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










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 7288, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33790356

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/genética , Autorrenovação Celular , Proteínas Cromossômicas não Histona/genética , Células-Tronco Hematopoéticas/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Leucemia Mieloide Aguda/metabolismo , Animais , Benzimidazóis/farmacologia , Proteínas de Ciclo Celular/deficiência , Células Cultivadas , Proteínas Cromossômicas não Histona/deficiência , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/fisiologia , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Coesinas
2.
J Biol Chem ; 296: 100189, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33334884

RESUMO

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.


Assuntos
Proteína Homeobox Nanog/genética , RNA Polimerase II/genética , Animais , Sistemas CRISPR-Cas , Linhagem Celular , Elementos Facilitadores Genéticos , Edição de Genes , Regulação da Expressão Gênica , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Ativação Transcricional
3.
Cancer Immunol Res ; 7(10): 1647-1662, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31515257

RESUMO

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.


Assuntos
Inflamação/imunologia , Células Matadoras Naturais/imunologia , Melanoma Experimental/imunologia , MicroRNAs/genética , Linfócitos T Citotóxicos/imunologia , Fator 6 Associado a Receptor de TNF/metabolismo , Fator de Transcrição AP-1/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Células Cultivadas , Citocinas/metabolismo , Feminino , Humanos , Inflamação/metabolismo , Inflamação/patologia , Masculino , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/imunologia , MicroRNAs/metabolismo , Transdução de Sinais , Ubiquitina/metabolismo , Ubiquitinação
4.
Cell Rep ; 18(7): 1713-1726, 2017 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-28199843

RESUMO

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.


Assuntos
Proteínas Correpressoras/metabolismo , Histona Desacetilase 2/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Proteína Homeobox Nanog/metabolismo , Proteínas Repressoras/metabolismo , Animais , Reprogramação Celular/genética , Reprogramação Celular/fisiologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox/genética , Genoma/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Camundongos , Complexo Correpressor Histona Desacetilase e Sin3 , Transcrição Gênica/genética
5.
Methods Mol Biol ; 1468: 91-109, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27662872

RESUMO

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.


Assuntos
Imunoprecipitação da Cromatina/métodos , Elementos Facilitadores Genéticos , Análise de Sequência de RNA/métodos , Animais , Biologia Computacional/métodos , Regulação da Expressão Gênica , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Regiões Promotoras Genéticas , RNA Longo não Codificante/genética , Reação em Cadeia da Polimerase em Tempo Real , Transcrição Gênica
6.
Cell Rep ; 17(1): 19-28, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27681417

RESUMO

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.


Assuntos
Elementos Facilitadores Genéticos , Células-Tronco Embrionárias Humanas/metabolismo , Proteína Homeobox Nanog/genética , Células-Tronco Pluripotentes/metabolismo , Proteínas/genética , RNA Longo não Codificante/genética , Animais , Sistemas CRISPR-Cas , Proteínas Cromossômicas não Histona , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Endonucleases/genética , Endonucleases/metabolismo , Edição de Genes , Regulação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Humanos , Camundongos , Células NIH 3T3 , Proteína Homeobox Nanog/metabolismo , Células-Tronco Pluripotentes/citologia , Cultura Primária de Células , Proteínas/metabolismo , RNA Longo não Codificante/metabolismo , Transcrição Gênica
7.
Artigo em Inglês | MEDLINE | ID: mdl-27087855

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-23962787

RESUMO

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.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Hipertensão/genética , Miócitos Cardíacos/metabolismo , RNA Mensageiro/genética , Receptores da Neurocinina-1/genética , Substância P/genética , Remodelação Ventricular , Animais , Modelos Animais de Doenças , Feminino , Ventrículos do Coração/embriologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/fisiopatologia , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Miócitos Cardíacos/patologia , Reação em Cadeia da Polimerase , Gravidez , Prenhez , RNA Mensageiro/biossíntese , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Receptores da Neurocinina-1/biossíntese , Substância P/biossíntese , Função Ventricular Esquerda
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...