers that is marked by H3K122ac but lacks H3K27ac. This work suggests that, to identify enhancers, a more comprehensive analysis of histone acetylation is required than has previously been considered.


Asunto(s)
Elementos de Facilitación Genéticos , Histonas/metabolismo , Acetilación , Inmunoprecipitación de Cromatina , Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Histonas/química , Humanos , Células K562 , Lisina/metabolismo
17.
Nucleic Acids Res ; 42(14): 9021-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25056311

RESUMEN

Trithorax and polycomb group proteins are generally thought to antagonize one another. The trithorax family member MLL (myeloid/lymphoid or mixed-lineage leukemia) is presumed to activate Hox expression, counteracting polycomb-mediated repression. PC4 and SF2 interacting protein 1 (PSIP1)/p75, also known as LEDGF, whose PWWP domain binds to H3K36me3, interacts with MLL and tethers MLL fusion proteins to HOXA9 in leukaemias. Here we show, unexpectedly, that Psip1/p75 regulates homeotic genes by recruiting not only MLL complexes, but also the polycomb group protein Bmi1. In Psip1(-/-) cells binding of Mll1/2, Bmi1 and the co-repressor Ctbp1 at Hox loci are all abrogated and Hoxa and Hoxd mRNA expression increased. Our data not only reveal a potential mechanism of action for Psip1 in the regulation of Hox genes but also suggest an unexpected interplay between proteins usually considered as transcriptional activators and repressors.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Regulación de la Expresión Génica , Genes Homeobox , N-Metiltransferasa de Histona-Lisina/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Oxidorreductasas de Alcohol/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/fisiología
18.
Development ; 139(17): 3157-67, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22872084

RESUMEN

A late phase of HoxD activation is crucial for the patterning and growth of distal structures across the anterior-posterior (A-P) limb axis of mammals. Polycomb complexes and chromatin compaction have been shown to regulate Hox loci along the main body axis in embryonic development, but the extent to which they have a role in limb-specific HoxD expression, an evolutionary adaptation defined by the activity of distal enhancer elements that drive expression of 5' Hoxd genes, has yet to be fully elucidated. We reveal two levels of chromatin topology that differentiate distal limb A-P HoxD activity. Using both immortalised cell lines derived from posterior and anterior regions of distal E10.5 mouse limb buds, and analysis in E10.5 dissected limb buds themselves, we show that there is a loss of polycomb-catalysed H3K27me3 histone modification and a chromatin decompaction over HoxD in the distal posterior limb compared with anterior. Moreover, we show that the global control region (GCR) long-range enhancer spatially colocalises with the 5' HoxD genomic region specifically in the distal posterior limb. This is consistent with the formation of a chromatin loop between 5' HoxD and the GCR regulatory module at the time and place of distal limb bud development when the GCR participates in initiating Hoxd gene quantitative collinearity and Hoxd13 expression. This is the first example of A-P differences in chromatin compaction and chromatin looping in the development of the mammalian secondary body axis (limb).


Asunto(s)
Tipificación del Cuerpo/fisiología , Ensamble y Desensamble de Cromatina/fisiología , Extremidades/embriología , Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Animales , Western Blotting , Línea Celular , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica/genética , Histonas/metabolismo , Procesamiento de Imagen Asistido por Computador , Hibridación Fluorescente in Situ , Ratones , Microscopía Fluorescente , Proteínas del Grupo Polycomb , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/metabolismo
19.
PLoS Genet ; 8(5): e1002717, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22615581

RESUMEN

Increasing evidence suggests that chromatin modifications have important roles in modulating constitutive or alternative splicing. Here we demonstrate that the PWWP domain of the chromatin-associated protein Psip1/Ledgf can specifically recognize tri-methylated H3K36 and that, like this histone modification, the Psip1 short (p52) isoform is enriched at active genes. We show that the p52, but not the long (p75), isoform of Psip1 co-localizes and interacts with Srsf1 and other proteins involved in mRNA processing. The level of H3K36me3 associated Srsf1 is reduced in Psip1 mutant cells and alternative splicing of specific genes is affected. Moreover, we show altered Srsf1 distribution around the alternatively spliced exons of these genes in Psip1 null cells. We propose that Psip1/p52, through its binding to both chromatin and splicing factors, might act to modulate splicing.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Empalme Alternativo/genética , Histonas , Isoformas de Proteínas , Factores de Transcripción/genética , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , Fibroblastos/citología , Regulación de la Expresión Génica , Histonas/genética , Histonas/metabolismo , Metilación , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Empalme Serina-Arginina , Factores de Transcripción/metabolismo
20.
PLoS One ; 7(4): e34848, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496869

RESUMEN

The essential histone variant H2A.Z localises to both active and silent chromatin sites. In embryonic stem cells (ESCs), H2A.Z is also reported to co-localise with polycomb repressive complex 2 (PRC2) at developmentally silenced genes. The mechanism of H2A.Z targeting is not clear, but a role for the PRC2 component Suz12 has been suggested. Given this association, we wished to determine if polycomb functionally directs H2A.Z incorporation in ESCs. We demonstrate that the PRC1 component Ring1B interacts with multiple complexes in ESCs. Moreover, we show that although the genomic distribution of H2A.Z co-localises with PRC2, Ring1B and with the presence of CpG islands, H2A.Z still blankets polycomb target loci in the absence of Suz12, Eed (PRC2) or Ring1B (PRC1). Therefore we conclude that H2A.Z accumulates at developmentally silenced genes in ESCs in a polycomb independent manner.


Asunto(s)
Células Madre Embrionarias/metabolismo , Genes del Desarrollo , Histonas/metabolismo , Proteínas Represoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Cromatina/metabolismo , Islas de CpG , Sitios Genéticos , Ratones , Complejo Represivo Polycomb 1 , Complejo Represivo Polycomb 2 , Proteínas del Grupo Polycomb , Regiones Promotoras Genéticas , Proteínas Represoras/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA