Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.419
Filter
Add more filters

Publication year range
1.
Cell ; 182(6): 1441-1459.e21, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32888430

ABSTRACT

Throughout a 24-h period, the small intestine (SI) is exposed to diurnally varying food- and microbiome-derived antigenic burdens but maintains a strict immune homeostasis, which when perturbed in genetically susceptible individuals, may lead to Crohn disease. Herein, we demonstrate that dietary content and rhythmicity regulate the diurnally shifting SI epithelial cell (SIEC) transcriptional landscape through modulation of the SI microbiome. We exemplify this concept with SIEC major histocompatibility complex (MHC) class II, which is diurnally modulated by distinct mucosal-adherent SI commensals, while supporting downstream diurnal activity of intra-epithelial IL-10+ lymphocytes regulating the SI barrier function. Disruption of this diurnally regulated diet-microbiome-MHC class II-IL-10-epithelial barrier axis by circadian clock disarrangement, alterations in feeding time or content, or epithelial-specific MHC class II depletion leads to an extensive microbial product influx, driving Crohn-like enteritis. Collectively, we highlight nutritional features that modulate SI microbiome, immunity, and barrier function and identify dietary, epithelial, and immune checkpoints along this axis to be potentially exploitable in future Crohn disease interventions.


Subject(s)
Crohn Disease/microbiology , Epithelial Cells/metabolism , Gastrointestinal Microbiome , Histocompatibility Antigens Class II/metabolism , Intestine, Small/immunology , Intestine, Small/microbiology , Transcriptome/genetics , Animals , Anti-Bacterial Agents/pharmacology , Circadian Clocks/physiology , Crohn Disease/immunology , Crohn Disease/metabolism , Diet , Epithelial Cells/cytology , Epithelial Cells/immunology , Flow Cytometry , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/genetics , Gene Expression Profiling , Histocompatibility Antigens Class II/genetics , Homeostasis , In Situ Hybridization, Fluorescence , Interleukin-10/metabolism , Interleukin-10/pharmacology , Intestine, Small/physiology , Lymphocytes , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Periodicity , T-Lymphocytes/immunology , Transcriptome/physiology
2.
Cell ; 178(6): 1493-1508.e20, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31474370

ABSTRACT

Clinical benefits of cytokine blockade in ileal Crohn's disease (iCD) are limited to a subset of patients. Here, we applied single-cell technologies to iCD lesions to address whether cellular heterogeneity contributes to treatment resistance. We found that a subset of patients expressed a unique cellular module in inflamed tissues that consisted of IgG plasma cells, inflammatory mononuclear phagocytes, activated T cells, and stromal cells, which we named the GIMATS module. Analysis of ligand-receptor interaction pairs identified a distinct network connectivity that likely drives the GIMATS module. Strikingly, the GIMATS module was also present in a subset of patients in four independent iCD cohorts (n = 441), and its presence at diagnosis correlated with failure to achieve durable corticosteroid-free remission upon anti-TNF therapy. These results emphasize the limitations of current diagnostic assays and the potential for single-cell mapping tools to identify novel biomarkers of treatment response and tailored therapeutic opportunities.


Subject(s)
Crohn Disease/therapy , Cytokines/immunology , Intestines/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Crohn Disease/immunology , Crohn Disease/pathology , Humans , Immunotherapy/methods , Phagocytes/pathology , Single-Cell Analysis , Stromal Cells/pathology , T-Lymphocytes/pathology
3.
Nat Immunol ; 22(4): 471-484, 2021 04.
Article in English | MEDLINE | ID: mdl-33664518

ABSTRACT

The diversity of regulatory T (Treg) cells in health and in disease remains unclear. Individuals with colorectal cancer harbor a subpopulation of RORγt+ Treg cells with elevated expression of ß-catenin and pro-inflammatory properties. Here we show progressive expansion of RORγt+ Treg cells in individuals with inflammatory bowel disease during inflammation and early dysplasia. Activating Wnt-ß-catenin signaling in human and murine Treg cells was sufficient to recapitulate the disease-associated increase in the frequency of RORγt+ Treg cells coexpressing multiple pro-inflammatory cytokines. Binding of the ß-catenin interacting partner, TCF-1, to DNA overlapped with Foxp3 binding at enhancer sites of pro-inflammatory pathway genes. Sustained Wnt-ß-catenin activation induced newly accessible chromatin sites in these genes and upregulated their expression. These findings indicate that TCF-1 and Foxp3 together limit the expression of pro-inflammatory genes in Treg cells. Activation of ß-catenin signaling interferes with this function and promotes the disease-associated RORγt+ Treg phenotype.


Subject(s)
Cell Proliferation , Cellular Reprogramming , Colitis, Ulcerative/metabolism , Colitis-Associated Neoplasms/metabolism , Crohn Disease/metabolism , Epigenesis, Genetic , Lymphocyte Activation , T-Lymphocytes, Regulatory/metabolism , Wnt Signaling Pathway , Animals , Case-Control Studies , Cells, Cultured , Colitis, Ulcerative/genetics , Colitis, Ulcerative/immunology , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/metabolism , Humans , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , T Cell Transcription Factor 1 , T-Lymphocytes, Regulatory/immunology
4.
Nat Immunol ; 20(8): 970-979, 2019 08.
Article in English | MEDLINE | ID: mdl-31235952

ABSTRACT

Inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis are characterized by uncontrolled activation of intestinal immune cells in a genetically susceptible host. Due to the progressive and destructive nature of the inflammatory process in IBD, complications such as fibrosis, stenosis or cancer are frequently observed, which highlights the need for effective anti-inflammatory therapy. Studies have identified altered trafficking of immune cells and pathogenic immune cell circuits as crucial drivers of mucosal inflammation and tissue destruction in IBD. A defective gut barrier and microbial dysbiosis induce such accumulation and local activation of immune cells, which results in a pro-inflammatory cytokine loop that overrides anti-inflammatory signals and causes chronic intestinal inflammation. This Review discusses pathogenic cytokine responses of immune cells as well as immune cell trafficking as a rational basis for new translational therapies in IBD.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Cytokines/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Animals , Cell Movement/immunology , Colitis, Ulcerative/therapy , Crohn Disease/therapy , Dysbiosis , Humans , Inflammation/pathology , Intestinal Mucosa/cytology , Lymphocyte Activation/immunology , Mice , T-Lymphocytes/immunology
5.
Nat Immunol ; 19(12): 1403-1414, 2018 12.
Article in English | MEDLINE | ID: mdl-30397350

ABSTRACT

Repair of tissue damaged during inflammatory processes is key to the return of local homeostasis and restoration of epithelial integrity. Here we describe CD161+ regulatory T (Treg) cells as a distinct, highly suppressive population of Treg cells that mediate wound healing. These Treg cells were enriched in intestinal lamina propria, particularly in Crohn's disease. CD161+ Treg cells had an all-trans retinoic acid (ATRA)-regulated gene signature, and CD161 expression on Treg cells was induced by ATRA, which directly regulated the CD161 gene. CD161 was co-stimulatory, and ligation with the T cell antigen receptor induced cytokines that accelerated the wound healing of intestinal epithelial cells. We identified a transcription-factor network, including BACH2, RORγt, FOSL2, AP-1 and RUNX1, that controlled expression of the wound-healing program, and found a CD161+ Treg cell signature in Crohn's disease mucosa associated with reduced inflammation. These findings identify CD161+ Treg cells as a population involved in controlling the balance between inflammation and epithelial barrier healing in the gut.


Subject(s)
Intestinal Mucosa/immunology , NK Cell Lectin-Like Receptor Subfamily B/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Tretinoin/immunology , Wound Healing/immunology , Crohn Disease/immunology , Humans
6.
Nat Immunol ; 19(3): 246-254, 2018 03.
Article in English | MEDLINE | ID: mdl-29358708

ABSTRACT

Defective autophagy is linked to diseases such as rheumatoid arthritis, lupus and inflammatory bowel disease (IBD). However, the mechanisms by which autophagy limits inflammation remain poorly understood. Here we found that loss of the autophagy-related gene Atg16l1 promoted accumulation of the adaptor TRIF and downstream signaling in macrophages. Multiplex proteomic profiling identified SQSTM1 and Tax1BP1 as selective autophagy-related receptors that mediated the turnover of TRIF. Knockdown of Tax1bp1 increased production of the cytokines IFN-ß and IL-1ß. Mice lacking Atg16l1 in myeloid cells succumbed to lipopolysaccharide-mediated sepsis but enhanced their clearance of intestinal Salmonella typhimurium in an interferon receptor-dependent manner. Human macrophages with the Crohn's disease-associated Atg16l1 variant T300A exhibited more production of IFN-ß and IL-1ß. An elevated interferon-response gene signature was observed in patients with IBD who were resistant to treatment with an antibody to the cytokine TNF. These findings identify selective autophagy as a key regulator of signaling via the innate immune system.


Subject(s)
Adaptor Proteins, Vesicular Transport/immunology , Autophagy/immunology , Immunity, Innate/immunology , Inflammation/immunology , Animals , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Crohn Disease/immunology , Female , Humans , Macrophages/immunology , Male , Mice , Mice, Transgenic , Signal Transduction/immunology
7.
Immunity ; 54(12): 2795-2811.e9, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34788601

ABSTRACT

Lymphangitis and the formation of tertiary lymphoid organs (TLOs) in the mesentery are features of Crohn's disease. Here, we examined the genesis of these TLOs and their impact on disease progression. Whole-mount and intravital imaging of the ileum and ileum-draining collecting lymphatic vessels (CLVs) draining to mesenteric lymph nodes from TNFΔARE mice, a model of ileitis, revealed TLO formation at valves of CLVs. TLOs obstructed cellular and molecular outflow from the gut and were sites of lymph leakage and backflow. Tumor necrosis factor (TNF) neutralization begun at early stages of TLO formation restored lymph transport. However, robustly developed, chronic TLOs resisted regression and restoration of flow after TNF neutralization. TNF stimulation of cultured lymphatic endothelial cells reprogrammed responses to oscillatory shear stress, preventing the induction of valve-associated genes. Disrupted transport of immune cells, driven by loss of valve integrity and TLO formation, may contribute to the pathology of Crohn's disease.


Subject(s)
Crohn Disease/immunology , Endothelial Cells/immunology , Ileum/immunology , Lymph/metabolism , Lymphatic Vessels/immunology , Mesentery/immunology , Tertiary Lymphoid Structures/immunology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Humans , Ileitis , Lymphangitis , Mice , Mice, Knockout , Stress, Mechanical
8.
Immunity ; 54(8): 1728-1744.e7, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34343498

ABSTRACT

Inflammatory bowel disease (IBD) mainly includes Crohn's disease (CD) and ulcerative colitis (UC). Immune disorders play an essential role in the pathogenesis of these two IBDs, but the differences in the immune microenvironment of the colon and their underlying mechanisms remain poorly investigated. Here we examined the immunological features and metabolic microenvironment of untreated individuals with IBD by multiomics analyses. Modulation of CD-specific metabolites, particularly reduced selenium, can obviously shape type 1 T helper (Th1) cell differentiation, which is specifically enriched in CD. Selenium supplementation suppressed the symptoms and onset of CD and Th1 cell differentiation via selenoprotein W (SELW)-mediated cellular reactive oxygen species scavenging. SELW promoted purine salvage pathways and inhibited one-carbon metabolism by recruiting an E3 ubiquitin ligase, tripartite motif-containing protein 21, which controlled the stability of serine hydroxymethyltransferase 2. Our work highlights selenium as an essential regulator of T cell responses and potential therapeutic targets in CD.


Subject(s)
Antioxidants/pharmacology , Crohn Disease/drug therapy , Crohn Disease/immunology , Selenium/pharmacology , Selenoprotein W/metabolism , Th1 Cells/cytology , Cell Differentiation/immunology , Cell Polarity , Colon/immunology , Colon/pathology , Glycine Hydroxymethyltransferase/metabolism , Humans , Reactive Oxygen Species/metabolism , Ribonucleoproteins/metabolism , Th1 Cells/immunology , Ubiquitin-Protein Ligases/metabolism
9.
Immunity ; 54(5): 856-858, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979584

ABSTRACT

Intestinal microbiome perturbation characterizes Crohn's disease (CD), though specific contributors to pathophysiology remain elusive. In a recent issue of Science, Jain et al. show that Debaryomyces hansenii impairs intestinal healing in mice via effects on type I interferon signaling and chemokine CCL5 expression in macrophages and that it is also prevalent in the inflamed mucosa of CD patients.


Subject(s)
Crohn Disease/immunology , Crohn Disease/microbiology , Intestinal Mucosa/microbiology , Wound Healing/immunology , Animals , Chemokine CCL5/immunology , Gastrointestinal Microbiome/immunology , Humans , Interferon Type I/immunology , Intestinal Mucosa/immunology , Macrophages/immunology , Mice , Mycoses/immunology , Mycoses/microbiology , Saccharomycetales/immunology , Signal Transduction/immunology
10.
Cell ; 158(5): 1000-1010, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25171403

ABSTRACT

Specific members of the intestinal microbiota dramatically affect inflammatory bowel disease (IBD) in mice. In humans, however, identifying bacteria that preferentially affect disease susceptibility and severity remains a major challenge. Here, we used flow-cytometry-based bacterial cell sorting and 16S sequencing to characterize taxa-specific coating of the intestinal microbiota with immunoglobulin A (IgA-SEQ) and show that high IgA coating uniquely identifies colitogenic intestinal bacteria in a mouse model of microbiota-driven colitis. We then used IgA-SEQ and extensive anaerobic culturing of fecal bacteria from IBD patients to create personalized disease-associated gut microbiota culture collections with predefined levels of IgA coating. Using these collections, we found that intestinal bacteria selected on the basis of high coating with IgA conferred dramatic susceptibility to colitis in germ-free mice. Thus, our studies suggest that IgA coating identifies inflammatory commensals that preferentially drive intestinal disease. Targeted elimination of such bacteria may reduce, reverse, or even prevent disease development.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Immunoglobulin A/immunology , Microbiota , Animals , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Crohn Disease/microbiology , Crohn Disease/pathology , DNA, Bacterial/analysis , Dysbiosis/immunology , Dysbiosis/microbiology , Humans , Inflammasomes/immunology , Inflammation/immunology , Inflammation/microbiology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S/analysis , Specific Pathogen-Free Organisms
11.
Trends Immunol ; 45(6): 470-481, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38782626

ABSTRACT

Skip lesions are an enigmatic spatial feature characterizing Crohn's disease (CD). They comprise inflamed and adjacent non-inflamed tissue sections with a clear demarcation. Currently, spatial features of the human gastrointestinal (GI) system lack clarity regarding the organization of microbes, mucus, tissue, and host cells during inflammation. New technologies with multiplexing abilities and innovative approaches provide ways of examining the spatial organization of inflamed and non-inflamed tissues in CD, which may open new avenues for diagnosis, prognosis, and treatment. In this review, we present evidence of the relevance of spatial context in patients with CD and the methods and ideas recently published in studies of spatiality during inflammation. With this review, we aim to provide inspiration for further research to address existing gaps.


Subject(s)
Crohn Disease , Crohn Disease/immunology , Crohn Disease/pathology , Humans , Animals , Inflammation/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/pathology
12.
Immunity ; 47(6): 1182-1196.e10, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29262351

ABSTRACT

CD4+ T cells are tightly regulated by microbiota in the intestine, but whether intestinal T cells interface with host-derived metabolites is less clear. Here, we show that CD4+ T effector (Teff) cells upregulated the xenobiotic transporter, Mdr1, in the ileum to maintain homeostasis in the presence of bile acids. Whereas wild-type Teff cells upregulated Mdr1 in the ileum, those lacking Mdr1 displayed mucosal dysfunction and induced Crohn's disease-like ileitis following transfer into Rag1-/- hosts. Mdr1 mitigated oxidative stress and enforced homeostasis in Teff cells exposed to conjugated bile acids (CBAs), a class of liver-derived emulsifying agents that actively circulate through the ileal mucosa. Blocking ileal CBA reabsorption in transferred Rag1-/- mice restored Mdr1-deficient Teff cell homeostasis and attenuated ileitis. Further, a subset of ileal Crohn's disease patients displayed MDR1 loss of function. Together, these results suggest that coordinated interaction between mucosal Teff cells and CBAs in the ileum regulate intestinal immune homeostasis.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/immunology , Bile Acids and Salts/immunology , CD4-Positive T-Lymphocytes/immunology , Crohn Disease/immunology , Ileitis/immunology , Intestinal Mucosa/immunology , ATP Binding Cassette Transporter, Subfamily B, Member 1/deficiency , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Acridines/pharmacology , Adult , Animals , Bile Acids and Salts/metabolism , Bile Acids and Salts/pharmacology , Biological Transport , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/pathology , Crohn Disease/genetics , Crohn Disease/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Homeostasis/immunology , Humans , Ileitis/genetics , Ileitis/pathology , Ileum/immunology , Ileum/pathology , Immunity, Mucosal , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Oxidative Stress , Signal Transduction , Tetrahydroisoquinolines/pharmacology
13.
J Immunol ; 212(11): 1843-1854, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38568091

ABSTRACT

Intraepithelial lymphocytes (IELs) are T cells important for the maintenance of barrier integrity in the intestine. Colon IELs are significantly reduced in both MyD88-deficient mice and those lacking an intact microbiota, suggesting that MyD88-mediated detection of bacterial products is important for the recruitment and/or retention of these cells. Here, using conditionally deficient MyD88 mice, we show that myeloid cells are the key mediators of TCRαß+ IEL recruitment to the colon. Upon exposure to luminal bacteria, myeloid cells produce sphingosine-1-phosphate (S1P) in a MyD88-dependent fashion. TCRαß+ IEL recruitment may be blocked using the S1P receptor antagonist FTY720, confirming the importance of S1P in the recruitment of TCRαß+ IELs to the colon epithelium. Finally, using the TNFΔARE/+ model of Crohn's-like bowel inflammation, we show that disruption of colon IEL recruitment through myeloid-specific MyD88 deficiency results in reduced pathology. Our results illustrate one mechanism for recruitment of a subset of IELs to the colon.


Subject(s)
Colon , Intestinal Mucosa , Intraepithelial Lymphocytes , Lysophospholipids , Mice, Knockout , Myeloid Cells , Myeloid Differentiation Factor 88 , Receptors, Antigen, T-Cell, alpha-beta , Sphingosine , Animals , Lysophospholipids/metabolism , Mice , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Colon/immunology , Myeloid Differentiation Factor 88/metabolism , Myeloid Cells/immunology , Myeloid Cells/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Mice, Inbred C57BL , Fingolimod Hydrochloride/pharmacology , Crohn Disease/immunology
14.
Gastroenterology ; 166(5): 826-841.e19, 2024 05.
Article in English | MEDLINE | ID: mdl-38266738

ABSTRACT

BACKGROUND & AIMS: Incapacitated regulatory T cells (Tregs) contribute to immune-mediated diseases. Inflammatory Tregs are evident during human inflammatory bowel disease; however, mechanisms driving the development of these cells and their function are not well understood. Therefore, we investigated the role of cellular metabolism in Tregs relevant to gut homeostasis. METHODS: Using human Tregs, we performed mitochondrial ultrastructural studies via electron microscopy and confocal imaging, biochemical and protein analyses using proximity ligation assay, immunoblotting, mass cytometry and fluorescence-activated cell sorting, metabolomics, gene expression analysis, and real-time metabolic profiling utilizing the Seahorse XF analyzer. We used a Crohn's disease single-cell RNA sequencing dataset to infer the therapeutic relevance of targeting metabolic pathways in inflammatory Tregs. We examined the superior functionality of genetically modified Tregs in CD4+ T-cell-induced murine colitis models. RESULTS: Mitochondria-endoplasmic reticulum appositions, known to mediate pyruvate entry into mitochondria via voltage-dependent anion channel 1 (VDAC1), are abundant in Tregs. VDAC1 inhibition perturbed pyruvate metabolism, eliciting sensitization to other inflammatory signals reversible by membrane-permeable methyl pyruvate supplementation. Notably, interleukin (IL) 21 diminished mitochondria-endoplasmic reticulum appositions, resulting in enhanced enzymatic function of glycogen synthase kinase 3 ß, a putative negative regulator of VDAC1, and a hypermetabolic state that amplified Treg inflammatory response. Methyl pyruvate and glycogen synthase kinase 3 ß pharmacologic inhibitor (LY2090314) reversed IL21-induced metabolic rewiring and inflammatory state. Moreover, IL21-induced metabolic genes in Tregs in vitro were enriched in human Crohn's disease intestinal Tregs. Adoptively transferred Il21r-/- Tregs efficiently rescued murine colitis in contrast to wild-type Tregs. CONCLUSIONS: IL21 triggers metabolic dysfunction associated with Treg inflammatory response. Inhibiting IL21-induced metabolism in Tregs may mitigate CD4+ T-cell-driven chronic intestinal inflammation.


Subject(s)
Colitis , Mitochondria , Animals , Humans , Mice , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Chronic Disease , Colitis/immunology , Colitis/metabolism , Colitis/pathology , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Interleukins/metabolism , Interleukins/pharmacology , Mice, Inbred C57BL , Mitochondria/metabolism , T-Lymphocytes, Regulatory/immunology , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 1/genetics
15.
Gastroenterology ; 167(2): 315-332, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38490347

ABSTRACT

BACKGROUND & AIMS: Patients with inflammatory bowel disease (IBD) frequently develop extraintestinal manifestations (EIMs) that contribute substantially to morbidity. We assembled the largest multicohort data set to date to investigate the clinical, serologic, and genetic factors associated with EIM complications in IBD. METHODS: Data were available in 12,083 unrelated European ancestry IBD cases with presence or absence of EIMs (eg, ankylosing spondylitis [ankylosing spondylitis and sacroiliitis], primary sclerosing cholangitis [PSC], peripheral arthritis, and skin and ocular manifestations) across 4 cohorts (Cedars-Sinai Medical Center, National Institute for Diabetes and Digestive and Kidney Diseases IBD Genetics Consortium, Sinai Helmsley Alliance for Research Excellence Consortium, and Risk Stratification and Identification of Immunogenetic and Microbial Markers of Rapid Disease Progression in Children with Crohn's Disease cohort). Clinical and serologic parameters were analyzed by means of univariable and multivariable regression analyses using a mixed-effects model. Within-case logistic regression was performed to assess genetic associations. RESULTS: Most EIMs occurred more commonly in female subjects (overall EIM: P = 9.0E-05, odds ratio [OR], 1.2; 95% CI, 1.1-1.4), with CD (especially colonic disease location; P = 9.8E-09, OR, 1.7; 95% CI, 1.4-2.0), and in subjects who required surgery (both CD and UC; P = 3.6E-19, OR, 1.7; 95% CI, 1.5-1.9). Smoking increased risk of EIMs except for PSC, where there was a "protective" effect. Multiple serologic associations were observed, including with PSC (anti-nuclear cytoplasmic antibody; IgG and IgA, anti-Saccharomyces cerevisiae antibodies; and anti-flagellin) and any EIM (anti-nuclear cytoplasmic antibody; IgG and IgA, anti-Saccharomyces cerevisiae antibodies; and anti-Pseudomonas fluorescens-associated sequence). We identified genome-wide significant associations within major histocompatibility complex (ankylosing spondylitis and sacroiliitis, P = 1.4E-15; OR, 2.5; 95% CI, 2.0-3.1; PSC, P = 2.7E-10; OR, 2.8; 95% CI, 2.0-3.8; ocular, P = 2E-08, OR, 3.6; 95% CI, 2.3-5.6; and overall EIM, P = 8.4E-09; OR, 2.2; 95% CI, 1.7-2.9) and CPEB4 (skin, P = 2.7E-08; OR, 1.5; 95% CI, 1.3-1.8). Genetic associations implicated tumor necrosis factor, JAK-STAT, and IL6 as potential targets for EIMs. Contrary to previous reports, only 2% of our subjects had multiple EIMs and most co-occurrences were negatively correlated. CONCLUSIONS: We have identified demographic, clinical, and genetic associations with EIMs that revealed underlying mechanisms and implicated novel and existing drug targets-important steps toward a more personalized approach to IBD management.


Subject(s)
Cholangitis, Sclerosing , Colitis, Ulcerative , Crohn Disease , Humans , Female , Male , Adult , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/genetics , Cholangitis, Sclerosing/diagnosis , Cholangitis, Sclerosing/complications , Middle Aged , Colitis, Ulcerative/immunology , Colitis, Ulcerative/genetics , Colitis, Ulcerative/diagnosis , Crohn Disease/immunology , Crohn Disease/genetics , Crohn Disease/diagnosis , Adolescent , Risk Factors , Child , Spondylitis, Ankylosing/genetics , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/diagnosis , Spondylitis, Ankylosing/complications , Genetic Predisposition to Disease , Young Adult , Sex Factors , Skin Diseases/etiology , Skin Diseases/immunology , Skin Diseases/genetics , Eye Diseases/etiology , Eye Diseases/immunology , Eye Diseases/diagnosis , Eye Diseases/genetics , Eye Diseases/epidemiology , Phenotype , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/diagnosis , Logistic Models , Aged
16.
Nat Immunol ; 14(9): 927-36, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23892723

ABSTRACT

Mutations that result in loss of function of Nod2, an intracellular receptor for bacterial peptidoglycan, are associated with Crohn's disease. Here we found that the E3 ubiquitin ligase Pellino3 was an important mediator in the Nod2 signaling pathway. Pellino3-deficient mice had less induction of cytokines after engagement of Nod2 and had exacerbated disease in various experimental models of colitis. Furthermore, expression of Pellino3 was lower in the colons of patients with Crohn's disease. Pellino3 directly bound to the kinase RIP2 and catalyzed its ubiquitination. Loss of Pellino3 led to attenuation of Nod2-induced ubiquitination of RIP2 and less activation of the transcription factor NF-κB and mitogen-activated protein kinases (MAPKs). Our findings identify RIP2 as a substrate for Pellino3 and Pellino3 as an important mediator in the Nod2 pathway and regulator of intestinal inflammation.


Subject(s)
Colitis/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Ubiquitin-Protein Ligases/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Citrobacter rodentium/immunology , Colitis/genetics , Colitis/immunology , Colitis/microbiology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Male , Mice , Mice, Knockout , Middle Aged , Protein Binding , Protein Interaction Domains and Motifs , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Young Adult
17.
Nat Immunol ; 14(3): 221-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23334791

ABSTRACT

Innate lymphoid cells (ILCs) are effectors of innate immunity and regulators of tissue modeling. Recently identified ILC populations have a cytokine expression pattern that resembles that of the helper T cell subsets T(H)2, T(H)17 and T(H)22. Here we describe a distinct ILC subset similar to T(H)1 cells, which we call 'ILC1'. ILC1 cells expressed the transcription factor T-bet and responded to interleukin 12 (IL-12) by producing interferon-γ (IFN-γ). ILC1 cells were distinct from natural killer (NK) cells as they lacked perforin, granzyme B and the NK cell markers CD56, CD16 and CD94, and could develop from RORγt(+) ILC3 under the influence of IL-12. The frequency of the ILC1 subset was much higher in inflamed intestine of people with Crohn's disease, which indicated a role for these IFN-γ-producing ILC1 cells in the pathogenesis of gut mucosal inflammation.


Subject(s)
Crohn Disease/immunology , Interleukin-12/metabolism , Intestinal Mucosa/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , T-Box Domain Proteins/biosynthesis , Animals , CD56 Antigen/analysis , Cell Differentiation , Cells, Cultured , Colitis/immunology , Cytokines/immunology , Cytokines/metabolism , Granzymes/analysis , Humans , Inflammation , Interferon-gamma/biosynthesis , Intestinal Mucosa/metabolism , Intestines/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/metabolism , Lymphocytes/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily D/analysis , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Perforin/analysis , Receptors, IgG/analysis , Th1 Cells/immunology , Th1 Cells/metabolism
18.
Immunity ; 44(5): 1227-39, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27178470

ABSTRACT

Inflammatory intestinal diseases are characterized by abnormal immune responses and affect distinct locations of the gastrointestinal tract. Although the role of several immune subsets in driving intestinal pathology has been studied, a system-wide approach that simultaneously interrogates all major lineages on a single-cell basis is lacking. We used high-dimensional mass cytometry to generate a system-wide view of the human mucosal immune system in health and disease. We distinguished 142 immune subsets and through computational applications found distinct immune subsets in peripheral blood mononuclear cells and intestinal biopsies that distinguished patients from controls. In addition, mucosal lymphoid malignancies were readily detected as well as precursors from which these likely derived. These findings indicate that an integrated high-dimensional analysis of the entire immune system can identify immune subsets associated with the pathogenesis of complex intestinal disorders. This might have implications for diagnostic procedures, immune-monitoring, and treatment of intestinal diseases and mucosal malignancies.


Subject(s)
Celiac Disease/immunology , Crohn Disease/immunology , Image Cytometry/methods , Intestinal Mucosa/immunology , Lymphocyte Subsets/immunology , Lymphocytes/immunology , Lymphocytes/physiology , Lymphoma, T-Cell/immunology , Adult , Aged , Celiac Disease/diagnosis , Cohort Studies , Computational Biology , Crohn Disease/diagnosis , Female , HEK293 Cells , Humans , Immunologic Tests , Lymphoma, T-Cell/diagnosis , Male , Middle Aged , Monitoring, Immunologic , Organ Specificity , Single-Cell Analysis
19.
Nature ; 568(7752): 405-409, 2019 04.
Article in English | MEDLINE | ID: mdl-30944470

ABSTRACT

Interleukin (IL)-2 is a pleiotropic cytokine that is necessary to prevent chronic inflammation in the gastrointestinal tract1-4. The protective effects of IL-2 involve the generation, maintenance and function of regulatory T (Treg) cells4-8, and the use of low doses of IL-2 has emerged as a potential therapeutic strategy for patients with inflammatory bowel disease9. However, the cellular and molecular pathways that control the production of IL-2 in the context of intestinal health are undefined. Here we show, in a mouse model, that IL-2 is acutely required to maintain Treg cells and immunological homeostasis throughout the gastrointestinal tract. Notably, lineage-specific deletion of IL-2 in T cells did not reduce Treg cells in the small intestine. Unbiased analyses revealed that, in the small intestine, group-3 innate lymphoid cells (ILC3s) are the dominant cellular source of IL-2, which is induced selectively by IL-1ß. Macrophages in the small intestine produce IL-1ß, and activation of this pathway involves MYD88- and NOD2-dependent sensing of the microbiota. Our loss-of-function studies show that ILC3-derived IL-2 is essential for maintaining Treg cells, immunological homeostasis and oral tolerance to dietary antigens in the small intestine. Furthermore, production of IL-2 by ILC3s was significantly reduced in the small intestine of patients with Crohn's disease, and this correlated with lower frequencies of Treg cells. Our results reveal a previously unappreciated pathway in which a microbiota- and IL-1ß-dependent axis promotes the production of IL-2 by ILC3s to orchestrate immune regulation in the intestine.


Subject(s)
Immunity, Innate/immunology , Interleukin-2/immunology , Intestines/cytology , Intestines/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens/administration & dosage , Antigens/immunology , Crohn Disease/immunology , Crohn Disease/metabolism , Crohn Disease/pathology , Female , Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Humans , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Interleukin-2/deficiency , Interleukin-2/metabolism , Intestine, Small/cytology , Intestine, Small/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Nod2 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism , T-Lymphocytes, Regulatory/classification , T-Lymphocytes, Regulatory/metabolism
20.
Genes Immun ; 25(3): 209-218, 2024 06.
Article in English | MEDLINE | ID: mdl-38789829

ABSTRACT

The pathogenesis of Crohn's disease (CD) involves abnormal immune cell infiltration and dysregulated immune response. Therefore, thorough research on immune cell abnormalities in CD is crucial for improved treatment of this disease. Single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data of CD were obtained from the Gene Expression Omnibus (GEO) database. Cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT), weighted gene co-expression network analysis (WGCNA), protein-protein interaction (PPI) networks evaluated the proportion of immune infiltrating cells, constructed co-expression network and identified key genes, respectively. Based on the dataset (GSE134809), 15 cell clusters were defined and labeled as different cell types. Among the 11 modules, the yellow module had the closest relationship with plasma cells (cluster 5). Confirmed using RNA sequencing and IHC assay, the expression of COL5A2 in CD samples was higher than that in control samples. Furthermore, the COL5A2 protein expression remarkably decreased in the group of patients who responded to anti-tumor necrosis factor (TNF) treatments, compared to the non-response group. The comprehensive analyses described here provided novel insight into the landscape of CD-associated immune environment. In addition, COL5A2 were identified as potential diagnostic indicators for CD, as well as promising predictive markers for CD patients.


Subject(s)
Collagen Type V , Crohn Disease , Crohn Disease/immunology , Crohn Disease/genetics , Humans , Collagen Type V/genetics , Collagen Type V/immunology , Protein Interaction Maps , Biomarkers , Gene Regulatory Networks
SELECTION OF CITATIONS
SEARCH DETAIL