Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38766054

ABSTRACT

Identifying the causal variants and mechanisms that drive complex traits and diseases remains a core problem in human genetics. The majority of these variants have individually weak effects and lie in non-coding gene-regulatory elements where we lack a complete understanding of how single nucleotide alterations modulate transcriptional processes to affect human phenotypes. To address this, we measured the activity of 221,412 trait-associated variants that had been statistically fine-mapped using a Massively Parallel Reporter Assay (MPRA) in 5 diverse cell-types. We show that MPRA is able to discriminate between likely causal variants and controls, identifying 12,025 regulatory variants with high precision. Although the effects of these variants largely agree with orthogonal measures of function, only 69% can plausibly be explained by the disruption of a known transcription factor (TF) binding motif. We dissect the mechanisms of 136 variants using saturation mutagenesis and assign impacted TFs for 91% of variants without a clear canonical mechanism. Finally, we provide evidence that epistasis is prevalent for variants in close proximity and identify multiple functional variants on the same haplotype at a small, but important, subset of trait-associated loci. Overall, our study provides a systematic functional characterization of likely causal common variants underlying complex and molecular human traits, enabling new insights into the regulatory grammar underlying disease risk.

2.
Blood ; 139(16): 2534-2546, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35030251

ABSTRACT

Master regulators, such as the hematopoietic transcription factor (TF) GATA1, play an essential role in orchestrating lineage commitment and differentiation. However, the precise mechanisms by which such TFs regulate transcription through interactions with specific cis-regulatory elements remain incompletely understood. Here, we describe a form of congenital hemolytic anemia caused by missense mutations in an intrinsically disordered region of GATA1, with a poorly understood role in transcriptional regulation. Through integrative functional approaches, we demonstrate that these mutations perturb GATA1 transcriptional activity by partially impairing nuclear localization and selectively altering precise chromatin occupancy by GATA1. These alterations in chromatin occupancy and concordant chromatin accessibility changes alter faithful gene expression, with failure to both effectively silence and activate select genes necessary for effective terminal red cell production. We demonstrate how disease-causing mutations can reveal regulatory mechanisms that enable the faithful genomic targeting of master TFs during cellular differentiation.


Subject(s)
Anemia , GATA1 Transcription Factor , Cell Differentiation/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Erythropoiesis/genetics , GATA1 Transcription Factor/genetics , GATA1 Transcription Factor/metabolism , Humans
3.
Elife ; 72018 12 21.
Article in English | MEDLINE | ID: mdl-30575519

ABSTRACT

Changes in DNA methylation are involved in development, disease, and the response to environmental conditions. However, not all regulatory elements are functionally methylation-dependent (MD). Here, we report a method, mSTARR-seq, that assesses the causal effects of DNA methylation on regulatory activity at hundreds of thousands of fragments (millions of CpG sites) simultaneously. Using mSTARR-seq, we identify thousands of MD regulatory elements in the human genome. MD activity is partially predictable using sequence and chromatin state information, and distinct transcription factors are associated with higher activity in unmethylated versus methylated DNA. Further, pioneer TFs linked to higher activity in the methylated state appear to drive demethylation of experimentally methylated sites. MD regulatory elements also predict methylation-gene expression relationships across individuals, where they are 1.6x enriched among sites with strong negative correlations. mSTARR-seq thus provides a map of MD regulatory activity in the human genome and facilitates interpretation of differential methylation studies.


Subject(s)
Chromatin/metabolism , DNA Methylation , Epigenesis, Genetic , Genome, Human , Sequence Analysis, DNA/statistics & numerical data , Transcription Factors/genetics , Chromatin/chemistry , Computational Biology/methods , CpG Islands , High-Throughput Nucleotide Sequencing , Humans , K562 Cells , Regulatory Sequences, Nucleic Acid , Sequence Analysis, DNA/methods , Transcription Factors/metabolism
4.
Nat Commun ; 9(1): 5317, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30575722

ABSTRACT

Environmental stimuli commonly act via changes in gene regulation. Human-genome-scale assays to measure such responses are indirect or require knowledge of the transcription factors (TFs) involved. Here, we present the use of human genome-wide high-throughput reporter assays to measure environmentally-responsive regulatory element activity. We focus on responses to glucocorticoids (GCs), an important class of pharmaceuticals and a paradigmatic genomic response model. We assay GC-responsive regulatory activity across >108 unique DNA fragments, covering the human genome at >50×. Those assays directly detected thousands of GC-responsive regulatory elements genome-wide. We then validate those findings with measurements of transcription factor occupancy, histone modifications, chromatin accessibility, and gene expression. We also detect allele-specific environmental responses. Notably, the assays did not require knowledge of GC response mechanisms. Thus, this technology can be used to agnostically quantify genomic responses for which the underlying mechanism remains unknown.


Subject(s)
Gene Expression Regulation/drug effects , Genome, Human , Glucocorticoids/pharmacology , Regulatory Elements, Transcriptional/drug effects , Gene-Environment Interaction , High-Throughput Screening Assays , Humans
5.
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
6.
Cell Syst ; 7(2): 146-160.e7, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30031775

ABSTRACT

The glucocorticoid receptor (GR) is a hormone-inducible transcription factor involved in metabolic and anti-inflammatory gene expression responses. To investigate what controls interactions between GR binding sites and their target genes, we used in situ Hi-C to generate high-resolution, genome-wide maps of chromatin interactions before and after glucocorticoid treatment. We found that GR binding to the genome typically does not cause new chromatin interactions to target genes but instead acts through chromatin interactions that already exist prior to hormone treatment. Both glucocorticoid-induced and glucocorticoid-repressed genes increased interactions with distal GR binding sites. In addition, while glucocorticoid-induced genes increased interactions with transcriptionally active chromosome compartments, glucocorticoid-repressed genes increased interactions with transcriptionally silent compartments. Lastly, while the architectural DNA-binding proteins CTCF and RAD21 were bound to most chromatin interactions, we found that glucocorticoid-responsive chromatin interactions were depleted for CTCF binding but enriched for RAD21. Together, these findings offer new insights into the mechanisms underlying GC-mediated gene activation and repression.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Glucocorticoids/metabolism , Receptors, Glucocorticoid/metabolism , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Cycle Proteins , Cell Line , Chromatin/genetics , DNA-Binding Proteins , Genome, Human , Humans , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Binding
7.
PLoS Comput Biol ; 14(1): e1005896, 2018 01.
Article in English | MEDLINE | ID: mdl-29337990

ABSTRACT

Transcriptome-wide time series expression profiling is used to characterize the cellular response to environmental perturbations. The first step to analyzing transcriptional response data is often to cluster genes with similar responses. Here, we present a nonparametric model-based method, Dirichlet process Gaussian process mixture model (DPGP), which jointly models data clusters with a Dirichlet process and temporal dependencies with Gaussian processes. We demonstrate the accuracy of DPGP in comparison to state-of-the-art approaches using hundreds of simulated data sets. To further test our method, we apply DPGP to published microarray data from a microbial model organism exposed to stress and to novel RNA-seq data from a human cell line exposed to the glucocorticoid dexamethasone. We validate our clusters by examining local transcription factor binding and histone modifications. Our results demonstrate that jointly modeling cluster number and temporal dependencies can reveal shared regulatory mechanisms. DPGP software is freely available online at https://github.com/PrincetonUniversity/DP_GP_cluster.


Subject(s)
Cluster Analysis , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , A549 Cells , Algorithms , Cell Line, Tumor , Computational Biology , Computer Simulation , Dexamethasone/chemistry , Gene Expression Profiling , Glucocorticoids/chemistry , Histones/chemistry , Humans , Hydrogen Bonding , Hydrogen Peroxide/chemistry , Lung Neoplasms/drug therapy , Models, Biological , Normal Distribution , Oligonucleotide Array Sequence Analysis , Sequence Analysis, RNA , Time Factors , Transcription Factors/chemistry
8.
Transcription ; 8(4): 261-267, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28598247

ABSTRACT

Gene regulation is fundamentally important for the coordination of diverse biologic processes including homeostasis and responses to developmental and environmental stimuli. Transcription factor (TF) binding sites are one of the major functional subunits of gene regulation. They are arranged in cis-regulatory modules (CRMs) that can be more active than the sum of their individual effects. Recently, we described a mechanism of glucocorticoid (GC)-induced gene regulation in which the glucocorticoid receptor (GR) binds coordinately to multiple CRMs that are 10s of kilobases apart in the genome. In those results, the minority of GR binding sites appear to involve direct TF:DNA interactions. Meanwhile, other GR binding sites in a cluster interact with those direct binding sites to tune their gene regulatory activity. Here, we consider the implications of those and related results in the context of existing models of gene regulation. Based on our analyses, we propose that the billboard and regulatory grammar models of cis-regulatory element activity be expanded to consider the influence of long-range interactions between cis-regulatory modules.


Subject(s)
DNA/chemistry , Transcriptional Activation , Binding Sites , DNA/metabolism , Models, Genetic , Receptors, Glucocorticoid/metabolism , Regulon
9.
Curr Opin Genet Dev ; 43: 38-45, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27984826

ABSTRACT

Genetic variation in gene regulatory elements contributes to diverse human diseases, ranging from rare and severe developmental defects to common and complex diseases such as obesity and diabetes. Early examples of regulatory mechanisms of human diseases involve large chromosomal rearrangements that change the regulatory connections within the genome. Single nucleotide variants in regulatory elements can also contribute to disease, potentially via demonstrated associations with changes in transcription factor binding, enhancer activity, post-translational histone modifications, long-range enhancer-promoter interactions, or RNA polymerase recruitment. Establishing causality between non-coding genetic variants, gene regulation, and disease has recently become more feasible with advances in genome-editing and epigenome-editing technologies. As establishing causal regulatory mechanisms of diseases becomes routine, functional annotation of target genes is likely to emerge as a major bottleneck for translation into patient benefits. In this review, we discuss the history and recent advances in understanding the regulatory mechanisms of human disease, and new challenges likely to be encountered once establishing those mechanisms becomes rote.


Subject(s)
Gene Expression Regulation/genetics , Genetic Predisposition to Disease , Genome, Human/genetics , Regulatory Sequences, Nucleic Acid/genetics , DNA-Directed RNA Polymerases/genetics , Enhancer Elements, Genetic , Humans , Polymorphism, Genetic , Promoter Regions, Genetic
10.
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
11.
Genome Res ; 25(8): 1206-14, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26084464

ABSTRACT

We report a novel high-throughput method to empirically quantify individual-specific regulatory element activity at the population scale. The approach combines targeted DNA capture with a high-throughput reporter gene expression assay. As demonstration, we measured the activity of more than 100 putative regulatory elements from 95 individuals in a single experiment. In agreement with previous reports, we found that most genetic variants have weak effects on distal regulatory element activity. Because haplotypes are typically maintained within but not between assayed regulatory elements, the approach can be used to identify causal regulatory haplotypes that likely contribute to human phenotypes. Finally, we demonstrate the utility of the method to functionally fine map causal regulatory variants in regions of high linkage disequilibrium identified by expression quantitative trait loci (eQTL) analyses.


Subject(s)
Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Regulatory Sequences, Nucleic Acid , Computational Biology/methods , Genome, Human , Haplotypes , Humans , Patient-Specific Modeling , Quantitative Trait Loci
12.
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
13.
Nat Biotechnol ; 33(5): 510-7, 2015 May.
Article in English | MEDLINE | ID: mdl-25849900

ABSTRACT

Technologies that enable targeted manipulation of epigenetic marks could be used to precisely control cell phenotype or interrogate the relationship between the epigenome and transcriptional control. Here we describe a programmable, CRISPR-Cas9-based acetyltransferase consisting of the nuclease-null dCas9 protein fused to the catalytic core of the human acetyltransferase p300. The fusion protein catalyzes acetylation of histone H3 lysine 27 at its target sites, leading to robust transcriptional activation of target genes from promoters and both proximal and distal enhancers. Gene activation by the targeted acetyltransferase was highly specific across the genome. In contrast to previous dCas9-based activators, the acetyltransferase activates genes from enhancer regions and with an individual guide RNA. We also show that the core p300 domain can be fused to other programmable DNA-binding proteins. These results support targeted acetylation as a causal mechanism of transactivation and provide a robust tool for manipulating gene regulation.


Subject(s)
CRISPR-Cas Systems/genetics , Enhancer Elements, Genetic , Epigenomics/methods , Acetyltransferases/genetics , E1A-Associated p300 Protein/genetics , HEK293 Cells , Humans , Promoter Regions, Genetic , RNA Editing/genetics , RNA, Guide, Kinetoplastida/genetics , Transcriptional Activation
14.
Nat Neurosci ; 18(5): 647-56, 2015 May.
Article in English | MEDLINE | ID: mdl-25849986

ABSTRACT

To identify chromatin mechanisms of neuronal differentiation, we characterized chromatin accessibility and gene expression in cerebellar granule neurons (CGNs) of the developing mouse. We used DNase-seq to map accessibility of cis-regulatory elements and RNA-seq to profile transcript abundance across postnatal stages of neuronal differentiation in vivo and in culture. We observed thousands of chromatin accessibility changes as CGNs differentiated, and verified, using H3K27ac ChIP-seq, reporter gene assays and CRISPR-mediated activation, that many of these regions function as neuronal enhancers. Motif discovery in differentially accessible chromatin regions suggested a previously unknown role for the Zic family of transcription factors in CGN maturation. We confirmed the association of Zic with these elements by ChIP-seq and found, using knockdown, that Zic1 and Zic2 are required for coordinating mature neuronal gene expression patterns. Together, our data reveal chromatin dynamics at thousands of gene regulatory elements that facilitate the gene expression patterns necessary for neuronal differentiation and function.


Subject(s)
Cerebellar Cortex/growth & development , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental , Neurogenesis/genetics , Neurons/cytology , Transcription Factors/physiology , Animals , Cerebellar Cortex/embryology , Cerebellar Cortex/metabolism , Chromatin/ultrastructure , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Expression Profiling , Genes, Reporter , Histones/metabolism , Mice , Molecular Sequence Data , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic
15.
ACS Synth Biol ; 4(6): 689-99, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25494287

ABSTRACT

Genetic reprogramming holds great potential for disease modeling, drug screening, and regenerative medicine. Genetic reprogramming of mammalian cells is typically achieved by forced expression of natural transcription factors that control master gene networks and cell lineage specification. However, in many instances, the natural transcription factors do not induce a sufficiently robust response to completely reprogram cell phenotype. In this study, we demonstrate that protein engineering of the master transcription factor MyoD can enhance the conversion of human dermal fibroblasts and adult stem cells to a skeletal myocyte phenotype. Fusion of potent transcriptional activation domains to MyoD led to increased myogenic gene expression, myofiber formation, cell fusion, and global reprogramming of the myogenic gene network. This work supports a general strategy for synthetically enhancing the direct conversion between cell types that can be applied in both synthetic biology and regenerative medicine.


Subject(s)
MyoD Protein/metabolism , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Amino Acid Sequence , Cell Lineage , Cell Transdifferentiation , Cellular Reprogramming , Dermis/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Gene Regulatory Networks , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Molecular Sequence Data , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , MyoD Protein/chemistry , MyoD Protein/genetics , Protein Engineering , Protein Structure, Tertiary
16.
Nat Methods ; 10(10): 973-6, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23892895

ABSTRACT

Technologies for engineering synthetic transcription factors have enabled many advances in medical and scientific research. In contrast to existing methods based on engineering of DNA-binding proteins, we created a Cas9-based transactivator that is targeted to DNA sequences by guide RNA molecules. Coexpression of this transactivator and combinations of guide RNAs in human cells induced specific expression of endogenous target genes, demonstrating a simple and versatile approach for RNA-guided gene activation.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats , Protein Engineering/methods , RNA Editing , Transcription Factors/genetics , Transcriptional Activation , HEK293 Cells , High-Throughput Nucleotide Sequencing , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Ribonucleases/genetics , RNA, Small Untranslated
17.
Proc Natl Acad Sci U S A ; 110(23): 9356-61, 2013 Jun 04.
Article in English | MEDLINE | ID: mdl-23690579

ABSTRACT

Most of the airways of the human lung are lined by an epithelium made up of ciliated and secretory luminal cells and undifferentiated basal progenitor cells. The integrity of this epithelium and its ability to act as a selective barrier are critical for normal lung function. In other epithelia, there is evidence that transcription factors of the evolutionarily conserved grainyheadlike (GRHL) family play key roles in coordinating multiple cellular processes required for epithelial morphogenesis, differentiation, remodeling, and repair. However, only a few target genes have been identified, and little is known about GRHL function in the adult lung. Here we focus on the role of GRHL2 in primary human bronchial epithelial cells, both as undifferentiated progenitors and as they differentiate in air-liquid interface culture into an organized mucociliary epithelium with transepithelial resistance. Using a dominant-negative protein or shRNA to inhibit GRHL2, we follow changes in epithelial phenotype and gene transcription using RNA sequencing or microarray analysis. We identify several hundreds of genes that are directly or indirectly regulated by GRHL2 in both undifferentiated cells and air-liquid interface cultures. Using ChIP sequencing to map sites of GRHL2 binding in the basal cells, we identify 7,687 potential primary targets and confirm that GRHL2 binding is strongly enriched near GRHL2-regulated genes. Taken together, the results support the hypothesis that GRHL2 plays a key role in regulating many physiological functions of human airway epithelium, including those involving cell morphogenesis, adhesion, and motility.


Subject(s)
DNA-Binding Proteins/metabolism , Epithelial Cells/cytology , Respiratory Mucosa/physiology , Transcription Factors/metabolism , Cell Adhesion/genetics , Cell Adhesion/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Chromatin Immunoprecipitation , DNA-Binding Proteins/antagonists & inhibitors , Epithelial Cells/metabolism , Gene Expression Regulation/physiology , Genetic Vectors , Humans , Immunohistochemistry , Lentivirus , Microarray Analysis , Morphogenesis/genetics , Morphogenesis/physiology , RNA, Small Interfering/pharmacology , Real-Time Polymerase Chain Reaction , Respiratory Mucosa/metabolism , Sequence Analysis, RNA , Transcription Factors/antagonists & inhibitors
18.
Genome Res ; 18(8): 1238-46, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18436892

ABSTRACT

Silencers and enhancer-blockers (EBs) are cis-acting, negative regulatory elements (NREs) that control interactions between promoters and enhancers. Although relatively uncharacterized in terms of biological mechanisms, these elements are likely to be abundant in the genome. We developed an experimental strategy to identify silencers and EBs using transient transfection assays. A known insulator and EB from the chicken beta-globin locus, cHS4, served as a control element for these assays. We examined 47 sequences from a 1.8-Mb region of human chromosome 7 for silencer and EB activities. The majority of functional elements displayed directional and promoter-specific activities. A limited number of sequences acted in a dual manner, as both silencers and EBs. We examined genomic data, epigenetic modifications, and sequence motifs within these regions. Strong silencer elements contained a novel CT-rich motif, often in multiple copies. Deletion of the motif from three regions caused a measurable loss of silencing ability in these sequences. Moreover, five duplicate occurrences of this motif were identified in the cHS4 insulator. These motifs provided an explanation for an uncharacterized silencing activity we measured in the insulator element. Overall, we identified 15 novel NREs, which contribute new insights into the prevalence and composition of sequences that negatively regulate gene expression.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Insulator Elements , Silencer Elements, Transcriptional , Animals , Binding Sites , CCCTC-Binding Factor , Cell Line , Chickens/genetics , Chromosomes, Human, Pair 7 , DNA-Binding Proteins/metabolism , Enhancer Elements, Genetic , Genomics , Humans , Repressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...