Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Nat Immunol ; 18(10): 1104-1116, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28825701

ABSTRACT

Cross-regulation of Toll-like receptor (TLR) responses by cytokines is essential for effective host defense, avoidance of toxicity and homeostasis, but the underlying mechanisms are not well understood. Our comprehensive epigenomics approach to the analysis of human macrophages showed that the proinflammatory cytokines TNF and type I interferons induced transcriptional cascades that altered chromatin states to broadly reprogram responses induced by TLR4. TNF tolerized genes encoding inflammatory molecules to prevent toxicity while preserving the induction of genes encoding antiviral and metabolic molecules. Type I interferons potentiated the inflammatory function of TNF by priming chromatin to prevent the silencing of target genes of the transcription factor NF-κB that encode inflammatory molecules. The priming of chromatin enabled robust transcriptional responses to weak upstream signals. Similar chromatin regulation occurred in human diseases. Our findings reveal that signaling crosstalk between interferons and TNF is integrated at the level of chromatin to reprogram inflammatory responses, and identify previously unknown functions and mechanisms of action of these cytokines.


Subject(s)
Epigenesis, Genetic , Inflammation/etiology , Inflammation/metabolism , Interferon Type I/metabolism , Macrophages/immunology , Macrophages/metabolism , Tumor Necrosis Factor-alpha/metabolism , Binding Sites , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Cluster Analysis , Computational Biology/methods , Cytokines/genetics , Cytokines/metabolism , Epigenomics/methods , Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Lipopolysaccharides/immunology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , Monocytes/immunology , Monocytes/metabolism , Nucleotide Motifs , Promoter Regions, Genetic , Protein Binding , Protein Transport , Signal Transduction , Toll-Like Receptor 4/metabolism , Transcription Factors/metabolism
2.
Immunity ; 51(2): 241-257.e9, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31303399

ABSTRACT

Cytokine tumor necrosis factor (TNF)-mediated macrophage polarization is important for inflammatory disease pathogenesis, but the mechanisms regulating polarization are not clear. We performed transcriptomic and epigenomic analysis of the TNF response in primary human macrophages and revealed late-phase activation of SREBP2, the master regulator of cholesterol biosynthesis genes. TNF stimulation extended the genomic profile of SREBP2 occupancy to include binding to and activation of inflammatory and interferon response genes independently of its functions in sterol metabolism. Genetic ablation of SREBP function shifted the balance of macrophage polarization from an inflammatory to a reparative phenotype in peritonitis and skin wound healing models. Genetic ablation of SREBP activity in myeloid cells or topical pharmacological inhibition of SREBP improved skin wound healing under homeostatic and chronic inflammatory conditions. Our results identify a function and mechanism of action for SREBPs in augmenting TNF-induced macrophage activation and inflammation and open therapeutic avenues for promoting wound repair.


Subject(s)
Inflammation/metabolism , Macrophages/immunology , Peritonitis/metabolism , Receptors, G-Protein-Coupled/metabolism , Skin Diseases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Differentiation , Cells, Cultured , Disease Models, Animal , Epigenomics , Female , Humans , Macrophage Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA, Small Interfering/genetics , Receptors, G-Protein-Coupled/genetics , Transcriptome , Wound Healing
3.
Immunity ; 47(2): 235-250.e4, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813657

ABSTRACT

Mechanisms by which interferon (IFN)-γ activates genes to promote macrophage activation are well studied, but little is known about mechanisms and functions of IFN-γ-mediated gene repression. We used an integrated transcriptomic and epigenomic approach to analyze chromatin accessibility, histone modifications, transcription-factor binding, and gene expression in IFN-γ-primed human macrophages. IFN-γ suppressed basal expression of genes corresponding to an "M2"-like homeostatic and reparative phenotype. IFN-γ repressed genes by suppressing the function of enhancers enriched for binding by transcription factor MAF. Mechanistically, IFN-γ disassembled a subset of enhancers by inducing coordinate suppression of binding by MAF, lineage-determining transcription factors, and chromatin accessibility. Genes associated with MAF-binding enhancers were suppressed in macrophages isolated from rheumatoid-arthritis patients, revealing a disease-associated signature of IFN-γ-mediated repression. These results identify enhancer inactivation and disassembly as a mechanism of IFN-γ-mediated gene repression and reveal that MAF regulates the macrophage enhancer landscape and is suppressed by IFN-γ to augment macrophage activation.


Subject(s)
Arthritis, Rheumatoid/immunology , Chromatin Assembly and Disassembly , Interferon-gamma/metabolism , Macrophages/immunology , Proto-Oncogene Proteins c-maf/metabolism , Cell Differentiation , Cell Lineage , Cells, Cultured , Cytokines/metabolism , Enhancer Elements, Genetic/genetics , Gene Expression Regulation , Histones/metabolism , Humans , Protein Binding , Proto-Oncogene Proteins c-maf/genetics , Transcriptome
4.
Immunity ; 47(1): 66-79.e5, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723554

ABSTRACT

Hypoxia augments inflammatory responses and osteoclastogenesis by incompletely understood mechanisms. We identified COMMD1 as a cell-intrinsic negative regulator of osteoclastogenesis that is suppressed by hypoxia. In human macrophages, COMMD1 restrained induction of NF-κB signaling and a transcription factor E2F1-dependent metabolic pathway by the cytokine RANKL. Downregulation of COMMD1 protein expression by hypoxia augmented RANKL-induced expression of inflammatory and E2F1 target genes and downstream osteoclastogenesis. E2F1 targets included glycolysis and metabolic genes including CKB that enabled cells to meet metabolic demands in challenging environments, as well as inflammatory cytokine-driven target genes. Expression quantitative trait locus analysis linked increased COMMD1 expression with decreased bone erosion in rheumatoid arthritis. Myeloid deletion of Commd1 resulted in increased osteoclastogenesis in arthritis and inflammatory osteolysis models. These results identify COMMD1 and an E2F-metabolic pathway as key regulators of osteoclastogenic responses under pathological inflammatory conditions and provide a mechanism by which hypoxia augments inflammation and bone destruction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Arthritis, Rheumatoid/immunology , Macrophages/immunology , Osteogenesis/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cells, Cultured , Disease Models, Animal , E2F1 Transcription Factor/metabolism , Female , Humans , Hypoxia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , NF-kappa B/metabolism , RNA, Small Interfering/genetics , Signal Transduction
6.
Mol Ther ; 31(2): 435-453, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36184851

ABSTRACT

Treating osteoporosis and associated bone fractures remains challenging for drug development in part due to potential off-target side effects and the requirement for long-term treatment. Here, we identify recombinant adeno-associated virus (rAAV)-mediated gene therapy as a complementary approach to existing osteoporosis therapies, offering long-lasting targeting of multiple targets and/or previously undruggable intracellular non-enzymatic targets. Treatment with a bone-targeted rAAV carrying artificial microRNAs (miRNAs) silenced the expression of WNT antagonists, schnurri-3 (SHN3), and sclerostin (SOST), and enhanced WNT/ß-catenin signaling, osteoblast function, and bone formation. A single systemic administration of rAAVs effectively reversed bone loss in both postmenopausal and senile osteoporosis. Moreover, the healing of bone fracture and critical-sized bone defects was also markedly improved by systemic injection or transplantation of AAV-bound allograft bone to the osteotomy sites. Collectively, our data demonstrate the clinical potential of bone-specific gene silencers to treat skeletal disorders of low bone mass and impaired fracture repair.


Subject(s)
Fractures, Bone , Osteoporosis , Humans , Adaptor Proteins, Signal Transducing/genetics , Osteoporosis/genetics , Osteoporosis/therapy , Fractures, Bone/genetics , Fractures, Bone/therapy , Bone and Bones , Genetic Therapy
7.
Nat Immunol ; 12(7): 607-15, 2011 May 22.
Article in English | MEDLINE | ID: mdl-21602809

ABSTRACT

Endotoxin tolerance, a key mechanism for suppressing excessive inflammatory cytokine production, is induced by prior exposure of macrophages to Toll-like receptor (TLR) ligands. Induction of cross-tolerance to endotoxin by endogenous cytokines has not been investigated. Here we show that prior exposure to tumor necrosis factor (TNF) induced a tolerant state in macrophages, with less cytokine production after challenge with lipopolysaccharide (LPS) and protection from LPS-induced death. TNF-induced cross-tolerization was mediated by suppression of LPS-induced signaling and chromatin remodeling. TNF-induced cross-tolerance was dependent on the kinase GSK3, which suppressed chromatin accessibility and promoted rapid termination of signaling via the transcription factor NF-κB by augmenting negative feedback by the signaling inhibitors A20 and IκBα. Our results demonstrate an unexpected homeostatic function for TNF and a GSK3-mediated mechanism for the prevention of prolonged and excessive inflammation.


Subject(s)
Endotoxins/immunology , Glycogen Synthase Kinase 3/immunology , Macrophages/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Chromatin Assembly and Disassembly/drug effects , Chromatin Assembly and Disassembly/immunology , Cysteine Endopeptidases/immunology , Cysteine Endopeptidases/metabolism , Cytokines/biosynthesis , Cytokines/immunology , Female , I-kappa B Proteins/immunology , I-kappa B Proteins/metabolism , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/immunology , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha , NF-kappa B/immunology , NF-kappa B/metabolism , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3
8.
Nature ; 526(7571): 112-7, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26367794

ABSTRACT

The extent to which low-frequency (minor allele frequency (MAF) between 1-5%) and rare (MAF ≤ 1%) variants contribute to complex traits and disease in the general population is mainly unknown. Bone mineral density (BMD) is highly heritable, a major predictor of osteoporotic fractures, and has been previously associated with common genetic variants, as well as rare, population-specific, coding variants. Here we identify novel non-coding genetic variants with large effects on BMD (ntotal = 53,236) and fracture (ntotal = 508,253) in individuals of European ancestry from the general population. Associations for BMD were derived from whole-genome sequencing (n = 2,882 from UK10K (ref. 10); a population-based genome sequencing consortium), whole-exome sequencing (n = 3,549), deep imputation of genotyped samples using a combined UK10K/1000 Genomes reference panel (n = 26,534), and de novo replication genotyping (n = 20,271). We identified a low-frequency non-coding variant near a novel locus, EN1, with an effect size fourfold larger than the mean of previously reported common variants for lumbar spine BMD (rs11692564(T), MAF = 1.6%, replication effect size = +0.20 s.d., Pmeta = 2 × 10(-14)), which was also associated with a decreased risk of fracture (odds ratio = 0.85; P = 2 × 10(-11); ncases = 98,742 and ncontrols = 409,511). Using an En1(cre/flox) mouse model, we observed that conditional loss of En1 results in low bone mass, probably as a consequence of high bone turnover. We also identified a novel low-frequency non-coding variant with large effects on BMD near WNT16 (rs148771817(T), MAF = 1.2%, replication effect size = +0.41 s.d., Pmeta = 1 × 10(-11)). In general, there was an excess of association signals arising from deleterious coding and conserved non-coding variants. These findings provide evidence that low-frequency non-coding variants have large effects on BMD and fracture, thereby providing rationale for whole-genome sequencing and improved imputation reference panels to study the genetic architecture of complex traits and disease in the general population.


Subject(s)
Bone Density/genetics , Fractures, Bone/genetics , Genome, Human/genetics , Homeodomain Proteins/genetics , Animals , Bone and Bones/metabolism , Disease Models, Animal , Europe/ethnology , Exome/genetics , Female , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Genomics , Genotype , Humans , Mice , Sequence Analysis, DNA , White People/genetics , Wnt Proteins/genetics
9.
Nat Immunol ; 9(4): 378-87, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345002

ABSTRACT

Rapid induction of inflammatory genes by tumor necrosis factor (TNF) has been well studied, but little is known about delayed and chronic TNF responses. Here we investigated the kinetics of primary macrophage responses to TNF and discovered that TNF initiates an interferon-beta-mediated autocrine loop that sustains expression of inflammatory genes and induces delayed expression of interferon-response genes such as those encoding the transcription factors STAT1 and IRF7, which enhance macrophage responses to stimulation of cytokines and Toll-like receptors. TNF-induced interferon-beta production depended on interferon-response factor 1, and downstream gene expression was mediated by synergy between small amounts of interferon-beta and canonical TNF-induced signals. Thus, TNF activates a 'feed-forward' loop that sustains inflammation but avoids the potential toxicity associated with the high interferon production induced by stimulation of Toll-like receptors.


Subject(s)
Autocrine Communication/immunology , Chemokines/biosynthesis , Chemokines/genetics , Gene Expression Regulation/immunology , Interferon Regulatory Factor-1/metabolism , Interferon Type I/genetics , STAT1 Transcription Factor/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Humans , Inflammation Mediators/physiology , Interferon Regulatory Factor-1/physiology , Interferon Type I/biosynthesis , Interferon Type I/physiology , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/physiology , Macrophages/metabolism , Macrophages/pathology , Mice , Time Factors
10.
Nat Immunol ; 9(2): 186-93, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18084294

ABSTRACT

Immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors modulate the amplitude and nature of macrophage responses to Toll-like receptor and cytokine receptor stimulation. However, the molecular mechanisms enabling this receptor crosstalk are not known. Here we investigated the function of the calcium-dependent kinases CaMK and Pyk2 'downstream' of ITAM-associated receptors in the regulation of cytokine-induced activation of Jak kinases and STAT transcription factors. CaMK and Pyk2 relayed signals from integrins and the ITAM-containing adaptor DAP12 to augment interleukin 10- and interferon-alpha-induced Jak activation and STAT1-dependent gene expression. CaMK inhibition suppressed STAT1-mediated interferon-alpha signaling in a mouse model of systemic lupus erythematosus. Our results associate Pyk2 and Jak kinases with the linkage of signals emanating from cytokine and heterologous ITAM-dependent receptors.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Focal Adhesion Kinase 2/metabolism , Interferon Type I/pharmacology , Janus Kinases/metabolism , Macrophages/drug effects , STAT1 Transcription Factor/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium/metabolism , Cells, Cultured , Humans , Macrophages/immunology , Membrane Proteins , Mice , Phosphorylation , Receptors, Immunologic/metabolism , Signal Transduction , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL