Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
PLoS Genet ; 20(2): e1011159, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38377146

ABSTRACT

Common genetic variants in the repressive GATA-family transcription factor (TF) TRPS1 locus are associated with breast cancer risk, and luminal breast cancer cell lines are particularly sensitive to TRPS1 knockout. We introduced an inducible degron tag into the native TRPS1 locus within a luminal breast cancer cell line to identify the direct targets of TRPS1 and determine how TRPS1 mechanistically regulates gene expression. We acutely deplete over 80 percent of TRPS1 from chromatin within 30 minutes of inducing degradation. We find that TRPS1 regulates transcription of hundreds of genes, including those related to estrogen signaling. TRPS1 directly regulates chromatin structure, which causes estrogen receptor alpha (ER) to redistribute in the genome. ER redistribution leads to both repression and activation of dozens of ER target genes. Downstream from these primary effects, TRPS1 depletion represses cell cycle-related gene sets and reduces cell doubling rate. Finally, we show that high TRPS1 activity, calculated using a gene expression signature defined by primary TRPS1-regulated genes, is associated with worse breast cancer patient prognosis. Taken together, these data suggest a model in which TRPS1 modulates the genomic distribution of ER, both activating and repressing transcription of genes related to cancer cell fitness.


Subject(s)
Breast Neoplasms , Chromatin , Fingers , Hair Diseases , Langer-Giedion Syndrome , Nose , Female , Humans , Breast Neoplasms/genetics , Chromatin/genetics , Estrogen Receptor alpha/genetics , Fingers/abnormalities , GATA Transcription Factors , Gene Expression , Genes, cdc , Nose/abnormalities , Repressor Proteins/genetics
2.
Genes Dev ; 33(19-20): 1441-1455, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31467088

ABSTRACT

Rapid perturbation of protein function permits the ability to define primary molecular responses while avoiding downstream cumulative effects of protein dysregulation. The auxin-inducible degron (AID) system was developed as a tool to achieve rapid and inducible protein degradation in nonplant systems. However, tagging proteins at their endogenous loci results in chronic auxin-independent degradation by the proteasome. To correct this deficiency, we expressed the auxin response transcription factor (ARF) in an improved inducible degron system. ARF is absent from previously engineered AID systems but is a critical component of native auxin signaling. In plants, ARF directly interacts with AID in the absence of auxin, and we found that expression of the ARF PB1 (Phox and Bem1) domain suppresses constitutive degradation of AID-tagged proteins. Moreover, the rate of auxin-induced AID degradation is substantially faster in the ARF-AID system. To test the ARF-AID system in a quantitative and sensitive manner, we measured genome-wide changes in nascent transcription after rapidly depleting the ZNF143 transcription factor. Transcriptional profiling indicates that ZNF143 activates transcription in cis and regulates promoter-proximal paused RNA polymerase density. Rapidly inducible degradation systems that preserve the target protein's native expression levels and patterns will revolutionize the study of biological systems by enabling specific and temporally defined protein dysregulation.


Subject(s)
Genetic Techniques , Proteins/metabolism , Proteolysis , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Indoleacetic Acids/pharmacology , Leupeptins/pharmacology , MCF-7 Cells , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Trans-Activators/genetics , Trans-Activators/metabolism
3.
Genome Res ; 33(3): 314-331, 2023 03.
Article in English | MEDLINE | ID: mdl-36810156

ABSTRACT

Adipocytes contribute to metabolic disorders such as obesity, diabetes, and atherosclerosis. Prior characterizations of the transcriptional network driving adipogenesis have overlooked transiently acting transcription factors (TFs), genes, and regulatory elements that are essential for proper differentiation. Moreover, traditional gene regulatory networks provide neither mechanistic details about individual regulatory element-gene relationships nor temporal information needed to define a regulatory hierarchy that prioritizes key regulatory factors. To address these shortcomings, we integrate kinetic chromatin accessibility (ATAC-seq) and nascent transcription (PRO-seq) data to generate temporally resolved networks that describe TF binding events and resultant effects on target gene expression. Our data indicate which TF families cooperate with and antagonize each other to regulate adipogenesis. Compartment modeling of RNA polymerase density quantifies how individual TFs mechanistically contribute to distinct steps in transcription. The glucocorticoid receptor activates transcription by inducing RNA polymerase pause release, whereas SP and AP-1 factors affect RNA polymerase initiation. We identify Twist2 as a previously unappreciated effector of adipocyte differentiation. We find that TWIST2 acts as a negative regulator of 3T3-L1 and primary preadipocyte differentiation. We confirm that Twist2 knockout mice have compromised lipid storage within subcutaneous and brown adipose tissue. Previous phenotyping of Twist2 knockout mice and Setleis syndrome Twist2 -/- patients noted deficiencies in subcutaneous adipose tissue. This network inference framework is a powerful and general approach for interpreting complex biological phenomena and can be applied to a wide range of cellular processes.


Subject(s)
Adipocytes , Gene Regulatory Networks , Twist-Related Protein 1 , Animals , Mice , Cell Line , Adipocytes/cytology , Adipocytes/metabolism , Transcription Factors/metabolism , Adipogenesis , Transcription, Genetic , Regulatory Elements, Transcriptional , Twist-Related Protein 1/metabolism
4.
Genes Dev ; 30(15): 1731-46, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27492368

ABSTRACT

The coordinated regulation of gene expression at the transcriptional level is fundamental to development and homeostasis. Inducible systems are invaluable when studying transcription because the regulatory process can be triggered instantaneously, allowing the tracking of ordered mechanistic events. Here, we use precision run-on sequencing (PRO-seq) to examine the genome-wide heat shock (HS) response in Drosophila and the function of two key transcription factors on the immediate transcription activation or repression of all genes regulated by HS. We identify the primary HS response genes and the rate-limiting steps in the transcription cycle that GAGA-associated factor (GAF) and HS factor (HSF) regulate. We demonstrate that GAF acts upstream of promoter-proximally paused RNA polymerase II (Pol II) formation (likely at the step of chromatin opening) and that GAF-facilitated Pol II pausing is critical for HS activation. In contrast, HSF is dispensable for establishing or maintaining Pol II pausing but is critical for the release of paused Pol II into the gene body at a subset of highly activated genes. Additionally, HSF has no detectable role in the rapid HS repression of thousands of genes.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/genetics , Gene Expression Regulation/genetics , Stress, Physiological/genetics , Transcription Factors/metabolism , Animals , Cell Line , DNA Polymerase II/metabolism , DNA-Binding Proteins/genetics , Drosophila/metabolism , Drosophila Proteins/genetics , Heat Shock Transcription Factors , Promoter Regions, Genetic/genetics , RNA Interference , Transcription Factors/genetics
5.
Rep Pract Oncol Radiother ; 28(6): 711-719, 2023.
Article in English | MEDLINE | ID: mdl-38515824

ABSTRACT

Background: The low incidence of myxofibrosarcoma (MFS) makes high power studies difficult to perform. Demographic and prognostic factors for MFS and how they differ from all extremity soft tissue sarcomas (STS) are not well understood. The purpose of this study was to characterize a large cohort of patients with MFS and evaluate epidemiologic and survival factors when compared to all STS. Materials and methods: We performed a retrospective review of the Surveillance, Epidemiology, and End Results (SEER) database from 2000 to 2015 to identify 1,440 patients diagnosed with MFS and 12,324 with STS. Survival curves were creased using Kaplan-Meier, and Cox regression analyses were performed to identify hazard ratios (HRs). Results: Overall survival was greater for STS than MFS (79% vs. 67%). Patients with MFS had a higher average age at diagnosis than STS (62 vs. 56), and older age was strongly associated with decreased survivorship for MFS (HR = 7.94). A greater proportion of patients under 30 diagnosed with MFS were female when compared to STS (49.6% vs. 45.4%). The incidence of MFS and STS increased over the 15-year period, with MFS increasing at a greater rate than STS (1.25% vs. 2.59%). Survival increased for patients diagnosed after 2008 for both STS (9.4%) and MFS (13.2%). Conclusions: There are differences between patient demographics and survival factors when comparing MFS to all STS. Monitoring changes in demographic and survival trends for rare STS subtypes like MFS is important to improve diagnostic algorithms and treatment options.

6.
J Biol Chem ; 296: 100097, 2021.
Article in English | MEDLINE | ID: mdl-33208463

ABSTRACT

Heat shock transcription factor 1 (HSF1) orchestrates cellular stress protection by activating or repressing gene transcription in response to protein misfolding, oncogenic cell proliferation, and other environmental stresses. HSF1 is tightly regulated via intramolecular repressive interactions, post-translational modifications, and protein-protein interactions. How these HSF1 regulatory protein interactions are altered in response to acute and chronic stress is largely unknown. To elucidate the profile of HSF1 protein interactions under normal growth and chronic and acutely stressful conditions, quantitative proteomics studies identified interacting proteins in the response to heat shock or in the presence of a poly-glutamine aggregation protein cell-based model of Huntington's disease. These studies identified distinct protein interaction partners of HSF1 as well as changes in the magnitude of shared interactions as a function of each stressful condition. Several novel HSF1-interacting proteins were identified that encompass a wide variety of cellular functions, including roles in DNA repair, mRNA processing, and regulation of RNA polymerase II. One HSF1 partner, CTCF, interacted with HSF1 in a stress-inducible manner and functions in repression of specific HSF1 target genes. Understanding how HSF1 regulates gene repression is a crucial question, given the dysregulation of HSF1 target genes in both cancer and neurodegeneration. These studies expand our understanding of HSF1-mediated gene repression and provide key insights into HSF1 regulation via protein-protein interactions.


Subject(s)
CCCTC-Binding Factor/metabolism , Gene Expression Regulation, Neoplastic , Heat Shock Transcription Factors/metabolism , Heat-Shock Response , Huntington Disease/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , CCCTC-Binding Factor/genetics , HEK293 Cells , Heat Shock Transcription Factors/genetics , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Protein Interaction Maps
7.
Mol Cell ; 56(2): 275-285, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25242143

ABSTRACT

Genomic footprinting has emerged as an unbiased discovery method for transcription factor (TF) occupancy at cognate DNA in vivo. A basic premise of footprinting is that sequence-specific TF-DNA interactions are associated with localized resistance to nucleases, leaving observable signatures of cleavage within accessible chromatin. This phenomenon is interpreted to imply protection of the critical nucleotides by the stably bound protein factor. However, this model conflicts with previous reports of many TFs exchanging with specific binding sites in living cells on a timescale of seconds. We show that TFs with short DNA residence times have no footprints at bound motif elements. Moreover, the nuclease cleavage profile within a footprint originates from the DNA sequence in the factor-binding site, rather than from the protein occupying specific nucleotides. These findings suggest a revised understanding of TF footprinting and reveal limitations in comprehensive reconstruction of the TF regulatory network using this approach.


Subject(s)
Base Sequence , DNA Footprinting , DNA/metabolism , Sequence Analysis, DNA , Transcription Factors/metabolism , Binding Sites/genetics , DNA/chemistry , DNA Cleavage , Deoxyribonuclease I/chemistry , Endodeoxyribonucleases/chemistry , Genomics , Humans , Protein Binding/genetics , Protein Structure, Tertiary , ROC Curve , Transcription Factors/chemistry
8.
Bioinformatics ; 36(9): 2926-2928, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31917388

ABSTRACT

SUMMARY: Nascent transcript measurements derived from run-on sequencing experiments are critical for the investigation of transcriptional mechanisms and regulatory networks. However, conventional mRNA gene annotations significantly differ from the boundaries of primary transcripts. New primary transcript annotations are needed to accurately interpret run-on data. We developed the primaryTranscriptAnnotation R package to infer the transcriptional start and termination sites of primary transcripts from genomic run-on data. We then used these inferred coordinates to annotate transcriptional units identified de novo. This package provides the novel utility to integrate data-driven primary transcript annotations with transcriptional unit coordinates identified in an unbiased manner. Highlighting the importance of using accurate primary transcript coordinates, we demonstrate that this new methodology increases the detection of differentially expressed transcripts and provides more accurate quantification of RNA polymerase pause indices. AVAILABILITY AND IMPLEMENTATION: https://github.com/WarrenDavidAnderson/genomicsRpackage/tree/master/primaryTranscriptAnnotation. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome , Genomics , Molecular Sequence Annotation , RNA, Messenger/genetics
9.
Nucleic Acids Res ; 46(12): e75, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29672735

ABSTRACT

A key challenge in quantitative ChIP combined with high-throughput sequencing (ChIP-seq) is the normalization of data in the presence of genome-wide changes in occupancy. Analysis-based normalization methods were developed for transcriptomic data and these are dependent on the underlying assumption that total transcription does not change between conditions. For genome-wide changes in transcription factor (TF) binding, these assumptions do not hold true. The challenges in normalization are confounded by experimental variability during sample preparation, processing and recovery. We present a novel normalization strategy utilizing an internal standard of unchanged peaks for reference. Our method can be readily applied to monitor genome-wide changes by ChIP-seq that are otherwise lost or misrepresented through analytical normalization. We compare our approach to normalization by total read depth and two alternative methods that utilize external experimental controls to study TF binding. We successfully resolve the key challenges in quantitative ChIP-seq analysis and demonstrate its application by monitoring the loss of Estrogen Receptor-alpha (ER) binding upon fulvestrant treatment, ER binding in response to estrodiol, ER mediated change in H4K12 acetylation and profiling ER binding in patient-derived xenographs. This is supported by an adaptable pipeline to normalize and quantify differential TF binding genome-wide and generate metrics for differential binding at individual sites.


Subject(s)
Chromatin Immunoprecipitation/standards , High-Throughput Nucleotide Sequencing/standards , Sequence Analysis, DNA/standards , Animals , Antibodies , CCCTC-Binding Factor/immunology , Drosophila melanogaster/genetics , Estrogen Receptor alpha/immunology , Estrogen Receptor alpha/metabolism , Histones/immunology , Histones/metabolism , Humans , MCF-7 Cells , Mice , Reference Standards
10.
Nucleic Acids Res ; 46(2): e9, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29126307

ABSTRACT

Coupling molecular biology to high-throughput sequencing has revolutionized the study of biology. Molecular genomics techniques are continually refined to provide higher resolution mapping of nucleic acid interactions and structure. Sequence preferences of enzymes can interfere with the accurate interpretation of these data. We developed seqOutBias to characterize enzymatic sequence bias from experimental data and scale individual sequence reads to correct intrinsic enzymatic sequence biases. SeqOutBias efficiently corrects DNase-seq, TACh-seq, ATAC-seq, MNase-seq and PRO-seq data. We show that seqOutBias correction facilitates identification of true molecular signatures resulting from transcription factors and RNA polymerase interacting with DNA.


Subject(s)
Algorithms , Computational Biology/methods , DNA/metabolism , Deoxyribonucleases/metabolism , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Bias , DNA/chemistry , DNA/genetics , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Deoxyribonucleases/genetics , Protein Binding , Reproducibility of Results , Transcription Factors/genetics , Transcription Factors/metabolism
11.
PLoS Genet ; 13(9): e1006761, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28957321

ABSTRACT

Genome-wide association studies (GWAS) have discovered thousands loci associated with disease risk and quantitative traits, yet most of the variants responsible for risk remain uncharacterized. The majority of GWAS-identified loci are enriched for non-coding single-nucleotide polymorphisms (SNPs) and defining the molecular mechanism of risk is challenging. Many non-coding causal SNPs are hypothesized to alter transcription factor (TF) binding sites as the mechanism by which they affect organismal phenotypes. We employed an integrative genomics approach to identify candidate TF binding motifs that confer breast cancer-specific phenotypes identified by GWAS. We performed de novo motif analysis of regulatory elements, analyzed evolutionary conservation of identified motifs, and assayed TF footprinting data to identify sequence elements that recruit TFs and maintain chromatin landscape in breast cancer-relevant tissue and cell lines. We identified candidate causal SNPs that are predicted to alter TF binding within breast cancer-relevant regulatory regions that are in strong linkage disequilibrium with significantly associated GWAS SNPs. We confirm that the TFs bind with predicted allele-specific preferences using CTCF ChIP-seq data. We used The Cancer Genome Atlas breast cancer patient data to identify ANKLE1 and ZNF404 as the target genes of candidate TF binding site SNPs in the 19p13.11 and 19q13.31 GWAS-identified loci. These SNPs are associated with the expression of ZNF404 and ANKLE1 in breast tissue. This integrative analysis pipeline is a general framework to identify candidate causal variants within regulatory regions and TF binding sites that confer phenotypic variation and disease risk.


Subject(s)
Breast Neoplasms/genetics , DNA-Binding Proteins/genetics , Endonucleases/genetics , Genome-Wide Association Study , Alleles , Breast Neoplasms/pathology , Chromatin/genetics , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Nucleotide Motifs/genetics , Polymorphism, Single Nucleotide , Protein Binding , Quantitative Trait Loci/genetics , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics
12.
Bioinformatics ; 34(16): 2867-2869, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29608647

ABSTRACT

Summary: Identification of functional transcription factors that regulate a given gene set is an important problem in gene regulation studies. Conventional approaches for identifying transcription factors, such as DNA sequence motif analysis, are unable to predict functional binding of specific factors and not sensitive enough to detect factors binding at distal enhancers. Here, we present binding analysis for regulation of transcription (BART), a novel computational method and software package for predicting functional transcription factors that regulate a query gene set or associate with a query genomic profile, based on more than 6000 existing ChIP-seq datasets for over 400 factors in human or mouse. This method demonstrates the advantage of utilizing publicly available data for functional genomics research. Availability and implementation: BART is implemented in Python and available at http://faculty.virginia.edu/zanglab/bart. Supplementary information: Supplementary data are available at Bioinformatics online.


Subject(s)
Epigenomics , Software , Transcription Factors/analysis , Animals , Databases, Genetic , Gene Expression Regulation , Humans , Mice , Sequence Analysis, DNA/methods , Transcription Factors/genetics
13.
PLoS Genet ; 11(3): e1005108, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25815464

ABSTRACT

Previous studies have shown that GAGA Factor (GAF) is enriched on promoters with paused RNA Polymerase II (Pol II), but its genome-wide function and mechanism of action remain largely uncharacterized. We assayed the levels of transcriptionally-engaged polymerase using global run-on sequencing (GRO-seq) in control and GAF-RNAi Drosophila S2 cells and found promoter-proximal polymerase was significantly reduced on a large subset of paused promoters where GAF occupancy was reduced by knock down. These promoters show a dramatic increase in nucleosome occupancy upon GAF depletion. These results, in conjunction with previous studies showing that GAF directly interacts with nucleosome remodelers, strongly support a model where GAF directs nucleosome displacement at the promoter and thereby allows the entry Pol II to the promoter and pause sites. This action of GAF on nucleosomes is at least partially independent of paused Pol II because intergenic GAF binding sites with little or no Pol II also show GAF-dependent nucleosome displacement. In addition, the insulator factor BEAF, the BEAF-interacting protein Chriz, and the transcription factor M1BP are strikingly enriched on those GAF-associated genes where pausing is unaffected by knock down, suggesting insulators or the alternative promoter-associated factor M1BP protect a subset of GAF-bound paused genes from GAF knock-down effects. Thus, GAF binding at promoters can lead to the local displacement of nucleosomes, but this activity can be restricted or compensated for when insulator protein or M1BP complexes also reside at GAF bound promoters.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , RNA Polymerase II/genetics , Transcription Factors/genetics , Transcription, Genetic , Animals , Binding Sites , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Eye Proteins/genetics , Eye Proteins/metabolism , Gene Knockdown Techniques , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Factors/metabolism
14.
Curr Neurol Neurosci Rep ; 17(3): 23, 2017 03.
Article in English | MEDLINE | ID: mdl-28283963

ABSTRACT

Traumatic brain injury (TBI) and traumatic spinal cord injury (SCI), collectively termed neurotrauma, are two parallel neurological conditions that can cause long-lasting neurological impairment and other comorbidities in patients, while at the same time, can create a high burden to society. To date, there are still no FDA-approved therapeutic interventions for either TBI or SCI. Recent advances in proteomic technologies, including tandem mass spectrometry, as well as imaging mass spectrometry, have enabled new approaches to study the differential proteome in TBI and SCI with the use of either animal disease models and/or biosamples from clinical observational studies. Thus, the applications of state-of-the-art proteomic method hold promises in shedding light on identifying clinically useful neurotrauma "biomarkers" and/or in identifying distinct and, otherwise, unobvious systems pathways or "key drivers" that can be further exploited as new therapeutic intervention targets.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Proteomics , Spinal Cord Injuries/metabolism , Animals , Biomarkers , Comorbidity , Disease Models, Animal , Humans , Mass Spectrometry
15.
PLoS Genet ; 10(9): e1004613, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25211228

ABSTRACT

Transcription factor binding to DNA in vivo causes the recruitment of chromatin modifiers that can cause changes in chromatin structure, including the modification of histone tails. We previously showed that estrogen receptor (ER) target gene activation is facilitated by peptidylarginine deiminase 2 (PAD2)-catalyzed histone H3R26 deimination (H3R26Cit). Here we report that the genomic distributions of ER and H3R26Cit in breast cancer cells are strikingly coincident, linearly correlated, and observed as early as 2 minutes following estradiol treatment. The H3R26Cit profile is unlike that of previously described histone modifications and is characterized by sharp, narrow peaks. Paired-end MNase ChIP-seq indicates that the charge-neutral H3R26Cit modification facilitates ER binding to DNA by altering the fine structure of the nucleosome. Clinically, we find that PAD2 and H3R26Cit levels correlate with ER expression in breast tumors and that high PAD2 expression is associated with increased survival in ER+ breast cancer patients. These findings provide insight into how transcription factors gain access to nucleosomal DNA and implicate PAD2 as a novel therapeutic target for ER+ breast cancer.


Subject(s)
Histones/metabolism , Nucleosomes/metabolism , Receptors, Estrogen/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Chromatin Assembly and Disassembly , Estrogens/metabolism , Estrogens/pharmacology , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Genomics , Humans , Hydrolases/genetics , Hydrolases/metabolism , MCF-7 Cells , Prognosis , Protein Binding , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases
16.
PLoS Genet ; 8(3): e1002610, 2012.
Article in English | MEDLINE | ID: mdl-22479205

ABSTRACT

DNA sequence and local chromatin landscape act jointly to determine transcription factor (TF) binding intensity profiles. To disentangle these influences, we developed an experimental approach, called protein/DNA binding followed by high-throughput sequencing (PB-seq), that allows the binding energy landscape to be characterized genome-wide in the absence of chromatin. We applied our methods to the Drosophila Heat Shock Factor (HSF), which inducibly binds a target DNA sequence element (HSE) following heat shock stress. PB-seq involves incubating sheared naked genomic DNA with recombinant HSF, partitioning the HSF-bound and HSF-free DNA, and then detecting HSF-bound DNA by high-throughput sequencing. We compared PB-seq binding profiles with ones observed in vivo by ChIP-seq and developed statistical models to predict the observed departures from idealized binding patterns based on covariates describing the local chromatin environment. We found that DNase I hypersensitivity and tetra-acetylation of H4 were the most influential covariates in predicting changes in HSF binding affinity. We also investigated the extent to which DNA accessibility, as measured by digital DNase I footprinting data, could be predicted from MNase-seq data and the ChIP-chip profiles for many histone modifications and TFs, and found GAGA element associated factor (GAF), tetra-acetylation of H4, and H4K16 acetylation to be the most predictive covariates. Lastly, we generated an unbiased model of HSF binding sequences, which revealed distinct biophysical properties of the HSF/HSE interaction and a previously unrecognized substructure within the HSE. These findings provide new insights into the interplay between the genomic sequence and the chromatin landscape in determining transcription factor binding intensity.


Subject(s)
Chromatin , DNA-Binding Proteins , Drosophila Proteins , Drosophila melanogaster , Transcription Factors/genetics , Acetylation , Animals , Binding Sites/genetics , Chromatin/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Deoxyribonuclease I/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation , Genome, Insect , Heat Shock Transcription Factors , Heat-Shock Response/genetics , High-Throughput Nucleotide Sequencing , Histones/genetics , Histones/metabolism , Transcription Factors/metabolism , Transcriptional Activation/genetics
17.
Proc Natl Acad Sci U S A ; 109(33): 13331-6, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22853951

ABSTRACT

Cofactors for estrogen receptor α (ERα) can modulate gene activity by posttranslationally modifying histone tails at target promoters. Here, we found that stimulation of ERα-positive cells with 17ß-estradiol (E2) promotes global citrullination of histone H3 arginine 26 (H3R26) on chromatin. Additionally, we found that the H3 citrulline 26 (H3Cit26) modification colocalizes with ERα at decondensed chromatin loci surrounding the estrogen-response elements of target promoters. Surprisingly, we also found that citrullination of H3R26 is catalyzed by peptidylarginine deiminase (PAD) 2 and not by PAD4 (which citrullinates H4R3). Further, we showed that PAD2 interacts with ERα after E2 stimulation and that inhibition of either PAD2 or ERα strongly suppresses E2-induced H3R26 citrullination and ERα recruitment at target gene promoters. Collectively, our data suggest that E2 stimulation induces the recruitment of PAD2 to target promoters by ERα, whereby PAD2 then citrullinates H3R26, which leads to local chromatin decondensation and transcriptional activation.


Subject(s)
Arginine/metabolism , Biocatalysis , Citrulline/metabolism , Estrogen Receptor alpha/metabolism , Histones/metabolism , Hydrolases/metabolism , Transcriptional Activation , Animals , Biocatalysis/drug effects , Cell Line, Tumor , Chromatin/metabolism , Estrogens/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Genome, Human/genetics , Humans , Mice , Nucleotide Motifs/genetics , Promoter Regions, Genetic/genetics , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases , Response Elements/genetics , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects
19.
bioRxiv ; 2024 May 15.
Article in English | MEDLINE | ID: mdl-38798607

ABSTRACT

Transcription factors bind to sequence motifs and act as activators or repressors. Transcription factors interface with a constellation of accessory cofactors to regulate distinct mechanistic steps to regulate transcription. We rapidly degraded the essential and ubiquitously expressed transcription factor ZNF143 to determine its function in the transcription cycle. ZNF143 facilitates RNA Polymerase initiation and activates gene expression. ZNF143 binds the promoter of nearly all its activated target genes. ZNF143 also binds near the site of genic transcription initiation to directly repress a subset of genes. Although ZNF143 stimulates initiation at ZNF143-repressed genes (i.e. those that increase expression upon ZNF143 depletion), the molecular context of binding leads to cis repression. ZNF143 competes with other more efficient activators for promoter access, physically occludes transcription initiation sites and promoter-proximal sequence elements, and acts as a molecular roadblock to RNA Polymerases during early elongation. The term context specific is often invoked to describe transcription factors that have both activation and repression functions. We define the context and molecular mechanisms of ZNF143-mediated cis activation and repression.

20.
PLoS Genet ; 6(9): e1001114, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20844575

ABSTRACT

Sequence-specific transcription factors (TFs) are critical for specifying patterns and levels of gene expression, but target DNA elements are not sufficient to specify TF binding in vivo. In eukaryotes, the binding of a TF is in competition with a constellation of other proteins, including histones, which package DNA into nucleosomes. We used the ChIP-seq assay to examine the genome-wide distribution of Drosophila Heat Shock Factor (HSF), a TF whose binding activity is mediated by heat shock-induced trimerization. HSF binds to 464 sites after heat shock, the vast majority of which contain HSF Sequence-binding Elements (HSEs). HSF-bound sequence motifs represent only a small fraction of the total HSEs present in the genome. ModENCODE ChIP-chip datasets, generated during non-heat shock conditions, were used to show that inducibly bound HSE motifs are associated with histone acetylation, H3K4 trimethylation, RNA Polymerase II, and coactivators, compared to HSE motifs that remain HSF-free. Furthermore, directly changing the chromatin landscape, from an inactive to an active state, permits inducible HSF binding. There is a strong correlation of bound HSEs to active chromatin marks present prior to induced HSF binding, indicating that an HSE's residence in "active" chromatin is a primary determinant of whether HSF can bind following heat shock.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Animals , Base Sequence , Binding Sites , Chromatin Immunoprecipitation , Consensus Sequence/genetics , DNA-Binding Proteins/deficiency , Drosophila Proteins/deficiency , Drosophila melanogaster/genetics , Heat Shock Transcription Factors , Molecular Sequence Data , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factors/deficiency , Transcriptional Activation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL