Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Publication year range
1.
Cell ; 140(6): 791-7, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20303870

ABSTRACT

The chronic autoimmune diseases include multiple complex genetic disorders. Recently, genome-wide association studies (GWAS) have identified a large number of major loci, with many associations shared between various autoimmune diseases. These associations highlight key roles for lymphocyte activation and prioritize specific cytokine pathways and mechanisms of host-microbe recognition. Despite success in identifying loci, comprehensive models of disease pathogenesis are currently lacking. Future efforts comparing association patterns between autoimmune diseases may be particularly illustrative. New genomic technologies applied to classic genetic studies involving twins, early onset cases, and phenotypic extremes may provide key insights into developmental and gene-environment interactions in autoimmunity.


Subject(s)
Autoimmune Diseases/genetics , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , Autoimmune Diseases/physiopathology , Bacteria/immunology , Cytokines/immunology , Genome-Wide Association Study , Humans , Lymphocyte Activation , Twin Studies as Topic
2.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Article in English | MEDLINE | ID: mdl-34353900

ABSTRACT

Balancing microbial-induced cytokines and microbial clearance is critical at mucosal sites such as the intestine. How the inflammatory bowel disease (IBD)-associated gene RNF186 regulates this balance is unclear. We found that macrophages from IBD-risk rs6426833 carriers in the RNF186 region showed reduced cytokines to stimulation through multiple pattern recognition receptors (PRRs). Upon stimulation of PRRs, the E3-ubiquitin ligase RNF186 promoted ubiquitination of signaling complex molecules shared across PRRs and those unique to select PRRs. Furthermore, RNF186 was required for PRR-initiated signaling complex assembly and downstream signaling. RNF186, along with its intact E3-ubiquitin ligase activity, was required for optimal PRR-induced antimicrobial reactive oxygen species, reactive nitrogen species, and autophagy pathways and intracellular bacterial clearance in human macrophages and for bacterial clearance in intestinal myeloid cells. Cells transfected with the rare RNF186-A64T IBD-risk variant and macrophages from common rs6426833 RNF186 IBD-risk carriers demonstrated a reduction in these RNF186-dependent outcomes. These studies identify mechanisms through which RNF186 regulates innate immunity and show that RNF186 IBD-risk variants demonstrate a loss of function in PRR-initiated outcomes.


Subject(s)
Inflammatory Bowel Diseases/pathology , Macrophages/metabolism , Macrophages/microbiology , Receptors, Pattern Recognition/metabolism , Ubiquitin-Protein Ligases/metabolism , Cytokines/metabolism , Humans , Immunity, Innate , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Intestines/cytology , Macrophages/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , NF-kappa B/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Polymorphism, Single Nucleotide , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
3.
Gastroenterology ; 162(6): 1602-1616.e6, 2022 05.
Article in English | MEDLINE | ID: mdl-35149024

ABSTRACT

Inflammatory bowel disease is characterized by defects in epithelial function and dysregulated inflammatory signaling by lamina propria mononuclear cells including macrophages and dendritic cells in response to microbiota. In this review, we focus on the role of pattern recognition receptors in the inflammatory response as well as epithelial barrier regulation. We explore cytokine networks that increase inflammation, regulate paracellular permeability, cause epithelial damage, up-regulate epithelial proliferation, and trigger restitutive processes. We focus on studies using patient samples as well as speculate on pathways that can be targeted to more holistically treat patients with inflammatory bowel disease.


Subject(s)
Inflammatory Bowel Diseases , Tight Junctions , Caco-2 Cells , Cytokines/metabolism , Epithelial Cells/metabolism , Humans , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestines , Myosin-Light-Chain Kinase/metabolism , Permeability , Tight Junctions/metabolism
4.
Immunity ; 40(5): 734-46, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24837102

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune homeostasis and cytokine secretion. Multiple loci are associated with IBD, but a functional explanation is missing for most. Here we found that pattern-recognition receptor (PRR)-induced cytokine secretion was diminished in human monocyte-derived dendritic cells (MDDC) from rs7282490 ICOSLG GG risk carriers. Homotypic interactions between the costimulatory molecule ICOS and the ICOS ligand on MDDCs amplified nucleotide-binding oligomerization domain 2 (NOD2)-initiated cytokine secretion. This amplification required arginine residues in the ICOSL cytoplasmic tail that recruited the adaptor protein RACK1 and the kinases PKC and JNK leading to PKC, MAPK, and NF-κB activation. MDDC from rs7282490 GG risk-carriers had reduced ICOSL expression and PRR-initiated signaling and this loss-of-function ICOSLG risk allele associated with an ileal Crohn's disease phenotype, similar to polymorphisms in NOD2. Taken together, ICOSL amplifies PRR-initiated outcomes, which might contribute to immune homeostasis.


Subject(s)
Crohn Disease/immunology , Dendritic Cells/immunology , Inducible T-Cell Co-Stimulator Ligand/immunology , Inducible T-Cell Co-Stimulator Protein/immunology , Receptors, Pattern Recognition/immunology , Cells, Cultured , Crohn Disease/genetics , Enzyme Activation/immunology , GTP-Binding Proteins/immunology , HL-60 Cells , Humans , Inducible T-Cell Co-Stimulator Ligand/genetics , Inducible T-Cell Co-Stimulator Protein/genetics , JNK Mitogen-Activated Protein Kinases/immunology , Macrophages/immunology , NF-kappa B/immunology , Neoplasm Proteins/immunology , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Phosphorylation/immunology , Polymorphism, Single Nucleotide , Protein Kinase C/immunology , RNA Interference , RNA, Small Interfering , Receptors for Activated C Kinase , Receptors, Cell Surface/immunology , Signal Transduction/genetics
5.
Nature ; 546(7660): 667-670, 2017 06 29.
Article in English | MEDLINE | ID: mdl-28636595

ABSTRACT

Rotavirus, a leading cause of severe gastroenteritis and diarrhoea in young children, accounts for around 215,000 deaths annually worldwide. Rotavirus specifically infects the intestinal epithelial cells in the host small intestine and has evolved strategies to antagonize interferon and NF-κB signalling, raising the question as to whether other host factors participate in antiviral responses in intestinal mucosa. The mechanism by which enteric viruses are sensed and restricted in vivo, especially by NOD-like receptor (NLR) inflammasomes, is largely unknown. Here we uncover and mechanistically characterize the NLR Nlrp9b that is specifically expressed in intestinal epithelial cells and restricts rotavirus infection. Our data show that, via RNA helicase Dhx9, Nlrp9b recognizes short double-stranded RNA stretches and forms inflammasome complexes with the adaptor proteins Asc and caspase-1 to promote the maturation of interleukin (Il)-18 and gasdermin D (Gsdmd)-induced pyroptosis. Conditional depletion of Nlrp9b or other inflammasome components in the intestine in vivo resulted in enhanced susceptibility of mice to rotavirus replication. Our study highlights an important innate immune signalling pathway that functions in intestinal epithelial cells and may present useful targets in the modulation of host defences against viral pathogens.


Subject(s)
Epithelial Cells/immunology , Epithelial Cells/virology , Inflammasomes/metabolism , Intestines/cytology , Receptors, G-Protein-Coupled/metabolism , Rotavirus Infections/immunology , Rotavirus Infections/virology , Rotavirus/immunology , Animals , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , DEAD-box RNA Helicases/metabolism , Epithelial Cells/metabolism , Female , Immunity, Innate , Inflammasomes/chemistry , Inflammasomes/genetics , Interleukin-18/immunology , Intestinal Mucosa/metabolism , Intestines/immunology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Phosphate-Binding Proteins , Pyroptosis , RNA, Double-Stranded/metabolism , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/immunology , Rotavirus/growth & development
7.
Gastroenterology ; 161(6): 1924-1939, 2021 12.
Article in English | MEDLINE | ID: mdl-34478742

ABSTRACT

BACKGROUNDS & AIMS: Increased permeability is implicated in the pathogenesis of intestinal disease. In vitro and in vivo studies have linked down-regulation of the scaffolding protein ZO-1, encoded by the TJP1 gene, to increased tight junction permeability. This has not, however, been tested in vivo. Here, we assessed the contributions of ZO-1 to in vivo epithelial barrier function and mucosal homeostasis. METHODS: Public Gene Expression Omnibus data sets and biopsy specimens from patients with inflammatory bowel disease (IBD) and healthy control individuals were analyzed. Tjp1f/f;vil-CreTg mice with intestinal epithelial-specific ZO-1 knockout (ZO-1KO.IEC) mice and Tjp1f/f mice littermates without Cre expression were studied using chemical and immune-mediated models of disease as well as colonic stem cell cultures. RESULTS: ZO-1 transcript and protein expression were reduced in biopsy specimens from patients with IBD. Despite mildly increased intestinal permeability, ZO-1KO.IEC mice were healthy and did not develop spontaneous disease. ZO-1KO.IEC mice were, however, hypersensitive to mucosal insults and displayed defective repair. Furthermore, ZO-1-deficient colonic epithelia failed to up-regulate proliferation in response to damage in vivo or Wnt signaling in vitro. ZO-1 was associated with centrioles in interphase cells and mitotic spindle poles during division. In the absence of ZO-1, mitotic spindles failed to correctly orient, resulting in mitotic catastrophe and abortive proliferation. ZO-1 is, therefore, critical for up-regulation of epithelial proliferation and successful completion of mitosis. CONCLUSIONS: ZO-1 makes critical, tight junction-independent contributions to Wnt signaling and mitotic spindle orientation. As a result, ZO-1 is essential for mucosal repair. We speculate that ZO-1 down-regulation may be one cause of ineffective mucosal healing in patients with IBD.


Subject(s)
Cell Proliferation , Colon/metabolism , Epithelial Cells/metabolism , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Mitosis , Zonula Occludens-1 Protein/metabolism , Animals , Cells, Cultured , Colon/pathology , Databases, Genetic , Disease Models, Animal , Epithelial Cells/pathology , Humans , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/pathology , Mice, Knockout , Permeability , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Spindle Apparatus/pathology , Wnt Signaling Pathway , Wound Healing , Zonula Occludens-1 Protein/genetics
8.
J Immunol ; 205(4): 1024-1038, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32690658

ABSTRACT

Common IRF5 genetic risk variants associated with multiple immune-mediated diseases are a major determinant of interindividual variability in pattern-recognition receptor (PRR)-induced cytokines in myeloid cells. However, how myeloid cell-intrinsic IRF5 regulates the multiple distinct checkpoints mediating T cell outcomes in vivo and IRF5-dependent mechanisms contributing to these distinct checkpoints are not well defined. Using an in vivo Ag-specific adoptive T cell transfer approach into Irf5-/- mice, we found that T cell-extrinsic IRF5 regulated T cell outcomes at multiple critical checkpoints, including chemokine-mediated T cell trafficking into lymph nodes and PDK1-dependent soluble Ag uptake, costimulatory molecule upregulation, and secretion of Th1 (IL-12)- and Th17 (IL-23, IL-1ß, and IL-6)-conditioning cytokines by myeloid cells, which then cross-regulated Th2 and regulatory T cells. IRF5 was required for PRR-induced MAPK and NF-κB activation, which, in turn, regulated these key outcomes in myeloid cells. Importantly, mice with IRF5 deleted from myeloid cells demonstrated T cell outcomes similar to those observed in Irf5-/- mice. Complementation of IL-12 and IL-23 was able to restore T cell differentiation both in vitro and in vivo in the context of myeloid cell-deficient IRF5. Finally, human monocyte-derived dendritic cells from IRF5 disease-associated genetic risk carriers leading to increased IRF5 expression demonstrated increased Ag uptake and increased PRR-induced costimulatory molecule expression and chemokine and cytokine secretion compared with monocyte-derived dendritic cells from low-expressing IRF5 genetic variant carriers. These data establish that myeloid cell-intrinsic IRF5 regulates multiple distinct checkpoints in T cell activation and differentiation and that these are modulated by IRF5 disease risk variants.


Subject(s)
Interferon Regulatory Factors/metabolism , Myeloid Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Differentiation/immunology , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/immunology , Humans , Interferon Regulatory Factors/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Myeloid Cells/immunology , Receptors, Pattern Recognition/immunology , Receptors, Pattern Recognition/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , Up-Regulation/immunology
9.
J Immunol ; 205(5): 1406-1418, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32661180

ABSTRACT

STAT proteins can regulate both pro- and anti-inflammatory cytokine signaling. Therefore, identifying consequences of modulating expression of a given STAT is ultimately critical for determining its potential as a therapeutic target and for defining the mechanisms through which immune-mediated disease variants in STAT genes contribute to disease pathogenesis. Genetic variants in the STAT1/STAT4 region are associated with multiple immune-mediated diseases, including inflammatory bowel disease (IBD). These diseases are characterized by dysregulated cytokine secretion in response to pattern-recognition receptor (PRR) stimulation. We found that the common IBD-associated rs1517352 C risk allele increased both STAT1 and STAT4 expression in human monocyte-derived macrophages (MDMs). We therefore hypothesized that the STAT1/STAT4 variant might regulate PRR-initiated responses in a complementary and cooperative manner because of the important role of autocrine/paracrine cytokines in modulating PRR-initiated signaling. STAT1 and STAT4 were required for PRR- and live bacterial-induced secretion of multiple cytokines. These outcomes were particularly dependent on PRR-initiated autocrine/paracrine IL-12-induced STAT4 activation to generate IFN-γ, with autocrine IFN-γ then signaling through STAT1. STAT1 and STAT4 also promoted bacterial-induced cytokines in intestinal myeloid cells and PRR-enhanced antimicrobial pathways in MDMs. Importantly, MDMs from rs1517352 C IBD risk allele carriers demonstrated increased TLR4-, IFN-γ- and IL-12-induced STAT1 and STAT4 phosphorylation and cytokine secretion and increased TLR4-enhanced antimicrobial pathways. Taken together, STAT1 and STAT4 expression is coregulated by a shared genetic region, and STAT1 /STAT4-immune disease-associated variants modulate IFN-γ- and IL-12-associated outcomes, and in turn, PRR-induced outcomes, highlighting that these genes cooperate to regulate pathways relevant to disease pathogenesis.


Subject(s)
Genetic Predisposition to Disease/genetics , Genetic Variation/immunology , Macrophages/metabolism , Receptors, Pattern Recognition/genetics , STAT1 Transcription Factor/genetics , STAT4 Transcription Factor/genetics , Alleles , Cell Line , Cytokines/genetics , Gene Expression/genetics , Humans , Inflammatory Bowel Diseases/genetics , Interferon-gamma/genetics , Interleukin-12/genetics , Myeloid Cells/metabolism , Phosphorylation/genetics , Signal Transduction/genetics
10.
Gastroenterology ; 159(3): 1051-1067, 2020 09.
Article in English | MEDLINE | ID: mdl-32693188

ABSTRACT

BACKGROUND & AIMS: Loss-of-function variants in the laccase domain containing 1 (LACC1) gene are associated with immune-mediated diseases, including inflammatory bowel disease. It is not clear how LACC1 balances defenses against intestinal bacteria vs intestinal inflammation or what cells are responsible for this balance in humans or mice. METHODS: Lacc1-/- mice and mice with myeloid-specific disruption of Lacc1 (Lacc1Δmye) were given oral Salmonella Typhimurium or dextran sodium sulfate. CD45RBhiCD4+T cells were transferred to Lacc1-/-Rag2-/- mice to induce colitis. Organs were collected and analyzed by histology and protein expression. Bone marrow-derived macrophages and dendritic cells, lamina propria macrophages, and mesenteric lymph node dendritic cells were examined. We performed assays to measure intestinal permeability, cell subsets, bacterial uptake and clearance, reactive oxygen species, nitrite production, autophagy, signaling, messenger RNA, and cytokine levels. RESULTS: Lacc1-/- mice developed more severe T-cell transfer colitis than wild-type mice and had an increased burden of bacteria in intestinal lymphoid organs, which expressed lower levels of T helper (Th) 1 and Th17 cytokines and higher levels of Th2 cytokines. Intestinal lymphoid organs from mice with deletion of LACC1 had an increased burden of bacteria after oral administration of S Typhimurium and after administration of dextran sodium sulfate compared with wild-type mice. In macrophages, expression of LACC1 was required for toll-like receptor-induced uptake of bacteria, which required PDK1, and of mitogen-activated protein kinase (MAPK)- and nuclear factor κB-dependent induction of reactive oxygen species, reactive nitrogen species, and autophagy. Expression of LACC1 by dendritic cells was required for increasing expression of Th1 and Th17 cytokines and reducing expression of Th2 cytokines upon coculture with CD4+ T cells. Mice with LACC1-deficient myeloid cells had an increased burden of bacteria and altered T-cell cytokines in intestinal lymphoid organs, similar to Lacc1-/- mice. Complementation of cytokines produced by myeloid cells to cocultures of LACC1-deficient myeloid cells and wild-type CD4+ T cells restored T-cell cytokine regulation. When S Typhimurium-infected Lacc1Δmye mice were injected with these myeloid cell-derived cytokines, intestinal tissues increased production of Th1 and Th17 cytokines, and bacteria were reduced. CONCLUSIONS: Disruption of Lacc1 in mice increases the burden of bacteria in intestinal lymphoid organs and intestinal inflammation after induction of chronic colitis. LACC1 expression by myeloid cells in mice is required to clear bacteria and to regulate adaptive T-cell responses against microbes.


Subject(s)
Colitis, Ulcerative/immunology , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Myeloid Cells/metabolism , Salmonella Infections/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Coculture Techniques , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Cytokines/metabolism , DNA-Binding Proteins/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Female , Host Microbial Interactions/immunology , Humans , Immunity, Mucosal , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Primary Cell Culture , Salmonella Infections/microbiology , Salmonella Infections/pathology , Salmonella typhimurium/immunology
11.
J Immunol ; 203(12): 3325-3338, 2019 12 15.
Article in English | MEDLINE | ID: mdl-31732533

ABSTRACT

Genetic variants in the STAT3/STAT5A/STAT5B region are associated with immune-mediated diseases, including inflammatory bowel disease (IBD). However, how STAT3 and STAT5 regulate the critical balance between pro- and anti-inflammatory cytokines and how common disease-associated genetic variants (e.g., rs12942547) in the region modulate this balance are incompletely understood. We found that upon pattern-recognition receptor (PRR) stimulation of human monocyte-derived macrophages (MDMs), decreasing STAT3, STAT5a, and STAT5b expression led to a progressive decrease in anti-inflammatory cytokines, whereas proinflammatory cytokines initially decreased but then increased when STAT3 or STAT5 expression fell below a critical threshold. Mechanisms regulating STAT3- and STAT5-dependent inflammatory cytokine outcomes included negative feedback from autocrine/paracrine IL-10, TGF-ß, IL-4, IL-13, IL-22, and TSLP secretion and SOCS1/SOCS2/SOCS3 induction. MDMs from rs12942547 AA disease-risk carriers demonstrated increased STAT3, STAT5a, and STAT5b expression and increased PRR-induced STAT3 and STAT5 phosphorylation relative to GG MDMs. Both pro- and anti-inflammatory cytokine secretion was decreased in MDMs from GG carriers, as STAT3, STAT5a, and STAT5b expression was above the threshold for reciprocal regulation of these cytokines. Taken together, we identify that the threshold of STAT3, STAT5a, and STAT5b expression determines if PRR-induced proinflammatory cytokines are increased or decreased, define mechanisms for this reciprocal regulation, and elucidate consequences for disease variants in the STAT3/STAT5A/STAT5B region, indicating that considering signaling thresholds and targeting specific cell types might be beneficial when evaluating therapeutic interventions in this pathway.


Subject(s)
Cytokines/metabolism , Inflammation Mediators/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Disease Susceptibility , Gene Expression , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Macrophages/immunology , Macrophages/metabolism , Phosphorylation , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/genetics
12.
J Immunol ; 202(3): 920-930, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30593537

ABSTRACT

Common IFN regulatory factor 5 (IRF5) variants associated with multiple immune-mediated diseases are a major determinant of interindividual variability in pattern recognition receptor (PRR)-induced cytokines in macrophages. PRR-initiated pathways also contribute to bacterial clearance, and dysregulation of bacterial clearance can contribute to immune-mediated diseases. However, the role of IRF5 in macrophage-mediated bacterial clearance is not well defined. Furthermore, it is unclear if macrophages from individuals who are carriers of low IRF5-expressing genetic variants associated with protection for immune-mediated diseases might be at a disadvantage in bacterial clearance. We found that IRF5 was required for optimal bacterial clearance in PRR-stimulated, M1-differentiated human macrophages. Mechanisms regulated by IRF5 included inducing reactive oxygen species through p40phox, p47phox and p67phox, NOS2, and autophagy through ATG5. Complementing these pathways in IRF5-deficient M1 macrophages restored bacterial clearance. Further, these antimicrobial pathways required the activation of IRF5-dependent MAPK, NF-κB, and Akt2 pathways. Importantly, relative to high IRF5-expressing rs2004640/rs2280714 TT/TT immune-mediated disease risk-carrier human macrophages, M1-differentiated GG/CC carrier macrophages demonstrated less reactive oxygen species, NOS2, and autophagy pathway induction and, consequently, reduced bacterial clearance. Increasing IRF5 expression to the rs2004640/rs2280714 TT/TT levels restored these antimicrobial pathways. We define mechanisms wherein common IRF5 genetic variants modulate bacterial clearance, thereby highlighting that immune-mediated disease risk IRF5 carriers might be relatively protected from microbial-associated diseases.


Subject(s)
Bacteria/immunology , Interferon Regulatory Factors/immunology , Macrophages/immunology , Macrophages/microbiology , Autophagy , Autophagy-Related Protein 5/genetics , Cell Differentiation , Cells, Cultured , Enterococcus faecalis/immunology , Genetic Predisposition to Disease , Genetic Variation , Humans , Interferon Regulatory Factors/genetics , Interferon-gamma/pharmacology , Nitric Oxide Synthase Type II/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/immunology , Salmonella enterica/immunology , Signal Transduction
13.
J Immunol ; 202(11): 3297-3308, 2019 06 01.
Article in English | MEDLINE | ID: mdl-31028123

ABSTRACT

Intestinal tissues are continuously exposed to microbial products that stimulate pattern-recognition receptors (PRRs). Ongoing PRR stimulation can confer epigenetic changes in macrophages, which can then regulate subsequent immune outcomes and adaptation to the local environment. Mechanisms leading to these changes are incompletely understood. We found that short-term stimulation of the PRR NOD2 in primary human monocyte-derived macrophages resulted in increased H3 and H4 acetylation of cytokine promoters, consistent with the increased cytokine secretion observed. However, with prolonged NOD2 stimulation, both the acetylation and cytokine secretion were dramatically decreased. Chronic NOD2 stimulation upregulated the transcription factors Twist1 and Twist2, which bound to the promoters of the histone deacetylases HDAC1 and HDAC3 and induced HDAC1 and HDAC3 expression. HDAC1 and HDAC3 then mediated histone deacetylation at cytokine promoters and, in turn, cytokine downregulation under these conditions. Similar regulation was observed upon chronic stimulation of multiple PRRs. Consistent with the chronic microbial exposure in the intestinal environment, TWIST1, TWIST2, HDAC1, and HDAC3 were upregulated in human intestinal relative to peripheral macrophages. Importantly, complementing HDAC1 and HDAC3 in Twist1/Twist2-deficient monocyte-derived macrophages restored the reduced histone acetylation on cytokine promoters and the decreased cytokine secretion with chronic NOD2 stimulation. Taken together, we identify mechanisms wherein Twist1 and Twist2 promote chromatin modifications, resulting in macrophage instruction and adaptation to conditions in the intestinal microenvironment.


Subject(s)
Macrophages/immunology , Twist-Related Protein 1/metabolism , Twist-Related Protein 2/metabolism , Acetylation , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/metabolism , Humans , Mice , Mice, Inbred C57BL , Monocytes/cytology , Promoter Regions, Genetic/genetics , Protein Binding , Receptors, Pattern Recognition/metabolism , Twist-Related Protein 1/genetics , Twist-Related Protein 2/genetics
14.
Gut ; 69(2): 264-273, 2020 02.
Article in English | MEDLINE | ID: mdl-31097538

ABSTRACT

OBJECTIVE: The interleukin (IL)23 pathway contributes to IBD pathogenesis and is being actively studied as a therapeutic target in patients with IBD. Unexpected outcomes in these therapeutic trials have highlighted the importance of understanding the cell types and mechanisms through which IL23 regulates immune outcomes. How IL23 regulates macrophage outcomes and the consequences of the IL23R R381Q IBD-protective variant on macrophages are not well defined; macrophages are key players in IBD pathogenesis and inflammation. DESIGN: We analysed protein and RNA expression, signalling and localisation in human monocyte-derived macrophages (MDMs) through western blot, ELISA, real-time PCR, flow cytometry, immunoprecipitation and microscopy. RESULTS: IL23R was critical for optimal levels of pattern-recognition receptor (PRR)-induced signalling and cytokines in human MDMs. In contrast to the coreceptor IL12Rß1, IL23 induced dynamic IL23R cell surface regulation and this required clathrin and dynamin-mediated endocytosis and endocytic recycling-dependent pathways; these pathways were essential for IL23R-mediated outcomes. The IBD-protective IL23R R381Q variant showed distinct outcomes. Relative to IL23R R381, HeLa cells expressing IL23R Q381 showed decreased IL23R recycling and reduced assembly of IL23R Q381 with Janus kinase/signal transducer and activator of transcription pathway members. In MDMs from IL23R Q381 carriers, IL23R accumulated in late endosomes and lysosomes on IL23 treatment and cells demonstrated decreased IL23R- and PRR-induced signalling and cytokines relative to IL23R R381 MDMs. CONCLUSION: Macrophage-mediated inflammatory pathways are key contributors to IBD pathogenesis, and we identify an autocrine/paracrine IL23 requirement in PRR-initiated human macrophage outcomes and in human intestinal myeloid cells, establish that IL23R undergoes ligand-induced recycling, define mechanisms regulating IL23R-induced signalling and determine how the IBD-protective IL23R R381Q variant modulates these processes.


Subject(s)
Cytokines/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Receptors, Interleukin/immunology , Autocrine Communication/immunology , Endocytosis/immunology , Endosomes/immunology , Genetic Variation , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/prevention & control , Interleukin-23/immunology , Janus Kinase 2/metabolism , Paracrine Communication/immunology , Receptors, Interleukin/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction/immunology
15.
Genes Immun ; 20(7): 577-588, 2019 09.
Article in English | MEDLINE | ID: mdl-30692607

ABSTRACT

Genome-wide association studies have identified ~170 loci associated with Crohn's disease (CD) and defining which genes drive these association signals is a major challenge. The primary aim of this study was to define which CD locus genes are most likely to be disease related. We developed a gene prioritization regression model (GPRM) by integrating complementary mRNA expression datasets, including bulk RNA-Seq from the terminal ileum of 302 newly diagnosed, untreated CD patients and controls, and in stimulated monocytes. Transcriptome-wide association and co-expression network analyses were performed on the ileal RNA-Seq datasets, identifying 40 genome-wide significant genes. Co-expression network analysis identified a single gene module, which was substantially enriched for CD locus genes and most highly expressed in monocytes. By including expression-based and epigenetic information, we refined likely CD genes to 2.5 prioritized genes per locus from an average of 7.8 total genes. We validated our model structure using cross-validation and our prioritization results by protein-association network analyses, which demonstrated significantly higher CD gene interactions for prioritized compared with non-prioritized genes. Although individual datasets cannot convey all of the information relevant to a disease, combining data from multiple relevant expression-based datasets improves prediction of disease genes and helps to further understanding of disease pathogenesis.


Subject(s)
Crohn Disease/genetics , Monocytes/pathology , Sequence Analysis, DNA/methods , Adolescent , Algorithms , Case-Control Studies , Child , Child, Preschool , Crohn Disease/metabolism , Female , Gene Regulatory Networks/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Humans , Male , Monocytes/metabolism , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Software , Transcriptome/genetics
16.
Gastroenterology ; 152(2): 374-388.e4, 2017 02.
Article in English | MEDLINE | ID: mdl-27780712

ABSTRACT

Insights into the pathogenesis of inflammatory bowel diseases (IBDs) have provided important information for the development of therapeutics. Levels of interleukin 23 (IL23) and T-helper (Th) 17 cell pathway molecules are increased in inflamed intestinal tissues of patients with IBD. Loss-of-function variants of the IL23-receptor gene (IL23R) protect against IBD, and, in animals, blocking IL23 reduces the severity of colitis. These findings indicated that the IL23 and Th17 cell pathways might be promising targets for the treatment of IBD. Clinical trials have investigated the effects of agents designed to target distinct levels of the IL23 and Th17 cell pathways, and the results are providing insights into IBD pathogenesis and additional strategies for modulating these pathways. Strategies to reduce levels of proinflammatory cytokines more broadly and increase anti-inflammatory mechanisms also are emerging for the treatment of IBD. The results from trials targeting these immune system pathways have provided important lessons for future trials. Findings indicate the importance of improving approaches to integrate patient features and biomarkers of response with selection of therapeutics.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Interleukin-23/immunology , Th17 Cells/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Cytokines/immunology , Humans , Inflammatory Bowel Diseases/immunology , Janus Kinases/immunology , Lysophospholipids/immunology , Molecular Targeted Therapy , Oligonucleotides/therapeutic use , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Signal Transduction , Smad7 Protein/immunology , Sphingosine/analogs & derivatives , Sphingosine/immunology , Transforming Growth Factor beta/immunology , Ustekinumab/therapeutic use
17.
J Immunol ; 197(9): 3695-3704, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27664279

ABSTRACT

JAK2 genetic variants are associated with inflammatory bowel disease (IBD) and JAK inhibitors are being evaluated for therapy targeting immune-mediated diseases, including IBD. As JAK pathway-mediated cytokine regulation varies across cell types and stimulation conditions, we examined how JAK signaling and IBD-associated JAK2 variants regulate distinct acute and chronic microbial product exposure outcomes in human myeloid cells, consistent with the conditions of initial entry and ongoing intestinal tissue residence, respectively. Macrophages from controls and ulcerative colitis patients carrying the IBD-risk rs10758669 CC genotype showed increased JAK2 expression and nucleotide-binding oligomerization domain 2-induced JAK2 phosphorylation relative to AA carriers. Interestingly, the threshold of JAK2 expression and signaling determined pattern-recognition receptor (PRR)-induced outcomes; whereas anti-inflammatory cytokines progressively decreased with lower JAK2 expression, proinflammatory cytokines switched from decreased to increased secretion below a certain JAK2 expression threshold. Low JAK2-expressing rs10758669 AA macrophages were above this threshold; consequently, both PRR-induced pro- and anti-inflammatory cytokines were decreased. However, relative to rs10758669 CC risk carriers, AA carrier macrophages switched to increased nucleotide-binding oligomerization domain 2-induced proinflammatory cytokines at lower therapeutically used JAK inhibitor doses. Importantly, JAK inhibitors increased proinflammatory cytokines secreted by peripheral macrophages following chronic PRR stimulation and by human intestinal myeloid cells following exposure to intestinal pathogens. Mechanistically, the decreased response to and secretion of autocrine/paracrine IL-10, IL-4, IL-22 and thymic stromal lymphopoietin regulated these JAK-dependent outcomes in myeloid cells. Taken together, the JAK signaling threshold determines whether PRR-induced pro- and anti-inflammatory cytokines are reciprocally regulated in myeloid cells; consideration of JAK2 genotype and targeting of specific cell types might improve JAK-targeted therapy in immune-mediated diseases.


Subject(s)
Inflammatory Bowel Diseases/immunology , Janus Kinase 2/metabolism , Macrophages/physiology , Adult , Alleles , Cells, Cultured , Cytokines/metabolism , Female , Genotype , Humans , Inflammation Mediators/metabolism , Inflammatory Bowel Diseases/genetics , Janus Kinase 2/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide , Protein Kinase Inhibitors/therapeutic use , Signal Transduction , Treatment Outcome
18.
Nature ; 491(7423): 259-63, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-23075849

ABSTRACT

Chronic mucosal inflammation and tissue damage predisposes patients to the development of colorectal cancer. This association could be explained by the hypothesis that the same factors and pathways important for wound healing also promote tumorigenesis. A sensor of tissue damage should induce these factors to promote tissue repair and regulate their action to prevent development of cancer. Interleukin 22 (IL-22), a cytokine of the IL-10 superfamily, has an important role in colonic epithelial cell repair, and its levels are increased in the blood and intestine of inflammatory bowel disease patients. This cytokine can be neutralized by the soluble IL-22 receptor, known as the IL-22 binding protein (IL-22BP, also known as IL22RA2); however, the significance of endogenous IL-22BP in vivo and the pathways that regulate this receptor are unknown. Here we describe that IL-22BP has a crucial role in controlling tumorigenesis and epithelial cell proliferation in the colon. IL-22BP is highly expressed by dendritic cells in the colon in steady-state conditions. Sensing of intestinal tissue damage via the NLRP3 or NLRP6 inflammasomes led to an IL-18-dependent downregulation of IL-22BP, thereby increasing the ratio of IL-22/IL-22BP. IL-22, which is induced during intestinal tissue damage, exerted protective properties during the peak of damage, but promoted tumour development if uncontrolled during the recovery phase. Thus, the IL-22-IL-22BP axis critically regulates intestinal tissue repair and tumorigenesis in the colon.


Subject(s)
Cell Transformation, Neoplastic , Inflammasomes/metabolism , Intestinal Mucosa/metabolism , Intestines/pathology , Receptors, Interleukin/metabolism , Animals , Colitis/complications , Colitis/metabolism , Colitis/pathology , Colon/metabolism , Colon/pathology , Colonic Neoplasms/complications , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Down-Regulation , Epithelial Cells/metabolism , Epithelial Cells/pathology , Genes, APC , Interleukin-18/metabolism , Interleukins/deficiency , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Knockout , Receptors, Interleukin/deficiency , Receptors, Interleukin/genetics , Time Factors , Weight Loss , Interleukin-22
19.
Nature ; 491(7422): 119-24, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23128233

ABSTRACT

Crohn's disease and ulcerative colitis, the two common forms of inflammatory bowel disease (IBD), affect over 2.5 million people of European ancestry, with rising prevalence in other populations. Genome-wide association studies and subsequent meta-analyses of these two diseases as separate phenotypes have implicated previously unsuspected mechanisms, such as autophagy, in their pathogenesis and showed that some IBD loci are shared with other inflammatory diseases. Here we expand on the knowledge of relevant pathways by undertaking a meta-analysis of Crohn's disease and ulcerative colitis genome-wide association scans, followed by extensive validation of significant findings, with a combined total of more than 75,000 cases and controls. We identify 71 new associations, for a total of 163 IBD loci, that meet genome-wide significance thresholds. Most loci contribute to both phenotypes, and both directional (consistently favouring one allele over the course of human history) and balancing (favouring the retention of both alleles within populations) selection effects are evident. Many IBD loci are also implicated in other immune-mediated disorders, most notably with ankylosing spondylitis and psoriasis. We also observe considerable overlap between susceptibility loci for IBD and mycobacterial infection. Gene co-expression network analysis emphasizes this relationship, with pathways shared between host responses to mycobacteria and those predisposing to IBD.


Subject(s)
Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Host-Pathogen Interactions , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/microbiology , Mycobacterium/immunology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/physiopathology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/microbiology , Crohn Disease/physiopathology , Genome, Human/genetics , Haplotypes/genetics , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/physiopathology , Mycobacterium/pathogenicity , Mycobacterium Infections/genetics , Mycobacterium Infections/microbiology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/pathogenicity , Phenotype , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results
20.
Proc Natl Acad Sci U S A ; 112(33): 10461-6, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26240347

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by dysregulated host:microbial interactions and cytokine production. Host pattern recognition receptors (PRRs) are critical in regulating these interactions. Multiple genetic loci are associated with IBD, but altered functions for most, including in the rs713875 MTMR3/HORMAD2/LIF/OSM region, are unknown. We identified a previously undefined role for myotubularin-related protein 3 (MTMR3) in amplifying PRR-induced cytokine secretion in human macrophages and defined MTMR3-initiated mechanisms contributing to this amplification. MTMR3 decreased PRR-induced phosphatidylinositol 3-phosphate (PtdIns3P) and autophagy levels, thereby increasing PRR-induced caspase-1 activation, autocrine IL-1ß secretion, NFκB signaling, and, ultimately, overall cytokine secretion. This MTMR3-mediated regulation required the N-terminal pleckstrin homology-GRAM domain and Cys413 within the phosphatase domain of MTMR3. In MTMR3-deficient macrophages, reducing the enhanced autophagy or restoring NFκB signaling rescued PRR-induced cytokines. Macrophages from rs713875 CC IBD risk carriers demonstrated increased MTMR3 expression and, in turn, decreased PRR-induced PtdIns3P and autophagy and increased PRR-induced caspase-1 activation, signaling, and cytokine secretion. Thus, the rs713875 IBD risk polymorphism increases MTMR3 expression, which modulates PRR-induced outcomes, ultimately leading to enhanced PRR-induced cytokines.


Subject(s)
Autophagy , Caspase 1/metabolism , Cytokines/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Signal Transduction , Alleles , Enzyme Activation , Gene Expression Regulation , Genetic Predisposition to Disease , Genotype , Homeostasis , Humans , Inflammation/metabolism , Inflammatory Bowel Diseases/genetics , Leukocytes, Mononuclear/cytology , Ligands , Macrophages/metabolism , Monocytes/cytology , Protein Structure, Tertiary , RNA, Small Interfering/metabolism , Risk Factors , Toll-Like Receptors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL