Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 166(5): 1269-1281.e19, 2016 Aug 25.
Article in English | MEDLINE | ID: mdl-27565349

ABSTRACT

The glucocorticoid receptor (GR) binds the human genome at >10,000 sites but only regulates the expression of hundreds of genes. To determine the functional effect of each site, we measured the glucocorticoid (GC) responsive activity of nearly all GR binding sites (GBSs) captured using chromatin immunoprecipitation (ChIP) in A549 cells. 13% of GBSs assayed had GC-induced activity. The responsive sites were defined by direct GR binding via a GC response element (GRE) and exclusively increased reporter-gene expression. Meanwhile, most GBSs lacked GC-induced reporter activity. The non-responsive sites had epigenetic features of steady-state enhancers and clustered around direct GBSs. Together, our data support a model in which clusters of GBSs observed with ChIP-seq reflect interactions between direct and tethered GBSs over tens of kilobases. We further show that those interactions can synergistically modulate the activity of direct GBSs and may therefore play a major role in driving gene activation in response to GCs.


Subject(s)
Genome, Human , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Transcriptional Activation , A549 Cells , Binding Sites/drug effects , Chromatin Immunoprecipitation , Dexamethasone/metabolism , Dexamethasone/pharmacology , Genes, Reporter , Glucocorticoids/pharmacology , Humans , Protein Binding/drug effects , Response Elements
2.
Cell ; 151(5): 951-63, 2012 Nov 21.
Article in English | MEDLINE | ID: mdl-23178118

ABSTRACT

The inactive X chromosome's (Xi) physical territory is microscopically devoid of transcriptional hallmarks and enriched in silencing-associated modifications. How these microscopic signatures relate to specific Xi sequences is unknown. Therefore, we profiled Xi gene expression and chromatin states at high resolution via allele-specific sequencing in mouse trophoblast stem cells. Most notably, X-inactivated transcription start sites harbored distinct epigenetic signatures relative to surrounding Xi DNA. These sites displayed H3-lysine27-trimethylation enrichment and DNaseI hypersensitivity, similar to autosomal Polycomb targets, yet excluded Pol II and other transcriptional hallmarks, similar to nontranscribed genes. CTCF bound X-inactivated and escaping genes, irrespective of measured chromatin boundaries. Escape from X inactivation occurred within, and X inactivation was maintained exterior to, the area encompassed by Xist in cells subject to imprinted and random X inactivation. The data support a model whereby inactivation of specific regulatory elements, rather than a simple chromosome-wide separation from transcription machinery, governs gene silencing over the Xi.


Subject(s)
Gene Silencing , Regulatory Elements, Transcriptional , X Chromosome Inactivation , Animals , CCCTC-Binding Factor , Chromatin/metabolism , Deoxyribonuclease I/metabolism , Histone Code , Long Interspersed Nucleotide Elements , Mice , Polycomb-Group Proteins/metabolism , RNA Polymerase II/metabolism , Repressor Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Trophoblasts/cytology
3.
Proc Natl Acad Sci U S A ; 121(11): e2317430121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437540

ABSTRACT

Brown-and-white giant pandas (hereafter brown pandas) are distinct coat color mutants found exclusively in the Qinling Mountains, Shaanxi, China. However, its genetic mechanism has remained unclear since their discovery in 1985. Here, we identified the genetic basis for this coat color variation using a combination of field ecological data, population genomic data, and a CRISPR-Cas9 knockout mouse model. We de novo assembled a long-read-based giant panda genome and resequenced the genomes of 35 giant pandas, including two brown pandas and two family trios associated with a brown panda. We identified a homozygous 25-bp deletion in the first exon of Bace2, a gene encoding amyloid precursor protein cleaving enzyme, as the most likely genetic basis for brown-and-white coat color. This deletion was further validated using PCR and Sanger sequencing of another 192 black giant pandas and CRISPR-Cas9 edited knockout mice. Our investigation revealed that this mutation reduced the number and size of melanosomes of the hairs in knockout mice and possibly in the brown panda, further leading to the hypopigmentation. These findings provide unique insights into the genetic basis of coat color variation in wild animals.


Subject(s)
Ursidae , Animals , Mice , Ursidae/genetics , Peptide Hydrolases , Amyloid beta-Protein Precursor , Animals, Wild , Mice, Knockout
4.
Genome Res ; 32(6): 1183-1198, 2022 06.
Article in English | MEDLINE | ID: mdl-35609992

ABSTRACT

Over a thousand different transcription factors (TFs) bind with varying occupancy across the human genome. Chromatin immunoprecipitation (ChIP) can assay occupancy genome-wide, but only one TF at a time, limiting our ability to comprehensively observe the TF occupancy landscape, let alone quantify how it changes across conditions. We developed TF occupancy profiler (TOP), a Bayesian hierarchical regression framework, to profile genome-wide quantitative occupancy of numerous TFs using data from a single chromatin accessibility experiment (DNase- or ATAC-seq). TOP is supervised, and its hierarchical structure allows it to predict the occupancy of any sequence-specific TF, even those never assayed with ChIP. We used TOP to profile the quantitative occupancy of hundreds of sequence-specific TFs at sites throughout the genome and examined how their occupancies changed in multiple contexts: in approximately 200 human cell types, through 12 h of exposure to different hormones, and across the genetic backgrounds of 70 individuals. TOP enables cost-effective exploration of quantitative changes in the landscape of TF binding.


Subject(s)
Chromatin , Transcription Factors , Bayes Theorem , Binding Sites/genetics , Chromatin/genetics , Genome, Human , Humans , Protein Binding , Transcription Factors/metabolism
5.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35179181

ABSTRACT

The epicardium is a mesothelial tissue layer that envelops the heart. Cardiac injury activates dynamic gene expression programs in epicardial tissue, which in zebrafish enables subsequent regeneration through paracrine and vascularizing effects. To identify tissue regeneration enhancer elements (TREEs) that control injury-induced epicardial gene expression during heart regeneration, we profiled transcriptomes and chromatin accessibility in epicardial cells purified from regenerating zebrafish hearts. We identified hundreds of candidate TREEs, which are defined by increased chromatin accessibility of non-coding elements near genes with increased expression during regeneration. Several of these candidate TREEs were incorporated into stable transgenic lines, with five out of six elements directing injury-induced epicardial expression but not ontogenetic epicardial expression in larval hearts. Whereas two independent TREEs linked to the gene gnai3 showed similar functional features of gene regulation in transgenic lines, two independent ncam1a-linked TREEs directed distinct spatiotemporal domains of epicardial gene expression. Thus, multiple TREEs linked to a regeneration gene can possess either matching or complementary regulatory controls. Our study provides a new resource and principles for understanding the regulation of epicardial genetic programs during heart regeneration. This article has an associated 'The people behind the papers' interview.


Subject(s)
Enhancer Elements, Genetic/genetics , Heart/physiology , Pericardium/metabolism , Regeneration/physiology , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Chromatin/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Gene Expression Regulation , Larva/growth & development , Larva/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/genetics , Neural Cell Adhesion Molecules/metabolism , Pericardium/cytology , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
6.
Development ; 147(14)2020 07 30.
Article in English | MEDLINE | ID: mdl-32665240

ABSTRACT

To identify candidate tissue regeneration enhancer elements (TREEs) important for zebrafish fin regeneration, we performed ATAC-seq from bulk tissue or purified fibroblasts of uninjured and regenerating caudal fins. We identified tens of thousands of DNA regions from each sample type with dynamic accessibility during regeneration, and assigned these regions to proximal genes with corresponding expression changes by RNA-seq. To determine whether these profiles reveal bona fide TREEs, we tested the sufficiency and requirements of several sequences in stable transgenic lines and mutant lines with homozygous deletions. These experiments validated new non-coding regulatory sequences near induced and/or essential genes during fin regeneration, including fgf20a, mdka and cx43, identifying distinct domains of directed expression for each confirmed TREE. Whereas deletion of the previously identified LEN enhancer abolished detectable induction of the nearby leptin b gene during regeneration, deletions of enhancers linked to fgf20a, mdka and cx43 had no effect or partially reduced gene expression. Our study generates a new resource for dissecting the regulatory mechanisms of appendage generation and reveals a range of requirements for individual TREEs in control of regeneration programs.


Subject(s)
Animal Fins/metabolism , Enhancer Elements, Genetic/genetics , Regeneration/physiology , Zebrafish/metabolism , Animal Fins/physiology , Animals , Animals, Genetically Modified/metabolism , Chromatin/metabolism , Chromatin Assembly and Disassembly , Connexin 43/genetics , Connexin 43/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression , Leptin/genetics , Leptin/metabolism , Midkine/genetics , Midkine/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
7.
BMC Infect Dis ; 23(1): 300, 2023 May 08.
Article in English | MEDLINE | ID: mdl-37158831

ABSTRACT

BACKGROUND: Standard treatment for drug-susceptible tuberculosis (DS-TB) includes a multidrug regimen requiring at least 6 months of treatment, and this lengthy treatment easily leads to poor adherence. There is an urgent need to simplify and shorten treatment regimens to reduce interruption and adverse event rates, improve compliance, and reduce costs. METHODS: ORIENT is a multicenter, randomized controlled, open-label, phase II/III, non-inferiority trial involving DS-TB patients to evaluate the safety and efficacy of short-term regimens compared with the standardized six-month treatment regimen. In stage 1, corresponding to a phase II trial, a total of 400 patients are randomly divided into four arms, stratified by site and the presence of lung cavitation. Investigational arms include 3 short-term regimens with rifapentine 10 mg/kg, 15 mg/kg, and 20 mg/kg, while the control arm uses the standardized six-month treatment regimen. A combination of rifapentine, isoniazid, pyrazinamide, and moxifloxacin is administered for 17 or 26 weeks in rifapentine arms, while a 26-week regimen containing rifampicin, isoniazid, pyrazinamide, and ethambutol is applied in the control arm. After the safety and preliminary effectiveness analysis of patients in stage 1, the control arm and the investigational arm meeting the conditions will enter into stage 2, which is equivalent to a phase III trial and will be expanded to recruit DS-TB patients. If all investigational arms do not meet the safety conditions, stage 2 will be canceled. In stage 1, the primary safety endpoint is permanent regimen discontinuation at 8 weeks after the first dose. The primary efficacy endpoint is the proportion of favorable outcomes at 78 weeks after the first dose for both two stages. DISCUSSION: This trial will contribute to the optimal dose of rifapentine in the Chinese population and suggest the feasibility of the short-course treatment regimen containing high-dose rifapentine and moxifloxacin for DS-TB. TRIAL REGISTRATION: The trial has been registered on ClinicalTrials.gov on 28 May 2022 with the identifier NCT05401071.


Subject(s)
Rifampin , Tuberculosis , Humans , Rifampin/adverse effects , Isoniazid/adverse effects , Pyrazinamide , Moxifloxacin/therapeutic use , Tuberculosis/drug therapy , Randomized Controlled Trials as Topic , Multicenter Studies as Topic , Clinical Trials, Phase II as Topic
8.
Genome Res ; 28(10): 1577-1588, 2018 10.
Article in English | MEDLINE | ID: mdl-30139769

ABSTRACT

Cis-regulatory elements (CRE), short DNA sequences through which transcription factors (TFs) exert regulatory control on gene expression, are postulated to be the major sites of causal sequence variation underlying the genetics of complex traits and diseases. We present integrative analyses, combining high-throughput genomic and epigenomic data with sequence-based computations, to identify the causal transcriptional components in a given tissue. We use data on adult human hearts to demonstrate that (1) sequence-based predictions detect numerous, active, tissue-specific CREs missed by experimental observations, (2) learned sequence features identify the cognate TFs, (3) CRE variants are specifically associated with cardiac gene expression, and (4) a significant fraction of the heritability of exemplar cardiac traits (QT interval, blood pressure, pulse rate) is attributable to these variants. This general systems approach can thus identify candidate causal variants and the components of gene regulatory networks (GRN) to enable understanding of the mechanisms of complex disorders on a tissue- or cell-type basis.


Subject(s)
Myocardium/metabolism , Regulatory Elements, Transcriptional , Sequence Analysis, DNA/methods , Transcription Factors/genetics , Adult , Epigenomics , Gene Expression , Genetic Variation , High-Throughput Nucleotide Sequencing , Humans , Organ Specificity
9.
Genome Res ; 28(9): 1272-1284, 2018 09.
Article in English | MEDLINE | ID: mdl-30097539

ABSTRACT

Glucocorticoids are potent steroid hormones that regulate immunity and metabolism by activating the transcription factor (TF) activity of glucocorticoid receptor (GR). Previous models have proposed that DNA binding motifs and sites of chromatin accessibility predetermine GR binding and activity. However, there are vast excesses of both features relative to the number of GR binding sites. Thus, these features alone are unlikely to account for the specificity of GR binding and activity. To identify genomic and epigenetic contributions to GR binding specificity and the downstream changes resultant from GR binding, we performed hundreds of genome-wide measurements of TF binding, epigenetic state, and gene expression across a 12-h time course of glucocorticoid exposure. We found that glucocorticoid treatment induces GR to bind to nearly all pre-established enhancers within minutes. However, GR binds to only a small fraction of the set of accessible sites that lack enhancer marks. Once GR is bound to enhancers, a combination of enhancer motif composition and interactions between enhancers then determines the strength and persistence of GR binding, which consequently correlates with dramatic shifts in enhancer activation. Over the course of several hours, highly coordinated changes in TF binding and histone modification occupancy occur specifically within enhancers, and these changes correlate with changes in the expression of nearby genes. Following GR binding, changes in the binding of other TFs precede changes in chromatin accessibility, suggesting that other TFs are also sensitive to genomic features beyond that of accessibility.


Subject(s)
Enhancer Elements, Genetic , Histone Code , Nucleotide Motifs , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Cell Line, Tumor , Epigenesis, Genetic , Humans , Protein Binding , Transcription Factors/metabolism
10.
Genome Res ; 27(7): 1195-1206, 2017 07.
Article in English | MEDLINE | ID: mdl-28385711

ABSTRACT

Microbiota influence diverse aspects of intestinal physiology and disease in part by controlling tissue-specific transcription of host genes. However, host genomic mechanisms mediating microbial control of intestinal gene expression are poorly understood. Hepatocyte nuclear factor 4 (HNF4) is the most ancient family of nuclear receptor transcription factors with important roles in human metabolic and inflammatory bowel diseases, but a role in host response to microbes is unknown. Using an unbiased screening strategy, we found that zebrafish Hnf4a specifically binds and activates a microbiota-suppressed intestinal epithelial transcriptional enhancer. Genetic analysis revealed that zebrafish hnf4a activates nearly half of the genes that are suppressed by microbiota, suggesting microbiota negatively regulate Hnf4a. In support, analysis of genomic architecture in mouse intestinal epithelial cells disclosed that microbiota colonization leads to activation or inactivation of hundreds of enhancers along with drastic genome-wide reduction of HNF4A and HNF4G occupancy. Interspecies meta-analysis suggested interactions between HNF4A and microbiota promote gene expression patterns associated with human inflammatory bowel diseases. These results indicate a critical and conserved role for HNF4A in maintaining intestinal homeostasis in response to microbiota.


Subject(s)
Gastrointestinal Microbiome , Gene Expression Regulation , Hepatocyte Nuclear Factor 4/biosynthesis , Inflammatory Bowel Diseases , Zebrafish Proteins/biosynthesis , Zebrafish , Animals , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice , Species Specificity , Zebrafish/metabolism , Zebrafish/microbiology
11.
Development ; 144(4): 720-730, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087634

ABSTRACT

A current goal of molecular biology is to identify transcriptional networks that regulate cell differentiation. However, identifying functional gene regulatory elements has been challenging in the context of developing tissues where material is limited and cell types are mixed. To identify regulatory sites during sex determination, we subjected Sertoli cells from mouse fetal testes to DNaseI-seq and ChIP-seq for H3K27ac. DNaseI-seq identified putative regulatory sites around genes enriched in Sertoli and pregranulosa cells; however, active enhancers marked by H3K27ac were enriched proximal to only Sertoli-enriched genes. Sequence analysis identified putative binding sites of known and novel transcription factors likely controlling Sertoli cell differentiation. As a validation of this approach, we identified a novel Sertoli cell enhancer upstream of Wt1, and used it to drive expression of a transgenic reporter in Sertoli cells. This work furthers our understanding of the complex genetic network that underlies sex determination and identifies regions that potentially harbor non-coding mutations underlying disorders of sexual development.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Regulatory Elements, Transcriptional , Sertoli Cells/metabolism , Animals , Binding Sites , Cell Differentiation , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic , Genes, Reporter , Genome , Histones/metabolism , Homozygote , Male , Mice , Mutation , Promoter Regions, Genetic , Sex Determination Processes , Testis/embryology , Transgenes
12.
Nucleic Acids Res ; 45(20): 11684-11699, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-28977539

ABSTRACT

Our current understanding of cellular transdifferentiation systems is limited. It is oftentimes unknown, at a genome-wide scale, how much transdifferentiated cells differ quantitatively from both the starting cells and the target cells. Focusing on transdifferentiation of primary human skin fibroblasts by forced expression of myogenic transcription factor MyoD, we performed quantitative analyses of gene expression and chromatin accessibility profiles of transdifferentiated cells compared to fibroblasts and myoblasts. In this system, we find that while many of the early muscle marker genes are reprogrammed, global gene expression and accessibility changes are still incomplete when compared to myoblasts. In addition, we find evidence of epigenetic memory in the transdifferentiated cells, with reminiscent features of fibroblasts being visible both in chromatin accessibility and gene expression. Quantitative analyses revealed a continuum of changes in chromatin accessibility induced by MyoD, and a strong correlation between chromatin-remodeling deficiencies and incomplete gene expression reprogramming. Classification analyses identified genetic and epigenetic features that distinguish reprogrammed from non-reprogrammed sites, and suggested ways to potentially improve transdifferentiation efficiency. Our approach for combining gene expression, DNA accessibility, and protein-DNA binding data to quantify and characterize the efficiency of cellular transdifferentiation on a genome-wide scale can be applied to any transdifferentiation system.


Subject(s)
Cell Transdifferentiation/genetics , Cellular Reprogramming/genetics , Chromatin Assembly and Disassembly/genetics , MyoD Protein/genetics , Blotting, Western , Cells, Cultured , Chromatin/genetics , Chromatin/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling/methods , Gene Ontology , HEK293 Cells , Humans , Microscopy, Confocal , MyoD Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/cytology
13.
Genome Res ; 25(8): 1158-69, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26025803

ABSTRACT

Genome engineering technologies based on the CRISPR/Cas9 and TALE systems are enabling new approaches in science and biotechnology. However, the specificity of these tools in complex genomes and the role of chromatin structure in determining DNA binding are not well understood. We analyzed the genome-wide effects of TALE- and CRISPR-based transcriptional activators in human cells using ChIP-seq to assess DNA-binding specificity and RNA-seq to measure the specificity of perturbing the transcriptome. Additionally, DNase-seq was used to assess genome-wide chromatin remodeling that occurs as a result of their action. Our results show that these transcription factors are highly specific in both DNA binding and gene regulation and are able to open targeted regions of closed chromatin independent of gene activation. Collectively, these results underscore the potential for these technologies to make precise changes to gene expression for gene and cell therapies or fundamental studies of gene function.


Subject(s)
CRISPR-Cas Systems , Chromatin/chemistry , DNA-Binding Proteins/metabolism , DNA/metabolism , Transcription Factors/metabolism , Binding Sites , Chromatin Assembly and Disassembly , DNA/chemistry , DNA-Binding Proteins/chemistry , Gene Expression Regulation , Genetic Engineering/methods , Genome, Human , HEK293 Cells , Humans , Sequence Analysis, DNA , Sequence Analysis, RNA , Transcription Factors/chemistry
14.
Nat Methods ; 12(12): 1143-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26501517

ABSTRACT

Epigenome editing with the CRISPR (clustered, regularly interspaced, short palindromic repeats)-Cas9 platform is a promising technology for modulating gene expression to direct cell phenotype and to dissect the causal epigenetic mechanisms of gene regulation. Fusions of nuclease-inactive dCas9 to the Krüppel-associated box (KRAB) repressor (dCas9-KRAB) can silence target gene expression, but the genome-wide specificity and the extent of heterochromatin formation catalyzed by dCas9-KRAB are not known. We targeted dCas9-KRAB to the HS2 enhancer, a distal regulatory element that orchestrates the expression of multiple globin genes, and observed highly specific induction of H3K9 trimethylation (H3K9me3) at the enhancer and decreased chromatin accessibility of both the enhancer and its promoter targets. Targeted epigenetic modification of HS2 silenced the expression of multiple globin genes, with minimal off-target changes in global gene expression. These results demonstrate that repression mediated by dCas9-KRAB is sufficiently specific to disrupt the activity of individual enhancers via local modification of the epigenome.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Epigenesis, Genetic , Epigenomics/methods , Regulatory Elements, Transcriptional/genetics , Enhancer Elements, Genetic , Gene Expression Regulation, Viral , Globins/genetics , HEK293 Cells , Humans , K562 Cells , Lentivirus/genetics , RNA, Guide, Kinetoplastida/genetics , Repressor Proteins/genetics , Viral Proteins/genetics
15.
Nature ; 489(7414): 75-82, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22955617

ABSTRACT

DNase I hypersensitive sites (DHSs) are markers of regulatory DNA and have underpinned the discovery of all classes of cis-regulatory elements including enhancers, promoters, insulators, silencers and locus control regions. Here we present the first extensive map of human DHSs identified through genome-wide profiling in 125 diverse cell and tissue types. We identify ∼2.9 million DHSs that encompass virtually all known experimentally validated cis-regulatory sequences and expose a vast trove of novel elements, most with highly cell-selective regulation. Annotating these elements using ENCODE data reveals novel relationships between chromatin accessibility, transcription, DNA methylation and regulatory factor occupancy patterns. We connect ∼580,000 distal DHSs with their target promoters, revealing systematic pairing of different classes of distal DHSs and specific promoter types. Patterning of chromatin accessibility at many regulatory regions is organized with dozens to hundreds of co-activated elements, and the transcellular DNase I sensitivity pattern at a given region can predict cell-type-specific functional behaviours. The DHS landscape shows signatures of recent functional evolutionary constraint. However, the DHS compartment in pluripotent and immortalized cells exhibits higher mutation rates than that in highly differentiated cells, exposing an unexpected link between chromatin accessibility, proliferative potential and patterns of human variation.


Subject(s)
Chromatin/genetics , Chromatin/metabolism , DNA/genetics , Encyclopedias as Topic , Genome, Human/genetics , Molecular Sequence Annotation , Regulatory Sequences, Nucleic Acid/genetics , DNA Footprinting , DNA Methylation , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/metabolism , Evolution, Molecular , Genomics , Humans , Mutation Rate , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Initiation Site , Transcription, Genetic
16.
PLoS Genet ; 11(1): e1004885, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25569532

ABSTRACT

Cellular stresses activate the tumor suppressor p53 protein leading to selective binding to DNA response elements (REs) and gene transactivation from a large pool of potential p53 REs (p53REs). To elucidate how p53RE sequences and local chromatin context interact to affect p53 binding and gene transactivation, we mapped genome-wide binding localizations of p53 and H3K4me3 in untreated and doxorubicin (DXR)-treated human lymphoblastoid cells. We examined the relationships among p53 occupancy, gene expression, H3K4me3, chromatin accessibility (DNase 1 hypersensitivity, DHS), ENCODE chromatin states, p53RE sequence, and evolutionary conservation. We observed that the inducible expression of p53-regulated genes was associated with the steady-state chromatin status of the cell. Most highly inducible p53-regulated genes were suppressed at baseline and marked by repressive histone modifications or displayed CTCF binding. Comparison of p53RE sequences residing in different chromatin contexts demonstrated that weaker p53REs resided in open promoters, while stronger p53REs were located within enhancers and repressed chromatin. p53 occupancy was strongly correlated with similarity of the target DNA sequences to the p53RE consensus, but surprisingly, inversely correlated with pre-existing nucleosome accessibility (DHS) and evolutionary conservation at the p53RE. Occupancy by p53 of REs that overlapped transposable element (TE) repeats was significantly higher (p<10-7) and correlated with stronger p53RE sequences (p<10-110) relative to nonTE-associated p53REs, particularly for MLT1H, LTR10B, and Mer61 TEs. However, binding at these elements was generally not associated with transactivation of adjacent genes. Occupied p53REs located in L2-like TEs were unique in displaying highly negative PhyloP scores (predicted fast-evolving) and being associated with altered H3K4me3 and DHS levels. These results underscore the systematic interaction between chromatin status and p53RE context in the induced transactivation response. This p53 regulated response appears to have been tuned via evolutionary processes that may have led to repression and/or utilization of p53REs originating from primate-specific transposon elements.


Subject(s)
Chromatin/genetics , Response Elements/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Animals , Binding Sites , Chromatin/drug effects , Chromosome Structures/drug effects , Chromosome Structures/genetics , DNA Transposable Elements , Doxorubicin/administration & dosage , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase , Humans , Nucleosomes/genetics , Promoter Regions, Genetic , Protein Binding , Tumor Suppressor Protein p53/metabolism
17.
Hum Mol Genet ; 24(19): 5433-50, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26206884

ABSTRACT

SOX10 is required for melanocyte development and maintenance, and has been linked to melanoma initiation and progression. However, the molecular mechanisms by which SOX10 guides the appropriate gene expression programs necessary to promote the melanocyte lineage are not fully understood. Here we employ genetic and epigenomic analysis approaches to uncover novel genomic targets and previously unappreciated molecular roles of SOX10 in melanocytes. Through global analysis of SOX10-binding sites and epigenetic characteristics of chromatin states, we uncover an extensive catalog of SOX10 targets genome-wide. Our findings reveal that SOX10 predominantly engages 'open' chromatin regions and binds to distal regulatory elements, including novel and previously known melanocyte enhancers. Integrated chromatin occupancy and transcriptome analysis suggest a role for SOX10 in both transcriptional activation and repression to regulate functionally distinct classes of genes. We demonstrate that distinct epigenetic signatures and cis-regulatory sequence motifs predicted to bind putative co-regulatory transcription factors define SOX10-activated and SOX10-repressed target genes. Collectively, these findings uncover a central role of SOX10 as a global regulator of gene expression in the melanocyte lineage by targeting diverse regulatory pathways.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Melanocytes/metabolism , Oligonucleotide Array Sequence Analysis/methods , SOXE Transcription Factors/metabolism , Animals , Binding Sites , Cell Line , Chromatin/genetics , Chromatin/metabolism , Epigenomics/methods , Melanocytes/cytology , Mice , SOXE Transcription Factors/chemistry , SOXE Transcription Factors/genetics
18.
Hum Mol Genet ; 24(22): 6552-63, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26307087

ABSTRACT

Exfoliation syndrome (XFS) is a common, age-related, systemic fibrillinopathy. It greatly increases risk of exfoliation glaucoma (XFG), a major worldwide cause of irreversible blindness. Coding variants in the lysyl oxidase-like 1 (LOXL1) gene are strongly associated with XFS in all studied populations, but a functional role for these variants has not been established. To identify additional candidate functional variants, we sequenced the entire LOXL1 genomic locus (∼40 kb) in 50 indigenous, black South African XFS cases and 50 matched controls. The variants with the strongest evidence of association were located in a well-defined 7-kb region bounded by the 3'-end of exon 1 and the adjacent region of intron 1 of LOXL1. We replicated this finding in US Caucasian (91 cases/1031 controls), German (771 cases/1365 controls) and Japanese (1484 cases/1188 controls) populations. The region of peak association lies upstream of LOXL1-AS1, a long non-coding RNA (lncRNA) encoded on the opposite strand of LOXL1. We show that this region contains a promoter and, importantly, that the strongly associated XFS risk alleles in the South African population are functional variants that significantly modulate the activity of this promoter. LOXL1-AS1 expression is also significantly altered in response to oxidative stress in human lens epithelial cells and in response to cyclic mechanical stress in human Schlemm's canal endothelial cells. Taken together, these findings support a functional role for the LOXL1-AS1 lncRNA in cellular stress response and suggest that dysregulation of its expression by genetic risk variants plays a key role in XFS pathogenesis.


Subject(s)
Amino Acid Oxidoreductases/genetics , Exfoliation Syndrome/genetics , RNA, Long Noncoding/genetics , Aged , Alleles , Case-Control Studies , Exfoliation Syndrome/metabolism , Female , Gene Expression , Gene Frequency , Genetic Predisposition to Disease , Humans , Male , Oxidative Stress/genetics , Polymorphism, Single Nucleotide , Promoter Regions, Genetic
19.
BMC Genomics ; 17(1): 887, 2016 11 07.
Article in English | MEDLINE | ID: mdl-27821050

ABSTRACT

BACKGROUND: The transcription factor SOX10 is essential for all stages of Schwann cell development including myelination. SOX10 cooperates with other transcription factors to activate the expression of key myelin genes in Schwann cells and is therefore a context-dependent, pro-myelination transcription factor. As such, the identification of genes regulated by SOX10 will provide insight into Schwann cell biology and related diseases. While genome-wide studies have successfully revealed SOX10 target genes, these efforts mainly focused on myelinating stages of Schwann cell development. We propose that less-biased approaches will reveal novel functions of SOX10 outside of myelination. RESULTS: We developed a stringent, computational-based screen for genome-wide identification of SOX10 response elements. Experimental validation of a pilot set of predicted binding sites in multiple systems revealed that SOX10 directly regulates a previously unreported alternative promoter at SOX6, which encodes a transcription factor that inhibits glial cell differentiation. We further explored the utility of our computational approach by combining it with DNase-seq analysis in cultured Schwann cells and previously published SOX10 ChIP-seq data from rat sciatic nerve. Remarkably, this analysis enriched for genomic segments that map to loci involved in the negative regulation of gliogenesis including SOX5, SOX6, NOTCH1, HMGA2, HES1, MYCN, ID4, and ID2. Functional studies in Schwann cells revealed that: (1) all eight loci are expressed prior to myelination and down-regulated subsequent to myelination; (2) seven of the eight loci harbor validated SOX10 binding sites; and (3) seven of the eight loci are down-regulated upon repressing SOX10 function. CONCLUSIONS: Our computational strategy revealed a putative novel function for SOX10 in Schwann cells, which suggests a model where SOX10 activates the expression of genes that inhibit myelination during non-myelinating stages of Schwann cell development. Importantly, the computational and functional datasets we present here will be valuable for the study of transcriptional regulation, SOX protein function, and glial cell biology.


Subject(s)
Cell Differentiation , Neuroglia/cytology , Neuroglia/metabolism , SOXE Transcription Factors/metabolism , Base Sequence , Cell Differentiation/genetics , Consensus Sequence , Conserved Sequence , Exons , Genomics/methods , High-Throughput Nucleotide Sequencing , Promoter Regions, Genetic , Regulatory Elements, Transcriptional , Response Elements , SOXE Transcription Factors/chemistry , SOXE Transcription Factors/genetics , Schwann Cells/metabolism
20.
Genome Res ; 23(7): 1118-29, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23657885

ABSTRACT

DNase-seq is primarily used to identify nucleosome-depleted DNase I hypersensitive (DHS) sites genome-wide that correspond to active regulatory elements. However, ≈ 40 yr ago it was demonstrated that DNase I also digests with a ≈ 10-bp periodicity around nucleosomes matching the exposure of the DNA minor groove as it wraps around histones. Here, we use DNase-seq data from 49 samples representing diverse cell types to reveal this digestion pattern at individual loci and predict genomic locations where nucleosome rotational positioning, the orientation of DNA with respect to the histone surface, is stably maintained. We call these regions DNase I annotated regions of nucleosome stability (DARNS). Compared to MNase-seq experiments, we show DARNS correspond well to annotated nucleosomes. Interestingly, many DARNS are positioned over only one side of annotated nucleosomes, suggesting that the periodic digestion pattern attenuates over the nucleosome dyad. DARNS reproduce the arrangement of nucleosomes around transcription start sites and are depleted at ubiquitous DHS sites. We also generated DARNS from multiple lymphoblast cell line (LCL) samples. We found that LCL DARNS were enriched at DHS sites present in most of the original 49 samples but absent in LCLs, while multi-cell-type DARNS were enriched at LCL-specific DHS sites. This indicates that variably open DHS sites are often occupied by rotationally stable nucleosomes in cell types where the DHS site is closed. DARNS provide additional information about precise DNA orientation within individual nucleosomes not available from other nucleosome positioning assays and contribute to understanding the role of chromatin in gene regulation.


Subject(s)
DNA/genetics , DNA/metabolism , Deoxyribonucleases/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , Binding Sites , Cell Line , Deoxyribonuclease I/metabolism , Genetic Loci , Genomics , Humans , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL