Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
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
2.
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
3.
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
4.
Mol Cell ; 62(3): 422-431, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-27153539

RESUMEN

The role of cytosine methylation in the structure and function of enhancers is not well understood. In this study, we investigate the role of DNA methylation at enhancers by comparing the epigenomes of the HCT116 cell line and its highly demethylated derivative, DKO1. Unlike promoters, a portion of regular and super- or stretch enhancers show active H3K27ac marks co-existing with extensive DNA methylation, demonstrating the unexpected presence of bivalent chromatin in both cultured and uncultured cells. Furthermore, our findings also show that bivalent regions have fewer nucleosome-depleted regions and transcription factor-binding sites than monovalent regions. Reduction of DNA methylation genetically or pharmacologically leads to a decrease of the H3K27ac mark. Thus, DNA methylation plays an unexpected dual role at enhancer regions, being anti-correlated focally at transcription factor-binding sites but positively correlated globally with the active H3K27ac mark to ensure structural enhancer integrity.


Asunto(s)
Metilación de ADN , Elementos de Facilitación Genéticos , Epigénesis Genética , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Acetilación , Sitios de Unión , Ensamble y Desensamble de Cromatina , Citosina , Células HCT116 , Histonas/genética , Humanos , Factores de Tiempo
5.
Circulation ; 138(12): 1224-1235, 2018 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-29950403

RESUMEN

BACKGROUND: Genetic diversity and the heterogeneous nature of cardiac fibroblasts (CFbs) have hindered characterization of the molecular mechanisms that regulate cardiac fibrosis. The Hybrid Mouse Diversity Panel offers a valuable tool to examine genetically diverse cardiac fibroblasts and their role in fibrosis. METHODS: Three strains of mice (C57BL/6J, C3H/HeJ, and KK/HlJ) were selected from the Hybrid Mouse Diversity Panel and treated with either isoproterenol (ISO) or saline by an intraperitoneally implanted osmotic pump. After 21 days, cardiac function and levels of fibrosis were measured by echocardiography and trichrome staining, respectively. Activation and proliferation of CFbs were measured by in vitro and in vivo assays under normal and injury conditions. RNA sequencing was done on isolated CFbs from each strain. Results were analyzed by Ingenuity Pathway Analysis and validated by reverse transcription-qPCR, immunohistochemistry, and ELISA. RESULTS: ISO treatment in C57BL/6J, C3H/HeJ, and KK/HlJ mice resulted in minimal, moderate, and extensive levels of fibrosis, respectively (n=7-8 hearts per condition). Isolated CFbs treated with ISO exhibited strain-specific increases in the levels of activation but showed comparable levels of proliferation. Similar results were found in vivo, with fibroblast activation, and not proliferation, correlating with the differential levels of cardiac fibrosis after ISO treatment. RNA sequencing revealed that CFbs from each strain exhibit unique gene expression changes in response to ISO. We identified Ltbp2 as a commonly upregulated gene after ISO treatment. Expression of LTBP2 was elevated and specifically localized in the fibrotic regions of the myocardium after injury in mice and in human heart failure patients. CONCLUSIONS: This study highlights the importance of genetic variation in cardiac fibrosis by using multiple inbred mouse strains to characterize CFbs and their response to ISO treatment. Our data suggest that, although fibroblast activation is a response that parallels the extent of scar formation, proliferation may not necessarily correlate with levels of fibrosis. In addition, by comparing CFbs from multiple strains, we identified pathways as potential therapeutic targets and LTBP2 as a marker for fibrosis, with relevance to patients with underlying myocardial fibrosis.


Asunto(s)
Cardiomiopatías/genética , Cardiomiopatías/patología , Proliferación Celular , Fibroblastos/patología , Variación Genética , Proteínas de Unión a TGF-beta Latente/genética , Animales , Cardiomiopatías/inducido químicamente , Cardiomiopatías/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Fibrosis , Predisposición Genética a la Enfermedad , Isoproterenol , Proteínas de Unión a TGF-beta Latente/metabolismo , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Fenotipo , Especificidad de la Especie , Transcriptoma
6.
Hum Mol Genet ; 26(17): 3253-3270, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28595297

RESUMEN

Fibroblast Growth Factor (FGF) signaling promotes self-renewal in progenitor cells by encouraging proliferation and inhibiting cellular senescence. Yet, these beneficial effects can be hijacked by disease-causing mutations in FGF receptor (FGFR) during embryogenesis. By studying dominant FGFR2 mutations that are germline in bent bone dysplasia syndrome (BBDS), we reveal a mechanistic connection between FGFR2, ribosome biogenesis, and cellular stress that links cell fate determination to disease pathology. We previously showed that FGFR2 mutations in BBDS, which amplify nucleolar targeting of FGFR2, activate ribosomal DNA (rDNA) transcription and delay differentiation in osteoprogenitor cells and patient-derived bone. Here we find that the BBDS mutations augment the ability of FGFR2 to recruit histone-remodeling factors that epigenetically activate transcriptionally silent rDNA. Nucleolar morphology is controlled by chromatin structure, and the high levels of euchromatic rDNA induced by the BBDS mutations direct nucleolar disorganization, alter ribosome biogenesis, and activate the Rpl11-Mdm2-p53 nucleolar stress response pathway. Inhibition of p53 in cells expressing the FGFR2 mutations in BBDS rescues delayed osteoblast differentiation, suggesting that p53 activation is an essential pathogenic factor in, and potential therapeutic target for, BBDS. This work establishes rDNA as developmentally regulated loci that receive direct input from FGF signaling to balance self-renewal and cell fate determination.


Asunto(s)
Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Enfermedades del Desarrollo Óseo/genética , Enfermedades del Desarrollo Óseo/metabolismo , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , ADN Ribosómico/metabolismo , Humanos , Mutación , Osteoblastos/metabolismo , Osteogénesis/genética , Fragmentos de Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ribosomas/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo
7.
Genome Res ; 25(4): 467-77, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25747664

RESUMEN

The holistic role of DNA methylation in the organization of the cancer epigenome is not well understood. Here we perform a comprehensive, high-resolution analysis of chromatin structure to compare the landscapes of HCT116 colon cancer cells and a DNA methylation-deficient derivative. The NOMe-seq accessibility assay unexpectedly revealed symmetrical and transcription-independent nucleosomal phasing across active, poised, and inactive genomic elements. DNA methylation abolished this phasing primarily at enhancers and CpG island (CGI) promoters, with little effect on insulators and non-CGI promoters. Abolishment of DNA methylation led to the context-specific reestablishment of the poised and active states of normal colon cells, which were marked in methylation-deficient cells by distinct H3K27 modifications and the presence of either well-phased nucleosomes or nucleosome-depleted regions, respectively. At higher-order genomic scales, we found that long, H3K9me3-marked domains had lower accessibility, consistent with a more compact chromatin structure. Taken together, our results demonstrate the nuanced and context-dependent role of DNA methylation in the functional, multiscale organization of cancer epigenomes.


Asunto(s)
Cromatina/genética , Neoplasias del Colon/genética , Metilación de ADN/genética , Línea Celular Tumoral , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , ADN (Citosina-5-)-Metiltransferasas/genética , Epigénesis Genética , Células HCT116 , Histonas/genética , Humanos , Nucleosomas/genética , Regiones Promotoras Genéticas/genética , ADN Metiltransferasa 3B
8.
Genome Res ; 24(4): 545-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24515120

RESUMEN

Extracellular cues play critical roles in the establishment of the epigenome during development and may also contribute to epigenetic perturbations found in disease states. The direct role of the local tissue environment on the post-development human epigenome, however, remains unclear due to limitations in studies of human subjects. Here, we use an isogenic human ileal neobladder surgical model and compare global DNA methylation levels of intestinal epithelial cells pre- and post-neobladder construction using the Infinium HumanMethylation450 BeadChip. Our study is the first to quantify the effect of environmental cues on the human epigenome and show that the local tissue environment directly modulates DNA methylation patterns in normal differentiated cells in vivo. In the neobladder, the intestinal epithelial cells lose their tissue-specific epigenetic landscape in a time-dependent manner following the tissue's exposure to a bladder environment. We find that de novo methylation of many intestine-specific enhancers occurs at the rate of 0.41% per month (P < 0.01, Pearson = 0.71), while demethylation of primarily non-intestine-specific transcribed regions occurs at the rate of -0.37% per month (P < 0.01, Pearson = -0.57). The dynamic resetting of the DNA methylome in the neobladder not only implicates local environmental cues in the shaping and maintenance of the epigenome but also illustrates an unexpected cross-talk between the epigenome and the cellular environment.


Asunto(s)
Diferenciación Celular/genética , Metilación de ADN/genética , Epigénesis Genética , Intestinos/crecimiento & desarrollo , Anciano , Islas de CpG , Genoma Humano , Humanos , Intestinos/cirugía , Intestinos/trasplante , Persona de Mediana Edad , Trasplante de Tejidos
9.
Genome Res ; 22(12): 2497-506, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22960375

RESUMEN

DNA methylation and nucleosome positioning work together to generate chromatin structures that regulate gene expression. Nucleosomes are typically mapped using nuclease digestion requiring significant amounts of material and varying enzyme concentrations. We have developed a method (NOMe-seq) that uses a GpC methyltransferase (M.CviPI) and next generation sequencing to generate a high resolution footprint of nucleosome positioning genome-wide using less than 1 million cells while retaining endogenous DNA methylation information from the same DNA strand. Using a novel bioinformatics pipeline, we show a striking anti-correlation between nucleosome occupancy and DNA methylation at CTCF regions that is not present at promoters. We further show that the extent of nucleosome depletion at promoters is directly correlated to expression level and can accommodate multiple nucleosomes and provide genome-wide evidence that expressed non-CpG island promoters are nucleosome-depleted. Importantly, NOMe-seq obtains DNA methylation and nucleosome positioning information from the same DNA molecule, giving the first genome-wide DNA methylation and nucleosome positioning correlation at the single molecule, and thus, single cell level, that can be used to monitor disease progression and response to therapy.


Asunto(s)
Mapeo Cromosómico , Metilación de ADN , ADN/genética , Epigenómica/métodos , Nucleosomas/genética , Alelos , Línea Celular , Ensamble y Desensamble de Cromatina , Islas de CpG , Huella de ADN , Eliminación de Gen , Regulación de la Expresión Génica , Sitios Genéticos , Humanos , Metiltransferasas , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Alineación de Secuencia , Análisis de Secuencia de ADN
10.
Mol Ther ; 17(7): 1155-63, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19367256

RESUMEN

Neonatal gene therapy has the potential to ameliorate abnormalities before disease onset. Our gene knockout of arginase I (AI) deficiency is characterized by increasing hyperammonemia, neurological deterioration, and early death. We constructed a helper-dependent adenoviral vector (HDV) carrying AI and examined for correction of this defect. Neonates were administered 5 x 10(9) viral particles/g and analyzed for survival, arginase activity, and ammonia and amino acids levels. The life expectancy of arg(-/-) mice increased to 27 days while controls died at 14 days with hyperammonemia and in extremis. Death correlated with a decrease in viral DNA/RNA per cell as liver mass increased. Arginase assays demonstrated that vector-injected hepatocytes had ~20% activity of heterozygotes at 2 weeks of age. Hepatic arginine and ornithine in treated mice were similar to those of saline-injected heterozygotes at 2 weeks, whereas ammonia was normal. By 26 days, arginase activity in the treated arg(-/-) livers declined to <10%, and arginine and ornithine increased. Ammonia levels began increasing by day 25, suggesting the cause of death to be similar to that of uninjected arg(-/-) mice, albeit at a later time. These studies demonstrate that the AI deficient newborn mouse can be temporarily corrected and rescued using a HDV.


Asunto(s)
Adenoviridae/genética , Arginasa/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Hiperamonemia/terapia , Hiperargininemia/terapia , Aminoácidos/metabolismo , Amoníaco/metabolismo , Animales , Animales Recién Nacidos , Arginasa/metabolismo , Arginasa/fisiología , Ratones , Ratones Noqueados
11.
Nat Commun ; 10(1): 4154, 2019 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-31515496

RESUMEN

To better understand the impact of chromatin structure on regulation of the prostate cancer transcriptome, we develop high-resolution chromatin interaction maps in normal and prostate cancer cells using in situ Hi-C. By combining the in situ Hi-C data with active and repressive histone marks, CTCF binding sites, nucleosome-depleted regions, and transcriptome profiling, we identify topologically associating domains (TADs) that change in size and epigenetic states between normal and prostate cancer cells. Moreover, we identify normal and prostate cancer-specific enhancer-promoter loops and involved transcription factors. For example, we show that FOXA1 is enriched in prostate cancer-specific enhancer-promoter loop anchors. We also find that the chromatin structure surrounding the androgen receptor (AR) locus is altered in the prostate cancer cells with many cancer-specific enhancer-promoter loops. This creation of 3D epigenomic maps enables a better understanding of prostate cancer biology and mechanisms of gene regulation.


Asunto(s)
Epigenómica , Neoplasias de la Próstata/genética , Transcriptoma/genética , Línea Celular Tumoral , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Sitios Genéticos , Código de Histonas/genética , Humanos , Masculino , Regiones Promotoras Genéticas , Receptores Androgénicos/genética
12.
Methods Mol Biol ; 1708: 267-284, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29224149

RESUMEN

Various methodologies are available to interrogate specific components of epigenetic mechanisms such as DNA methylation or nucleosome occupancy at both the locus-specific and the genome-wide level. It has become increasingly clear, however, that comprehension of the functional interactions between epigenetic mechanisms is critical for understanding how cellular transcription programs are regulated or deregulated during normal and disease development. The Nucleosome Occupancy and Methylome sequencing (NOMe-seq) assay allows us to directly measure the relationship between DNA methylation and nucleosome occupancy by taking advantage of the methyltransferase M.CviPI, which methylates unprotected GpC dinucleotides to create a footprint of chromatin accessibility. This assay generates dual nucleosome occupancy and DNA methylation information at a single-DNA molecule resolution using as little as 200,000 cells and in as short as 15 min reaction time. DNA methylation levels and nucleosome occupancy status of genomic regions of interest can be subsequently interrogated by cloning PCR-amplified bisulfite DNA and sequencing individual clones. Alternatively, NOMe-seq can be combined with next-generation sequencing in order to generate an integrated global map of DNA methylation and nucleosome occupancy, which allows for comprehensive examination as to how these epigenetic components correlate with each other.


Asunto(s)
Metilación de ADN , Nucleosomas/metabolismo , Análisis de Secuencia de ADN/métodos , Islas de CpG , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metiltransferasas/metabolismo , Regiones Promotoras Genéticas
13.
Sci Adv ; 4(12): eaav8550, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30555922

RESUMEN

As part of PsychENCODE, we developed a three-dimensional (3D) epigenomic map of primary cultured neuronal cells derived from olfactory neuroepithelium (CNON). We mapped topologically associating domains and high-resolution chromatin interactions using Hi-C and identified regulatory elements using chromatin immunoprecipitation and nucleosome positioning assays. Using epigenomic datasets from biopsies of 63 living individuals, we found that epigenetic marks at distal regulatory elements are more variable than marks at proximal regulatory elements. By integrating genotype and metadata, we identified enhancers that have different levels corresponding to differences in genetic variation, gender, smoking, and schizophrenia. Motif searches revealed that many CNON enhancers are bound by neuronal-related transcription factors. Last, we combined 3D epigenomic maps and gene expression profiles to predict enhancer-target gene interactions on a genome-wide scale. This study not only provides a framework for understanding individual epigenetic variation using a primary cell model system but also contributes valuable data resources for epigenomic studies of neuronal epithelium.


Asunto(s)
Epigénesis Genética , Epigenómica , Regulación de la Expresión Génica , Neuronas Receptoras Olfatorias/metabolismo , Sitios de Unión , Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Biología Computacional/métodos , Elementos de Facilitación Genéticos , Epigenómica/métodos , Perfilación de la Expresión Génica , Variación Genética , Heterocromatina/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Motivos de Nucleótidos , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/metabolismo , Transcriptoma , Flujo de Trabajo
14.
Cell Rep ; 21(6): 1411-1417, 2017 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-29117547

RESUMEN

Prostate cancer (PCa) is the leading cancer among men in the United States, with genetic factors contributing to ∼42% of the susceptibility to PCa. We analyzed a PCa risk region located at 7p15.2 to gain insight into the mechanisms by which this noncoding region may affect gene regulation and contribute to PCa risk. We performed Hi-C analysis and demonstrated that this region has long-range interactions with the HOXA locus, located ∼873 kb away. Using the CRISPR/Cas9 system, we deleted a 4-kb region encompassing several PCa risk-associated SNPs and performed RNA-seq to investigate transcriptomic changes in prostate cells lacking the regulatory element. Our results suggest that the risk element affects the expression of HOXA13 and HOTTIP, but not other genes in the HOXA locus, via a repressive loop. Forced expression of HOXA13 was performed to gain further insight into the mechanisms by which this risk element affects PCa risk.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Proteínas Co-Represoras , Proteínas de Unión al ADN , Sitios Genéticos , Proteínas de Homeodominio/genética , Humanos , Masculino , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , ARN/química , ARN/aislamiento & purificación , ARN/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Riesgo , Análisis de Secuencia de ARN , Transcriptoma
15.
Sci Rep ; 7: 41947, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176797

RESUMEN

Chromatin is structurally involved in the transcriptional regulation of all genes. While the nucleosome positioning at RNA polymerase II (pol II) promoters has been extensively studied, less is known about the chromatin structure at pol III promoters in human cells. We use a high-resolution analysis to show substantial differences in chromatin structure of pol II and pol III promoters, and between subtypes of pol III genes. Notably, the nucleosome depleted region at the transcription start site of pol III genes extends past the termination sequences, resulting in nucleosome free gene bodies. The +1 nucleosome is located further downstream than at pol II genes and furthermore displays weak positioning. The variable position of the +1 location is seen not only within individual cell populations and between cell types, but also between different pol III promoter subtypes, suggesting that the +1 nucleosome may be involved in the transcriptional regulation of pol III genes. We find that expression and DNA methylation patterns correlate with distinct accessibility patterns, where DNA methylation associates with the silencing and inaccessibility at promoters. Taken together, this study provides the first high-resolution map of nucleosome positioning and occupancy at human pol III promoters at specific loci and genome wide.


Asunto(s)
Ensamble y Desensamble de Cromatina , Nucleosomas/química , Nucleosomas/genética , Regiones Promotoras Genéticas , ARN Polimerasa III/genética , Sitio de Iniciación de la Transcripción , Transcripción Genética , Metilación de ADN , Humanos , Células K562 , ARN Polimerasa II/genética
16.
Gene ; 612: 29-35, 2017 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-27847259

RESUMEN

Changes in ribosome biogenesis are tightly linked to cell growth, proliferation, and differentiation. The rate of ribosome biogenesis is established by RNA Pol I-mediated transcription of ribosomal RNA (rRNA). Thus, rRNA gene transcription is a key determinant of cell behavior. Here, we show that ribosome biogenesis is dynamically regulated during osteoblast differentiation. Upon osteoinduction, osteoprogenitor cells transiently silence a subset of rRNA genes through a reversible mechanism that is initiated through biphasic nucleolar depletion of UBF1 and then RNA Pol I. Nucleolar depletion of UBF1 is coincident with an increase in the number of silent but transcriptionally permissible rRNA genes. This increase in the number of silent rRNA genes reduces levels of ribosome biogenesis and subsequently, protein synthesis. Together these findings demonstrate that fluctuations in rRNA gene transcription are determined by nucleolar occupancy of UBF1 and closely coordinated with the early events necessary for acquisition of the osteoblast cell fate.


Asunto(s)
Diferenciación Celular , Osteoblastos/citología , Ribosomas/metabolismo , Células 3T3 , Animales , Ratones , Biosíntesis de Proteínas , ARN Ribosómico/genética , Transcripción Genética
17.
Cancer Cell ; 26(4): 577-90, 2014 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-25263941

RESUMEN

DNA methylation in promoters is well known to silence genes and is the presumed therapeutic target of methylation inhibitors. Gene body methylation is positively correlated with expression, yet its function is unknown. We show that 5-aza-2'-deoxycytidine treatment not only reactivates genes but decreases the overexpression of genes, many of which are involved in metabolic processes regulated by c-MYC. Downregulation is caused by DNA demethylation of the gene bodies and restoration of high levels of expression requires remethylation by DNMT3B. Gene body methylation may, therefore, be an unexpected therapeutic target for DNA methylation inhibitors, resulting in the normalization of gene overexpression induced during carcinogenesis. Our results provide direct evidence for a causal relationship between gene body methylation and transcription.


Asunto(s)
Metilación de ADN , Expresión Génica , División Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Humanos , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA