Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Proc Natl Acad Sci U S A ; 120(49): e2305773120, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-38011552

ABSTRACT

Exposure to stressful life events increases the risk for psychiatric disorders. Mechanistic insight into the genetic factors moderating the impact of stress can increase our understanding of disease processes. Here, we test 3,662 single nucleotide polymorphisms (SNPs) from preselected expression quantitative trait loci in massively parallel reporter assays to identify genetic variants that modulate the activity of regulatory elements sensitive to glucocorticoids, important mediators of the stress response. Of the tested SNP sequences, 547 were located in glucocorticoid-responsive regulatory elements of which 233 showed allele-dependent activity. Transcripts regulated by these functional variants were enriched for those differentially expressed in psychiatric disorders in the postmortem brain. Phenome-wide Mendelian randomization analysis in 4,439 phenotypes revealed potentially causal associations specifically in neurobehavioral traits, including major depression and other psychiatric disorders. Finally, a functional gene score derived from these variants was significantly associated with differences in the physiological stress response, suggesting that these variants may alter disease risk by moderating the individual set point of the stress response.


Subject(s)
Glucocorticoids , Mental Disorders , Humans , High-Throughput Screening Assays , Regulatory Sequences, Nucleic Acid , Quantitative Trait Loci , Mental Disorders/genetics , Polymorphism, Single Nucleotide , Genome-Wide Association Study , Genetic Predisposition to Disease
2.
Nucleic Acids Res ; 49(21): 12178-12195, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34850108

ABSTRACT

Embryonic stem cells (ESCs) can differentiate into any given cell type and therefore represent a versatile model to study the link between gene regulation and differentiation. To quantitatively assess the dynamics of enhancer activity during the early stages of murine ESC differentiation, we analyzed accessible genomic regions using STARR-seq, a massively parallel reporter assay. This resulted in a genome-wide quantitative map of active mESC enhancers, in pluripotency and during the early stages of differentiation. We find that only a minority of accessible regions is active and that such regions are enriched near promoters, characterized by specific chromatin marks, enriched for distinct sequence motifs, and modeling shows that active regions can be predicted from sequence alone. Regions that change their activity upon retinoic acid-induced differentiation are more prevalent at distal intergenic regions when compared to constitutively active enhancers. Further, analysis of differentially active enhancers verified the contribution of individual TF motifs toward activity and inducibility as well as their role in regulating endogenous genes. Notably, the activity of retinoic acid receptor alpha (RARα) occupied regions can either increase or decrease upon the addition of its ligand, retinoic acid, with the direction of the change correlating with spacing and orientation of the RARα consensus motif and the co-occurrence of additional sequence motifs. Together, our genome-wide enhancer activity map elucidates features associated with enhancer activity levels, identifies regulatory regions disregarded by computational prediction tools, and provides a resource for future studies into regulatory elements in mESCs.


Subject(s)
Mouse Embryonic Stem Cells/cytology , Receptors, Retinoic Acid/metabolism , Animals , Cell Differentiation , Chromosome Mapping , Enhancer Elements, Genetic , Mice
3.
Nucleic Acids Res ; 49(7): 3856-3875, 2021 04 19.
Article in English | MEDLINE | ID: mdl-33751115

ABSTRACT

The glucocorticoid (GR) and androgen (AR) receptors execute unique functions in vivo, yet have nearly identical DNA binding specificities. To identify mechanisms that facilitate functional diversification among these transcription factor paralogs, we studied them in an equivalent cellular context. Analysis of chromatin and sequence suggest that divergent binding, and corresponding gene regulation, are driven by different abilities of AR and GR to interact with relatively inaccessible chromatin. Divergent genomic binding patterns can also be the result of subtle differences in DNA binding preference between AR and GR. Furthermore, the sequence composition of large regions (>10 kb) surrounding selectively occupied binding sites differs significantly, indicating a role for the sequence environment in guiding AR and GR to distinct binding sites. The comparison of binding sites that are shared shows that the specificity paradox can also be resolved by differences in the events that occur downstream of receptor binding. Specifically, shared binding sites display receptor-specific enhancer activity, cofactor recruitment and changes in histone modifications. Genomic deletion of shared binding sites demonstrates their contribution to directing receptor-specific gene regulation. Together, these data suggest that differences in genomic occupancy as well as divergence in the events that occur downstream of receptor binding direct functional diversification among transcription factor paralogs.


Subject(s)
Chromatin/metabolism , DNA/metabolism , Receptors, Androgen/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Line, Tumor , Gene Expression Regulation , Humans , Protein Binding
4.
PLoS Genet ; 14(11): e1007793, 2018 11.
Article in English | MEDLINE | ID: mdl-30427832

ABSTRACT

The binding of transcription factors to short recognition sequences plays a pivotal role in controlling the expression of genes. The sequence and shape characteristics of binding sites influence DNA binding specificity and have also been implicated in modulating the activity of transcription factors downstream of binding. To quantitatively assess the transcriptional activity of tens of thousands of designed synthetic sites in parallel, we developed a synthetic version of STARR-seq (synSTARR-seq). We used the approach to systematically analyze how variations in the recognition sequence of the glucocorticoid receptor (GR) affect transcriptional regulation. Our approach resulted in the identification of a novel highly active functional GR binding sequence and revealed that sequence variation both within and flanking GR's core binding site can modulate GR activity without apparent changes in DNA binding affinity. Notably, we found that the sequence composition of variants with similar activity profiles was highly diverse. In contrast, groups of variants with similar activity profiles showed specific DNA shape characteristics indicating that DNA shape may be a better predictor of activity than DNA sequence. Finally, using single cell experiments with individual enhancer variants, we obtained clues indicating that the architecture of the response element can independently tune expression mean and cell-to cell variability in gene expression (noise). Together, our studies establish synSTARR as a powerful method to systematically study how DNA sequence and shape modulate transcriptional output and noise.


Subject(s)
DNA/genetics , Sequence Analysis, DNA/methods , Transcription, Genetic , Binding Sites/genetics , DNA/chemistry , DNA/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation , Genes, Reporter , Genes, Synthetic , Genetic Variation , Humans , Nucleic Acid Conformation , Protein Conformation , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Response Elements , Sequence Analysis, DNA/statistics & numerical data , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Nucleic Acids Res ; 46(6): 2868-2882, 2018 04 06.
Article in English | MEDLINE | ID: mdl-29385519

ABSTRACT

Genomic binding of transcription factors, like the glucocorticoid receptor (GR), is linked to the regulation of genes. However, as we show here, GR binding is a poor predictor of GR-dependent gene regulation even when taking the 3D organization of the genome into account. To connect GR binding sites to the regulation of genes in the endogenous genomic context, we turned to genome editing. By deleting GR binding sites, individually or in combination, we uncovered how cooperative interactions between binding sites contribute to the regulation of genes. Specifically, for the GR target gene GILZ, we show that the simultaneous presence of a cluster of GR binding sites is required for the activity of an individual enhancer and that the GR-dependent regulation of GILZ depends on multiple GR-bound enhancers. Further, by deleting GR binding sites that are shared between different cell types, we show how cell type-specific genome organization and enhancer-blocking can result in cell type-specific wiring of promoter-enhancer contacts. This rewiring allows an individual GR binding site shared between different cell types to direct the expression of distinct transcripts and thereby contributes to the cell type-specific consequences of glucocorticoid signaling.


Subject(s)
Enhancer Elements, Genetic/genetics , Genome/genetics , Genomics/methods , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , A549 Cells , Animals , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Dexamethasone/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glucocorticoids/pharmacology , Humans , Protein Binding
6.
J Immunol ; 199(1): 48-61, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28515280

ABSTRACT

Although glucocorticoids (GCs) are a mainstay in the clinical management of asthma, the target cells that mediate their therapeutic effects are unknown. Contrary to our expectation, we found that GC receptor (GR) expression in immune cells was dispensable for successful therapy of allergic airway inflammation (AAI) with dexamethasone. Instead, GC treatment was compromised in mice expressing a defective GR in the nonhematopoietic compartment or selectively lacking the GR in airway epithelial cells. Further, we found that an intact GR dimerization interface was a prerequisite for the suppression of AAI and airway hyperresponsiveness by GCs. Our observation that the ability of dexamethasone to modulate gene expression in airway epithelial cells coincided with its potency to resolve AAI supports a crucial role for transcriptional regulation by the GR in this cell type. Taken together, we identified an unknown mode of GC action in the treatment of allergic asthma that might help to develop more specific therapies in the future.


Subject(s)
Asthma/drug therapy , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Glucocorticoids/pharmacology , Receptors, Glucocorticoid/metabolism , Respiratory Mucosa/drug effects , Animals , Asthma/immunology , Asthma/physiopathology , Dexamethasone/therapeutic use , Disease Models, Animal , Epithelial Cells/immunology , Epithelial Cells/metabolism , Gene Expression Regulation , Glucocorticoids/therapeutic use , Inflammation/drug therapy , Inflammation/immunology , Mice , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/genetics , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Signal Transduction
7.
Nucleic Acids Res ; 45(4): 1805-1819, 2017 02 28.
Article in English | MEDLINE | ID: mdl-27903902

ABSTRACT

The genomic loci bound by the glucocorticoid receptor (GR), a hormone-activated transcription factor, show little overlap between cell types. To study the role of chromatin and sequence in specifying where GR binds, we used Bayesian modeling within the universe of accessible chromatin. Taken together, our results uncovered that although GR preferentially binds accessible chromatin, its binding is biased against accessible chromatin located at promoter regions. This bias can only be explained partially by the presence of fewer GR recognition sequences, arguing for the existence of additional mechanisms that interfere with GR binding at promoters. Therefore, we tested the role of H3K9ac, the chromatin feature with the strongest negative association with GR binding, but found that this correlation does not reflect a causative link. Finally, we find a higher percentage of promoter-proximal GR binding for genes regulated by GR across cell types than for cell type-specific target genes. Given that GR almost exclusively binds accessible chromatin, we propose that cell type-specific regulation by GR preferentially occurs via distal enhancers, whose chromatin accessibility is typically cell type-specific, whereas ubiquitous target gene regulation is more likely to result from binding to promoter regions, which are often accessible regardless of cell type examined.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin/genetics , Chromatin/metabolism , Gene Expression Regulation , Receptors, Glucocorticoid/metabolism , Animals , Base Sequence , Bayes Theorem , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Computational Biology/methods , Genome-Wide Association Study , Genomics , High-Throughput Nucleotide Sequencing , Mice , Nucleotide Motifs , Organ Specificity/genetics , Promoter Regions, Genetic , Protein Binding , p300-CBP Transcription Factors/genetics , p300-CBP Transcription Factors/metabolism
8.
Genome Res ; 25(6): 825-35, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25720775

ABSTRACT

The classical DNA recognition sequence of the glucocorticoid receptor (GR) appears to be present at only a fraction of bound genomic regions. To identify sequences responsible for recruitment of this transcription factor (TF) to individual loci, we turned to the high-resolution ChIP-exo approach. We exploited this signal by determining footprint profiles of TF binding at single-base-pair resolution using ExoProfiler, a computational pipeline based on DNA binding motifs. When applied to our GR and the few available public ChIP-exo data sets, we find that ChIP-exo footprints are protein- and recognition sequence-specific signatures of genomic TF association. Furthermore, we show that ChIP-exo captures information about TFs other than the one directly targeted by the antibody in the ChIP procedure. Consequently, the shape of the ChIP-exo footprint can be used to discriminate between direct and indirect (tethering to other DNA-bound proteins) DNA association of GR. Together, our findings indicate that the absence of classical recognition sequences can be explained by direct GR binding to a broader spectrum of sequences than previously known, either as a homodimer or as a heterodimer binding together with a member of the ETS or TEAD families of TFs, or alternatively by indirect recruitment via FOX or STAT proteins. ChIP-exo footprints also bring structural insights and locate DNA:protein cross-link points that are compatible with crystal structures of the studied TFs. Overall, our generically applicable footprint-based approach uncovers new structural and functional insights into the diverse ways of genomic cooperation and association of TFs.


Subject(s)
Chromatin Immunoprecipitation/methods , Genomics , Hepatocyte Nuclear Factor 3-alpha/genetics , Receptors, Glucocorticoid/genetics , CCCTC-Binding Factor , Cell Line, Tumor , Computational Biology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Profiling , Genetic Loci , HeLa Cells , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , K562 Cells , MCF-7 Cells , Protein Binding , Protein Conformation , Receptors, Glucocorticoid/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Sequence Analysis, DNA
9.
Nucleic Acids Res ; 44(13): 6142-56, 2016 07 27.
Article in English | MEDLINE | ID: mdl-27016732

ABSTRACT

Out of the myriad of potential DNA binding sites of the glucocorticoid receptor (GR) found in the human genome, only a cell-type specific minority is actually bound, indicating that the presence of a recognition sequence alone is insufficient to specify where GR binds. Cooperative interactions with other transcription factors (TFs) are known to contribute to binding specificity. Here, we reasoned that sequence signals preventing GR recruitment to certain loci provide an alternative means to confer specificity. Motif analyses uncovered candidate Negative Regulatory Sequences (NRSs) that interfere with genomic GR binding. Subsequent functional analyses demonstrated that NRSs indeed prevent GR binding to nearby response elements. We show that NRS activity is conserved across species, found in most tissues and that they also interfere with the genomic binding of other TFs. Interestingly, the effects of NRSs appear not to be a simple consequence of changes in chromatin accessibility. Instead, we find that NRSs interact with proteins found at sub-nuclear structures called paraspeckles and that these proteins might mediate the repressive effects of NRSs. Together, our studies suggest that the joint influence of positive and negative sequence signals partition the genome into regions where GR can bind and those where it cannot.


Subject(s)
Base Sequence/genetics , DNA-Binding Proteins/genetics , Receptors, Glucocorticoid/genetics , Response Elements/genetics , Binding Sites/genetics , Chromatin/genetics , DNA-Binding Proteins/metabolism , Humans , Promoter Regions, Genetic , Receptors, Glucocorticoid/metabolism , Transcription Factors/genetics
10.
Proc Natl Acad Sci U S A ; 110(44): 17826-31, 2013 Oct 29.
Article in English | MEDLINE | ID: mdl-24127590

ABSTRACT

In addition to guiding proteins to defined genomic loci, DNA can act as an allosteric ligand that influences protein structure and activity. Here we compared genome-wide binding, transcriptional regulation, and, using NMR, the conformation of two glucocorticoid receptor (GR) isoforms that differ by a single amino acid insertion in the lever arm, a domain that adopts DNA sequence-specific conformations. We show that these isoforms differentially regulate gene expression levels through two mechanisms: differential DNA binding and altered communication between GR domains. Our studies suggest a versatile role for DNA in both modulating GR activity and also in directing the use of GR isoforms. We propose that the lever arm is a "fulcrum" for bidirectional allosteric signaling, conferring conformational changes in the DNA reading head that influence DNA sequence selectivity, as well as conferring changes in the dimerization domain that connect functionally with remote regulatory surfaces, thereby influencing which genes are regulated and the magnitude of their regulation.


Subject(s)
Alternative Splicing/genetics , Amino Acids/genetics , Gene Expression Regulation/genetics , Models, Molecular , Protein Conformation , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/physiology , Cell Line, Tumor , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Humans , Immunoblotting , Microarray Analysis , Mutagenesis, Insertional/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Real-Time Polymerase Chain Reaction , Transcription, Genetic/genetics
12.
Adv Exp Med Biol ; 872: 59-81, 2015.
Article in English | MEDLINE | ID: mdl-26215990

ABSTRACT

One fascinating aspect of glucocorticoid signaling is their broad range of physiological and pharmacological effects. These effects are at least in part a consequence of transcriptional regulation by the glucocorticoid receptor (GR). Activation of GR by glucocorticoids results in tissue-specific changes in gene expression levels with some genes being activated whereas others are repressed. This raises two questions: First, how does GR regulate different subsets of target genes in different tissues? And second, how can GR both activate and repress the expression of genes?To answer these questions, this chapter will describe the function of the various "components" and how they cooperate to mediate the transcriptional responses to glucocorticoids. The first "component" is GR itself. The second "component" is the chromatin and its role in specifying where in the genome GR binds. Binding to the genome however is just the first step in regulating the expression of genes and transcriptional regulation by GR depends on the recruitment of coregulator proteins that either directly or indirectly influence the recruitment and or activity of RNA polymerase II. Ultimately, the integration of inputs including GR isoform, DNA sequence, chromatin and cooperation with coregulators determines which genes are regulated and the direction of their regulation.


Subject(s)
Gene Expression Regulation/physiology , Glucocorticoids/physiology , Transcription, Genetic/physiology , Animals , Humans , Protein Processing, Post-Translational , Receptors, Glucocorticoid/metabolism
13.
Genome Res ; 20(8): 1133-42, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20508142

ABSTRACT

Isogenic settings are routine in model organisms, yet remain elusive for genetic experiments on human cells. We describe the use of designed zinc finger nucleases (ZFNs) for efficient transgenesis without drug selection into the PPP1R12C gene, a "safe harbor" locus known as AAVS1. ZFNs enable targeted transgenesis at a frequency of up to 15% following transient transfection of both transformed and primary human cells, including fibroblasts and hES cells. When added to this locus, transgenes such as expression cassettes for shRNAs, small-molecule-responsive cDNA expression cassettes, and reporter constructs, exhibit consistent expression and sustained function over 50 cell generations. By avoiding random integration and drug selection, this method allows bona fide isogenic settings for high-throughput functional genomics, proteomics, and regulatory DNA analysis in essentially any transformed human cell type and in primary cells.


Subject(s)
Gene Transfer Techniques , Genome, Human , Genomics/methods , Regulatory Sequences, Nucleic Acid/genetics , Zinc Fingers/genetics , Base Sequence , Cell Line , Endonucleases/genetics , Genetic Loci , Humans , Molecular Sequence Data , Proteomics/methods
14.
Life Sci Alliance ; 4(10)2021 10.
Article in English | MEDLINE | ID: mdl-34446533

ABSTRACT

Glucocorticoids are stress hormones that elicit cellular responses by binding to the glucocorticoid receptor, a ligand-activated transcription factor. The exposure of cells to this hormone induces wide-spread changes in the chromatin landscape and gene expression. Previous studies have suggested that some of these changes are reversible whereas others persist even when the hormone is no longer around. However, when we examined chromatin accessibility in human airway epithelial cells after hormone washout, we found that the hormone-induced changes were universally reversed after 1 d. Moreover, priming of cells by a previous exposure to hormone, in general, did not alter the transcriptional response to a subsequent encounter of the same cue except for one gene, ZBTB16, that displays transcriptional memory manifesting itself as a more robust transcriptional response upon repeated hormone stimulation. Single-cell analysis revealed that the more robust response is driven by a higher probability of primed cells to activate ZBTB16 and by a subset of cells that express the gene at levels that are higher than the induction levels observed for naïve cells.


Subject(s)
Chromatin , Glucocorticoids/metabolism , Signal Transduction , Transcription, Genetic/genetics , A549 Cells , Animals , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , Humans , Rats , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Single-Cell Analysis
15.
Mol Cell Biol ; 27(7): 2442-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17261597

ABSTRACT

Ligand binding to the glucocorticoid receptor (GR) results in receptor binding to glucocorticoid response elements (GREs) and the formation of transcriptional regulatory complexes. Equally important, these complexes are continuously disassembled, with active processes driving GR off GREs. We found that co-chaperone p23-dependent disruption of GR-driven transcription depended on the ligand binding domain (LBD). Next, we examined the importance of the LBD and of ligand dissociation in GR-GRE dissociation in living cells. We showed in fluorescence recovery after photobleaching studies that dissociation of GR from GREs is faster in the absence of the LBD. Furthermore, GR interaction with a target promoter revealed ligand-specific exchange rates. However, using covalently binding ligands, we demonstrated that ligand dissociation is not required for receptor dissociation from GREs. Overall, these studies showed that activities impinging on the LBD regulate GR exchange with GREs but that the dissociation of GR from GREs is independent from ligand dissociation.


Subject(s)
Molecular Chaperones/metabolism , Receptors, Glucocorticoid/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , Cloning, Molecular , Fluorescence Recovery After Photobleaching , Humans , Ligands , Mice , Molecular Chaperones/genetics , Mutation , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Rats , Receptors, Glucocorticoid/genetics , Response Elements
16.
Life Sci Alliance ; 2(2)2019 04.
Article in English | MEDLINE | ID: mdl-30867223

ABSTRACT

The glucocorticoid receptor (GR), a hormone-activated transcription factor, binds to a myriad of genomic binding sites yet seems to regulate a much smaller number of genes. Genome-wide analysis of GR binding and gene regulation has shown that the likelihood of GR-dependent regulation increases with decreased distance of its binding to the transcriptional start site of a gene. To test if we can adopt this knowledge to expand the repertoire of GR target genes, we used CRISPR/Cas-mediated homology-directed repair to add a single GR-binding site directly upstream of the transcriptional start site of each of four genes. To our surprise, we found that the addition of a single GR-binding site can be enough to convert a gene into a GR target. The gain of GR-dependent regulation was observed for two of four genes analyzed and coincided with acquired GR binding at the introduced binding site. However, the gene-specific gain of GR-dependent regulation could not be explained by obvious differences in chromatin accessibility between converted genes and their non-converted counterparts. Furthermore, by introducing GR-binding sequences with different nucleotide compositions, we show that activation can be facilitated by distinct sequences without obvious differences in activity between the GR-binding sequence variants we tested. The approach to use genome engineering to build genomic response elements facilitates the generation of cell lines with tailored repertoires of GR-responsive genes and a framework to test and refine our understanding of the cis-regulatory logic of gene regulation by testing if engineered response elements behave as predicted.


Subject(s)
Gene Editing/methods , Receptors, Glucocorticoid/genetics , Response Elements/genetics , Animals , Binding Sites/genetics , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Gene Expression Regulation , Humans , Protein Binding/genetics , RNA-Seq , Rats , Regulatory Elements, Transcriptional/genetics , Transcription Initiation Site , Transcription, Genetic/genetics , Transcriptional Activation/genetics , Transfection
17.
J Steroid Biochem Mol Biol ; 195: 105485, 2019 12.
Article in English | MEDLINE | ID: mdl-31561002

ABSTRACT

Glucocorticoids (GCs) play an important role in controlling acute graft-versus-host disease (aGvHD), a frequent complication of allogeneic hematopoietic stem cell transplantation. The anti-inflammatory activity of GCs is mainly ascribed to the modulation of T cells and macrophages, for which reason a genetically induced GC resistance of either of these cell types causes aggravated aGvHD. Since only a few genes are currently known that are differentially regulated under these conditions, we analyzed the expression of 54 candidate genes in the inflamed small intestine of mice suffering from aGvHD when either allogeneic T cells or host myeloid cells were GC resistant using a microfluidic dynamic array platform for high-throughput quantitative PCR. The majority of genes categorized as cytokines (e.g. Il2, Il6), chemokines (e.g. Ccl2, Cxcl1), cell surface receptors (e.g. Fasl, Ctla4) and intracellular molecules (e.g. Dusp1, Arg1) were upregulated in mice transplanted with GC resistant allogeneic T cells. Moreover, the expression of several genes linked to energy metabolism (e.g. Glut1) was altered. Surprisingly, mice harboring GC resistant myeloid cells showed almost no changes in gene expression despite their fatal disease course after aGvHD induction. To identify additional genes in the inflamed small intestine that were affected by a GC resistance of allogeneic T cells, we performed an RNAseq analysis, which uncovered more than 500 differentially expressed transcripts (e.g. Cxcr6, Glut3, Otc, Aoc1, Il1r1, Sphk1) that were enriched for biological processes associated with inflammation and tissue disassembly. The changes in gene expression could be confirmed during full-blown disease but hardly any of them in the preclinical phase using high-throughput quantitative PCR. Further analysis of some of these genes revealed a highly selective expression pattern in T cells, intestinal epithelial cells and macrophages, which correlated with their regulation during disease progression. Collectively, we identified an altered gene expression profile caused by GC resistance of transplanted allogeneic T cells, which could help to define new targets for aGvHD therapy.


Subject(s)
Drug Resistance/genetics , Glucocorticoids , Graft vs Host Disease/genetics , Intestine, Small/metabolism , T-Lymphocytes/transplantation , Animals , Graft vs Host Disease/pathology , Intestine, Small/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Transcriptome
18.
Nat Commun ; 10(1): 3477, 2019 08 02.
Article in English | MEDLINE | ID: mdl-31375664

ABSTRACT

Oct4, along with Sox2 and Klf4 (SK), can induce pluripotency but structurally similar factors like Oct6 cannot. To decode why Oct4 has this unique ability, we compare Oct4-binding, accessibility patterns and transcriptional waves with Oct6 and an Oct4 mutant defective in the dimerization with Sox2 (Oct4defSox2). We find that initial silencing of the somatic program proceeds indistinguishably with or without Oct4. Oct6 mitigates the mesenchymal-to-epithelial transition and derails reprogramming. These effects are a consequence of differences in genome-wide binding, as the early binding profile of Oct4defSox2 resembles Oct4, whilst Oct6 does not bind pluripotency enhancers. Nevertheless, in the Oct6-SK condition many otherwise Oct4-bound locations become accessible but chromatin opening is compromised when Oct4defSox2 occupies these sites. We find that Sox2 predominantly facilitates chromatin opening, whilst Oct4 serves an accessory role. Formation of Oct4/Sox2 heterodimers is essential for pluripotency establishment; however, reliance on Oct4/Sox2 heterodimers declines during pluripotency maintenance.


Subject(s)
Cellular Reprogramming/genetics , Chromatin/metabolism , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Epithelial-Mesenchymal Transition/genetics , Fibroblasts , Induced Pluripotent Stem Cells/physiology , Kruppel-Like Factor 4 , Mice, Transgenic , Mutation , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-6/metabolism , Primary Cell Culture , Protein Multimerization/genetics , SOXB1 Transcription Factors/genetics , Time Factors
19.
Genome Biol ; 20(1): 227, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31699133

ABSTRACT

We present the software Condition-specific Regulatory Units Prediction (CRUP) to infer from epigenetic marks a list of regulatory units consisting of dynamically changing enhancers with their target genes. The workflow consists of a novel pre-trained enhancer predictor that can be reliably applied across cell types and species, solely based on histone modification ChIP-seq data. Enhancers are subsequently assigned to different conditions and correlated with gene expression to derive regulatory units. We thoroughly test and then apply CRUP to a rheumatoid arthritis model, identifying enhancer-gene pairs comprising known disease genes as well as new candidate genes.


Subject(s)
Enhancer Elements, Genetic , Software , Animals , Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Chromatin Immunoprecipitation Sequencing , Histone Code , Mice
20.
Mol Cell Biol ; 38(12)2018 06 15.
Article in English | MEDLINE | ID: mdl-29581184

ABSTRACT

The transcription factor GATA2 is required for expansion and differentiation of hematopoietic stem cells (HSCs). In mesenchymal stem cells (MSCs), GATA2 blocks adipogenesis, but its biological relevance and underlying genomic events are unknown. We report a dual function of GATA2 in bone homeostasis. GATA2 in MSCs binds near genes involved in skeletal system development and colocalizes with motifs for FOX and HOX transcription factors, known regulators of skeletal development. Ectopic GATA2 blocks osteoblastogenesis by interfering with SMAD1/5/8 activation. MSC-specific deletion of GATA2 in mice increases the numbers and differentiation capacity of bone-derived precursors, resulting in elevated bone formation. Surprisingly, MSC-specific GATA2 deficiency impairs the trabecularization and mechanical strength of bone, involving reduced MSC expression of the osteoclast inhibitor osteoprotegerin and increased osteoclast numbers. Thus, GATA2 affects bone turnover via MSC-autonomous and indirect effects. By regulating bone trabecularization, GATA2 expression in the osteogenic lineage may contribute to the anatomical and cellular microenvironment of the HSC niche required for hematopoiesis.


Subject(s)
Bone and Bones/metabolism , GATA2 Transcription Factor/genetics , Gene Expression Regulation, Developmental/genetics , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Osteogenesis/genetics , 3T3 Cells , Animals , Binding Sites/genetics , Bone Marrow Cells/metabolism , Cell Differentiation/genetics , Cell Line , Cellular Microenvironment/genetics , Fractures, Bone/genetics , GATA2 Deficiency/genetics , GATA2 Deficiency/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Nuclear Proteins/metabolism , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL