Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 17(8): 2087-2100, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27851970

RESUMEN

The t(8;21) acute myeloid leukemia (AML)-associated oncoprotein AML1-ETO disrupts normal hematopoietic differentiation. Here, we have investigated its effects on the transcriptome and epigenome in t(8,21) patient cells. AML1-ETO binding was found at promoter regions of active genes with high levels of histone acetylation but also at distal elements characterized by low acetylation levels and binding of the hematopoietic transcription factors LYL1 and LMO2. In contrast, ERG, FLI1, TAL1, and RUNX1 bind at all AML1-ETO-occupied regulatory regions, including those of the AML1-ETO gene itself, suggesting their involvement in regulating AML1-ETO expression levels. While expression of AML1-ETO in myeloid differentiated induced pluripotent stem cells (iPSCs) induces leukemic characteristics, overexpression increases cell death. We find that expression of wild-type transcription factors RUNX1 and ERG in AML is required to prevent this oncogene overexpression. Together our results show that the interplay of the epigenome and transcription factors prevents apoptosis in t(8;21) AML cells.


Asunto(s)
Apoptosis/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Hematopoyesis/genética , Leucemia Mieloide Aguda/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína 1 Compañera de Translocación de RUNX1/metabolismo , Translocación Genética , Acetilación , Secuencia de Bases , Línea Celular Tumoral , Linaje de la Célula/genética , Supervivencia Celular/genética , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Técnicas de Silenciamiento del Gen , Genoma Humano , Histona Desacetilasas/metabolismo , Humanos , Leucemia Mieloide Aguda/patología , Oncogenes , Regiones Promotoras Genéticas , Unión Proteica/genética , Regulador Transcripcional ERG/metabolismo
2.
Cell ; 167(5): 1354-1368.e14, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863248

RESUMEN

Innate immune memory is the phenomenon whereby innate immune cells such as monocytes or macrophages undergo functional reprogramming after exposure to microbial components such as lipopolysaccharide (LPS). We apply an integrated epigenomic approach to characterize the molecular events involved in LPS-induced tolerance in a time-dependent manner. Mechanistically, LPS-treated monocytes fail to accumulate active histone marks at promoter and enhancers of genes in the lipid metabolism and phagocytic pathways. Transcriptional inactivity in response to a second LPS exposure in tolerized macrophages is accompanied by failure to deposit active histone marks at promoters of tolerized genes. In contrast, ß-glucan partially reverses the LPS-induced tolerance in vitro. Importantly, ex vivo ß-glucan treatment of monocytes from volunteers with experimental endotoxemia re-instates their capacity for cytokine production. Tolerance is reversed at the level of distal element histone modification and transcriptional reactivation of otherwise unresponsive genes. VIDEO ABSTRACT.


Asunto(s)
Tolerancia Inmunológica , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Sepsis/inmunología , Transcripción Genética , beta-Glucanos/inmunología , Diferenciación Celular , Metilación de ADN , Epigenómica , Redes Reguladoras de Genes , Código de Histonas , Humanos , Inmunidad Innata , Memoria Inmunológica , Macrófagos/citología , Monocitos/citología , Sepsis/genética
3.
Cell ; 167(5): 1415-1429.e19, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863252

RESUMEN

Many common variants have been associated with hematological traits, but identification of causal genes and pathways has proven challenging. We performed a genome-wide association analysis in the UK Biobank and INTERVAL studies, testing 29.5 million genetic variants for association with 36 red cell, white cell, and platelet properties in 173,480 European-ancestry participants. This effort yielded hundreds of low frequency (<5%) and rare (<1%) variants with a strong impact on blood cell phenotypes. Our data highlight general properties of the allelic architecture of complex traits, including the proportion of the heritable component of each blood trait explained by the polygenic signal across different genome regulatory domains. Finally, through Mendelian randomization, we provide evidence of shared genetic pathways linking blood cell indices with complex pathologies, including autoimmune diseases, schizophrenia, and coronary heart disease and evidence suggesting previously reported population associations between blood cell indices and cardiovascular disease may be non-causal.


Asunto(s)
Variación Genética , Estudio de Asociación del Genoma Completo , Células Madre Hematopoyéticas/metabolismo , Enfermedades del Sistema Inmune/genética , Alelos , Diferenciación Celular , Predisposición Genética a la Enfermedad , Células Madre Hematopoyéticas/patología , Humanos , Enfermedades del Sistema Inmune/patología , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Población Blanca/genética
4.
Science ; 345(6204): 1251086, 2014 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-25258085

RESUMEN

Monocyte differentiation into macrophages represents a cornerstone process for host defense. Concomitantly, immunological imprinting of either tolerance or trained immunity determines the functional fate of macrophages and susceptibility to secondary infections. We characterized the transcriptomes and epigenomes in four primary cell types: monocytes and in vitro-differentiated naïve, tolerized, and trained macrophages. Inflammatory and metabolic pathways were modulated in macrophages, including decreased inflammasome activation, and we identified pathways functionally implicated in trained immunity. ß-glucan training elicits an exclusive epigenetic signature, revealing a complex network of enhancers and promoters. Analysis of transcription factor motifs in deoxyribonuclease I hypersensitive sites at cell-type-specific epigenetic loci unveiled differentiation and treatment-specific repertoires. Altogether, we provide a resource to understand the epigenetic changes that underlie innate immunity in humans.


Asunto(s)
Diferenciación Celular/genética , Epigénesis Genética , Inmunidad Innata/genética , Macrófagos/citología , Monocitos/citología , Animales , Sitios de Unión/genética , Desoxirribonucleasa I/química , Impresión Genómica , Humanos , Memoria Inmunológica , Inflamasomas/genética , Inflamasomas/inmunología , Macrófagos/inmunología , Ratones , Monocitos/inmunología , Factores de Transcripción/metabolismo , beta-Glucanos/inmunología
5.
Genome Res ; 24(3): 401-10, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24336765

RESUMEN

During embryonic development, maintenance of cell identity and lineage commitment requires the Polycomb-group PRC2 complex, which catalyzes histone H3 lysine 27 trimethylation (H3K27me3). However, the developmental origins of this regulation are unknown. Here we show that H3K27me3 enrichment increases from blastula stages onward in embryos of the Western clawed frog (Xenopus tropicalis) within constrained domains strictly defined by sequence. Strikingly, although PRC2 also binds widely to active enhancers, H3K27me3 is only deposited at a small subset of these sites. Using a Support Vector Machine algorithm, these sequences can be predicted accurately on the basis of DNA sequence alone, with a sequence signature conserved between humans, frogs, and fish. These regions correspond to the subset of blastula-stage DNA methylation-free domains that are depleted for activating promoter motifs, and enriched for motifs of developmental factors. These results imply a genetic-default model in which a preexisting absence of DNA methylation is the major determinant of H3K27 methylation when not opposed by transcriptional activation. The sequence and motif signatures reveal the hierarchical and genetically inheritable features of epigenetic cross-talk that impose constraints on Polycomb regulation and guide H3K27 methylation during the exit of pluripotency.


Asunto(s)
Blástula/metabolismo , Núcleo Celular/genética , Gástrula/metabolismo , Histonas/metabolismo , Complejo Represivo Polycomb 2/fisiología , Proteínas de Xenopus/genética , Xenopus/embriología , Animales , Secuencia de Bases , Secuencia Conservada , Metilación de ADN , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Procesamiento Proteico-Postraduccional , Máquina de Vectores de Soporte , Xenopus/genética , Xenopus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...