Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 7288, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33790356

RESUMEN

Acute myeloid leukemia (AML) is a high-risk malignancy characterized by a diverse spectrum of somatic genetic alterations. The mechanisms by which these mutations contribute to leukemia development and how this informs the use of targeted therapies is critical to improving outcomes for patients. Importantly, how to target loss-of-function mutations has been a critical challenge in precision medicine. Heterozygous inactivating mutations in cohesin complex genes contribute to AML in adults by increasing the self-renewal capacity of hematopoietic stem and progenitor cells (HSPCs) by altering PRC2 targeting to induce HOXA9 expression, a key self-renewal transcription factor. Here we sought to delineate the epigenetic mechanism underpinning the enhanced self-renewal conferred by cohesin-haploinsufficiency. First, given the substantial difference in the mutational spectrum between pediatric and adult AML patients, we first sought to identify if HOXA9 was also elevated in children. Next, using primary HSPCs as a model we demonstrate that abnormal self-renewal due to cohesin loss is blocked by DOT1L inhibition. In cohesin-depleted cells, DOT1L inhibition is associated with H3K79me2 depletion and a concomitant increase in H3K27me3. Importantly, we find that there are cohesin-dependent gene expression changes that promote a leukemic profile, including HoxA overexpression, that are preferentially reversed by DOT1L inhibition. Our data further characterize how cohesin mutations contribute to AML development, identifying DOT1L as a potential therapeutic target for adult and pediatric AML patients harboring cohesin mutations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Autorrenovación de las Células , Proteínas Cromosómicas no Histona/genética , Células Madre Hematopoyéticas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , Leucemia Mieloide Aguda/metabolismo , Animales , Bencimidazoles/farmacología , Proteínas de Ciclo Celular/deficiencia , Células Cultivadas , Proteínas Cromosómicas no Histona/deficiencia , Inhibidores Enzimáticos/farmacología , Epigénesis Genética , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/fisiología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Cohesinas
2.
J Biol Chem ; 296: 100189, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33334884

RESUMEN

Transcriptional enhancers have been defined by their ability to operate independent of distance and orientation in plasmid-based reporter assays of gene expression. At present, histone marks are used to identify and define enhancers but do not consider the endogenous role of an enhancer in the context of native chromatin. We employed a combination of genomic editing, single cell analyses, and sequencing approaches to investigate a Nanog-associated cis-regulatory element, which has been reported by others to be either an alternative promoter or a super-enhancer. We first demonstrate both distance and orientation independence in native chromatin, eliminating the issues raised with plasmid-based approaches. We next demonstrate that the dominant super-enhancer modulates Nanog globally and operates by recruiting and/or initiating RNA Polymerase II. Our studies have important implications to how transcriptional enhancers are defined and how they regulate gene expression.


Asunto(s)
Proteína Homeótica Nanog/genética , ARN Polimerasa II/genética , Animales , Sistemas CRISPR-Cas , Línea Celular , Elementos de Facilitación Genéticos , Edición Génica , Regulación de la Expresión Génica , Ratones , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Activación Transcripcional
3.
Cancer Immunol Res ; 7(10): 1647-1662, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31515257

RESUMEN

Natural killer (NK) cells generate proinflammatory cytokines that are required to contain infections and tumor growth. However, the posttranscriptional mechanisms that regulate NK cell functions are not fully understood. Here, we define the role of the microRNA cluster known as Mirc11 (which includes miRNA-23a, miRNA-24a, and miRNA-27a) in NK cell-mediated proinflammatory responses. Absence of Mirc11 did not alter the development or the antitumor cytotoxicity of NK cells. However, loss of Mirc11 reduced generation of proinflammatory factors in vitro and interferon-γ-dependent clearance of Listeria monocytogenes or B16F10 melanoma in vivo by NK cells. These functional changes resulted from Mirc11 silencing ubiquitin modifiers A20, Cbl-b, and Itch, allowing TRAF6-dependent activation of NF-κB and AP-1. Lack of Mirc11 caused increased translation of A20, Cbl-b, and Itch proteins, resulting in deubiquitylation of scaffolding K63 and addition of degradative K48 moieties on TRAF6. Collectively, our results describe a function of Mirc11 that regulates generation of proinflammatory cytokines from effector lymphocytes.


Asunto(s)
Inflamación/inmunología , Células Asesinas Naturales/inmunología , Melanoma Experimental/inmunología , MicroARNs/genética , Linfocitos T Citotóxicos/inmunología , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción AP-1/metabolismo , Factor de Transcripción ReIA/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/inmunología , MicroARNs/metabolismo , Transducción de Señal , Ubiquitina/metabolismo , Ubiquitinación
4.
Cell Rep ; 18(7): 1713-1726, 2017 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-28199843

RESUMEN

Although SIN3A is required for the survival of early embryos and embryonic stem cells (ESCs), the role of SIN3A in the maintenance and establishment of pluripotency remains unclear. Here, we find that the SIN3A/HDAC corepressor complex maintains ESC pluripotency and promotes the generation of induced pluripotent stem cells (iPSCs). Members of the SIN3A/HDAC corepressor complex are enriched in an extended NANOG interactome and function in transcriptional coactivation in ESCs. We also identified a critical role for SIN3A and HDAC2 in efficient reprogramming of somatic cells. Mechanistically, NANOG and SIN3A co-occupy transcriptionally active pluripotency genes in ESCs and also co-localize extensively at their genome-wide targets in pre-iPSCs. Additionally, both factors are required to directly induce a synergistic transcriptional program wherein pluripotency genes are activated and reprogramming barrier genes are repressed. Our findings indicate a transcriptional regulatory role for a major HDAC-containing complex in promoting pluripotency.


Asunto(s)
Proteínas Co-Represoras/metabolismo , Histona Desacetilasa 2/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteínas Represoras/metabolismo , Animales , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Genes Homeobox/genética , Genoma/genética , Células Madre Pluripotentes Inducidas/fisiología , Ratones , Complejo Correpresor Histona Desacetilasa y Sin3 , Transcripción Genética/genética
5.
Methods Mol Biol ; 1468: 91-109, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27662872

RESUMEN

Recent work has shown that RNA polymerase II-mediated transcription at distal cis-regulatory elements serves as a mark of highly active enhancers. Production of noncoding RNAs at enhancers, termed eRNAs, correlates with higher expression of genes that the enhancer interacts with; hence, eRNAs provide a new tool to model gene activity in normal and disease tissues. Moreover, this unique class of noncoding RNA has diverse roles in transcriptional regulation. Transcribed enhancers can be identified by a common signature of epigenetic marks by overlaying a series of genome-wide chromatin immunoprecipitation and RNA sequencing datasets. A computational approach to filter non-enhancer elements and other classes of noncoding RNAs is essential to not cloud downstream analysis. Here we present a protocol that combines wet and dry bench methods to accurately identify transcribed enhancers genome-wide as well as an experimental procedure to validate these datasets.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Elementos de Facilitación Genéticos , Análisis de Secuencia de ARN/métodos , Animales , Biología Computacional/métodos , Regulación de la Expresión Génica , Ratones , Células Madre Embrionarias de Ratones/citología , Regiones Promotoras Genéticas , ARN Largo no Codificante/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transcripción Genética
6.
Cell Rep ; 17(1): 19-28, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681417

RESUMEN

Super-enhancers are tissue-specific cis-regulatory elements that drive expression of genes associated with cell identity and malignancy. A cardinal feature of super-enhancers is that they are transcribed to produce enhancer-derived RNAs (eRNAs). It remains unclear whether super-enhancers robustly activate genes in situ and whether their functions are attributable to eRNAs or the DNA element. CRISPR/Cas9 was used to systematically delete three discrete super-enhancers at the Nanog locus in embryonic stem cells, revealing functional differences in Nanog transcriptional regulation. One distal super-enhancer 45 kb upstream of Nanog (-45 enhancer) regulates both nearest neighbor genes, Nanog and Dppa3. Interestingly, eRNAs produced at the -45 enhancer specifically regulate Dppa3 expression by stabilizing looping of the -45 enhancer and Dppa3. Our work illustrates that genomic editing is required to determine enhancer function and points to a method to selectively target a subset of super-enhancer-regulated genes by depleting eRNAs.


Asunto(s)
Elementos de Facilitación Genéticos , Células Madre Embrionarias Humanas/metabolismo , Proteína Homeótica Nanog/genética , Células Madre Pluripotentes/metabolismo , Proteínas/genética , ARN Largo no Codificante/genética , Animales , Sistemas CRISPR-Cas , Proteínas Cromosómicas no Histona , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Endonucleasas/genética , Endonucleasas/metabolismo , Edición Génica , Regulación de la Expresión Génica , Células Madre Embrionarias Humanas/citología , Humanos , Ratones , Células 3T3 NIH , Proteína Homeótica Nanog/metabolismo , Células Madre Pluripotentes/citología , Cultivo Primario de Células , Proteínas/metabolismo , ARN Largo no Codificante/metabolismo , Transcripción Genética
7.
Artículo en Inglés | MEDLINE | ID: mdl-27087855

RESUMEN

BACKGROUND: The cohesin complex consists of multiple core subunits that play critical roles in mitosis and transcriptional regulation. The cohesin-associated protein Wapal plays a central role in off-loading cohesin to facilitate sister chromatid separation, but its role in regulating mammalian gene expression is not understood. We used embryonic stem cells as a model, given that the well-defined transcriptional regulatory circuits were established through master transcription factors and epigenetic pathways that regulate their ability to maintain a pluripotent state. RESULTS: RNAi-mediated depletion of Wapal causes a loss of pluripotency, phenocopying loss of core cohesin subunits. Using chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq), we determine that Wapal occupies genomic sites distal to genes in combination with CTCF and core cohesin subunits such as Rad21. Interestingly, genomic sites occupied by Wapal appear enriched for cohesin, implying that Wapal does not off-load cohesin at regions it occupies. Wapal depletion induces derepression of Polycomb group (PcG) target genes without altering total levels of Polycomb-mediated histone modifications, implying that PcG enzymatic activity is preserved. By integrating ChIP-seq and gene expression changes data, we identify that Wapal binding is enriched at the promoters of PcG-silenced genes and is required for proper Polycomb repressive complex 2 (PRC2) recruitment. Lastly, we demonstrate that Wapal is required for the interaction of a distal cis-regulatory element (CRE) with the c-Fos promoter. CONCLUSIONS: Collectively, this work indicates that Wapal plays a critical role in silencing of PcG target genes through the interaction of distal CREs with promoters.

8.
Int J Cardiol ; 168(5): 4643-51, 2013 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-23962787

RESUMEN

BACKGROUND: Substance P is a sensory nerve neuropeptide located near coronary vessels in the heart. Therefore, substance P may be one of the first mediators released in the heart in response to hypertension, and can contribute to adverse myocardial remodeling via interactions with the neurokinin-1 receptor. We asked: 1) whether substance P promoted cardiac hypertrophy, including the expression of fetal genes known to be re-expressed during pathological hypertrophy; and 2) the extent to which substance P regulated collagen production and fibrosis. METHODS AND RESULTS: Spontaneously hypertensive rats (SHR) were treated with the neurokinin-1 receptor antagonist L732138 (5mg/kg/d) from 8 to 24 weeks of age. Age-matched WKY served as controls. The gene encoding substance P, TAC1, was up-regulated as blood pressure increased in SHR. Fetal gene expression by cardiomyocytes was increased in SHR and was prevented by L732138. Cardiac fibrosis also occurred in the SHR and was prevented by L732138. Endothelin-1 was up-regulated in the SHR and this was prevented by L732138. In isolated cardiac fibroblasts, substance P transiently up-regulated several genes related to cell-cell adhesion, cell-matrix adhesion, and extracellular matrix regulation, however, no changes in fibroblast function were observed. CONCLUSIONS: Substance P activation of the neurokinin-1 receptor induced expression of fetal genes related to pathological hypertrophy in the hypertensive heart. Additionally, activation of the neurokinin-1 receptor was critical to the development of cardiac fibrosis. Since no functional changes were induced in isolated cardiac fibroblasts by substance P, we conclude that substance P mediates fibrosis via up-regulation of endothelin-1.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Hipertensión/genética , Miocitos Cardíacos/metabolismo , ARN Mensajero/genética , Receptores de Neuroquinina-1/genética , Sustancia P/genética , Remodelación Ventricular , Animales , Modelos Animales de Enfermedad , Femenino , Ventrículos Cardíacos/embriología , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Hipertensión/metabolismo , Hipertensión/fisiopatología , Miocitos Cardíacos/patología , Reacción en Cadena de la Polimerasa , Embarazo , Preñez , ARN Mensajero/biosíntesis , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Receptores de Neuroquinina-1/biosíntesis , Sustancia P/biosíntesis , Función Ventricular Izquierda
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...