Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-38711367

RESUMEN

Hi-C data are commonly normalized using single sample processing methods, with focus on comparisons between regions within a given contact map. Here, we aim to compare contact maps across different samples. We demonstrate that unwanted variation, of likely technical origin, is present in Hi-C data with replicates from different individuals, and that properties of this unwanted variation change across the contact map. We present band-wise normalization and batch correction, a method for normalization and batch correction of Hi-C data and show that it substantially improves comparisons across samples, including in a quantitative trait loci analysis as well as differential enrichment across cell types.


Asunto(s)
Sitios de Carácter Cuantitativo , Humanos , Biología Computacional
2.
PLoS Genet ; 19(6): e1010759, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37289818

RESUMEN

Gene regulation is highly cell type-specific and understanding the function of non-coding genetic variants associated with complex traits requires molecular phenotyping at cell type resolution. In this study we performed single nucleus ATAC-seq (snATAC-seq) and genotyping in peripheral blood mononuclear cells from 13 individuals. Clustering chromatin accessibility profiles of 96,002 total nuclei identified 17 immune cell types and sub-types. We mapped chromatin accessibility QTLs (caQTLs) in each immune cell type and sub-type using individuals of European ancestry which identified 6,901 caQTLs at FDR < .10 and 4,220 caQTLs at FDR < .05, including those obscured from assays of bulk tissue such as with divergent effects on different cell types. For 3,941 caQTLs we further annotated putative target genes of variant activity using single cell co-accessibility, and caQTL variants were significantly correlated with the accessibility level of linked gene promoters. We fine-mapped loci associated with 16 complex immune traits and identified immune cell caQTLs at 622 candidate causal variants, including those with cell type-specific effects. At the 6q15 locus associated with type 1 diabetes, in line with previous reports, variant rs72928038 was a naïve CD4+ T cell caQTL linked to BACH2 and we validated the allelic effects of this variant on regulatory activity in Jurkat T cells. These results highlight the utility of snATAC-seq for mapping genetic effects on accessible chromatin in specific cell types.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina , Cromatina , Humanos , Cromatina/genética , Herencia Multifactorial , Leucocitos Mononucleares , Sitios de Carácter Cuantitativo/genética
3.
Cell ; 186(7): 1493-1511.e40, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-37001506

RESUMEN

Understanding how genetic variants impact molecular phenotypes is a key goal of functional genomics, currently hindered by reliance on a single haploid reference genome. Here, we present the EN-TEx resource of 1,635 open-access datasets from four donors (∼30 tissues × âˆ¼15 assays). The datasets are mapped to matched, diploid genomes with long-read phasing and structural variants, instantiating a catalog of >1 million allele-specific loci. These loci exhibit coordinated activity along haplotypes and are less conserved than corresponding, non-allele-specific ones. Surprisingly, a deep-learning transformer model can predict the allele-specific activity based only on local nucleotide-sequence context, highlighting the importance of transcription-factor-binding motifs particularly sensitive to variants. Furthermore, combining EN-TEx with existing genome annotations reveals strong associations between allele-specific and GWAS loci. It also enables models for transferring known eQTLs to difficult-to-profile tissues (e.g., from skin to heart). Overall, EN-TEx provides rich data and generalizable models for more accurate personal functional genomics.


Asunto(s)
Epigenoma , Sitios de Carácter Cuantitativo , Estudio de Asociación del Genoma Completo , Genómica , Fenotipo , Polimorfismo de Nucleótido Simple
5.
Genomics ; 114(3): 110330, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35278615

RESUMEN

Primary hepatocytes are widely used in the pharmaceutical industry to screen drug candidates for hepatotoxicity, but hepatocytes quickly dedifferentiate and lose their mature metabolic function in culture. Attempts have been made to better recapitulate the in vivo liver environment in culture, but the full spectrum of signals required to maintain hepatocyte function ex vivo remains elusive. To elucidate molecular changes that accompany, and may contribute to dedifferentiation of hepatocytes ex vivo, we performed lineage tracing and comprehensive profiling of alterations in their gene expression profiles and chromatin landscape during culture. First, using genetically tagged hepatocytes we demonstrate that expression of the fetal gene alpha-fetoprotein in cultured hepatocytes comes from cells that previously expressed the mature gene albumin, and not from a population of albumin-negative precursor cells, proving mature hepatocytes undergo true dedifferentiation in culture. Next we studied the dedifferentiation process in detail through bulk RNA-sequencing of hepatocytes cultured over an extended period. We identified three distinct phases of dedifferentiation: an early phase, where mature hepatocyte genes are rapidly downregulated in a matter of hours; a middle phase, where fetal genes are activated; and a late phase, where initially rare contaminating non-parenchymal cells proliferate, taking over the culture. Lastly, to better understand the signaling events that result in the rapid downregulation of mature genes in hepatocytes, we examined changes in chromatin accessibility in these cells during the first 24 h of culture using Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq). We find that drastic and rapid changes in chromatin accessibility occur immediately upon the start of culture. Using binding motif analysis of the areas of open chromatin sharing similar temporal profiles, we identify several candidate transcription factors potentially involved in the dedifferentiation of primary hepatocytes in culture.


Asunto(s)
Hepatocitos , Hígado , Células Cultivadas , Hepatocitos/metabolismo , Albúminas , Cromatina/genética
6.
Hum Genomics ; 15(1): 44, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34256850

RESUMEN

BACKGROUND: Previous research in autism and other neurodevelopmental disorders (NDDs) has indicated an important contribution of protein-coding (coding) de novo variants (DNVs) within specific genes. The role of de novo noncoding variation has been observable as a general increase in genetic burden but has yet to be resolved to individual functional elements. In this study, we assessed whole-genome sequencing data in 2671 families with autism (discovery cohort of 516 families, replication cohort of 2155 families). We focused on DNVs in enhancers with characterized in vivo activity in the brain and identified an excess of DNVs in an enhancer named hs737. RESULTS: We adapted the fitDNM statistical model to work in noncoding regions and tested enhancers for excess of DNVs in families with autism. We found only one enhancer (hs737) with nominal significance in the discovery (p = 0.0172), replication (p = 2.5 × 10-3), and combined dataset (p = 1.1 × 10-4). Each individual with a DNV in hs737 had shared phenotypes including being male, intact cognitive function, and hypotonia or motor delay. Our in vitro assessment of the DNVs showed they all reduce enhancer activity in a neuronal cell line. By epigenomic analyses, we found that hs737 is brain-specific and targets the transcription factor gene EBF3 in human fetal brain. EBF3 is genome-wide significant for coding DNVs in NDDs (missense p = 8.12 × 10-35, loss-of-function p = 2.26 × 10-13) and is widely expressed in the body. Through characterization of promoters bound by EBF3 in neuronal cells, we saw enrichment for binding to NDD genes (p = 7.43 × 10-6, OR = 1.87) involved in gene regulation. Individuals with coding DNVs have greater phenotypic severity (hypotonia, ataxia, and delayed development syndrome [HADDS]) in comparison to individuals with noncoding DNVs that have autism and hypotonia. CONCLUSIONS: In this study, we identify DNVs in the hs737 enhancer in individuals with autism. Through multiple approaches, we find hs737 targets the gene EBF3 that is genome-wide significant in NDDs. By assessment of noncoding variation and the genes they affect, we are beginning to understand their impact on gene regulatory networks in NDDs.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Hipotonía Muscular/genética , Trastornos del Neurodesarrollo/genética , Factores de Transcripción/genética , Trastorno Autístico/epidemiología , Trastorno Autístico/patología , Elementos de Facilitación Genéticos/genética , Exoma/genética , Femenino , Redes Reguladoras de Genes/genética , Humanos , Masculino , Hipotonía Muscular/epidemiología , Hipotonía Muscular/patología , Mutación/genética , Trastornos del Neurodesarrollo/epidemiología , Trastornos del Neurodesarrollo/patología , Neuronas/metabolismo , Neuronas/patología
7.
Nature ; 594(7863): 398-402, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34012112

RESUMEN

Genetic risk variants that have been identified in genome-wide association studies of complex diseases are primarily non-coding1. Translating these risk variants into mechanistic insights requires detailed maps of gene regulation in disease-relevant cell types2. Here we combined two approaches: a genome-wide association study of type 1 diabetes (T1D) using 520,580 samples, and the identification of candidate cis-regulatory elements (cCREs) in pancreas and peripheral blood mononuclear cells using single-nucleus assay for transposase-accessible chromatin with sequencing (snATAC-seq) of 131,554 nuclei. Risk variants for T1D were enriched in cCREs that were active in T cells and other cell types, including acinar and ductal cells of the exocrine pancreas. Risk variants at multiple T1D signals overlapped with exocrine-specific cCREs that were linked to genes with exocrine-specific expression. At the CFTR locus, the T1D risk variant rs7795896 mapped to a ductal-specific cCRE that regulated CFTR; the risk allele reduced transcription factor binding, enhancer activity and CFTR expression in ductal cells. These findings support a role for the exocrine pancreas in the pathogenesis of T1D and highlight the power of large-scale genome-wide association studies and single-cell epigenomics for understanding the cellular origins of complex disease.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Epigenómica , Predisposición Genética a la Enfermedad , Análisis de la Célula Individual , Cromatina/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Femenino , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Inmunidad/genética , Masculino , Conductos Pancreáticos/metabolismo , Conductos Pancreáticos/patología
8.
Nat Genet ; 53(4): 455-466, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33795864

RESUMEN

Single-nucleus assay for transposase-accessible chromatin using sequencing (snATAC-seq) creates new opportunities to dissect cell type-specific mechanisms of complex diseases. Since pancreatic islets are central to type 2 diabetes (T2D), we profiled 15,298 islet cells by using combinatorial barcoding snATAC-seq and identified 12 clusters, including multiple alpha, beta and delta cell states. We cataloged 228,873 accessible chromatin sites and identified transcription factors underlying lineage- and state-specific regulation. We observed state-specific enrichment of fasting glucose and T2D genome-wide association studies for beta cells and enrichment for other endocrine cell types. At T2D signals localized to islet-accessible chromatin, we prioritized variants with predicted regulatory function and co-accessibility with target genes. A causal T2D variant rs231361 at the KCNQ1 locus had predicted effects on a beta cell enhancer co-accessible with INS and genome editing in embryonic stem cell-derived beta cells affected INS levels. Together our findings demonstrate the power of single-cell epigenomics for interpreting complex disease genetics.


Asunto(s)
Cromatina/química , Diabetes Mellitus Tipo 2/genética , Células Secretoras de Glucagón/metabolismo , Células Secretoras de Insulina/metabolismo , Canal de Potasio KCNQ1/genética , Células Secretoras de Polipéptido Pancreático/metabolismo , Células Secretoras de Somatostatina/metabolismo , Glucemia/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Epigenómica , Ayuno , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Células Secretoras de Glucagón/patología , Secuenciación de Nucleótidos de Alto Rendimiento , Células Madre Embrionarias Humanas/citología , Humanos , Células Secretoras de Insulina/patología , Canal de Potasio KCNQ1/metabolismo , Familia de Multigenes , Células Secretoras de Polipéptido Pancreático/patología , Polimorfismo Genético , Análisis de la Célula Individual , Células Secretoras de Somatostatina/patología , Factores de Transcripción/clasificación , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
11.
Nature ; 583(7818): 744-751, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728240

RESUMEN

The Encyclopedia of DNA Elements (ENCODE) project has established a genomic resource for mammalian development, profiling a diverse panel of mouse tissues at 8 developmental stages from 10.5 days after conception until birth, including transcriptomes, methylomes and chromatin states. Here we systematically examined the state and accessibility of chromatin in the developing mouse fetus. In total we performed 1,128 chromatin immunoprecipitation with sequencing (ChIP-seq) assays for histone modifications and 132 assay for transposase-accessible chromatin using sequencing (ATAC-seq) assays for chromatin accessibility across 72 distinct tissue-stages. We used integrative analysis to develop a unified set of chromatin state annotations, infer the identities of dynamic enhancers and key transcriptional regulators, and characterize the relationship between chromatin state and accessibility during developmental gene regulation. We also leveraged these data to link enhancers to putative target genes and demonstrate tissue-specific enrichments of sequence variants associated with disease in humans. The mouse ENCODE data sets provide a compendium of resources for biomedical researchers and achieve, to our knowledge, the most comprehensive view of chromatin dynamics during mammalian fetal development to date.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Desarrollo Fetal/genética , Histonas/metabolismo , Anotación de Secuencia Molecular , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/química , Secuenciación de Inmunoprecipitación de Cromatina , Enfermedad/genética , Elementos de Facilitación Genéticos/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Variación Genética , Histonas/química , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos/genética , Reproducibilidad de los Resultados , Transposasas/metabolismo
12.
Nature ; 583(7818): 752-759, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728242

RESUMEN

Cytosine DNA methylation is essential for mammalian development but understanding of its spatiotemporal distribution in the developing embryo remains limited1,2. Here, as part of the mouse Encyclopedia of DNA Elements (ENCODE) project, we profiled 168 methylomes from 12 mouse tissues or organs at 9 developmental stages from embryogenesis to adulthood. We identified 1,808,810 genomic regions that showed variations in CG methylation by comparing the methylomes of different tissues or organs from different developmental stages. These DNA elements predominantly lose CG methylation during fetal development, whereas the trend is reversed after birth. During late stages of fetal development, non-CG methylation accumulated within the bodies of key developmental transcription factor genes, coinciding with their transcriptional repression. Integration of genome-wide DNA methylation, histone modification and chromatin accessibility data enabled us to predict 461,141 putative developmental tissue-specific enhancers, the human orthologues of which were enriched for disease-associated genetic variants. These spatiotemporal epigenome maps provide a resource for studies of gene regulation during tissue or organ progression, and a starting point for investigating regulatory elements that are involved in human developmental disorders.


Asunto(s)
Metilación de ADN , Epigenoma , Feto/embriología , Feto/metabolismo , Animales , Animales Recién Nacidos , Cromatina/genética , Cromatina/metabolismo , Enfermedad/genética , Regulación hacia Abajo , Elementos de Facilitación Genéticos/genética , Represión Epigenética , Femenino , Silenciador del Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Anotación de Secuencia Molecular , Polimorfismo de Nucleótido Simple , Análisis Espacio-Temporal
13.
Nature ; 583(7818): 699-710, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32728249

RESUMEN

The human and mouse genomes contain instructions that specify RNAs and proteins and govern the timing, magnitude, and cellular context of their production. To better delineate these elements, phase III of the Encyclopedia of DNA Elements (ENCODE) Project has expanded analysis of the cell and tissue repertoires of RNA transcription, chromatin structure and modification, DNA methylation, chromatin looping, and occupancy by transcription factors and RNA-binding proteins. Here we summarize these efforts, which have produced 5,992 new experimental datasets, including systematic determinations across mouse fetal development. All data are available through the ENCODE data portal (https://www.encodeproject.org), including phase II ENCODE1 and Roadmap Epigenomics2 data. We have developed a registry of 926,535 human and 339,815 mouse candidate cis-regulatory elements, covering 7.9 and 3.4% of their respective genomes, by integrating selected datatypes associated with gene regulation, and constructed a web-based server (SCREEN; http://screen.encodeproject.org) to provide flexible, user-defined access to this resource. Collectively, the ENCODE data and registry provide an expansive resource for the scientific community to build a better understanding of the organization and function of the human and mouse genomes.


Asunto(s)
ADN/genética , Bases de Datos Genéticas , Genoma/genética , Genómica , Anotación de Secuencia Molecular , Sistema de Registros , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Cromatina/genética , Cromatina/metabolismo , ADN/química , Huella de ADN , Metilación de ADN/genética , Momento de Replicación del ADN , Desoxirribonucleasa I/metabolismo , Genoma Humano , Histonas/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas de Unión al ARN/genética , Transcripción Genética/genética , Transposasas/metabolismo
14.
Genome Biol ; 20(1): 255, 2019 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-31779666

RESUMEN

BACKGROUND: The 3-dimensional (3D) conformation of chromatin inside the nucleus is integral to a variety of nuclear processes including transcriptional regulation, DNA replication, and DNA damage repair. Aberrations in 3D chromatin conformation have been implicated in developmental abnormalities and cancer. Despite the importance of 3D chromatin conformation to cellular function and human health, little is known about how 3D chromatin conformation varies in the human population, or whether DNA sequence variation between individuals influences 3D chromatin conformation. RESULTS: To address these questions, we perform Hi-C on lymphoblastoid cell lines from 20 individuals. We identify thousands of regions across the genome where 3D chromatin conformation varies between individuals and find that this variation is often accompanied by variation in gene expression, histone modifications, and transcription factor binding. Moreover, we find that DNA sequence variation influences several features of 3D chromatin conformation including loop strength, contact insulation, contact directionality, and density of local cis contacts. We map hundreds of quantitative trait loci associated with 3D chromatin features and find evidence that some of these same variants are associated at modest levels with other molecular phenotypes as well as complex disease risk. CONCLUSION: Our results demonstrate that common DNA sequence variants can influence 3D chromatin conformation, pointing to a more pervasive role for 3D chromatin conformation in human phenotypic variation than previously recognized.


Asunto(s)
Secuencia de Bases , Variación Genética , Genoma Humano , Conformación de Ácido Nucleico , Epigenoma , Humanos , Sitios de Carácter Cuantitativo , Transcriptoma
15.
Nat Commun ; 10(1): 1784, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992455

RESUMEN

The incomplete identification of structural variants (SVs) from whole-genome sequencing data limits studies of human genetic diversity and disease association. Here, we apply a suite of long-read, short-read, strand-specific sequencing technologies, optical mapping, and variant discovery algorithms to comprehensively analyze three trios to define the full spectrum of human genetic variation in a haplotype-resolved manner. We identify 818,054 indel variants (<50 bp) and 27,622 SVs (≥50 bp) per genome. We also discover 156 inversions per genome and 58 of the inversions intersect with the critical regions of recurrent microdeletion and microduplication syndromes. Taken together, our SV callsets represent a three to sevenfold increase in SV detection compared to most standard high-throughput sequencing studies, including those from the 1000 Genomes Project. The methods and the dataset presented serve as a gold standard for the scientific community allowing us to make recommendations for maximizing structural variation sensitivity for future genome sequencing studies.


Asunto(s)
Genoma Humano/genética , Variación Estructural del Genoma , Genómica/métodos , Haplotipos/genética , Algoritmos , Mapeo Cromosómico/métodos , Bases de Datos Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mutación INDEL , Secuenciación Completa del Genoma/métodos
16.
Cell ; 175(5): 1228-1243.e20, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30392959

RESUMEN

Genetic drivers of cancer can be dysregulated through epigenetic modifications of DNA. Although the critical role of DNA 5-methylcytosine (5mC) in the regulation of transcription is recognized, the functions of other non-canonical DNA modifications remain obscure. Here, we report the identification of novel N6-methyladenine (N6-mA) DNA modifications in human tissues and implicate this epigenetic mark in human disease, specifically the highly malignant brain cancer glioblastoma. Glioblastoma markedly upregulated N6-mA levels, which co-localized with heterochromatic histone modifications, predominantly H3K9me3. N6-mA levels were dynamically regulated by the DNA demethylase ALKBH1, depletion of which led to transcriptional silencing of oncogenic pathways through decreasing chromatin accessibility. Targeting the N6-mA regulator ALKBH1 in patient-derived human glioblastoma models inhibited tumor cell proliferation and extended the survival of tumor-bearing mice, supporting this novel DNA modification as a potential therapeutic target for glioblastoma. Collectively, our results uncover a novel epigenetic node in cancer through the DNA modification N6-mA.


Asunto(s)
Adenina/análogos & derivados , Neoplasias Encefálicas/patología , Metilación de ADN , Glioblastoma/patología , Adenina/análisis , Adenina/química , Adulto , Anciano , Histona H2a Dioxigenasa, Homólogo 1 de AlkB/antagonistas & inhibidores , Histona H2a Dioxigenasa, Homólogo 1 de AlkB/genética , Histona H2a Dioxigenasa, Homólogo 1 de AlkB/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidad , Hipoxia de la Célula , Niño , Epigenómica , Femenino , Glioblastoma/metabolismo , Glioblastoma/mortalidad , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Estimación de Kaplan-Meier , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
17.
Nat Neurosci ; 21(7): 1015, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29497140

RESUMEN

In the version of this article initially published online, the accession code was given as GSE1000333. The correct code is GSE100033. The error has been corrected in the print, HTML and PDF versions of the article.

18.
Nat Neurosci ; 21(3): 432-439, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29434377

RESUMEN

Analysis of chromatin accessibility can reveal transcriptional regulatory sequences, but heterogeneity of primary tissues poses a significant challenge in mapping the precise chromatin landscape in specific cell types. Here we report single-nucleus ATAC-seq, a combinatorial barcoding-assisted single-cell assay for transposase-accessible chromatin that is optimized for use on flash-frozen primary tissue samples. We apply this technique to the mouse forebrain through eight developmental stages. Through analysis of more than 15,000 nuclei, we identify 20 distinct cell populations corresponding to major neuronal and non-neuronal cell types. We further define cell-type-specific transcriptional regulatory sequences, infer potential master transcriptional regulators and delineate developmental changes in forebrain cellular composition. Our results provide insight into the molecular and cellular dynamics that underlie forebrain development in the mouse and establish technical and analytical frameworks that are broadly applicable to other heterogeneous tissues.


Asunto(s)
Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Prosencéfalo/crecimiento & desarrollo , Animales , Línea Celular , Proteínas de Unión al ADN , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Neuronas/fisiología , Proteínas Nucleares/metabolismo , Embarazo , Prosencéfalo/citología , Prosencéfalo/metabolismo , Análisis de la Célula Individual
19.
Proc Natl Acad Sci U S A ; 114(9): E1633-E1640, 2017 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-28193886

RESUMEN

Accurate enhancer identification is critical for understanding the spatiotemporal transcriptional regulation during development as well as the functional impact of disease-related noncoding genetic variants. Computational methods have been developed to predict the genomic locations of active enhancers based on histone modifications, but the accuracy and resolution of these methods remain limited. Here, we present an algorithm, regulatory element prediction based on tissue-specific local epigenetic marks (REPTILE), which integrates histone modification and whole-genome cytosine DNA methylation profiles to identify the precise location of enhancers. We tested the ability of REPTILE to identify enhancers previously validated in reporter assays. Compared with existing methods, REPTILE shows consistently superior performance across diverse cell and tissue types, and the enhancer locations are significantly more refined. We show that, by incorporating base-resolution methylation data, REPTILE greatly improves upon current methods for annotation of enhancers across a variety of cell and tissue types. REPTILE is available at https://github.com/yupenghe/REPTILE/.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Epigénesis Genética/genética , Regulación de la Expresión Génica/genética , Algoritmos , Biología Computacional/métodos , Metilación de ADN/genética , Epigenómica/métodos , Genómica/métodos , Código de Histonas/genética , Histonas/genética , Humanos , Transcripción Genética/genética
20.
Nat Commun ; 7: 12923, 2016 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-27703156

RESUMEN

Whole-genome sequencing is identifying growing numbers of non-coding variants in human disease studies, but the lack of accurate functional annotations prevents their interpretation. We describe the genome-wide landscape of distant-acting enhancers active in the developing and adult human heart, an organ whose impairment is a predominant cause of mortality and morbidity. Using integrative analysis of >35 epigenomic data sets from mouse and human pre- and postnatal hearts we created a comprehensive reference of >80,000 putative human heart enhancers. To illustrate the importance of enhancers in the regulation of genes involved in heart disease, we deleted the mouse orthologs of two human enhancers near cardiac myosin genes. In both cases, we observe in vivo expression changes and cardiac phenotypes consistent with human heart disease. Our study provides a comprehensive catalogue of human heart enhancers for use in clinical whole-genome sequencing studies and highlights the importance of enhancers for cardiac function.


Asunto(s)
Elementos de Facilitación Genéticos , Corazón/fisiología , Animales , Ecocardiografía , Epigenómica , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma Humano , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...