Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 40(7)2020 03 16.
Article in English | MEDLINE | ID: mdl-31932484

ABSTRACT

Peroxisome proliferator-activated receptor γ (PPARγ) is a nuclear receptor essential for adipocyte development and the maintenance of the alternatively polarized macrophage phenotype. Biochemical studies have established that as an obligate heterodimer with retinoid X receptor (RXR), PPARγ binds directly repeated nuclear receptor half sites spaced by one nucleotide (direct repeat 1 [DR1]). However, it has not been analyzed systematically and genome-wide how cis factors such as the sequences of DR1s and adjacent sequences and trans factors such as cobinding lineage-determining transcription factors (LDTFs) contribute to the direct binding of PPARγ in different cellular contexts. We developed a novel motif optimization approach using sequence composition and chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) densities from macrophages and adipocytes to complement de novo motif enrichment analysis and to define and classify high-affinity binding sites. We found that approximately half of the PPARγ cistrome represents direct DNA binding; both half sites can be extended upstream, and these are typically not of equal strength within a DR1. Strategically positioned LDTFs have greater impact on PPARγ binding than the quality of DR1, and the presence of the extension of DR1 provides a remarkable synergy with LDTFs. This approach of considering not only nucleotide frequencies but also their contribution to protein binding in a cellular context is applicable to other transcription factors.


Subject(s)
Adipocytes/cytology , DNA-Binding Proteins/metabolism , Macrophages/cytology , PPAR gamma/metabolism , Retinoid X Receptors/metabolism , Animals , Cells, Cultured , Chromatin Immunoprecipitation , DNA-Binding Proteins/genetics , High-Throughput Nucleotide Sequencing , Male , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , Phosphoproteins/genetics , Repetitive Sequences, Nucleic Acid/genetics , Transcription Factors/genetics
2.
Nucleic Acids Res ; 47(6): 2778-2792, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30799488

ABSTRACT

The concept of tissue-specific gene expression posits that lineage-determining transcription factors (LDTFs) determine the open chromatin profile of a cell via collaborative binding, providing molecular beacons to signal-dependent transcription factors (SDTFs). However, the guiding principles of LDTF binding, chromatin accessibility and enhancer activity have not yet been systematically evaluated. We sought to study these features of the macrophage genome by the combination of experimental (ChIP-seq, ATAC-seq and GRO-seq) and computational approaches. We show that Random Forest and Support Vector Regression machine learning methods can accurately predict chromatin accessibility using the binding patterns of the LDTF PU.1 and four other key TFs of macrophages (IRF8, JUNB, CEBPA and RUNX1). Any of these TFs alone were not sufficient to predict open chromatin, indicating that TF binding is widespread at closed or weakly opened chromatin regions. Analysis of the PU.1 cistrome revealed that two-thirds of PU.1 binding occurs at low accessible chromatin. We termed these sites labelled regulatory elements (LREs), which may represent a dormant state of a future enhancer and contribute to macrophage cellular plasticity. Collectively, our work demonstrates the existence of LREs occupied by various key TFs, regulating specific gene expression programs triggered by divergent macrophage polarizing stimuli.


Subject(s)
Chromatin Assembly and Disassembly/physiology , Macrophages/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism , Animals , Cells, Cultured , Computational Biology , Gene Expression Regulation/physiology , Genome , Machine Learning , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Staining and Labeling/methods , Transcriptional Activation/physiology
3.
Immunity ; 49(4): 615-626.e6, 2018 10 16.
Article in English | MEDLINE | ID: mdl-30332629

ABSTRACT

Macrophages polarize into distinct phenotypes in response to complex environmental cues. We found that the nuclear receptor PPARγ drove robust phenotypic changes in macrophages upon repeated stimulation with interleukin (IL)-4. The functions of PPARγ on macrophage polarization in this setting were independent of ligand binding. Ligand-insensitive PPARγ bound DNA and recruited the coactivator P300 and the architectural protein RAD21. This established a permissive chromatin environment that conferred transcriptional memory by facilitating the binding of the transcriptional regulator STAT6 and RNA polymerase II, leading to robust production of enhancer and mRNAs upon IL-4 re-stimulation. Ligand-insensitive PPARγ binding controlled the expression of an extracellular matrix remodeling-related gene network in macrophages. Expression of these genes increased during muscle regeneration in a mouse model of injury, and this increase coincided with the detection of IL-4 and PPARγ in the affected tissue. Thus, a predominantly ligand-insensitive PPARγ:RXR cistrome regulates progressive and/or reinforcing macrophage polarization.


Subject(s)
Epigenesis, Genetic/immunology , Epigenomics/methods , Gene Expression Regulation/immunology , Macrophage Activation/immunology , Macrophages/immunology , PPAR gamma/immunology , Animals , Cell Line , Cells, Cultured , Interleukin-4/immunology , Interleukin-4/pharmacology , Ligands , Macrophage Activation/genetics , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism
4.
Nucleic Acids Res ; 46(9): 4425-4439, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29506156

ABSTRACT

Retinoid X receptor (RXR) is an obligate heterodimeric partner of several nuclear receptors (NRs), and as such a central component of NR signaling regulating the immune and metabolic phenotype of macrophages. Importantly, the binding motifs of RXR heterodimers are enriched in the tissue-selective open chromatin regions of resident macrophages, suggesting roles in subtype specification. Recent genome-wide studies revealed that RXR binds to thousands of sites in the genome, but the mechanistic details how the cistrome is established and serves ligand-induced transcriptional activity remained elusive. Here we show that IL-4-mediated macrophage plasticity results in a greatly extended RXR cistrome via both direct and indirect actions of the transcription factor STAT6. Activation of STAT6 leads to chromatin remodeling and RXR recruitment to de novo enhancers. In addition, STAT6 triggers a secondary transcription factor wave, including PPARγ. PPARγ appears to be indispensable for the development of RXR-bound de novo enhancers, whose activities can be modulated by the ligands of the PPARγ:RXR heterodimer conferring ligand selective cellular responses. Collectively, these data reveal the mechanisms leading to the dynamic extension of the RXR cistrome and identify the lipid-sensing enhancer sets responsible for the appearance of ligand-preferred gene signatures in alternatively polarized macrophages.


Subject(s)
Interleukin-4/physiology , Macrophages/metabolism , PPAR gamma/metabolism , Retinoid X Receptors/metabolism , STAT6 Transcription Factor/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Gene Regulatory Networks , Ligands , Macrophages/enzymology , Male , Mice, Inbred C57BL , Mice, Knockout , RNA Polymerase II/metabolism , Retinoid X Receptors/genetics , Signal Transduction
5.
Immunity ; 48(1): 75-90.e6, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29343442

ABSTRACT

The molecular basis of signal-dependent transcriptional activation has been extensively studied in macrophage polarization, but our understanding remains limited regarding the molecular determinants of repression. Here we show that IL-4-activated STAT6 transcription factor is required for the direct transcriptional repression of a large number of genes during in vitro and in vivo alternative macrophage polarization. Repression results in decreased lineage-determining transcription factor, p300, and RNA polymerase II binding followed by reduced enhancer RNA expression, H3K27 acetylation, and chromatin accessibility. The repressor function of STAT6 is HDAC3 dependent on a subset of IL-4-repressed genes. In addition, STAT6-repressed enhancers show extensive overlap with the NF-κB p65 cistrome and exhibit decreased responsiveness to lipopolysaccharide after IL-4 stimulus on a subset of genes. As a consequence, macrophages exhibit diminished inflammasome activation, decreased IL-1ß production, and pyroptosis. Thus, the IL-4-STAT6 signaling pathway establishes an alternative polarization-specific epigenenomic signature resulting in dampened macrophage responsiveness to inflammatory stimuli.


Subject(s)
Interleukin-4/metabolism , Macrophages/metabolism , STAT6 Transcription Factor/metabolism , Animals , Blotting, Western , Cell Line , Enhancer Elements, Genetic , Flow Cytometry , Gene Expression Regulation , Inflammasomes/metabolism , Laser Scanning Cytometry , Lipopolysaccharides/pharmacology , Macrophages/physiology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Pyroptosis/genetics , Signal Transduction/genetics , Signal Transduction/physiology
6.
Mol Cell Endocrinol ; 471: 51-62, 2018 08 15.
Article in English | MEDLINE | ID: mdl-28778663

ABSTRACT

Retinoid X Receptors (RXRs) are unique and enigmatic members of the nuclear receptor (NR) family with extensive and complex biological functions in cellular differentiation. On the one hand, RXRs through permissive heterodimerization with other NRs are able to integrate multiple lipid signaling pathways and are believed to play a central role to coordinate the development of the central nervous system. On the other hand, RXRs may have heterodimer-independent functions as well. Therefore, a more RXR-centric analysis is warranted to identify its genomic binding sites and regulated gene networks, which are orchestrating the earliest events in neuronal differentiation. Recently developed genome-wide approaches allow systematic analyses of the RXR-driven neural differentiation. Here we applied next generation sequencing-based methodology to track the dynamic redistribution of the RXR cistrome along the path of embryonic stem cell to glutamatergic neuron differentiation. We identified Retinoic Acid Receptor (RAR) and Liver X Receptor (LXR) as dominant heterodimeric partners of RXR in these cellular stages. Our data presented here characterize the RAR:RXR and LXR:RXR-mediated transcriptional program in embryonic stem cells, neural progenitors and terminally differentiated neurons. Considering the growing evidence for dysregulated RXR-mediated signaling in neurodegenerative disorders, such as Alzheimer's Disease or Amyotrophic Lateral Sclerosis, the data presented here will be also a valuable resource for the field of neuro(patho)biology.


Subject(s)
Cell Lineage/genetics , Gene Expression Regulation , Neurogenesis/genetics , Protein Multimerization , Retinoid X Receptors/metabolism , Transcription, Genetic , Animals , Base Sequence , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Gene Expression Regulation/drug effects , Homeodomain Proteins/metabolism , Ligands , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Neurogenesis/drug effects , Protein Binding/drug effects , Time Factors , Transcription, Genetic/drug effects , Transcriptome/drug effects , Transcriptome/genetics , Tretinoin/pharmacology
7.
Biochim Biophys Acta Gene Regul Mech ; 1861(1): 14-28, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29133016

ABSTRACT

MicroRNAs are important components of the post-transcriptional fine-tuning of macrophage gene expression in physiological and pathological conditions. However, the mechanistic underpinnings and the cis-acting genomic factors of how macrophage polarizing signals induce miRNA expression changes are not well characterized. Therefore, we systematically evaluated the transcriptional basis underlying the inflammation-mediated regulation of macrophage microRNome using the combination of different next generation sequencing datasets. We investigated the LPS-induced expression changes at mature miRNA and pri-miRNA levels in mouse macrophages utilizing a small RNA-seq method and publicly available GRO-seq dataset, respectively. Next, we identified an enhancer set associated with LPS-responsive pri-miRNAs based on publicly available H3K4 mono-methylation-specific ChIP-seq and GRO-seq datasets. This enhancer set was further characterized by the combination of publicly available ChIP and ATAC-seq datasets. Finally, direct interactions between the miR-155-coding genomic region and its distal regulatory elements were identified using a 3C-seq approach. Our analysis revealed 15 robustly LPS-regulated miRNAs at the transcriptional level. In addition, we found that these miRNA genes are associated with an inflammation-responsive enhancer network. Based on NFκB-p65 and JunB transcription factor binding, we showed two distinct enhancer subsets associated with LPS-activated miRNAs that possess distinct epigenetic characteristics and LPS-responsiveness. Finally, our 3C-seq analysis revealed the LPS-induced extensive reorganization of the pri-miR-155-associated functional chromatin domain as well as chromatin loop formation between LPS-responsive enhancers and the promoter region. Our genomic approach successfully combines various genome-wide datasets and allows the identification of the putative regulatory elements controlling miRNA expression in classically activated macrophages.


Subject(s)
Gene Regulatory Networks/genetics , Inflammation/genetics , MicroRNAs/genetics , Transcription, Genetic , Animals , Chromatin/drug effects , Chromatin/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , High-Throughput Nucleotide Sequencing , Humans , Inflammation/chemically induced , Inflammation/pathology , Lipopolysaccharides/toxicity , Mice , Promoter Regions, Genetic/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factor RelA/genetics
8.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(12): 1575-1586, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28774822

ABSTRACT

During cold-exposure 'beige' adipocytes with increased mitochondrial content are activated in white adipose tissue (WAT). These cells, similarly to brown adipose tissue (BAT), dissipate stored chemical energy in the form of heat with the help of uncoupling protein 1 (UCP1). We investigated the effect of tissue transglutaminase (TG2) ablation on the function of ATs in mice. Although TG2+/+ and TG2-/- mice had the same amount of WAT and BAT, we found that TG2+/+ animals could tolerate acute cold exposure for 4h, whereas TG2-/- mice only for 3h. Both TG2-/- and TG2+/+ animals used up half of the triacylglycerol content of subcutaneous WAT (SCAT) after 3h treatment; however, TG2-/- mice still possessed markedly whiter and higher amount of gonadal WAT (GONAT) as reflected in the larger size of adipocytes and lower free fatty acid levels in serum. Furthermore, lower expression of 'beige' marker genes such as UCP1, TBX1 and TNFRFS9 was observed after cold exposure in GONAT of TG2-/- mice, paralleled with a lower level of UCP1 protein and a decreased mitochondrial content. The detected changes in gene expression of Resistin and Adiponectin did not provoke glucose intolerance in the investigated TG2-/- mice, and TG2 deletion did not influence adrenaline, noradrenaline, glucagon and insulin production. Our data suggest that TG2 has a tissue-specific role in GONAT function and browning, which becomes apparent under acute cold exposure.


Subject(s)
Acclimatization , Adipose Tissue, White/metabolism , Cold Temperature , Fatty Acids/metabolism , GTP-Binding Proteins/deficiency , Testis/metabolism , Transglutaminases/deficiency , Adiponectin/biosynthesis , Adiponectin/genetics , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/cytology , Animals , Fatty Acids/genetics , Male , Mice , Mice, Knockout , Protein Glutamine gamma Glutamyltransferase 2 , Resistin/biosynthesis , Resistin/genetics , Testis/cytology
9.
Int J Mol Sci ; 17(8)2016 Jul 30.
Article in English | MEDLINE | ID: mdl-27483259

ABSTRACT

With the increasing number of patients affected with metabolic diseases such as type 2 diabetes, obesity, atherosclerosis and insulin resistance, academic researchers and pharmaceutical companies are eager to better understand metabolic syndrome and develop new drugs for its treatment. Many studies have focused on the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ), which plays a crucial role in adipogenesis and lipid metabolism. These studies have been able to connect this transcription factor to several human metabolic diseases. Due to obvious limitations concerning experimentation in humans, animal models-mainly mouse models-have been generated to investigate the role of PPARγ in different tissues. This review focuses on the metabolic features of human and mouse PPARγ-related diseases and the utility of the mouse as a model.


Subject(s)
Disease Models, Animal , Metabolic Diseases/physiopathology , PPAR gamma/metabolism , Animals , Humans , Mice
10.
Mol Cell ; 63(4): 647-661, 2016 08 18.
Article in English | MEDLINE | ID: mdl-27499297

ABSTRACT

Cell type specification relies on the capacity of undifferentiated cells to properly respond to specific differentiation-inducing signals. Using genomic approaches along with loss- and gain-of-function genetic models, we identified OCT4-dependent mechanisms that provide embryonic stem cells with the means to customize their response to external cues. OCT4 binds a large set of low-accessible genomic regions. At these sites, OCT4 is required for proper enhancer and gene activation by recruiting co-regulators and RAR:RXR or ß-catenin, suggesting an unexpected collaboration between the lineage-determining transcription factor and these differentiation-initiating, signal-dependent transcription factors. As a proof of concept, we demonstrate that overexpression of OCT4 in a kidney cell line is sufficient for signal-dependent activation of otherwise unresponsive genes in these cells. Our results uncover OCT4 as an integral and necessary component of signal-regulated transcriptional processes required for tissue-specific responses.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Wnt Signaling Pathway , Animals , Binding Sites , Cell Differentiation/drug effects , Cellular Reprogramming , Embryonic Stem Cells/drug effects , Gene Expression Regulation , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/drug effects , Promoter Regions, Genetic , RNA Interference , Retinoic Acid Receptor alpha/genetics , Retinoic Acid Receptor alpha/metabolism , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Transcription, Genetic , Transfection , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects
11.
Methods Mol Biol ; 1341: 407-15, 2016.
Article in English | MEDLINE | ID: mdl-25762296

ABSTRACT

Obesity and its comorbidity incidence have increased worldwide during the past 10 years. In consequence, researchers have drawn their attention to the understanding of adipocyte differentiation. Several cellular model systems have been established; however no efficient protocol could be developed so far to differentiate the pluripotent embryonic stem cells to adipocytes. In this chapter, we describe a detailed protocol that is optimized for mouse embryonic stem cells. The result of this differentiation is a homogenous adipocyte monolayer culture that can be used for several applications including developmental and pharmacological research.


Subject(s)
Adipocytes/cytology , Adipogenesis , Cell Culture Techniques/methods , Mouse Embryonic Stem Cells/cytology , Animals , Cell Line , Mice
12.
Eur J Clin Invest ; 45(9): 964-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26251129

ABSTRACT

BACKGROUND: Systems biology approaches have become indispensable tools in biomedical and basic research. These data integrating bioinformatic methods gained prominence after high-throughput technologies became available to investigate complex cellular processes, such as transcriptional regulation and protein-protein interactions, on a scale that had not been studied before. Immunology is one of the medical fields that systems biology impacted profoundly due to the plasticity of cell types involved and the accessibility of a wide range of experimental models. MATERIALS AND METHODS: In this review, we summarize the most important recent genomewide studies exploring the function of peroxisome proliferator-activated receptor γ in macrophages and dendritic cells. PPARγ ChIP-seq experiments were performed in adipocytes derived from embryonic stem cells to complement the existing data sets and to provide comparators to macrophage data. Finally, lists of regulated genes generated from such experiments were analysed with bioinformatics and system biology approaches. RESULTS: We show that genomewide studies utilizing high-throughput data acquisition methods made it possible to gain deeper insights into the role of PPARγ in these immune cell types. We also demonstrate that analysis and visualization of data using network-based approaches can be used to identify novel genes and functions regulated by the receptor. CONCLUSIONS: The example of PPARγ in macrophages and dendritic cells highlights the crucial importance of systems biology approaches in establishing novel cellular functions for long-known signaling pathways.


Subject(s)
Dendritic Cells/immunology , Macrophages/immunology , PPAR gamma/immunology , Systems Biology , Adipocytes/immunology , High-Throughput Nucleotide Sequencing , Humans , PPAR gamma/genetics , Signal Transduction
13.
Stem Cell Res ; 13(1): 88-97, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24858493

ABSTRACT

Adipocyte differentiation and function have become the major research targets due to the increasing interest in obesity and related metabolic conditions. Although, late stages of adipogenesis have been extensively studied, the early phases remain poorly understood. Here we present that supplementing ascorbic acid (AsA) to the adipogenic differentiation cocktail enables the robust and efficient differentiation of mouse embryonic stem cells (mESCs) to mature adipocytes. Such ESC-derived adipocytes mimic the gene-expression profile of subcutaneous isolated adipocytes in vivo remarkably well, much closer than 3T3-L1 derived ones. Moreover, the differentiated cells are in a monolayer, allowing a broad range of genome-wide studies in early and late stages of adipocyte differentiation to be performed.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Ascorbic Acid/pharmacology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Adipocytes/metabolism , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cytological Techniques/methods , Embryonic Stem Cells/metabolism , Mice
14.
Semin Cell Dev Biol ; 24(10-12): 716-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24184382

ABSTRACT

Cellular metabolism is underpinning physiological processes in all cells. These include housekeeping functions as well as specific activities unique to a particular cell type. A growing number of studies in various experimental models indicate that metabolism is tightly connected to embryonic development as well. It is also emerging that metabolic processes have regulatory roles and by changing metabolism, cellular processes and even fates can be influenced. Nuclear receptors (NRs) are transcription factors, responding to changes in metabolites and are implicated in diverse biological processes such as embryonic development, differentiation, metabolism and cancer. Therefore, NRs are key links between metabolism and cell fate decisions. In this review, we introduce ESRRß, DAX-1 and LRH-1 as putative regulators of metabolism in pluripotent embryonic stem cells. We also discuss the role of TR4, NGF1ß, LXRß and RARs in stemness. In addition, we summarize our current understanding of the potential roles of NRs in cancer stem cells.


Subject(s)
Neoplastic Stem Cells/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stem Cells/metabolism , Animals , Humans , Models, Biological , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...