Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(1): 8-9, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34995510

RESUMEN

Hsieh et al. (2022) reveal that carbon starvation elicits an unexpected compensatory reallocation of histone acetylation to establish an adaptive gene expression program, demonstrating how chromatin may integrate cellular carbon flow via histone acetylation with gene regulation.


Asunto(s)
Cromatina , Histonas , Acetilación , Carbono , Cromatina/genética , Expresión Génica , Histonas/genética , Histonas/metabolismo , Orgánulos/metabolismo
2.
Cell ; 150(3): 590-605, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863011

RESUMEN

Endothelium in embryonic hematopoietic tissues generates hematopoietic stem/progenitor cells; however, it is unknown how its unique potential is specified. We show that transcription factor Scl/Tal1 is essential for both establishing the hematopoietic transcriptional program in hemogenic endothelium and preventing its misspecification to a cardiomyogenic fate. Scl(-/-) embryos activated a cardiac transcriptional program in yolk sac endothelium, leading to the emergence of CD31+Pdgfrα+ cardiogenic precursors that generated spontaneously beating cardiomyocytes. Ectopic cardiogenesis was also observed in Scl(-/-) hearts, where the disorganized endocardium precociously differentiated into cardiomyocytes. Induction of mosaic deletion of Scl in Scl(fl/fl)Rosa26Cre-ER(T2) embryos revealed a cell-intrinsic, temporal requirement for Scl to prevent cardiomyogenesis from endothelium. Scl(-/-) endothelium also upregulated the expression of Wnt antagonists, which promoted rapid cardiomyocyte differentiation of ectopic cardiogenic cells. These results reveal unexpected plasticity in embryonic endothelium such that loss of a single master regulator can induce ectopic cardiomyogenesis from endothelial cells.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Endotelio Vascular/embriología , Corazón/embriología , Proteínas Proto-Oncogénicas/metabolismo , Animales , Cadherinas/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Hemangioblastos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Mesodermo/metabolismo , Ratones , Miocitos Cardíacos/citología , Placenta/irrigación sanguínea , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Embarazo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína 1 de la Leucemia Linfocítica T Aguda , Factores de Transcripción/metabolismo , Saco Vitelino/irrigación sanguínea
3.
Mol Cell ; 73(2): 250-263.e5, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30527662

RESUMEN

Metazoan chromosomes are sequentially partitioned into topologically associating domains (TADs) and then into smaller sub-domains. One class of sub-domains, insulated neighborhoods, are proposed to spatially sequester and insulate the enclosed genes through self-association and chromatin looping. However, it has not been determined functionally whether promoter-enhancer interactions and gene regulation are broadly restricted to within these loops. Here, we employed published datasets from murine embryonic stem cells (mESCs) to identify insulated neighborhoods that confine promoter-enhancer interactions and demarcate gene regulatory regions. To directly address the functionality of these regions, we depleted estrogen-related receptor ß (Esrrb), which binds the Mediator co-activator complex, to impair enhancers of genes within 222 insulated neighborhoods without causing mESC differentiation. Esrrb depletion reduces Mediator binding, promoter-enhancer looping, and expression of both nascent RNA and mRNA within the insulated neighborhoods without significantly affecting the flanking genes. Our data indicate that insulated neighborhoods represent functional regulons in mammalian genomes.


Asunto(s)
Cromosomas de los Mamíferos , Elementos de Facilitación Genéticos , Elementos Aisladores , Células Madre Embrionarias de Ratones/fisiología , Regiones Promotoras Genéticas , Transcripción Genética , Animales , Sitios de Unión , Factor de Unión a CCCTC/genética , Factor de Unión a CCCTC/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Bases de Datos Genéticas , Regulación hacia Abajo , Ratones , Unión Proteica , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Cohesinas
4.
PLoS Biol ; 21(10): e3002371, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37889915

RESUMEN

Perez and Sarkies uncover histones as methyl group repositories in normal and cancer human cells, shedding light on an intriguing function of histone methylation in optimizing the cellular methylation potential independently of gene regulation.


Asunto(s)
Histonas , Neoplasias , Humanos , Histonas/metabolismo , Metilación , Regulación de la Expresión Génica , Neoplasias/genética , Histona Metiltransferasas/metabolismo
5.
J Biol Chem ; 300(6): 107314, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38657861

RESUMEN

The copper reductase activity of histone H3 suggests undiscovered characteristics within the protein. Here, we investigated the function of leucine 126 (H3L126), which occupies an axial position relative to the copper binding. Typically found as methionine or leucine in copper-binding proteins, the axial ligand influences the reduction potential of the bound ion, modulating its tendency to accept or yield electrons. We found that mutation of H3L126 to methionine (H3L126M) enhanced the enzymatic activity of native yeast nucleosomes in vitro and increased intracellular levels of Cu1+, leading to improved copper-dependent activities including mitochondrial respiration and growth in oxidative media with low copper. Conversely, H3L126 to histidine (H3L126H) mutation decreased nucleosome's enzymatic activity and adversely affected copper-dependent activities in vivo. Our findings demonstrate that H3L126 fine-tunes the copper reductase activity of nucleosomes and highlights the utility of nucleosome enzymatic activity as a novel paradigm to uncover previously unnoticed features of histones.

6.
Nature ; 576(7786): 281-286, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31776511

RESUMEN

Limited knowledge of the mechanisms that govern the self-renewal of human haematopoietic stem cells (HSCs), and why this fails in culture, have impeded the expansion of HSCs for transplantation1. Here we identify MLLT3 (also known as AF9) as a crucial regulator of HSCs that is highly enriched in human fetal, neonatal and adult HSCs, but downregulated in culture. Depletion of MLLT3 prevented the maintenance of transplantable human haematopoietic stem or progenitor cells (HSPCs) in culture, whereas stabilizing MLLT3 expression in culture enabled more than 12-fold expansion of transplantable HSCs that provided balanced multilineage reconstitution in primary and secondary mouse recipients. Similar to endogenous MLLT3, overexpressed MLLT3 localized to active promoters in HSPCs, sustained levels of H3K79me2 and protected the HSC transcriptional program in culture. MLLT3 thus acts as HSC maintenance factor that links histone reader and modifying activities to modulate HSC gene expression, and may provide a promising approach to expand HSCs for transplantation.


Asunto(s)
Autorrenovación de las Células , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteínas Nucleares/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Proteínas Nucleares/genética , Unión Proteica
7.
Mol Cell ; 67(4): 594-607.e4, 2017 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-28735899

RESUMEN

Pervasive transcription initiates from cryptic promoters and is observed in eukaryotes ranging from yeast to mammals. The Set2-Rpd3 regulatory system prevents cryptic promoter function within expressed genes. However, conserved systems that control pervasive transcription within intergenic regions have not been well established. Here we show that Mot1, Ino80 chromatin remodeling complex (Ino80C), and NC2 co-localize on chromatin and coordinately suppress pervasive transcription in S. cerevisiae and murine embryonic stem cells (mESCs). In yeast, all three proteins bind subtelomeric heterochromatin through a Sir3-stimulated mechanism and to euchromatin via a TBP-stimulated mechanism. In mESCs, the proteins bind to active and poised TBP-bound promoters along with promoters of polycomb-silenced genes apparently lacking TBP. Depletion of Mot1, Ino80C, or NC2 by anchor away in yeast or RNAi in mESCs leads to near-identical transcriptome phenotypes, with new subtelomeric transcription in yeast, and greatly increased pervasive transcription in both yeast and mESCs.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Células Madre Embrionarias/enzimología , Fosfoproteínas/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/enzimología , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Sitios de Unión , Línea Celular , Proteínas de Unión al ADN , Eucromatina/genética , Eucromatina/metabolismo , Regulación Fúngica de la Expresión Génica , Silenciador del Gen , Genotipo , Heterocromatina/genética , Heterocromatina/metabolismo , Fenotipo , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Proteínas Represoras/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/genética , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/metabolismo , Factores Asociados con la Proteína de Unión a TATA/genética , Proteína de Unión a TATA-Box/genética , Proteína de Unión a TATA-Box/metabolismo , Factor de Transcripción TFIID , Factores de Transcripción/genética , Transfección
8.
Genes Dev ; 31(3): 241-246, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28270516

RESUMEN

Chromobox homolog 3 (Cbx3/heterochromatin protein 1γ [HP1γ]) stimulates cell differentiation, but its mechanism is unknown. We found that Cbx3 binds to gene promoters upon differentiation of murine embryonic stem cells (ESCs) to neural progenitor cells (NPCs) and recruits the Mediator subunit Med26. RNAi knockdown of either Cbx3 or Med26 inhibits neural differentiation while up-regulating genes involved in mesodermal lineage decisions. Thus, Cbx3 and Med26 together ensure the fidelity of lineage specification by enhancing the expression of neural genes and down-regulating genes specific to alternative fates.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proteínas Cromosómicas no Histona/metabolismo , Células Madre Embrionarias/citología , Regulación de la Expresión Génica , Complejo Mediador/metabolismo , Células-Madre Neurales/citología , Animales , Células Cultivadas , Proteínas Cromosómicas no Histona/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/genética , Quinasa 8 Dependiente de Ciclina/genética , Quinasa 8 Dependiente de Ciclina/metabolismo , Células Madre Embrionarias/metabolismo , Complejo Mediador/genética , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Células-Madre Neurales/metabolismo , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética
9.
Mol Cell ; 61(1): 27-38, 2016 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-26669263

RESUMEN

Gene activation in metazoans is accompanied by the presence of histone variants H2AZ and H3.3 within promoters and enhancers. It is not known, however, what protein deposits H3.3 into chromatin or whether variant chromatin plays a direct role in gene activation. Here we show that chromatin containing acetylated H2AZ and H3.3 stimulates transcription in vitro. Analysis of the Pol II pre-initiation complex on immobilized chromatin templates revealed that the E1A binding protein p400 (EP400) was bound preferentially to and required for transcription stimulation by acetylated double-variant chromatin. EP400 also stimulated H2AZ/H3.3 deposition into promoters and enhancers and influenced transcription in vivo at a step downstream of the Mediator complex. EP400 efficiently exchanged recombinant histones H2A and H3.1 with H2AZ and H3.3, respectively, in a chromatin- and ATP-stimulated manner in vitro. Our data reveal that EP400 deposits H3.3 into chromatin alongside H2AZ and contributes to gene regulation after PIC assembly.


Asunto(s)
Ensamble y Desensamble de Cromatina , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Regiones Promotoras Genéticas , Activación Transcripcional , Acetilación , Adenosina Trifosfato/metabolismo , Sitios de Unión , Línea Celular Tumoral , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Genes Reporteros , Histonas/genética , Humanos , Interferencia de ARN , ARN Polimerasa II/metabolismo , Factores de Tiempo , Transfección
10.
Nat Mater ; 21(10): 1191-1199, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35927431

RESUMEN

Cell reprogramming has wide applications in tissue regeneration, disease modelling and personalized medicine. In addition to biochemical cues, mechanical forces also contribute to the modulation of the epigenetic state and a variety of cell functions through distinct mechanisms that are not fully understood. Here we show that millisecond deformation of the cell nucleus caused by confinement into microfluidic channels results in wrinkling and transient disassembly of the nuclear lamina, local detachment of lamina-associated domains in chromatin and a decrease of histone methylation (histone H3 lysine 9 trimethylation) and DNA methylation. These global changes in chromatin at the early stage of cell reprogramming boost the conversion of fibroblasts into neurons and can be partially reproduced by inhibition of histone H3 lysine 9 and DNA methylation. This mechanopriming approach also triggers macrophage reprogramming into neurons and fibroblast conversion into induced pluripotent stem cells, being thus a promising mechanically based epigenetic state modulation method for cell engineering.


Asunto(s)
Reprogramación Celular , Histonas , Núcleo Celular/metabolismo , Cromatina/metabolismo , Metilación de ADN , Epigénesis Genética , Histonas/genética , Histonas/metabolismo , Lisina/genética , Lisina/metabolismo
11.
Genes Dev ; 29(4): 350-5, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25691465

RESUMEN

Here we show that the Ino80 chromatin remodeling complex (Ino80C) directly prevents euchromatin from invading transcriptionally silent chromatin within intergenic regions and at the border of euchromatin and heterochromatin. Deletion of Ino80C subunits leads to increased H3K79 methylation and noncoding RNA polymerase II (Pol II) transcription centered at the Ino80C-binding sites. The effect of Ino80C is direct, as it blocks H3K79 methylation by Dot1 in vitro. Heterochromatin stimulates the binding of Ino80C in vitro and in vivo. Our data reveal that Ino80C serves as a general silencing complex that restricts transcription to gene units in euchromatin.


Asunto(s)
Cromatina/genética , Eucromatina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Sitios de Unión , Eucromatina/genética , Regulación Fúngica de la Expresión Génica , Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Metilación , Proteínas Nucleares/metabolismo , Unión Proteica , ARN Polimerasa II/metabolismo
12.
Development ; 146(19)2019 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-31558570

RESUMEN

Over the past few years, interest in chromatin and its evolution has grown. To further advance these interests, we organized a workshop with the support of The Company of Biologists to debate the current state of knowledge regarding the origin and evolution of chromatin. This workshop led to prospective views on the development of a new field of research that we term 'EvoChromo'. In this short Spotlight article, we define the breadth and expected impact of this new area of scientific inquiry on our understanding of both chromatin and evolution.


Asunto(s)
Cromatina/genética , Evolución Molecular , Animales , Genoma , Humanos
13.
Mol Cell ; 49(2): 310-21, 2013 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-23201122

RESUMEN

Differences in global levels of histone acetylation occur in normal and cancer cells, although the reason why cells regulate these levels has been unclear. Here we demonstrate a role for histone acetylation in regulating intracellular pH (pH(i)). As pH(i) decreases, histones are globally deacetylated by histone deacetylases (HDACs), and the released acetate anions are coexported with protons out of the cell by monocarboxylate transporters (MCTs), preventing further reductions in pH(i). Conversely, global histone acetylation increases as pH(i) rises, such as when resting cells are induced to proliferate. Inhibition of HDACs or MCTs decreases acetate export and lowers pH(i), particularly compromising pH(i) maintenance in acidic environments. Global deacetylation at low pH is reflected at a genomic level by decreased abundance and extensive redistribution of acetylation throughout the genome. Thus, acetylation of chromatin functions as a rheostat to regulate pH(i) with important implications for mechanism of action and therapeutic use of HDAC inhibitors.


Asunto(s)
Histonas/metabolismo , Líquido Intracelular/metabolismo , Procesamiento Proteico-Postraduccional , Acetatos , Acetilación , Metabolismo de los Hidratos de Carbono , Cromatina , Regulación de la Expresión Génica , Glucosa/fisiología , Glutamina/fisiología , Células HeLa , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Histonas/genética , Humanos , Concentración de Iones de Hidrógeno , Ácidos Hidroxámicos/farmacología , Transportadores de Ácidos Monocarboxílicos/metabolismo , Niacinamida/farmacología , Ácido Pirúvico/metabolismo , Análisis de Secuencia de ARN , Transcriptoma
14.
EMBO J ; 34(6): 759-77, 2015 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-25564442

RESUMEN

Scl/Tal1 confers hemogenic competence and prevents ectopic cardiomyogenesis in embryonic endothelium by unknown mechanisms. We discovered that Scl binds to hematopoietic and cardiac enhancers that become epigenetically primed in multipotent cardiovascular mesoderm, to regulate the divergence of hematopoietic and cardiac lineages. Scl does not act as a pioneer factor but rather exploits a pre-established epigenetic landscape. As the blood lineage emerges, Scl binding and active epigenetic modifications are sustained in hematopoietic enhancers, whereas cardiac enhancers are decommissioned by removal of active epigenetic marks. Our data suggest that, rather than recruiting corepressors to enhancers, Scl prevents ectopic cardiogenesis by occupying enhancers that cardiac factors, such as Gata4 and Hand1, use for gene activation. Although hematopoietic Gata factors bind with Scl to both activated and repressed genes, they are dispensable for cardiac repression, but necessary for activating genes that enable hematopoietic stem/progenitor cell development. These results suggest that a unique subset of enhancers in lineage-specific genes that are accessible for regulators of opposing fates during the time of the fate decision provide a platform where the divergence of mutually exclusive fates is orchestrated.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Elementos de Facilitación Genéticos/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/citología , Mesodermo/embriología , Mioblastos Cardíacos/citología , Proteínas Proto-Oncogénicas/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Biblioteca de Genes , Células Madre Hematopoyéticas/fisiología , Humanos , Mesodermo/metabolismo , Análisis por Micromatrices , Modelos Biológicos , Datos de Secuencia Molecular , Mioblastos Cardíacos/fisiología , Análisis de Secuencia de ARN , Proteína 1 de la Leucemia Linfocítica T Aguda
15.
J Biol Chem ; 292(32): 13197-13204, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28637866

RESUMEN

The endoplasmic reticulum (ER)-mitochondria encounter structure (ERMES) is a protein complex that physically tethers the two organelles to each other and creates the physical basis for communication between them. ERMES functions in lipid exchange between the ER and mitochondria, protein import into mitochondria, and maintenance of mitochondrial morphology and genome. Here, we report that ERMES is also required for iron homeostasis. Loss of ERMES components activates an Aft1-dependent iron deficiency response even in iron-replete conditions, leading to accumulation of excess iron inside the cell. This function is independent of known ERMES roles in calcium regulation, phospholipid biosynthesis, or effects on mitochondrial morphology. A mutation in the vacuolar protein sorting 13 (VPS13) gene that rescues the glycolytic phenotype of ERMES mutants suppresses the iron deficiency response and iron accumulation. Our findings reveal that proper communication between the ER and mitochondria is required for appropriate maintenance of cellular iron levels.


Asunto(s)
Retículo Endoplásmico/metabolismo , Hierro/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Modelos Biológicos , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/fisiología , Alelos , Sustitución de Aminoácidos , Retículo Endoplásmico/química , Metabolismo Energético , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Homeostasis , Hierro/análisis , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Mitocondrias/química , Mutación Puntual , Transporte de Proteínas , ARN de Hongos/química , ARN de Hongos/metabolismo , ARN Mensajero/química , ARN Mensajero/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/crecimiento & desarrollo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Análisis de Secuencia de ARN , Espectrofotometría Atómica
16.
J Biol Chem ; 291(30): 15428-46, 2016 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-27226577

RESUMEN

Transcriptome remodeling in heart disease occurs through the coordinated actions of transcription factors, histone modifications, and other chromatin features at pathology-associated genes. The extent to which genome-wide chromatin reorganization also contributes to the resultant changes in gene expression remains unknown. We examined the roles of two chromatin structural proteins, Ctcf (CCCTC-binding factor) and Hmgb2 (high mobility group protein B2), in regulating pathologic transcription and chromatin remodeling. Our data demonstrate a reciprocal relationship between Hmgb2 and Ctcf in controlling aspects of chromatin structure and gene expression. Both proteins regulate each others' expression as well as transcription in cardiac myocytes; however, only Hmgb2 does so in a manner that involves global reprogramming of chromatin accessibility. We demonstrate that the actions of Hmgb2 on local chromatin accessibility are conserved across genomic loci, whereas the effects on transcription are loci-dependent and emerge in concert with histone modification and other chromatin features. Finally, although both proteins share gene targets, Hmgb2 and Ctcf, neither binds these genes simultaneously nor do they physically colocalize in myocyte nuclei. Our study uncovers a previously unknown relationship between these two ubiquitous chromatin proteins and provides a mechanistic explanation for how Hmgb2 regulates gene expression and cellular phenotype. Furthermore, we provide direct evidence for structural remodeling of chromatin on a genome-wide scale in the setting of cardiac disease.


Asunto(s)
Cromatina/metabolismo , Regulación de la Expresión Génica/fisiología , Proteína HMGB2/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Represoras/metabolismo , Animales , Factor de Unión a CCCTC , Cromatina/genética , Epigenómica , Femenino , Células HEK293 , Proteína HMGB2/genética , Células HeLa , Humanos , Ratones , Proteínas Represoras/genética
17.
J Immunol ; 192(8): 3981-9, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24639349

RESUMEN

Modern immune therapies (PD-1/PD-L1 and CTLA-4 checkpoints blockade and adoptive cell transfer) have remarkably improved the response rates of metastatic melanoma. These modalities rely on the killing potential of CTL as proximal mediator of antimelanoma responses. Mechanisms of tumor resistance to and the predominant cytotoxic pathway(s) used by melanoma-reactive CTL are important outcome determinants. We hypothesized that downmodulation of death receptors (DRs) in addition to aberrant apoptotic signaling might confer resistance to death signals delivered by CTL. To test these two hypotheses, we used an in vitro model of MART CTL-resistant melanoma sublines. TCR-transgenic and patient-derived CTLs used the TRAIL cytotoxic pathway through DR5. Furthermore, recombinant human TRAIL and drozitumab (anti-DR5 agonistic mAb) were used to explicitly verify the contribution of the DR5/TRAIL pathway in killing melanomas. CTL resistance was due to DR5 downregulation and an inverted ratio of pro- to antiapoptotic molecules, both of which were reversed by the histone deacetylase inhibitor suberoylanilide hydroxanic acid. Apoptosis negative (c-IAP-2 and Bcl-xL) and positive (DR5) regulators were potential incriminators partly regulating CTL sensitivity. These preclinical findings suggest that exposure to this chromatin remodeling drug of immune-resistant melanomas can skew toward an intracellular proapoptotic milieu, increase DR expression, and overcome acquired immune resistance.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Melanoma/inmunología , Melanoma/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Apoptosis/genética , Caspasa 3/metabolismo , Línea Celular Tumoral , Citotoxicidad Inmunológica/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación Neoplásica de la Expresión Génica , Humanos , Isoantígenos/genética , Isoantígenos/inmunología , Melanoma/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Linfocitos T Citotóxicos/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Transducción Genética
18.
Proc Natl Acad Sci U S A ; 110(31): 12655-60, 2013 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-23852730

RESUMEN

The positive transcription elongation factor b (P-TEFb) is involved in physiological and pathological events including inflammation, cancer, AIDS, and cardiac hypertrophy. The balance between its active and inactive form is tightly controlled to ensure cellular integrity. We report that the transcriptional repressor CTIP2 is a major modulator of P-TEFb activity. CTIP2 copurifies and interacts with an inactive P-TEFb complex containing the 7SK snRNA and HEXIM1. CTIP2 associates directly with HEXIM1 and, via the loop 2 of the 7SK snRNA, with P-TEFb. In this nucleoprotein complex, CTIP2 significantly represses the Cdk9 kinase activity of P-TEFb. Accordingly, we show that CTIP2 inhibits large sets of P-TEFb- and 7SK snRNA-sensitive genes. In hearts of hypertrophic cardiomyopathic mice, CTIP2 controls P-TEFb-sensitive pathways involved in the establishment of this pathology. Overexpression of the ß-myosin heavy chain protein contributes to the pathological cardiac wall thickening. The inactive P-TEFb complex associates with CTIP2 at the MYH7 gene promoter to repress its activity. Taken together, our results strongly suggest that CTIP2 controls P-TEFb function in physiological and pathological conditions.


Asunto(s)
Cardiomegalia/metabolismo , Factor B de Elongación Transcripcional Positiva/metabolismo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Miosinas Cardíacas/genética , Miosinas Cardíacas/metabolismo , Cardiomegalia/genética , Cardiomegalia/patología , Quinasa 9 Dependiente de la Ciclina/genética , Quinasa 9 Dependiente de la Ciclina/metabolismo , Células HEK293 , Humanos , Ratones , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Factor B de Elongación Transcripcional Positiva/genética , Estructura Secundaria de Proteína , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Proteínas Represoras/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética
19.
Genome Res ; 22(7): 1212-21, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22499665

RESUMEN

Adenovirus small e1a oncoprotein causes ~70% reduction in cellular levels of histone H3 lysine 18 acetylation (H3K18ac). It is unclear, however, where this dramatic reduction occurs genome-wide. ChIP-sequencing revealed that by 24 h after expression, e1a erases 95% of H3K18ac peaks in normal, contact-inhibited fibroblasts and replaces them with one-third as many at new genomic locations. The H3K18ac peaks at promoters and intergenic regions of genes with fibroblast-related functions are eliminated after infection, and new H3K18ac peaks are established at promoters of highly induced genes that regulate cell cycling and at new putative enhancers. Strikingly, the regions bound by the retinoblastoma family of proteins in contact-inhibited fibroblasts gain new peaks of H3K18ac in the e1a-expressing cells, including 55% of RB1-bound loci. In contrast, over half of H3K9ac peaks are similarly distributed before and after infection, independently of RB1. The strategic redistribution of H3K18ac by e1a highlights the importance of this modification for transcriptional activation and cellular transformation as well as functional differences between the RB-family member proteins.


Asunto(s)
Proteínas E1A de Adenovirus/metabolismo , Adenovirus Humanos/genética , Epigénesis Genética , Genoma Humano , Histonas/metabolismo , Acetilación , Proteínas E1A de Adenovirus/genética , Adenovirus Humanos/metabolismo , Adenovirus Humanos/patogenicidad , Ciclo Celular , Transformación Celular Viral , Células Cultivadas , Inmunoprecipitación de Cromatina , Fibroblastos/metabolismo , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Histonas/genética , Humanos , Pulmón/metabolismo , Pulmón/patología , Pulmón/virología , Anotación de Secuencia Molecular/métodos , Nucleosomas/genética , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Factores de Tiempo , Activación Transcripcional
20.
Nat Rev Genet ; 10(5): 290-4, 2009 05.
Artículo en Inglés | MEDLINE | ID: mdl-19290008

RESUMEN

The cancerous cellular state is associated with multiple epigenetic alterations, but elucidating the precise order of such alterations during tumorigenic progression and their contributions to the transformed phenotype remains a significant challenge in cancer biology. Here we discuss recent findings on how viral oncoproteins exploit specific epigenetic processes to coerce normal cells to replicate when they should remain quiescent - a hallmark of cancer. These findings may highlight roles of epigenetic processes in normal biology and shed light on epigenetic events occurring along the path of non-viral neoplastic transformation.


Asunto(s)
Transformación Celular Viral/genética , Epigénesis Genética/genética , Genoma , Proteínas Oncogénicas Virales/metabolismo , Animales , Humanos , Neoplasias/genética , Neoplasias/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA