Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cell ; 155(7): 1507-20, 2013 Dec 19.
Article in English | MEDLINE | ID: mdl-24360274

ABSTRACT

A key finding of the ENCODE project is that the enhancer landscape of mammalian cells undergoes marked alterations during ontogeny. However, the nature and extent of these changes are unclear. As part of the NIH Mouse Regulome Project, we here combined DNaseI hypersensitivity, ChIP-seq, and ChIA-PET technologies to map the promoter-enhancer interactomes of pluripotent ES cells and differentiated B lymphocytes. We confirm that enhancer usage varies widely across tissues. Unexpectedly, we find that this feature extends to broadly transcribed genes, including Myc and Pim1 cell-cycle regulators, which associate with an entirely different set of enhancers in ES and B cells. By means of high-resolution CpG methylomes, genome editing, and digital footprinting, we show that these enhancers recruit lineage-determining factors. Furthermore, we demonstrate that the turning on and off of enhancers during development correlates with promoter activity. We propose that organisms rely on a dynamic enhancer landscape to control basic cellular functions in a tissue-specific manner.


Subject(s)
B-Lymphocytes/metabolism , Embryonic Stem Cells/metabolism , Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Promoter Regions, Genetic , Regulon , Animals , Cell Lineage , Cells, Cultured , CpG Islands , DNA Methylation , Genetic Techniques , Mice , Organ Specificity , RNA, Long Noncoding/genetics , Transcription Factors/metabolism , Transcription, Genetic
2.
Trends Immunol ; 44(1): 32-43, 2023 01.
Article in English | MEDLINE | ID: mdl-36473794

ABSTRACT

Biological discovery has been driven by advances in throughput and resolution of analysis technologies. They have also created an indelible bias for snapshot-based knowledge. Even though recent methods such as multi-omics single-cell assays have empowered immunological investigations, they still provide snapshots of cellular behaviors and thus, have inherent limitations in reconstructing unsynchronized dynamic events across individual cells. Here, we present a rationale for how NF-κB may convey specificity of contextual information through subtle quantitative features of its signaling dynamics. The next frontier of predictive understanding should involve functional characterization of NF-κB signaling dynamics and their immunological implications. This may help solve the apparent paradox that a ubiquitously activated transcription factor can shape accurate responses to different immune challenges.


Subject(s)
NF-kappa B , Signal Transduction , Humans , NF-kappa B/metabolism , Gene Expression Regulation
3.
Immunity ; 47(2): 298-309.e5, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28801231

ABSTRACT

Despite the widespread use of glucocorticoids (GCs), their anti-inflammatory effects are not understood mechanistically. Numerous investigations have examined the effects of glucocorticoid receptor (GR) activation prior to inflammatory challenges. However, clinical situations are emulated by a GC intervention initiated in the midst of rampant inflammatory responses. To characterize the effects of a late GC treatment, we profiled macrophage transcriptional and chromatinscapes with Dexamethasone (Dex) treatment before or after stimulation by lipopolysaccharide (LPS). The late activation of GR had a similar gene-expression profile as from GR pre-activation, while ameliorating the disruption of metabolic genes. Chromatin occupancy of GR was not predictive of Dex-regulated gene expression, contradicting the "trans-repression by tethering" model. Rather, GR activation resulted in genome-wide blockade of NF-κB interaction with chromatin and directly induced inhibitors of NF-κB and AP-1. Our investigation using GC treatments with clinically relevant timing highlights mechanisms underlying GR actions for modulating the "inflamed epigenome."


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Inflammation/drug therapy , Macrophages/drug effects , Macrophages/immunology , Receptors, Glucocorticoid/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly and Disassembly , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Humans , Inflammation/immunology , Lipopolysaccharides/immunology , Macrophage Activation , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcriptome
4.
Cell ; 146(4): 544-54, 2011 Aug 19.
Article in English | MEDLINE | ID: mdl-21835447

ABSTRACT

The glucocorticoid receptor (GR), like other eukaryotic transcription factors, regulates gene expression by interacting with chromatinized DNA response elements. Photobleaching experiments in living cells indicate that receptors transiently interact with DNA on the time scale of seconds and predict that the response elements may be sparsely occupied on average. Here, we show that the binding of one receptor at the glucocorticoid response element (GRE) does not reduce the steady-state binding of another receptor variant to the same GRE. Mathematical simulations reproduce this noncompetitive state using short GR/GRE residency times and relatively long times between DNA binding events. At many genomic sites where GR binding causes increased chromatin accessibility, concurrent steady-state binding levels for the variant receptor are actually increased, a phenomenon termed assisted loading. Temporally sparse transcription factor-DNA interactions induce local chromatin reorganization, resulting in transient access for binding of secondary regulatory factors.


Subject(s)
Chromatin Assembly and Disassembly , Receptors, Glucocorticoid/metabolism , Response Elements , Adenosine Triphosphate/metabolism , Animals , Cell Line, Tumor , Mammary Tumor Virus, Mouse , Mice , Models, Biological , Monte Carlo Method , Nucleosomes/metabolism , Receptors, Estrogen/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
5.
PLoS Genet ; 18(11): e1010506, 2022 11.
Article in English | MEDLINE | ID: mdl-36441670

ABSTRACT

Short telomeres induce a DNA damage response (DDR) that evokes apoptosis and senescence in human cells. An extant question is the contribution of telomere dysfunction-induced DDR to the phenotypes observed in aging and telomere biology disorders. One candidate is RAP1, a telomere-associated protein that also controls transcription at extratelomeric regions. To distinguish these roles, we generated a knockin mouse carrying a mutated Rap1, which was incapable of binding telomeres and did not result in eroded telomeres or a DDR. Primary Rap1 knockin embryonic fibroblasts showed decreased RAP1 expression and re-localization away from telomeres, with an increased cytosolic distribution akin to that observed in human fibroblasts undergoing telomere erosion. Rap1 knockin mice were viable, but exhibited transcriptomic alterations, proinflammatory cytokine/chemokine signaling, reduced lifespan, and decreased healthspan with increased body weight/fasting blood glucose levels, spontaneous tumor incidence, and behavioral deficits. Taken together, our data present mechanisms distinct from telomere-induced DDR that underlie age-related phenotypes.


Subject(s)
Shelterin Complex , Telomere , Animals , Humans , Mice , Longevity , Phenotype , Telomere/genetics , Telomere Shortening
6.
Nucleic Acids Res ; 50(12): 6702-6714, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35713523

ABSTRACT

The rapid transcriptional response to the transcription factor, glucocorticoid receptor (GR), including gene activation or repression, is mediated by the spatial association of genes with multiple GR binding sites (GBSs) over large genomic distances. However, only a minority of the GBSs have independent GR-mediated activating capacity, and GBSs with independent repressive activity were rarely reported. To understand the positive and negative effects of GR we mapped the regulatory environment of its gene targets. We show that the chromatin interaction networks of GR-activated and repressed genes are spatially separated and vary in the features and configuration of their GBS and other non-GBS regulatory elements. The convergence of the KLF4 pathway in GR-activated domains and the STAT6 pathway in GR-repressed domains, impose opposite transcriptional effects to GR, independent of hormone application. Moreover, the ROR and Rev-erb transcription factors serve as positive and negative regulators, respectively, of GR-mediated gene activation. We found that the spatial crosstalk between GBSs and non-GBSs provides a physical platform for sequestering the Ep300 co-activator from non-GR regulatory loci in both GR-activated and -repressed gene compartments. While this allows rapid gene repression, Ep300 recruitment to GBSs is productive specifically in the activated compartments, thus providing the basis for gene induction.


Subject(s)
E1A-Associated p300 Protein , Gene Expression Regulation , Receptors, Glucocorticoid , Receptors, Glucocorticoid/genetics , Transcriptional Activation/genetics , Cell Line, Tumor , Humans , Animals , Mice , E1A-Associated p300 Protein/metabolism
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.
Immun Ageing ; 19(1): 20, 2022 May 17.
Article in English | MEDLINE | ID: mdl-35581646

ABSTRACT

NF-κB is generally recognized as an important regulator of ageing, through its roles in cellular senescence and inflammatory pathways. Activated in virtually all cell-cell communication networks of the immune system, NF-κB is thought to affect age-related defects of both innate and adaptive immune cells, relevant to inflamm-ageing and declining adaptive immunity, respectively. Moreover, the family of NF-κB proteins that exist as heterodimers and homodimers exert their function beyond the immune system. Given their involvement in diverse areas such as DNA damage to metabolism, NF-κB has the potential to serve as linkages between known hallmarks of ageing. However, the complexity of NF-κB dimer composition, dynamic signaling, and tissue-specific actions has received relatively little attention in ageing research. Here, we discuss some areas where further research may bear fruit in our understanding the impact of NF-κB in healthy ageing and longevity.

9.
J Immunol ; 201(2): 757-771, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29898962

ABSTRACT

Macrophage activation by bacterial LPS leads to induction of a complex inflammatory gene program dependent on numerous transcription factor families. The transcription factor Ikaros has been shown to play a critical role in lymphoid cell development and differentiation; however, its function in myeloid cells and innate immune responses is less appreciated. Using comprehensive genomic analysis of Ikaros-dependent transcription, DNA binding, and chromatin accessibility, we describe unexpected dual repressor and activator functions for Ikaros in the LPS response of murine macrophages. Consistent with the described function of Ikaros as transcriptional repressor, Ikzf1-/- macrophages showed enhanced induction for select responses. In contrast, we observed a dramatic defect in expression of many delayed LPS response genes, and chromatin immunoprecipitation sequencing analyses support a key role for Ikaros in sustained NF-κB chromatin binding. Decreased Ikaros expression in Ikzf1+/- mice and human cells dampens these Ikaros-enhanced inflammatory responses, highlighting the importance of quantitative control of Ikaros protein level for its activator function. In the absence of Ikaros, a constitutively open chromatin state was coincident with dysregulation of LPS-induced chromatin remodeling, gene expression, and cytokine responses. Together, our data suggest a central role for Ikaros in coordinating the complex macrophage transcriptional program in response to pathogen challenge.


Subject(s)
Chromatin/metabolism , Ikaros Transcription Factor/metabolism , Inflammation/immunology , Macrophages/physiology , Animals , Cell Differentiation , Chromatin Assembly and Disassembly , Gene Expression Regulation/immunology , Humans , Ikaros Transcription Factor/genetics , Inflammation/genetics , Lipopolysaccharides/immunology , Mice , Mice, Knockout , Promoter Regions, Genetic , Protein Binding , RAW 264.7 Cells
10.
Nat Methods ; 13(3): 222-228, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26914206

ABSTRACT

High-throughput sequencing technologies have allowed many gene locus-level molecular biology assays to become genome-wide profiling methods. DNA-cleaving enzymes such as DNase I have been used to probe accessible chromatin. The accessible regions contain functional regulatory sites, including promoters, insulators and enhancers. Deep sequencing of DNase-seq libraries and computational analysis of the cut profiles have been used to infer protein occupancy in the genome at the nucleotide level, a method introduced as 'digital genomic footprinting'. The approach has been proposed as an attractive alternative to the analysis of transcription factors (TFs) by chromatin immunoprecipitation followed by sequencing (ChIP-seq), and in theory it should overcome antibody issues, poor resolution and batch effects. Recent reports point to limitations of the DNase-based genomic footprinting approach and call into question the scope of detectable protein occupancy, especially for TFs with short-lived chromatin binding. The genomics community is grappling with issues concerning the utility of genomic footprinting and is reassessing the proposed approaches in terms of robust deliverables. Here we summarize the consensus as well as different views emerging from recent reports, and we describe the remaining issues and hurdles for genomic footprinting.


Subject(s)
Algorithms , Chromosome Mapping/methods , DNA Footprinting/methods , DNA/genetics , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans
11.
Mol Cell ; 43(1): 145-55, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21726817

ABSTRACT

Ligand-dependent transcription by the nuclear receptor glucocorticoid receptor (GR) is mediated by interactions with coregulators. The role of these interactions in determining selective binding of GR to regulatory elements remains unclear. Recent findings indicate that a large fraction of genomic GR binding coincides with chromatin that is accessible prior to hormone treatment, suggesting that receptor binding is dictated by proteins that maintain chromatin in an open state. Combining DNaseI accessibility and chromatin immunoprecipitation with high-throughput sequencing, we identify the activator protein 1 (AP1) as a major partner for productive GR-chromatin interactions. AP1 is critical for GR-regulated transcription and recruitment to co-occupied regulatory elements, illustrating an extensive AP1-GR interaction network. Importantly, the maintenance of baseline chromatin accessibility facilitates GR recruitment and is dependent on AP1 binding. We propose a model in which the basal occupancy of transcription factors acts to prime chromatin and direct inducible transcription factors to select regions in the genome.


Subject(s)
Chromatin/metabolism , Models, Genetic , Receptors, Glucocorticoid/metabolism , Transcription Factor AP-1/physiology , Animals , Binding Sites , Cell Line , Chromatin/chemistry , Gene Expression Regulation , Genome , Ligands , Mice , Receptors, Glucocorticoid/chemistry , Regulatory Elements, Transcriptional , Transcription Factor AP-1/chemistry
12.
Genome Res ; 25(2): 213-25, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25373146

ABSTRACT

Mitosis entails global alterations to chromosome structure and nuclear architecture, concomitant with transient silencing of transcription. How cells transmit transcriptional states through mitosis remains incompletely understood. While many nuclear factors dissociate from mitotic chromosomes, the observation that certain nuclear factors and chromatin features remain associated with individual loci during mitosis originated the hypothesis that such mitotically retained molecular signatures could provide transcriptional memory through mitosis. To understand the role of chromatin structure in mitotic memory, we performed the first genome-wide comparison of DNase I sensitivity of chromatin in mitosis and interphase, using a murine erythroblast model. Despite chromosome condensation during mitosis visible by microscopy, the landscape of chromatin accessibility at the macromolecular level is largely unaltered. However, mitotic chromatin accessibility is locally dynamic, with individual loci maintaining none, some, or all of their interphase accessibility. Mitotic reduction in accessibility occurs primarily within narrow, highly DNase hypersensitive sites that frequently coincide with transcription factor binding sites, whereas broader domains of moderate accessibility tend to be more stable. In mitosis, proximal promoters generally maintain their accessibility more strongly, whereas distal regulatory elements tend to lose accessibility. Large domains of DNA hypomethylation mark a subset of promoters that retain accessibility during mitosis and across many cell types in interphase. Erythroid transcription factor GATA1 exerts site-specific changes in interphase accessibility that are most pronounced at distal regulatory elements, but has little influence on mitotic accessibility. We conclude that features of open chromatin are remarkably stable through mitosis, but are modulated at the level of individual genes and regulatory elements.


Subject(s)
Chromatin Assembly and Disassembly , Chromosomes , Genome , Mitosis/genetics , Animals , Binding Sites , Cell Cycle/genetics , Cell Differentiation/genetics , Chromatin Immunoprecipitation , Computational Biology , DNA Methylation , Deoxyribonuclease I/metabolism , Erythroid Cells/cytology , Erythroid Cells/metabolism , GATA1 Transcription Factor/metabolism , High-Throughput Nucleotide Sequencing , Interphase/genetics , Mice , Mitosis/drug effects , Promoter Regions, Genetic , Protein Binding , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Transcription, Genetic
13.
Genome Res ; 25(6): 845-57, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25677181

ABSTRACT

Although physiological steroid levels are often pulsatile (ultradian), the genomic effects of this pulsatility are poorly understood. By utilizing glucocorticoid receptor (GR) signaling as a model system, we uncovered striking spatiotemporal relationships between receptor loading, lifetimes of the DNase I hypersensitivity sites (DHSs), long-range interactions, and gene regulation. We found that hormone-induced DHSs were enriched within ± 50 kb of GR-responsive genes and displayed a broad spectrum of lifetimes upon hormone withdrawal. These lifetimes dictate the strength of the DHS interactions with gene targets and contribute to gene regulation from a distance. Our results demonstrate that pulsatile and constant hormone stimulations induce unique, treatment-specific patterns of gene and regulatory element activation. These modes of activation have implications for corticosteroid function in vivo and for steroid therapies in various clinical settings.


Subject(s)
Chromatin/genetics , Glucocorticoids/pharmacology , Response Elements , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Chromatin/metabolism , Chromatin Immunoprecipitation , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Gene Expression Regulation , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Perilipin-4 , Protein Binding , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Sequence Analysis, DNA
14.
Bioessays ; 38(12): 1197-1208, 2016 12.
Article in English | MEDLINE | ID: mdl-27767214

ABSTRACT

Nuclear bodies contribute to non-random organization of the human genome and nuclear function. Using a major prototypical nuclear body, the Cajal body, as an example, we suggest that these structures assemble at specific gene loci located across the genome as a result of high transcriptional activity. Subsequently, target genes are physically clustered in close proximity in Cajal body-containing cells. However, Cajal bodies are observed in only a limited number of human cell types, including neuronal and cancer cells. Ultimately, Cajal body depletion perturbs splicing kinetics by reducing target small nuclear RNA (snRNA) transcription and limiting the levels of spliceosomal snRNPs, including their modification and turnover following each round of RNA splicing. As such, Cajal bodies are capable of shaping the chromatin interaction landscape and the transcriptome by influencing spliceosome kinetics. Future studies should concentrate on characterizing the direct influence of Cajal bodies upon snRNA gene transcriptional dynamics. Also see the video abstract here.


Subject(s)
Coiled Bodies/genetics , Genome, Human , Spliceosomes , Transcriptome , Coiled Bodies/metabolism , Humans
15.
Genome Res ; 23(3): 462-72, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23212947

ABSTRACT

Higher-order genome organization shows tissue-specific patterns. However, functional relevance and the mechanisms shaping the genome architecture are poorly understood. Here we report a profound shift from promiscuous to highly selective genome organization that accompanies the effector lineage choice of differentiating T cells. As multipotent naive cells receive antigenic signals and commit to a T helper (Th) pathway, the genome-wide contacts of a lineage-specific cytokine locus are preferentially enriched for functionally relevant genes. Despite the establishment of divergent interactomes and global reprogramming of transcription in Th1 versus Th2, the overall expression status of the contact genes is surprisingly similar between the two lineages. Importantly, during differentiation, the genomic contacts are retained and strengthened precisely at DNA binding sites of the specific lineage-determining STAT transcription factor. In cells from the specific STAT knock-out mouse, the signature cytokine locus is unable to shed the promiscuous contacts established in the naive T cells, indicating the importance of genomic STAT binding. Altogether, the global aggregation of STAT binding loci from genic and nongenic regions highlights a new role for differentiation-promoting transcription factors in direct specification of higher-order nuclear architecture through interacting with regulatory regions. Such subnuclear environments have significant implications for efficient functioning of the mature effector lymphocytes.


Subject(s)
Cell Differentiation , STAT4 Transcription Factor/metabolism , Th1 Cells/cytology , Th2 Cells/cytology , Animals , Binding Sites , Cell Lineage/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chromosomes/genetics , Chromosomes/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Library , Genetic Loci , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , STAT4 Transcription Factor/genetics
16.
Mol Cell ; 29(5): 611-24, 2008 Mar 14.
Article in English | MEDLINE | ID: mdl-18342607

ABSTRACT

The generality and spectrum of chromatin-remodeling requirements for nuclear receptor function are unknown. We have characterized glucocorticoid receptor (GR) binding events and chromatin structural transitions across GR-induced or -repressed genes. This analysis reveals that GR binding invariably occurs at nuclease-accessible sites (DHS). A remarkable diversity of mechanisms, however, render these sites available for GR binding. Accessibility of the GR binding sites is either constitutive or hormone inducible. Within each category, some DHS sites require the Brg1-containing Swi/Snf complex, but others are Brg1 independent, implicating a different remodeling complex. The H2A.Z histone variant is highly enriched at both inducible and constitutive DHS sites and is subject to exchange during hormone activation. The DHS profile is highly cell specific, implicating cell-selective organization of the chromatin landscape as a critical determinant of tissue-selective receptor function. Furthermore, the widespread requirement for chromatin remodeling supports the recent hypothesis that the rapid exchange of receptor proteins occurs during nucleosome reorganization.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Receptors, Glucocorticoid/metabolism , Animals , Cell Line , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Profiling , Histones/genetics , Histones/metabolism , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleoproteins/genetics , Nucleoproteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding , RNA/genetics , RNA/metabolism , Receptors, Glucocorticoid/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
17.
EMBO J ; 30(8): 1459-72, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21427703

ABSTRACT

Adipogenesis is tightly controlled by a complex network of transcription factors acting at different stages of differentiation. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein (C/EBP) family members are key regulators of this process. We have employed DNase I hypersensitive site analysis to investigate the genome-wide changes in chromatin structure that accompany the binding of adipogenic transcription factors. These analyses revealed a dramatic and dynamic modulation of the chromatin landscape during the first hours of adipocyte differentiation that coincides with cooperative binding of multiple early transcription factors (including glucocorticoid receptor, retinoid X receptor, Stat5a, C/EBPß and -δ) to transcription factor 'hotspots'. Our results demonstrate that C/EBPß marks a large number of these transcription factor 'hotspots' before induction of differentiation and chromatin remodelling and is required for their establishment. Furthermore, a subset of early remodelled C/EBP-binding sites persists throughout differentiation and is later occupied by PPARγ, indicating that early C/EBP family members, in addition to their well-established role in activation of PPARγ transcription, may act as pioneering factors for PPARγ binding.


Subject(s)
Adipogenesis/physiology , Chromatin Assembly and Disassembly , Transcription Factors/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Algorithms , Animals , Blotting, Western , CCAAT-Enhancer-Binding Protein-alpha/genetics , CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation , Chromatin Immunoprecipitation , Mice , RNA, Messenger/genetics , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Transcription Factors/genetics , Transcription, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
18.
EMBO J ; 30(15): 3028-39, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21701563

ABSTRACT

Cell-selective glucocorticoid receptor (GR) binding to distal regulatory elements is associated with cell type-specific regions of locally accessible chromatin. These regions can either pre-exist in chromatin (pre-programmed) or be induced by the receptor (de novo). Mechanisms that create and maintain these sites are not well understood. We observe a global enrichment of CpG density for pre-programmed elements, and implicate their demethylated state in the maintenance of open chromatin in a tissue-specific manner. In contrast, sites that are actively opened by GR (de novo) are characterized by low CpG density, and form a unique class of enhancers devoid of suppressive effect of agglomerated methyl-cytosines. Furthermore, treatment with glucocorticoids induces rapid changes in methylation levels at selected CpGs within de novo sites. Finally, we identify GR-binding elements with CpGs at critical positions, and show that methylation can affect GR-DNA interactions in vitro. The findings present a unique link between tissue-specific chromatin accessibility, DNA methylation and transcription factor binding and show that DNA methylation can be an integral component of gene regulation by nuclear receptors.


Subject(s)
DNA Methylation , DNA/metabolism , Enhancer Elements, Genetic , Receptors, Glucocorticoid/metabolism , Animals , Cell Line , Chromatin/metabolism , Mice , Protein Binding
19.
Genome Res ; 21(5): 697-706, 2011 May.
Article in English | MEDLINE | ID: mdl-21471403

ABSTRACT

The spatial organization of genes in the interphase nucleus plays an important role in establishment and regulation of gene expression. Contradicting results have been reported to date, with little consensus about the dynamics of nuclear organization and the features of the contact loci. In this study, we investigated the properties and dynamics of genomic loci that are in contact with glucocorticoid receptor (GR)-responsive loci. We took a systematic approach, combining genome-wide interaction profiling by the chromosome conformation capture on chip (4C) technology with expression, protein occupancy, and chromatin accessibility profiles. This approach allowed a comprehensive analysis of how distinct features of the linear genome are organized in the three-dimensional nuclear space in the context of rapid gene regulation. We found that the transcriptional response to GR occurs without dramatic nuclear reorganization. Moreover, contrary to the view of transcription-driven organization, even genes with opposite transcriptional responses colocalize. Regions contacting GR-regulated genes are not particularly enriched for GR-regulated loci or for any functional group of genes, suggesting that these subnuclear environments are not organized to respond to a specific factor. The contact regions are, however, highly enriched for DNase I-hypersensitive sites that comprehensively mark cell-type-specific regulatory sites. These findings indicate that the nucleus is pre-organized in a conformation allowing rapid transcriptional reprogramming, and this organization is significantly correlated with cell-type-specific chromatin sites accessible to regulatory factors. Numerous open chromatin loci may be arranged in nuclear domains that are poised to respond to diverse signals in general and to permit efficient gene regulation.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation , Proteins/genetics , Receptors, Glucocorticoid/chemistry , Receptors, Glucocorticoid/metabolism , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Line, Tumor , Cell Nucleus/genetics , Chromatin/metabolism , Dexamethasone/pharmacology , Epithelial Cells/chemistry , Epithelial Cells/ultrastructure , Mice , Oligonucleotide Array Sequence Analysis , Proteins/metabolism , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/genetics , Transcription, Genetic
20.
Cell Rep ; 43(3): 113940, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38483906

ABSTRACT

Individual cell sensing of external cues has evolved through the temporal patterns in signaling. Since nuclear factor κB (NF-κB) signaling dynamics have been examined using a single subunit, RelA, it remains unclear whether more information might be transmitted via other subunits. Using NF-κB double-knockin reporter mice, we monitored both canonical NF-κB subunits, RelA and c-Rel, simultaneously in single macrophages by quantitative live-cell imaging. We show that signaling features of RelA and c-Rel convey more information about the stimuli than those of either subunit alone. Machine learning is used to predict the ligand identity accurately based on RelA and c-Rel signaling features without considering the co-activated factors. Ligand discrimination is achieved through selective non-redundancy of RelA and c-Rel signaling dynamics, as well as their temporal coordination. These results suggest a potential role of c-Rel in fine-tuning immune responses and highlight the need for approaches that will elucidate the mechanisms regulating NF-κB subunit specificity.


Subject(s)
NF-kappa B , Proto-Oncogene Proteins c-rel , Mice , Animals , NF-kappa B/metabolism , Ligands , Proto-Oncogene Proteins c-rel/metabolism , Transcription Factor RelA/metabolism , Signal Transduction , Macrophages/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL