Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38766212

RESUMEN

Understanding the molecular mechanisms underlying tumorigenesis is crucial for developing effective cancer therapies. Here, we investigate the co-amplification of MED30 and MYC across diverse cancer types and its impact on oncogenic transcriptional programs. Transcriptional profiling of MYC and MED30 single or both overexpression/amplification revealed the over amount of MED30 lead MYC to a new transcriptional program that associate with poor prognosis. Mechanistically, MED30 overexpression/amplification recruits other Mediator components and binding of MYC to a small subset of novel genomic regulatory sites, changing the epigenetic marks and inducing the formation of new enhancers, which drive the expression of target genes crucial for cancer progression. In vivo studies in pancreatic ductal adenocarcinoma (PDAC) further validate the oncogenic potential of MED30, as its overexpression promotes tumor growth and can be attenuated by knockdown of MYC. Using another cancer type as an example, MED30 knockdown reduces tumor growth particularly in MYC high-expressed glioblastoma (GBM) cell lines. Overall, our study elucidates the critical role of MED30 overexpression in orchestrating oncogenic transcriptional programs and highlights its potential as a therapeutic target for MYC-amplified cancer.

3.
Nature ; 595(7867): 444-449, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34194047

RESUMEN

The size of the transcriptional program of long non-coding RNAs in the mammalian genome has engendered discussions about their biological roles1, particularly the promoter antisense (PAS) transcripts2,3. Here we report the development of an assay-referred to as chromatin isolation by RNA-Cas13a complex-to quantitatively detect the distribution of RNA in the genome. The assay revealed that PAS RNAs serve as a key gatekeeper of a broad transcriptional pause release program, based on decommissioning the 7SK small nuclear RNA-dependent inhibitory P-TEFb complex. Induction of PAS RNAs by liganded ERα led to a significant loss of H3K9me3 and the release of basally recruited HP1α and KAP1 on activated target gene promoters. This release was due to PAS RNA-dependent recruitment of H3K9me3 demethylases, which required interactions with a compact stem-loop structure in the PAS RNAs, an apparent feature of similarly regulated PAS RNAs. Activation of the ERα-bound MegaTrans enhancer, which is essential for robust pause release, required the recruitment of phosphorylated KAP1, with its transfer to the cognate promoters permitting 17ß-oestradiol-induced pause release and activation of the target gene. This study reveals a mechanism, based on RNA structure, that mediates the function of PAS RNAs in gene regulation.


Asunto(s)
Conformación de Ácido Nucleico , Regiones Promotoras Genéticas/genética , ARN sin Sentido/química , ARN sin Sentido/genética , Activación Transcripcional/genética , Línea Celular , Homólogo de la Proteína Chromobox 5/metabolismo , Proteína Sustrato Asociada a CrK , Receptor alfa de Estrógeno/metabolismo , Histonas/química , Histonas/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Ligandos , Factor B de Elongación Transcripcional Positiva/metabolismo , ARN Polimerasa II/metabolismo , Estabilidad del ARN , Proteína 28 que Contiene Motivos Tripartito/metabolismo
4.
Nature ; 595(7869): 735-740, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34040254

RESUMEN

The functional engagement between an enhancer and its target promoter ensures precise gene transcription1. Understanding the basis of promoter choice by enhancers has important implications for health and disease. Here we report that functional loss of a preferred promoter can release its partner enhancer to loop to and activate an alternative promoter (or alternative promoters) in the neighbourhood. We refer to this target-switching process as 'enhancer release and retargeting'. Genetic deletion, motif perturbation or mutation, and dCas9-mediated CTCF tethering reveal that promoter choice by an enhancer can be determined by the binding of CTCF at promoters, in a cohesin-dependent manner-consistent with a model of 'enhancer scanning' inside the contact domain. Promoter-associated CTCF shows a lower affinity than that at chromatin domain boundaries and often lacks a preferred motif orientation or a partnering CTCF at the cognate enhancer, suggesting properties distinct from boundary CTCF. Analyses of cancer mutations, data from the GTEx project and risk loci from genome-wide association studies, together with a focused CRISPR interference screen, reveal that enhancer release and retargeting represents an overlooked mechanism that underlies the activation of disease-susceptibility genes, as exemplified by a risk locus for Parkinson's disease (NUCKS1-RAB7L1) and three loci associated with cancer (CLPTM1L-TERT, ZCCHC7-PAX5 and PVT1-MYC).


Asunto(s)
Factor de Unión a CCCTC/genética , Elementos de Facilitación Genéticos , Predisposición Genética a la Enfermedad , Regiones Promotoras Genéticas , Sistemas CRISPR-Cas , Proteínas de Ciclo Celular/genética , Células Cultivadas , Cromatina , Proteínas Cromosómicas no Histona/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Células MCF-7 , Neoplasias/genética , Células-Madre Neurales , Oncogenes , Enfermedad de Parkinson/genética , Cohesinas
5.
Circulation ; 142(22): 2138-2154, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32933333

RESUMEN

BACKGROUND: Concentric and eccentric cardiac hypertrophy are associated with pressure and volume overload, respectively, in cardiovascular disease both conferring an increased risk of heart failure. These contrasting forms of hypertrophy are characterized by asymmetrical growth of the cardiac myocyte in mainly width or length, respectively. The molecular mechanisms determining myocyte preferential growth in width versus length remain poorly understood. Identification of the mechanisms governing asymmetrical myocyte growth could provide new therapeutic targets for the prevention or treatment of heart failure. METHODS: Primary adult rat ventricular myocytes, adeno-associated virus (AAV)-mediated gene delivery in mice, and human tissue samples were used to define a regulatory pathway controlling pathological myocyte hypertrophy. Chromatin immunoprecipitation assays with sequencing and precision nuclear run-on sequencing were used to define a transcriptional mechanism. RESULTS: We report that asymmetrical cardiac myocyte hypertrophy is modulated by SRF (serum response factor) phosphorylation, constituting an epigenomic switch balancing the growth in width versus length of adult ventricular myocytes in vitro and in vivo. SRF Ser103 phosphorylation is bidirectionally regulated by RSK3 (p90 ribosomal S6 kinase type 3) and PP2A (protein phosphatase 2A) at signalosomes organized by the scaffold protein mAKAPß (muscle A-kinase anchoring protein ß), such that increased SRF phosphorylation activates AP-1 (activator protein-1)-dependent enhancers that direct myocyte growth in width. AAV are used to express in vivo mAKAPß-derived RSK3 and PP2A anchoring disruptor peptides that block the association of the enzymes with the mAKAPß scaffold. Inhibition of RSK3 signaling prevents concentric cardiac remodeling induced by pressure overload, while inhibition of PP2A signaling prevents eccentric cardiac remodeling induced by myocardial infarction, in each case improving cardiac function. SRF Ser103 phosphorylation is significantly decreased in dilated human hearts, supporting the notion that modulation of the mAKAPß-SRF signalosome could be a new therapeutic approach for human heart failure. CONCLUSIONS: We have identified a new molecular switch, namely mAKAPß signalosome-regulated SRF phosphorylation, that controls a transcriptional program responsible for modulating changes in cardiac myocyte morphology that occur secondary to pathological stressors. Complementary AAV-based gene therapies constitute rationally-designed strategies for a new translational modality for heart failure.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Aumento de la Célula , Insuficiencia Cardíaca/metabolismo , Miocitos Cardíacos/metabolismo , Factor de Respuesta Sérica/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Adenoviridae/genética , Animales , Animales Recién Nacidos , Células Cultivadas , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/patología , Fosforilación/fisiología , Ratas , Ratas Sprague-Dawley
6.
Nat Struct Mol Biol ; 26(3): 193-203, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30833784

RESUMEN

A crucial feature of differentiated cells is the rapid activation of enhancer-driven transcriptional programs in response to signals. The potential contributions of physicochemical properties of enhancer assembly in signaling events remain poorly understood. Here we report that in human breast cancer cells, the acute 17ß-estradiol-dependent activation of functional enhancers requires assembly of an enhancer RNA-dependent ribonucleoprotein (eRNP) complex exhibiting properties of phase-separated condensates. Unexpectedly, while acute ligand-dependent assembly of eRNPs resulted in enhancer activation sensitive to chemical disruption of phase separation, chronically activated enhancers proved resistant to such disruption, with progressive maturation of eRNPs to a more gel-like state. Acute, but not chronic, stimulation resulted in ligand-induced, condensin-dependent changes in spatial chromatin conformation based on homotypic enhancer association, resulting in cooperative enhancer-activation events. Thus, distinct physicochemical properties of eRNP condensates on enhancers serve as determinants of rapid ligand-dependent alterations in chromosomal architecture and cooperative enhancer activation.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Estradiol/metabolismo , Ribonucleoproteínas/metabolismo , Activación Transcripcional/fisiología , Línea Celular Tumoral , Cromatina , Cromosomas/fisiología , Humanos , Células MCF-7 , Conformación Proteica , Transcripción Genética/genética , Activación Transcripcional/genética
7.
Nature ; 556(7702): 510-514, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29670286

RESUMEN

Enhancers for embryonic stem (ES) cell-expressed genes and lineage-determining factors are characterized by conventional marks of enhancer activation in ES cells1-3, but it remains unclear whether enhancers destined to regulate cell-type-restricted transcription units might also have distinct signatures in ES cells. Here we show that cell-type-restricted enhancers are 'premarked' and activated as transcription units by the binding of one or two ES cell transcription factors, although they do not exhibit traditional enhancer epigenetic marks in ES cells, thus uncovering the initial temporal origins of cell-type-restricted enhancers. This premarking is required for future cell-type-restricted enhancer activity in the differentiated cells, with the strength of the ES cell signature being functionally important for the subsequent robustness of cell-type-restricted enhancer activation. We have experimentally validated this model in macrophage-restricted enhancers and neural precursor cell (NPC)-restricted enhancers using ES cell-derived macrophages or NPCs, edited to contain specific ES cell transcription factor motif deletions. DNA hydroxyl-methylation of enhancers in ES cells, determined by ES cell transcription factors, may serve as a potential molecular memory for subsequent enhancer activation in mature macrophages. These findings suggest that the massive repertoire of cell-type-restricted enhancers are essentially hierarchically and obligatorily premarked by binding of a defining ES cell transcription factor in ES cells, dictating the robustness of enhancer activation in mature cells.


Asunto(s)
Diferenciación Celular/genética , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica/genética , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Animales , Epigénesis Genética , Femenino , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Especificidad de Órganos , Células Madre Pluripotentes/citología , Reproducibilidad de los Resultados
8.
Mol Cell ; 66(3): 321-331.e6, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475868

RESUMEN

The molecular mechanisms underlying the opposing functions of glucocorticoid receptors (GRs) and estrogen receptor α (ERα) in breast cancer development remain poorly understood. Here we report that, in breast cancer cells, liganded GR represses a large ERα-activated transcriptional program by binding, in trans, to ERα-occupied enhancers. This abolishes effective activation of these enhancers and their cognate target genes, and it leads to the inhibition of ERα-dependent binding of components of the MegaTrans complex. Consistent with the effects of SUMOylation on other classes of nuclear receptors, dexamethasone (Dex)-induced trans-repression of the estrogen E2 program appears to depend on GR SUMOylation, which leads to stable trans-recruitment of the GR-N-CoR/SMRT-HDAC3 corepressor complex on these enhancers. Together, these results uncover a mechanism by which competitive recruitment of DNA-binding nuclear receptors/transcription factors in trans to hot spot enhancers serves as an effective biological strategy for trans-repression, with clear implications for breast cancer and other diseases.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica , Receptor Cross-Talk , Receptores de Glucocorticoides/metabolismo , Transcripción Genética , Sitios de Unión , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Dexametasona/farmacología , Regulación hacia Abajo , Elementos de Facilitación Genéticos , Estradiol/farmacología , Receptor alfa de Estrógeno/agonistas , Receptor alfa de Estrógeno/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Células MCF-7 , Complejos Multiproteicos , Mutación , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Co-Represor 2 de Receptor Nuclear/genética , Co-Represor 2 de Receptor Nuclear/metabolismo , Unión Proteica , Interferencia de ARN , Receptor Cross-Talk/efectos de los fármacos , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/genética , Transducción de Señal , Sumoilación , Transcripción Genética/efectos de los fármacos , Transcriptoma , Transfección
9.
Neuron ; 86(3): 696-710, 2015 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-25892301

RESUMEN

One of the exceptional properties of the brain is its ability to acquire new knowledge through learning and to store that information through memory. The epigenetic mechanisms linking changes in neuronal transcriptional programs to behavioral plasticity remain largely unknown. Here, we identify the epigenetic signature of the neuronal enhancers required for transcriptional regulation of synaptic plasticity genes during memory formation, linking this to Reelin signaling. The binding of Reelin to its receptor, LRP8, triggers activation of this cohort of LRP8-Reelin-regulated neuronal (LRN) enhancers that serve as the ultimate convergence point of a novel synapse-to-nucleus pathway. Reelin simultaneously regulates NMDA-receptor transmission, which reciprocally permits the required γ-secretase-dependent cleavage of LRP8, revealing an unprecedented role for its intracellular domain in the regulation of synaptically generated signals. These results uncover an in vivo enhancer code serving as a critical molecular component of cognition and relevant to psychiatric disorders linked to defects in Reelin signaling.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/metabolismo , Condicionamiento Clásico/fisiología , Proteínas de la Matriz Extracelular/metabolismo , Proteínas Relacionadas con Receptor de LDL/metabolismo , Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Serina Endopeptidasas/metabolismo , Animales , Bicuculina/farmacología , Proteína de Unión a CREB/metabolismo , Moléculas de Adhesión Celular Neuronal/genética , Células Cultivadas , Embrión de Mamíferos , Inhibidores Enzimáticos/farmacología , Antagonistas de Aminoácidos Excitadores/farmacología , Proteínas de la Matriz Extracelular/genética , Histona Desacetilasas/metabolismo , Humanos , Proteínas Relacionadas con Receptor de LDL/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , N-Acetilglucosaminiltransferasas/genética , Proteínas del Tejido Nervioso/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Proteína Reelina , Serina Endopeptidasas/genética , Transducción de Señal/genética , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética
10.
Proc Natl Acad Sci U S A ; 112(5): 1380-5, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605944

RESUMEN

Substantial evidence supports the hypothesis that enhancers are critical regulators of cell-type determination, orchestrating both positive and negative transcriptional programs; however, the basic mechanisms by which enhancers orchestrate interactions with cognate promoters during activation and repression events remain incompletely understood. Here we report the required actions of LIM domain-binding protein 1 (LDB1)/cofactor of LIM homeodomain protein 2/nuclear LIM interactor, interacting with the enhancer-binding protein achaete-scute complex homolog 1, to mediate looping to target gene promoters and target gene regulation in corticotrope cells. LDB1-mediated enhancer:promoter looping appears to be required for both activation and repression of these target genes. Although LDB1-dependent activated genes are regulated at the level of transcriptional initiation, the LDB1-dependent repressed transcription units appear to be regulated primarily at the level of promoter pausing, with LDB1 regulating recruitment of metastasis-associated 1 family, member 2, a component of the nucleosome remodeling deacetylase complex, on these negative enhancers, required for the repressive enhancer function. These results indicate that LDB1-dependent looping events can deliver repressive cargo to cognate promoters to mediate promoter pausing events in a pituitary cell type.


Asunto(s)
Corticotrofos/fisiología , Proteínas de Unión al ADN/fisiología , Elementos de Facilitación Genéticos , Proteínas con Dominio LIM/fisiología , Regiones Promotoras Genéticas , Animales , Línea Celular , Proteínas de Unión al ADN/metabolismo , Proteínas con Dominio LIM/metabolismo , Ratones , Ratones Noqueados
11.
Cell ; 160(3): 367-80, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25619691

RESUMEN

The discovery that enhancers are regulated transcription units, encoding eRNAs, has raised new questions about the mechanisms of their activation. Here, we report an unexpected molecular mechanism that underlies ligand-dependent enhancer activation, based on DNA nicking to relieve torsional stress from eRNA synthesis. Using dihydrotestosterone (DHT)-induced binding of androgen receptor (AR) to prostate cancer cell enhancers as a model, we show rapid recruitment, within minutes, of DNA topoisomerase I (TOP1) to a large cohort of AR-regulated enhancers. Furthermore, we show that the DNA nicking activity of TOP1 is a prerequisite for robust eRNA synthesis and enhancer activation and is kinetically accompanied by the recruitment of ATR and the MRN complex, followed by additional components of DNA damage repair machinery to the AR-regulated enhancers. Together, our studies reveal a linkage between eRNA synthesis and ligand-dependent TOP1-mediated nicking-a strategy exerting quantitative effects on eRNA expression in regulating AR-bound enhancer-dependent transcriptional programs.


Asunto(s)
ADN-Topoisomerasas de Tipo I/metabolismo , Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Receptores Androgénicos/metabolismo , Línea Celular Tumoral , Roturas del ADN de Cadena Simple , Reparación del ADN , ADN-Topoisomerasas de Tipo I/genética , Proteínas de Unión al ADN/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Humanos , Proteína Homóloga de MRE11 , Factores de Transcripción/metabolismo , Transcripción Genética
12.
Cell ; 159(2): 358-73, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25303530

RESUMEN

Enhancers provide critical information directing cell-type-specific transcriptional programs, regulated by binding of signal-dependent transcription factors and their associated cofactors. Here, we report that the most strongly activated estrogen (E2)-responsive enhancers are characterized by trans-recruitment and in situ assembly of a large 1-2 MDa complex of diverse DNA-binding transcription factors by ERα at ERE-containing enhancers. We refer to enhancers recruiting these factors as mega transcription factor-bound in trans (MegaTrans) enhancers. The MegaTrans complex is a signature of the most potent functional enhancers and is required for activation of enhancer RNA transcription and recruitment of coactivators, including p300 and Med1. The MegaTrans complex functions, in part, by recruiting specific enzymatic machinery, exemplified by DNA-dependent protein kinase. Thus, MegaTrans-containing enhancers represent a cohort of functional enhancers that mediate a broad and important transcriptional program and provide a molecular explanation for transcription factor clustering and hotspots noted in the genome.


Asunto(s)
Elementos de Facilitación Genéticos , Receptor alfa de Estrógeno/metabolismo , Factores de Transcripción/metabolismo , Estrógenos/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Factor Nuclear 3-alfa del Hepatocito/metabolismo , Humanos , Complejos Multiproteicos/metabolismo
13.
Nature ; 516(7530): 267-71, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25252977

RESUMEN

Post-translational histone modifications have a critical role in regulating transcription, the cell cycle, DNA replication and DNA damage repair. The identification of new histone modifications critical for transcriptional regulation at initiation, elongation or termination is of particular interest. Here we report a new layer of regulation in transcriptional elongation that is conserved from yeast to mammals. This regulation is based on the phosphorylation of a highly conserved tyrosine residue, Tyr 57, in histone H2A and is mediated by the unsuspected tyrosine kinase activity of casein kinase 2 (CK2). Mutation of Tyr 57 in H2A in yeast or inhibition of CK2 activity impairs transcriptional elongation in yeast as well as in mammalian cells. Genome-wide binding analysis reveals that CK2α, the catalytic subunit of CK2, binds across RNA-polymerase-II-transcribed coding genes and active enhancers. Mutation of Tyr 57 causes a loss of H2B mono-ubiquitination as well as H3K4me3 and H3K79me3, histone marks associated with active transcription. Mechanistically, both CK2 inhibition and the H2A(Y57F) mutation enhance H2B deubiquitination activity of the Spt-Ada-Gcn5 acetyltransferase (SAGA) complex, suggesting a critical role of this phosphorylation in coordinating the activity of the SAGA complex during transcription. Together, these results identify a new component of regulation in transcriptional elongation based on CK2-dependent tyrosine phosphorylation of the globular domain of H2A.


Asunto(s)
Quinasa de la Caseína II/metabolismo , Histonas/química , Histonas/metabolismo , Elongación de la Transcripción Genética , Tirosina/metabolismo , Secuencia de Aminoácidos , Línea Celular , Secuencia Conservada , Histonas/genética , Humanos , Datos de Secuencia Molecular , Fosforilación , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Tirosina/química , Ubiquitinación/genética
14.
Nature ; 514(7521): 257-61, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25119036

RESUMEN

Homeodomain proteins, described 30 years ago, exert essential roles in development as regulators of target gene expression; however, the molecular mechanisms underlying transcriptional activity of homeodomain factors remain poorly understood. Here investigation of a developmentally required POU-homeodomain transcription factor, Pit1 (also known as Pou1f1), has revealed that, unexpectedly, binding of Pit1-occupied enhancers to a nuclear matrin-3-rich network/architecture is a key event in effective activation of the Pit1-regulated enhancer/coding gene transcriptional program. Pit1 association with Satb1 (ref. 8) and ß-catenin is required for this tethering event. A naturally occurring, dominant negative, point mutation in human PIT1(R271W), causing combined pituitary hormone deficiency, results in loss of Pit1 association with ß-catenin and Satb1 and therefore the matrin-3-rich network, blocking Pit1-dependent enhancer/coding target gene activation. This defective activation can be rescued by artificial tethering of the mutant R271W Pit1 protein to the matrin-3 network, bypassing the pre-requisite association with ß-catenin and Satb1 otherwise required. The matrin-3 network-tethered R271W Pit1 mutant, but not the untethered protein, restores Pit1-dependent activation of the enhancers and recruitment of co-activators, exemplified by p300, causing both enhancer RNA transcription and target gene activation. These studies have thus revealed an unanticipated homeodomain factor/ß-catenin/Satb1-dependent localization of target gene regulatory enhancer regions to a subnuclear architectural structure that serves as an underlying mechanism by which an enhancer-bound homeodomain factor effectively activates developmental gene transcriptional programs.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Proteínas de Homeodominio/genética , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Hipófisis/embriología , Hipófisis/metabolismo , Unión Proteica , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/metabolismo , Transcripción Genética/genética , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 111(25): 9235-40, 2014 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-24928520

RESUMEN

Understanding the mechanisms by which compounds discovered using cell-based phenotypic screening strategies might exert their effects would be highly augmented by new approaches exploring their potential interactions with the genome. For example, altered androgen receptor (AR) transcriptional programs, including castration resistance and subsequent chromosomal translocations, play key roles in prostate cancer pathological progression, making the quest for identification of new therapeutic agents and an understanding of their actions a continued priority. Here we report an approach that has permitted us to uncover the sites and mechanisms of action of a drug, referred to as "SD70," initially identified by phenotypic screening for inhibitors of ligand and genotoxic stress-induced translocations in prostate cancer cells. Based on synthesis of a derivatized form of SD70 that permits its application for a ChIP-sequencing-like approach, referred to as "Chem-seq," we were next able to efficiently map the genome-wide binding locations of this small molecule, revealing that it largely colocalized with AR on regulatory enhancers. Based on these observations, we performed the appropriate global analyses to ascertain that SD70 inhibits the androgen-dependent AR program, and prostate cancer cell growth, acting, at least in part, by functionally inhibiting the Jumonji domain-containing demethylase, KDM4C. Global location of candidate drugs represents a powerful strategy for new drug development by mapping genome-wide location of small molecules, a powerful adjunct to contemporary drug development strategies.


Asunto(s)
Antagonistas de Receptores Androgénicos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Proteínas de Neoplasias/genética , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Antineoplásicos , Línea Celular Tumoral , Análisis Mutacional de ADN/métodos , Humanos , Masculino , Ratones , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Translocación Genética
16.
Nature ; 500(7464): 598-602, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23945587

RESUMEN

Although recent studies have indicated roles of long non-coding RNAs (lncRNAs) in physiological aspects of cell-type determination and tissue homeostasis, their potential involvement in regulated gene transcription programs remains rather poorly understood. The androgen receptor regulates a large repertoire of genes central to the identity and behaviour of prostate cancer cells, and functions in a ligand-independent fashion in many prostate cancers when they become hormone refractory after initial androgen deprivation therapy. Here we report that two lncRNAs highly overexpressed in aggressive prostate cancer, PRNCR1 (also known as PCAT8) and PCGEM1, bind successively to the androgen receptor and strongly enhance both ligand-dependent and ligand-independent androgen-receptor-mediated gene activation programs and proliferation in prostate cancer cells. Binding of PRNCR1 to the carboxy-terminally acetylated androgen receptor on enhancers and its association with DOT1L appear to be required for recruitment of the second lncRNA, PCGEM1, to the androgen receptor amino terminus that is methylated by DOT1L. Unexpectedly, recognition of specific protein marks by PCGEM1-recruited pygopus 2 PHD domain enhances selective looping of androgen-receptor-bound enhancers to target gene promoters in these cells. In 'resistant' prostate cancer cells, these overexpressed lncRNAs can interact with, and are required for, the robust activation of both truncated and full-length androgen receptor, causing ligand-independent activation of the androgen receptor transcriptional program and cell proliferation. Conditionally expressed short hairpin RNA targeting these lncRNAs in castration-resistant prostate cancer cell lines strongly suppressed tumour xenograft growth in vivo. Together, these results indicate that these overexpressed lncRNAs can potentially serve as a required component of castration-resistance in prostatic tumours.


Asunto(s)
ARN Largo no Codificante/genética , Receptores Androgénicos/metabolismo , Activación Transcripcional/genética , Regulación hacia Arriba/genética , Animales , Castración , Línea Celular Tumoral , Proliferación Celular , Elementos de Facilitación Genéticos/genética , Humanos , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Regiones Promotoras Genéticas/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Factores de Transcripción/metabolismo
17.
Nature ; 498(7455): 516-20, 2013 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-23728302

RESUMEN

The functional importance of gene enhancers in regulated gene expression is well established. In addition to widespread transcription of long non-coding RNAs (lncRNAs) in mammalian cells, bidirectional ncRNAs are transcribed on enhancers, and are thus referred to as enhancer RNAs (eRNAs). However, it has remained unclear whether these eRNAs are functional or merely a reflection of enhancer activation. Here we report that in human breast cancer cells 17ß-oestradiol (E2)-bound oestrogen receptor α (ER-α) causes a global increase in eRNA transcription on enhancers adjacent to E2-upregulated coding genes. These induced eRNAs, as functional transcripts, seem to exert important roles for the observed ligand-dependent induction of target coding genes, increasing the strength of specific enhancer-promoter looping initiated by ER-α binding. Cohesin, present on many ER-α-regulated enhancers even before ligand treatment, apparently contributes to E2-dependent gene activation, at least in part by stabilizing E2/ER-α/eRNA-induced enhancer-promoter looping. Our data indicate that eRNAs are likely to have important functions in many regulated programs of gene transcription.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Estrógenos/farmacología , ARN no Traducido/genética , Activación Transcripcional/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Estradiol/farmacología , Receptor alfa de Estrógeno/metabolismo , Humanos , Ligandos , Células MCF-7 , Conformación de Ácido Nucleico/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN no Traducido/biosíntesis , ARN no Traducido/metabolismo , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Activación Transcripcional/genética , Cohesinas
18.
Nat Struct Mol Biol ; 19(11): 1168-75, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23064648

RESUMEN

Although liganded nuclear receptors have been established to regulate RNA polymerase II (Pol II)-dependent transcription units, their role in regulating Pol III-transcribed DNA repeats remains largely unknown. Here we report that ~2-3% of the ~100,000-200,000 total human DR2 Alu repeats located in proximity to activated Pol II transcription units are activated by the retinoic acid receptor (RAR) in human embryonic stem cells to generate Pol III-dependent RNAs. These transcripts are processed, initially in a DICER-dependent fashion, into small RNAs (~28-65 nt) referred to as repeat-induced RNAs that cause the degradation of a subset of crucial stem-cell mRNAs, including Nanog mRNA, which modulate exit from the proliferative stem-cell state. This regulation requires AGO3-dependent accumulation of processed DR2 Alu transcripts and the subsequent recruitment of AGO3-associated decapping complexes to the target mRNA. In this way, the RAR-dependent and Pol III-dependent DR2 Alu transcriptional events in stem cells functionally complement the Pol II-dependent neuronal transcriptional program.


Asunto(s)
Proteínas Argonautas/metabolismo , ARN Helicasas DEAD-box/metabolismo , Células Madre Embrionarias/fisiología , ARN Interferente Pequeño/metabolismo , Receptores de Ácido Retinoico/metabolismo , Ribonucleasa III/metabolismo , Transcripción Genética/fisiología , Elementos Alu/genética , Elementos Alu/fisiología , Secuencia de Bases , Northern Blotting , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , ADN Polimerasa III/fisiología , Células Madre Embrionarias/metabolismo , Humanos , Hibridación Fluorescente in Situ , Espectrometría de Masas , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN
19.
Mol Cell ; 46(1): 91-104, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22424771

RESUMEN

The association between hyperinflammatory states and numerous diseases is widely recognized, but our understanding of the molecular strategies that have evolved to prevent uncontrolled activation of inflammatory responses remains incomplete. Here, we report a critical, nontranscriptional role of GPS2 as a guardian against hyperstimulation of the TNF-α-induced gene program. GPS2 cytoplasmic actions are required to specifically modulate RIP1 ubiquitylation and JNK activation by inhibiting TRAF2/Ubc13 enzymatic activity. In vivo relevance of GPS2 anti-inflammatory role is confirmed by inhibition of TNF-α target genes in macrophages and by improved insulin signaling in the adipose tissue of aP2-GPS2 transgenic mice. As the nontranscriptional role is complemented by GPS2 functioning as positive and negative cofactor for nuclear receptors, in vivo overexpression also results in elevated circulating level of Resistin and development of hepatic steatosis. Together, these studies define GPS2 as a molecular guardian required for precise control of inflammatory responses involved in immunity and homeostasis.


Asunto(s)
Tejido Adiposo/metabolismo , Homeostasis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/metabolismo , Tejido Adiposo/inmunología , Animales , Línea Celular , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/inmunología , Proteínas Activadoras de GTPasa/metabolismo , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Insulina/genética , Insulina/inmunología , Insulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/inmunología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/inmunología , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Resistina/genética , Resistina/inmunología , Resistina/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/inmunología , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/genética , Ubiquitinación/inmunología
20.
Cell ; 147(4): 773-88, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22078878

RESUMEN

Although eukaryotic nuclei contain distinct architectural structures associated with noncoding RNAs (ncRNAs), their potential relationship to regulated transcriptional programs remains poorly understood. Here, we report that methylation/demethylation of Polycomb 2 protein (Pc2) controls relocation of growth-control genes between Polycomb bodies (PcGs) and interchromatin granules (ICGs) in response to growth signals. This movement is the consequence of binding of methylated and unmethylated Pc2 to the ncRNAs TUG1 and MALAT1/NEAT2, located in PcGs and ICGs, respectively. These ncRNAs mediate assembly of multiple corepressors/coactivators and can serve to switch mark recognition by "readers" of the histone code. Additionally, binding of NEAT2 to unmethylated Pc2 promotes E2F1 SUMOylation, leading to activation of the growth-control gene program. These observations delineate a molecular pathway linking the actions of subnuclear structure-specific ncRNAs and nonhistone protein methylation to relocation of transcription units in the three-dimensional space of the nucleus, thus achieving coordinated gene expression programs.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Línea Celular , Cromatina/metabolismo , Factor de Transcripción E2F1/metabolismo , Células HeLa , Humanos , Ligasas , Metilación , Metiltransferasas/metabolismo , Datos de Secuencia Molecular , Proteínas del Grupo Polycomb , ARN Largo no Codificante , Proteínas Represoras/química , Sumoilación , Transcripción Genética , Ubiquitina-Proteína Ligasas , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...