Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Nat Immunol ; 24(11): 1825-1838, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37735593

ABSTRACT

Noncoding genetic variation drives phenotypic diversity, but underlying mechanisms and affected cell types are incompletely understood. Here, investigation of effects of natural genetic variation on the epigenomes and transcriptomes of Kupffer cells derived from inbred mouse strains identified strain-specific environmental factors influencing Kupffer cell phenotypes, including leptin signaling in Kupffer cells from a steatohepatitis-resistant strain. Cell-autonomous and non-cell-autonomous effects of genetic variation were resolved by analysis of F1 hybrid mice and cells engrafted into an immunodeficient host. During homeostasis, non-cell-autonomous trans effects of genetic variation dominated control of Kupffer cells, while strain-specific responses to acute lipopolysaccharide injection were dominated by actions of cis-acting effects modifying response elements for lineage-determining and signal-dependent transcription factors. These findings demonstrate that epigenetic landscapes report on trans effects of genetic variation and serve as a resource for deeper analyses into genetic control of transcription in Kupffer cells and macrophages in vitro.


Subject(s)
Kupffer Cells , Transcriptome , Mice , Animals , Epigenome , Mice, Inbred C57BL , Genetic Variation
2.
Nat Immunol ; 24(7): 1188-1199, 2023 07.
Article in English | MEDLINE | ID: mdl-37322178

ABSTRACT

Spalt-like transcription factor 1 (SALL1) is a critical regulator of organogenesis and microglia identity. Here we demonstrate that disruption of a conserved microglia-specific super-enhancer interacting with the Sall1 promoter results in complete and specific loss of Sall1 expression in microglia. By determining the genomic binding sites of SALL1 and leveraging Sall1 enhancer knockout mice, we provide evidence for functional interactions between SALL1 and SMAD4 required for microglia-specific gene expression. SMAD4 binds directly to the Sall1 super-enhancer and is required for Sall1 expression, consistent with an evolutionarily conserved requirement of the TGFß and SMAD homologs Dpp and Mad for cell-specific expression of Spalt in the Drosophila wing. Unexpectedly, SALL1 in turn promotes binding and function of SMAD4 at microglia-specific enhancers while simultaneously suppressing binding of SMAD4 to enhancers of genes that become inappropriately activated in enhancer knockout microglia, thereby enforcing microglia-specific functions of the TGFß-SMAD signaling axis.


Subject(s)
Microglia , Transcription Factors , Animals , Mice , Binding Sites , DNA , Mice, Knockout , Microglia/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism
3.
Cell ; 173(7): 1796-1809.e17, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29779944

ABSTRACT

Non-coding genetic variation is a major driver of phenotypic diversity and allows the investigation of mechanisms that control gene expression. Here, we systematically investigated the effects of >50 million variations from five strains of mice on mRNA, nascent transcription, transcription start sites, and transcription factor binding in resting and activated macrophages. We observed substantial differences associated with distinct molecular pathways. Evaluating genetic variation provided evidence for roles of ∼100 TFs in shaping lineage-determining factor binding. Unexpectedly, a substantial fraction of strain-specific factor binding could not be explained by local mutations. Integration of genomic features with chromatin interaction data provided evidence for hundreds of connected cis-regulatory domains associated with differences in transcription factor binding and gene expression. This system and the >250 datasets establish a substantial new resource for investigation of how genetic variation affects cellular phenotypes.


Subject(s)
Genetic Variation , Macrophages/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Bone Marrow Cells/cytology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cluster Analysis , Enhancer Elements, Genetic/genetics , Female , Gene Expression Regulation/drug effects , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred NOD , Promoter Regions, Genetic , Protein Binding , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
4.
Nat Immunol ; 21(2): 221-231, 2020 02.
Article in English | MEDLINE | ID: mdl-31959980

ABSTRACT

The lung is inhabited by resident alveolar and interstitial macrophages as well as monocytic cells that survey lung tissues. Each cell type plays distinct functional roles under homeostatic and inflammatory conditions, but mechanisms establishing their molecular identities and functional potential remain poorly understood. In the present study, systematic evaluation of transcriptomes and open chromatin of alveolar macrophages (AMs), interstitial macrophages (IMs) and lung monocytes from two mouse strains enabled inference of common and cell-specific transcriptional regulators. We provide evidence that these factors drive selection of regulatory landscapes that specify distinct phenotypes of AMs and IMs and entrain qualitatively different responses to toll-like receptor 4 signaling in vivo. These studies reveal a striking divergence in a fundamental innate immune response pathway in AMs and establish a framework for further understanding macrophage diversity in the lung.


Subject(s)
Immunity, Innate/immunology , Lung/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Epigenesis, Genetic/immunology , Macrophages/cytology , Mice , Monocytes/cytology , Transcriptome/immunology
5.
Immunity ; 56(9): 2152-2171.e13, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37582369

ABSTRACT

Microglia phenotypes are highly regulated by the brain environment, but the transcriptional networks that specify the maturation of human microglia are poorly understood. Here, we characterized stage-specific transcriptomes and epigenetic landscapes of fetal and postnatal human microglia and acquired corresponding data in induced pluripotent stem cell (iPSC)-derived microglia, in cerebral organoids, and following engraftment into humanized mice. Parallel development of computational approaches that considered transcription factor (TF) co-occurrence and enhancer activity allowed prediction of shared and state-specific gene regulatory networks associated with fetal and postnatal microglia. Additionally, many features of the human fetal-to-postnatal transition were recapitulated in a time-dependent manner following the engraftment of iPSC cells into humanized mice. These data and accompanying computational approaches will facilitate further efforts to elucidate mechanisms by which human microglia acquire stage- and disease-specific phenotypes.


Subject(s)
Induced Pluripotent Stem Cells , Microglia , Humans , Mice , Animals , Gene Regulatory Networks , Brain , Gene Expression Regulation
6.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37751740

ABSTRACT

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Subject(s)
Osteoclasts , RNA , Humans , Mice , Animals , Co-Repressor Proteins/genetics , Osteoclasts/metabolism , RANK Ligand/genetics , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Gene Expression
7.
Immunity ; 52(6): 1057-1074.e7, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32362324

ABSTRACT

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macrophage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms controlling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages during diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These findings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited macrophages to acquire distinct gene expression programs and corresponding functions.


Subject(s)
Cellular Microenvironment/genetics , Cellular Reprogramming/genetics , Epigenesis, Genetic , Gene Expression Regulation , Myeloid Cells/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Biomarkers , Chromatin Immunoprecipitation Sequencing , Diet , Disease Models, Animal , Gene Expression Profiling , Gene Ontology , High-Throughput Nucleotide Sequencing , Kupffer Cells/immunology , Kupffer Cells/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/pathology , Organ Specificity/genetics , Organ Specificity/immunology , Protein Binding , Signal Transduction , Single-Cell Analysis
8.
Cell ; 159(6): 1327-40, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25480297

ABSTRACT

Macrophages reside in essentially all tissues of the body and play key roles in innate and adaptive immune responses. Distinct populations of tissue macrophages also acquire context-specific functions that are important for normal tissue homeostasis. To investigate mechanisms responsible for tissue-specific functions, we analyzed the transcriptomes and enhancer landscapes of brain microglia and resident macrophages of the peritoneal cavity. In addition, we exploited natural genetic variation as a genome-wide "mutagenesis" strategy to identify DNA recognition motifs for transcription factors that promote common or subset-specific binding of the macrophage lineage-determining factor PU.1. We find that distinct tissue environments drive divergent programs of gene expression by differentially activating a common enhancer repertoire and by inducing the expression of divergent secondary transcription factors that collaborate with PU.1 to establish tissue-specific enhancers. These findings provide insights into molecular mechanisms by which tissue environment influences macrophage phenotypes that are likely to be broadly applicable to other cell types.


Subject(s)
Enhancer Elements, Genetic , Macrophages/metabolism , Animals , Genetic Variation , Histone Code , Macrophages/cytology , Macrophages/immunology , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Inbred Strains , Transcription Factors/metabolism
9.
Immunity ; 51(4): 655-670.e8, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31587991

ABSTRACT

Tissue environment plays a powerful role in establishing and maintaining the distinct phenotypes of resident macrophages, but the underlying molecular mechanisms remain poorly understood. Here, we characterized transcriptomic and epigenetic changes in repopulating liver macrophages following acute Kupffer cell depletion as a means to infer signaling pathways and transcription factors that promote Kupffer cell differentiation. We obtained evidence that combinatorial interactions of the Notch ligand DLL4 and transforming growth factor-b (TGF-ß) family ligands produced by sinusoidal endothelial cells and endogenous LXR ligands were required for the induction and maintenance of Kupffer cell identity. DLL4 regulation of the Notch transcriptional effector RBPJ activated poised enhancers to rapidly induce LXRα and other Kupffer cell lineage-determining factors. These factors in turn reprogrammed the repopulating liver macrophage enhancer landscape to converge on that of the original resident Kupffer cells. Collectively, these findings provide a framework for understanding how macrophage progenitor cells acquire tissue-specific phenotypes.


Subject(s)
Kupffer Cells/physiology , Liver/metabolism , Macrophages/physiology , Myeloid Cells/physiology , Animals , Cell Differentiation , Cells, Cultured , Cellular Microenvironment , Cellular Reprogramming , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/cytology , Liver X Receptors/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Signal Transduction , Transforming Growth Factor beta/metabolism
10.
Cell ; 155(1): 200-214, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-24074869

ABSTRACT

Macrophage-mediated inflammation is a major contributor to obesity-associated insulin resistance. The corepressor NCoR interacts with inflammatory pathway genes in macrophages, suggesting that its removal would result in increased activity of inflammatory responses. Surprisingly, we find that macrophage-specific deletion of NCoR instead results in an anti-inflammatory phenotype along with robust systemic insulin sensitization in obese mice. We present evidence that derepression of LXRs contributes to this paradoxical anti-inflammatory phenotype by causing increased expression of genes that direct biosynthesis of palmitoleic acid and ω3 fatty acids. Remarkably, the increased ω3 fatty acid levels primarily inhibit NF-κB-dependent inflammatory responses by uncoupling NF-κB binding and enhancer/promoter histone acetylation from subsequent steps required for proinflammatory gene activation. This provides a mechanism for the in vivo anti-inflammatory insulin-sensitive phenotype observed in mice with macrophage-specific deletion of NCoR. Therapeutic methods to harness this mechanism could lead to a new approach to insulin-sensitizing therapies.


Subject(s)
Fatty Acids, Omega-3/metabolism , Insulin Resistance , Macrophages/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Orphan Nuclear Receptors/genetics , Animals , Liver X Receptors , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1/genetics
11.
Cell ; 151(1): 138-52, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23021221

ABSTRACT

Inflammation and macrophage foam cells are characteristic features of atherosclerotic lesions, but the mechanisms linking cholesterol accumulation to inflammation and LXR-dependent response pathways are poorly understood. To investigate this relationship, we utilized lipidomic and transcriptomic methods to evaluate the effect of diet and LDL receptor genotype on macrophage foam cell formation within the peritoneal cavities of mice. Foam cell formation was associated with significant changes in hundreds of lipid species and unexpected suppression, rather than activation, of inflammatory gene expression. We provide evidence that regulated accumulation of desmosterol underlies many of the homeostatic responses, including activation of LXR target genes, inhibition of SREBP target genes, selective reprogramming of fatty acid metabolism, and suppression of inflammatory-response genes, observed in macrophage foam cells. These observations suggest that macrophage activation in atherosclerotic lesions results from extrinsic, proinflammatory signals generated within the artery wall that suppress homeostatic and anti-inflammatory functions of desmosterol.


Subject(s)
Atherosclerosis/immunology , Cholesterol/biosynthesis , Desmosterol/metabolism , Foam Cells/metabolism , Lipid Metabolism , Transcriptome , Animals , Atherosclerosis/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Fatty Acids/metabolism , Foam Cells/immunology , Gene Knockdown Techniques , Leukocytes, Mononuclear/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 121(2): e2316104121, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38165941

ABSTRACT

The nuclear receptor corepressor (NCoR) forms a complex with histone deacetylase 3 (HDAC3) that mediates repressive functions of unliganded nuclear receptors and other transcriptional repressors by deacetylation of histone substrates. Recent studies provide evidence that NCoR/HDAC3 complexes can also exert coactivator functions in brown adipocytes by deacetylating and activating PPARγ coactivator 1α (PGC1α) and that signaling via receptor activator of nuclear factor kappa-B (RANK) promotes the formation of a stable NCoR/HDAC3/PGC1ß complex that coactivates nuclear factor kappa-B (NFκB)- and activator protein 1 (AP-1)-dependent genes required for osteoclast differentiation. Here, we demonstrate that activation of Toll-like receptor (TLR) 4, but not TLR3, the interleukin 4 (IL4) receptor nor the Type I interferon receptor, also promotes assembly of an NCoR/HDAC3/PGC1ß coactivator complex. Receptor-specific utilization of TNF receptor-associated factor 6 (TRAF6) and downstream activation of extracellular signal-regulated kinase 1 (ERK1) and TANK-binding kinase 1 (TBK1) accounts for the common ability of RANK and TLR4 to drive assembly of an NCoR/HDAC3/PGC1ß complex in macrophages. ERK1, the p65 component of NFκB, and the p300 histone acetyltransferase (HAT) are also components of the induced complex and are associated with local histone acetylation and transcriptional activation of TLR4-dependent enhancers and promoters. These observations identify a TLR4/TRAF6-dependent signaling pathway that converts NCoR from a corepressor of nuclear receptors to a coactivator of NFκB and AP-1 that may be relevant to functions of NCoR in other developmental and homeostatic processes.


Subject(s)
Histones , TNF Receptor-Associated Factor 6 , Transcriptional Activation , Co-Repressor Proteins/genetics , Histones/genetics , Histones/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor AP-1/metabolism , Toll-Like Receptor 4/metabolism , Signal Transduction , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism
13.
Nat Immunol ; 14(9): 893-900, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23959186

ABSTRACT

Intermediates in the cholesterol-biosynthetic pathway and oxysterol derivatives of cholesterol regulate diverse cellular processes. Recent studies have expanded the appreciation of their roles in controlling the functions of cells of the innate and adaptive immune systems. Here we review recent literature reporting on the biological functions of sterol intermediates and oxysterols, acting through transcription factors such as the liver X receptors (LXRs), sterol regulatory element-binding proteins (SREBPs) and the G protein-coupled receptor EBI2, in regulating the differentiation and population expansion of cells of the innate and adaptive immune systems, their responses to inflammatory mediators, their effects on the phagocytic functions of macrophages and their effects on antiviral activities and the migration of immune cells. Such findings have raised many new questions about the production of endogenous bioactive sterols and oxysterols and their mechanisms of action in the immune system.


Subject(s)
Immune System/cytology , Immune System/physiology , Sterols/metabolism , Animals , Humans
14.
Immunity ; 38(1): 106-18, 2013 Jan 24.
Article in English | MEDLINE | ID: mdl-23273843

ABSTRACT

Recent studies suggest that the sterol metabolic network participates in the interferon (IFN) antiviral response. However, the molecular mechanisms linking IFN with the sterol network and the identity of sterol mediators remain unknown. Here we report a cellular antiviral role for macrophage production of 25-hydroxycholesterol (cholest-5-en-3ß,25-diol, 25HC) as a component of the sterol metabolic network linked to the IFN response via Stat1. By utilizing quantitative metabolome profiling of all naturally occurring oxysterols upon infection or IFN-stimulation, we reveal 25HC as the only macrophage-synthesized and -secreted oxysterol. We show that 25HC can act at multiple levels as a potent paracrine inhibitor of viral infection for a broad range of viruses. We also demonstrate, using transcriptional regulatory-network analyses, genetic interventions and chromatin immunoprecipitation experiments that Stat1 directly coupled Ch25h regulation to IFN in macrophages. Our studies describe a physiological role for 25HC as a sterol-lipid effector of an innate immune pathway.


Subject(s)
Antiviral Agents/pharmacology , Hydroxycholesterols/metabolism , Interferons/pharmacology , Macrophages/immunology , Macrophages/metabolism , STAT1 Transcription Factor/metabolism , Animals , Binding Sites , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Bone Marrow Cells/virology , Gene Expression Regulation , Hydroxycholesterols/pharmacology , Liver X Receptors , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages/drug effects , Macrophages/virology , Mevalonic Acid/metabolism , Mice , Orphan Nuclear Receptors/metabolism , Promoter Regions, Genetic , Protein Binding , Steroid Hydroxylases/genetics , Virus Replication/drug effects
15.
Mol Cell ; 51(3): 310-25, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23932714

ABSTRACT

Recent studies suggest a hierarchical model in which lineage-determining factors act in a collaborative manner to select and prime cell-specific enhancers, thereby enabling signal-dependent transcription factors to bind and function in a cell-type-specific manner. Consistent with this model, TLR4 signaling primarily regulates macrophage gene expression through a pre-existing enhancer landscape. However, TLR4 signaling also induces priming of ∼3,000 enhancer-like regions de novo, enabling visualization of intermediates in enhancer selection and activation. Unexpectedly, we find that enhancer transcription precedes local mono- and dimethylation of histone H3 lysine 4 (H3K4me1/2). H3K4 methylation at de novo enhancers is primarily dependent on the histone methyltransferases Mll1, Mll2/4, and Mll3 and is significantly reduced by inhibition of RNA polymerase II elongation. Collectively, these findings suggest an essential role of enhancer transcription in H3K4me1/2 deposition at de novo enhancers that is independent of potential functions of the resulting eRNA transcripts.


Subject(s)
Enhancer Elements, Genetic , Macrophage Activation/genetics , Toll-Like Receptor 4/metabolism , Animals , Base Sequence , CCAAT-Enhancer-Binding Proteins/metabolism , Cells, Cultured , DNA Methylation , Gene Expression , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins/metabolism , RNA Polymerase II/antagonists & inhibitors , Sequence Analysis, DNA , Signal Transduction , Trans-Activators/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 115(20): E4680-E4689, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29632203

ABSTRACT

Activation of liver X receptors (LXRs) with synthetic agonists promotes reverse cholesterol transport and protects against atherosclerosis in mouse models. Most synthetic LXR agonists also cause marked hypertriglyceridemia by inducing the expression of sterol regulatory element-binding protein (SREBP)1c and downstream genes that drive fatty acid biosynthesis. Recent studies demonstrated that desmosterol, an intermediate in the cholesterol biosynthetic pathway that suppresses SREBP processing by binding to SCAP, also binds and activates LXRs and is the most abundant LXR ligand in macrophage foam cells. Here we explore the potential of increasing endogenous desmosterol production or mimicking its activity as a means of inducing LXR activity while simultaneously suppressing SREBP1c-induced hypertriglyceridemia. Unexpectedly, while desmosterol strongly activated LXR target genes and suppressed SREBP pathways in mouse and human macrophages, it had almost no activity in mouse or human hepatocytes in vitro. We further demonstrate that sterol-based selective modulators of LXRs have biochemical and transcriptional properties predicted of desmosterol mimetics and selectively regulate LXR function in macrophages in vitro and in vivo. These studies thereby reveal cell-specific discrimination of endogenous and synthetic regulators of LXRs and SREBPs, providing a molecular basis for dissociation of LXR functions in macrophages from those in the liver that lead to hypertriglyceridemia.


Subject(s)
Biomimetics , Desmosterol/pharmacology , Gene Expression Regulation/drug effects , Hepatocytes/metabolism , Liver X Receptors/metabolism , Macrophages/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Humans , Liver X Receptors/genetics , Macrophages/cytology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 1/genetics
17.
Mol Cell ; 48(1): 28-38, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-22921934

ABSTRACT

Regulation of genes that initiate and amplify inflammatory programs of gene expression is achieved by signal-dependent exchange of coregulator complexes that function to read, write, and erase specific histone modifications linked to transcriptional activation or repression. Here, we provide evidence for the role of trimethylated histone H4 lysine 20 (H4K20me3) as a repression checkpoint that restricts expression of toll-like receptor 4 (TLR4) target genes in macrophages. H4K20me3 is deposited at the promoters of a subset of these genes by the SMYD5 histone methyltransferase through its association with NCoR corepressor complexes. Signal-dependent erasure of H4K20me3 is required for effective gene activation and is achieved by NF-κB-dependent delivery of the histone demethylase PHF2. Liver X receptors antagonize TLR4-dependent gene activation by maintaining NCoR/SMYD5-mediated repression. These findings reveal a histone H4K20 trimethylation/demethylation strategy that integrates positive and negative signaling inputs that control immunity and homeostasis.


Subject(s)
Histones/metabolism , Inflammation/genetics , Inflammation/metabolism , Animals , Cell Line , Co-Repressor Proteins/metabolism , Drosophila/genetics , Drosophila/metabolism , Gene Expression Regulation , HEK293 Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Histones/chemistry , Humans , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Methylation , Mice , Models, Biological , NF-kappa B/metabolism , Promoter Regions, Genetic , Signal Transduction , Toll-Like Receptor 4/metabolism
18.
J Lipid Res ; 56(3): 722-736, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25598080

ABSTRACT

The spectrum of nonalcoholic fatty liver disease (NAFLD) includes steatosis, nonalcoholic steatohepatitis (NASH), and cirrhosis. Recognition and timely diagnosis of these different stages, particularly NASH, is important for both potential reversibility and limitation of complications. Liver biopsy remains the clinical standard for definitive diagnosis. Diagnostic tools minimizing the need for invasive procedures or that add information to histologic data are important in novel management strategies for the growing epidemic of NAFLD. We describe an "omics" approach to detecting a reproducible signature of lipid metabolites, aqueous intracellular metabolites, SNPs, and mRNA transcripts in a double-blinded study of patients with different stages of NAFLD that involves profiling liver biopsies, plasma, and urine samples. Using linear discriminant analysis, a panel of 20 plasma metabolites that includes glycerophospholipids, sphingolipids, sterols, and various aqueous small molecular weight components involved in cellular metabolic pathways, can be used to differentiate between NASH and steatosis. This identification of differential biomolecular signatures has the potential to improve clinical diagnosis and facilitate therapeutic intervention of NAFLD.


Subject(s)
Lipids/blood , Lipids/urine , Non-alcoholic Fatty Liver Disease , Polymorphism, Single Nucleotide , Adult , Biomarkers/metabolism , Biomarkers/urine , Double-Blind Method , Female , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/epidemiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/urine
19.
J Biol Chem ; 288(50): 35812-23, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24189069

ABSTRACT

25-Hydroxycholesterol (25OHC) is an enzymatically derived oxidation product of cholesterol that modulates lipid metabolism and immunity. 25OHC is synthesized in response to interferons and exerts broad antiviral activity by as yet poorly characterized mechanisms. To gain further insights into the basis for antiviral activity, we evaluated time-dependent responses of the macrophage lipidome and transcriptome to 25OHC treatment. In addition to altering specific aspects of cholesterol and sphingolipid metabolism, we found that 25OHC activates integrated stress response (ISR) genes and reprograms protein translation. Effects of 25OHC on ISR gene expression were independent of liver X receptors and sterol-response element-binding proteins and instead primarily resulted from activation of the GCN2/eIF2α/ATF4 branch of the ISR pathway. These studies reveal that 25OHC activates the integrated stress response, which may contribute to its antiviral activity.


Subject(s)
Hydroxycholesterols/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Oxidative Stress/drug effects , Protein Biosynthesis/drug effects , Transcription, Genetic/drug effects , Animals , Bone Marrow Cells/cytology , Cholesterol Esters/metabolism , Gene Expression Profiling , Hydroxycholesterols/metabolism , Liver X Receptors , Macrophages/cytology , Macrophages/virology , Mice , Mice, Inbred C57BL , Muromegalovirus/physiology , Orphan Nuclear Receptors/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingolipids/metabolism , Sterol Regulatory Element Binding Proteins/antagonists & inhibitors
20.
EMBO J ; 29(24): 4223-36, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21045807

ABSTRACT

The macrophage-mediated inflammatory response is a key etiologic component of obesity-related tissue inflammation and insulin resistance. The transcriptional factor FoxO1 is a key regulator of cell metabolism, cell cycle and cell death. Its activity is tightly regulated by the phosphoinositide-3-kinase-AKT (PI3K-Akt) pathway, which leads to phosphorylation, cytoplasmic retention and inactivation of FoxO1. Here, we show that FoxO1 promotes inflammation by enhancing Tlr4-mediated signalling in mature macrophages. By means of chromatin immunoprecipitation (ChIP) combined with massively parallel sequencing (ChIP-Seq), we show that FoxO1 binds to multiple enhancer-like elements within the Tlr4 gene itself, as well as to sites in a number of Tlr4 signalling pathway genes. While FoxO1 potentiates Tlr4 signalling, activation of the latter induces AKT and subsequently inactivates FoxO1, establishing a self-limiting mechanism of inflammation. Given the central role of macrophage Tlr4 in transducing extrinsic proinflammatory signals, the novel functions for FoxO1 in macrophages as a transcriptional regulator of the Tlr4 gene and its inflammatory pathway, highlights FoxO1 as a key molecular adaptor integrating inflammatory responses in the context of obesity and insulin resistance.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Macrophages/immunology , Signal Transduction , Toll-Like Receptor 4/biosynthesis , Animals , Cell Line , Chromatin Immunoprecipitation , DNA/metabolism , Enhancer Elements, Genetic , Forkhead Box Protein O1 , Inflammation Mediators/metabolism , Mice , Protein Binding , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL