Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Immunity ; 54(11): 2437-2439, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34758332

ABSTRACT

Microbiota-specific T cell responses have been identified for select microbes, but individual T cell receptor repertoire differences make characterizing responses across populations difficult. In this issue of Immunity, Muschaweck et al. establish a system allowing for reproducible responses between individual mice, a powerful tool for characterizing microbiota directed immunity.


Subject(s)
Microbiota , T-Lymphocytes , Animals , Mice , Receptors, Antigen, T-Cell
2.
Immunity ; 49(3): 389-391, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30231979

ABSTRACT

Intestinal homeostasis requires microbial recognition that results in appropriate responses to commensals and pathogens. In this issue of Immunity, Price et al. (2018) map the in vivo expression of five toll-like receptors (TLR) in intestinal epithelia, revealing distinct spatio-temporal expression patterns that shape responses to TLR ligands.


Subject(s)
Intestinal Mucosa , Toll-Like Receptors , Immunity , Ligands
3.
Immunity ; 49(6): 1077-1089.e5, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30552020

ABSTRACT

Inflammatory bowel disease (IBD) results from a dysregulated interaction between the microbiota and a genetically susceptible host. Genetic studies have linked TNFSF15 polymorphisms and its protein TNF-like ligand 1A (TL1A) with IBD, but the functional role of TL1A is not known. Here, we found that adherent IBD-associated microbiota induced TL1A release from CX3CR1+ mononuclear phagocytes (MNPs). Using cell-specific genetic deletion models, we identified an essential role for CX3CR1+MNP-derived TL1A in driving group 3 innate lymphoid cell (ILC3) production of interleukin-22 and mucosal healing during acute colitis. In contrast to this protective role in acute colitis, TL1A-dependent expression of co-stimulatory molecule OX40L in MHCII+ ILC3s during colitis led to co-stimulation of antigen-specific T cells that was required for chronic T cell colitis. These results identify a role for ILC3s in activating intestinal T cells and reveal a central role for TL1A in promoting ILC3 barrier immunity during colitis.


Subject(s)
Colitis/immunology , Immunity, Innate/immunology , Lymphocytes/immunology , Microbiota/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Adult , Aged , Animals , Colitis/genetics , Colitis/metabolism , Female , Humans , Immunity, Innate/genetics , Interleukins/genetics , Interleukins/immunology , Interleukins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocytes/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microbiota/physiology , Middle Aged , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Young Adult , Interleukin-22
4.
Immunity ; 49(1): 151-163.e5, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29980437

ABSTRACT

The intestinal barrier is vulnerable to damage by microbiota-induced inflammation that is normally restrained through mechanisms promoting homeostasis. Such disruptions contribute to autoimmune and inflammatory diseases including inflammatory bowel disease. We identified a regulatory loop whereby, in the presence of the normal microbiota, intestinal antigen-presenting cells (APCs) expressing the chemokine receptor CX3CR1 reduced expansion of intestinal microbe-specific T helper 1 (Th1) cells and promoted generation of regulatory T cells responsive to food antigens and the microbiota itself. We identified that disruption of the microbiota resulted in CX3CR1+ APC-dependent inflammatory Th1 cell responses with increased pathology after pathogen infection. Colonization with microbes that can adhere to the epithelium was able to compensate for intestinal microbiota loss, indicating that although microbial interactions with the epithelium can be pathogenic, they can also activate homeostatic regulatory mechanisms. Our results identify a cellular mechanism by which the microbiota limits intestinal inflammation and promotes tissue homeostasis.


Subject(s)
CX3C Chemokine Receptor 1/metabolism , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Mononuclear Phagocyte System/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Antigen Presentation , Bacterial Adhesion/immunology , Disease Models, Animal , Female , Homeostasis , Immune Tolerance , Immunity, Mucosal , Inflammation/immunology , Inflammatory Bowel Diseases/immunology , Interleukin-10/immunology , Interleukin-10/metabolism , Intestinal Mucosa/microbiology , Male , Mice , RAW 264.7 Cells
5.
Nature ; 594(7863): 413-417, 2021 06.
Article in English | MEDLINE | ID: mdl-33981034

ABSTRACT

Humans and their microbiota have coevolved a mutually beneficial relationship in which the human host provides a hospitable environment for the microorganisms and the microbiota provides many advantages for the host, including nutritional benefits and protection from pathogen infection1. Maintaining this relationship requires a careful immune balance to contain commensal microorganisms within the lumen while limiting inflammatory anti-commensal responses1,2. Antigen-specific recognition of intestinal microorganisms by T cells has previously been described3,4. Although the local environment shapes the differentiation of effector cells3-5 it is unclear how microbiota-specific T cells are educated in the thymus. Here we show that intestinal colonization in early life leads to the trafficking of microbial antigens from the intestine to the thymus by intestinal dendritic cells, which then induce the expansion of microbiota-specific T cells. Once in the periphery, microbiota-specific T cells have pathogenic potential or can protect against related pathogens. In this way, the developing microbiota shapes and expands the thymic and peripheral T cell repertoire, allowing for enhanced recognition of intestinal microorganisms and pathogens.


Subject(s)
Dendritic Cells/immunology , Gastrointestinal Microbiome/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Aging/immunology , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , CX3C Chemokine Receptor 1/metabolism , DNA, Bacterial/analysis , Dendritic Cells/metabolism , Escherichia coli/immunology , Female , Male , Mice , Organ Specificity , Salmonella/immunology , Symbiosis/immunology , Thymus Gland/metabolism
6.
Trends Immunol ; 40(3): 177-179, 2019 03.
Article in English | MEDLINE | ID: mdl-30718047

ABSTRACT

In a recent study, Harrison et al. (Science 2019;363;eaat6280) report that RORγt-expressing skin commensal-specific T cells rapidly respond to tissue wounding by producing type 2 T helper cell (Th2) cytokines in mice. The cells constitutively coexpress GATA-3 and type 2 cytokine mRNAs that are translated after injury. These T cells act as sentinels, linking T cell receptor (TCR) recognition of commensals, tissue damage, and wound repair.


Subject(s)
Microbiota/immunology , Th2 Cells/immunology , Wound Healing , Animals , Cytokines/metabolism , Humans , Immunity, Cellular , Mice , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Antigen, T-Cell/metabolism
7.
Immunity ; 36(2): 276-87, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22306017

ABSTRACT

Microbial penetration of the intestinal epithelial barrier triggers inflammatory responses that include induction of the bactericidal C-type lectin RegIIIγ. Systemic administration of flagellin, a bacterial protein that stimulates Toll-like receptor 5 (TLR5), induces epithelial expression of RegIIIγ and protects mice from intestinal colonization with antibiotic-resistant bacteria. Flagellin-induced RegIIIγ expression is IL-22 dependent, but how TLR signaling leads to IL-22 expression is incompletely defined. By using conditional depletion of lamina propria dendritic cell (LPDC) subsets, we demonstrated that CD103(+)CD11b(+) LPDCs, but not monocyte-derived CD103(-)CD11b(+) LPDCs, expressed high amounts of IL-23 after bacterial flagellin administration and drove IL-22-dependent RegIIIγ production. Maximal expression of IL-23 subunits IL-23p19 and IL-12p40 occurred within 60 min of exposure to flagellin. IL-23 subsequently induced a burst of IL-22 followed by sustained RegIIIγ expression. Thus, CD103(+)CD11b(+) LPDCs, in addition to promoting long-term tolerance to ingested antigens, also rapidly produce IL-23 in response to detection of flagellin in the lamina propria.


Subject(s)
Dendritic Cells/immunology , Flagellin/immunology , Interleukin-23/biosynthesis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Dendritic Cells/classification , Flagellin/administration & dosage , Immunity, Innate , Immunity, Mucosal , Integrin alpha Chains/metabolism , Interleukin-23/deficiency , Interleukin-23/genetics , Interleukins/biosynthesis , Interleukins/deficiency , Interleukins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatitis-Associated Proteins , Proteins/genetics , Signal Transduction/immunology , Toll-Like Receptor 5/deficiency , Toll-Like Receptor 5/genetics , Toll-Like Receptor 5/metabolism , Up-Regulation , Interleukin-22
8.
Nature ; 494(7435): 116-20, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23334413

ABSTRACT

The intestinal microbiota has a critical role in immune system and metabolic homeostasis, but it must be tolerated by the host to avoid inflammatory responses that can damage the epithelial barrier separating the host from the luminal contents. Breakdown of this regulation and the resulting inappropriate immune response to commensals are thought to lead to the development of inflammatory bowel diseases such as Crohn's disease and ulcerative colitis. We proposed that the intestinal immune system is instructed by the microbiota to limit responses to luminal antigens. Here we demonstrate in mice that, at steady state, the microbiota inhibits the transport of both commensal and pathogenic bacteria from the lumen to a key immune inductive site, the mesenteric lymph nodes (MLNs). However, in the absence of Myd88 or under conditions of antibiotic-induced dysbiosis, non-invasive bacteria were trafficked to the MLNs in a CCR7-dependent manner, and induced both T-cell responses and IgA production. Trafficking was carried out by CX(3)CR1(hi) mononuclear phagocytes, an intestinal-cell population previously reported to be non-migratory. These findings define a central role for commensals in regulating the migration to the MLNs of CX(3)CR1(hi) mononuclear phagocytes endowed with the ability to capture luminal bacteria, thereby compartmentalizing the intestinal immune response to avoid inflammation.


Subject(s)
Immunity, Mucosal/immunology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Mesentery/immunology , Metagenome/physiology , Phagocytes/metabolism , Receptors, Chemokine/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Antigens, Bacterial/immunology , CX3C Chemokine Receptor 1 , Cell Movement , Dendritic Cells/cytology , Dendritic Cells/immunology , Immunity, Mucosal/drug effects , Immunoglobulin A/immunology , Inflammation/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Metagenome/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/deficiency , Myeloid Differentiation Factor 88/metabolism , Phagocytes/cytology , Phagocytes/immunology , Phagocytes/microbiology , Phagocytosis , Receptors, CCR7/deficiency , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Salmonella/cytology , Salmonella/drug effects , Salmonella/immunology , T-Lymphocytes/immunology
9.
JCI Insight ; 8(3)2023 02 08.
Article in English | MEDLINE | ID: mdl-36538527

ABSTRACT

Chronic exposure to high-fat diets (HFD) worsens intestinal disease pathology, but acute effects of HFD in tissue damage remain unclear. Here, we used short-term HFD feeding in a model of intestinal injury and found sustained damage with increased cecal dead neutrophil accumulation, along with dietary lipid accumulation. Neutrophil depletion rescued enhanced pathology. Macrophages from HFD-treated mice showed reduced capacity to engulf dead neutrophils. Macrophage clearance of dead neutrophils activates critical barrier repair and antiinflammatory pathways, including IL-10, which was lost after acute HFD feeding and intestinal injury. IL-10 overexpression restored intestinal repair after HFD feeding and intestinal injury. Macrophage exposure to lipids from the HFD prevented tethering and uptake of apoptotic cells and Il10 induction. Milk fat globule-EGF factor 8 (MFGE8) is a bridging molecule that facilitates macrophage uptake of dead cells. MFGE8 also facilitates lipid uptake, and we demonstrate that dietary lipids interfere with MFGE8-mediated macrophage apoptotic neutrophil uptake and subsequent Il10 production. Our findings demonstrate that HFD promotes intestinal pathology by interfering with macrophage clearance of dead neutrophils, leading to unresolved tissue damage.


Subject(s)
Diet, High-Fat , Interleukin-10 , Mice , Animals , Intestines , Macrophages/physiology , Lipids
10.
J Exp Med ; 220(2)2023 02 06.
Article in English | MEDLINE | ID: mdl-36367776

ABSTRACT

Immune checkpoint blockade (ICB) has revolutionized cancer treatment, yet quality of life and continuation of therapy can be constrained by immune-related adverse events (irAEs). Limited understanding of irAE mechanisms hampers development of approaches to mitigate their damage. To address this, we examined whether mice gained sensitivity to anti-CTLA-4 (αCTLA-4)-mediated toxicity upon disruption of gut homeostatic immunity. We found αCTLA-4 drove increased inflammation and colonic tissue damage in mice with genetic predisposition to intestinal inflammation, acute gastrointestinal infection, transplantation with a dysbiotic fecal microbiome, or dextran sodium sulfate administration. We identified an immune signature of αCTLA-4-mediated irAEs, including colonic neutrophil accumulation and systemic interleukin-6 (IL-6) release. IL-6 blockade combined with antibiotic treatment reduced intestinal damage and improved αCTLA-4 therapeutic efficacy in inflammation-prone mice. Intestinal immune signatures were validated in biopsies from patients with ICB colitis. Our work provides new preclinical models of αCTLA-4 intestinal irAEs, mechanistic insights into irAE development, and potential approaches to enhance ICB efficacy while mitigating irAEs.


Subject(s)
Colitis , Interleukin-6 , Mice , Animals , Quality of Life , Colitis/pathology , Immunotherapy , Inflammation
11.
Gut Microbes ; 14(1): 2014772, 2022.
Article in English | MEDLINE | ID: mdl-34989321

ABSTRACT

The gut microbiota is essential for maintenance and repair of the intestinal epithelial barrier. As shifts in both intestinal epithelial barrier function and microbiota composition are found in inflammatory bowel disease patients, it is critical to understand the role of distinct bacteria in regulating barrier repair. We identified a mouse commensal E. coli isolate, GDAR2-2, that protects mice from Citrobacter rodentium infection and dextran sulfate sodium-induced colitis. Colonization with GDAR2-2 in mice resulted in expansion of CX3CR1+ mononuclear phagocytes, including CX3CR1+ macrophages/dendritic cells and monocytes, along with IL-22-secreting type 3 innate lymphoid cells and improved epithelial barrier function. In vitro co-culture of macrophages with GDAR2-2 resulted in IL-1ß production. In vivo, protection after GDAR2-2 colonization was lost after depletion of CX3CR1+ MNPs, or blockade of IL-1ß or IL-22. We further identified human commensal E. coli isolates that similarly protect mice from C. rodentium infection through CX3CR1+ MNP and IL-1ß production. Together, these findings demonstrate an unexpected role for commensal bacteria in promoting IL-1ß secretion to support intestinal barrier repair.


Subject(s)
Colitis/metabolism , Colitis/physiopathology , Gastrointestinal Microbiome , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Symbiosis , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Colitis/genetics , Colitis/microbiology , Humans , Interleukin-18/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL
12.
Gut Microbes ; 14(1): 2119054, 2022.
Article in English | MEDLINE | ID: mdl-36062329

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic life-long inflammatory disease affecting almost 2 million Americans. Although new biologic therapies have been developed, the standard medical treatment fails to selectively control the dysregulated immune pathways involved in chronic colonic inflammation. Further, IBD patients with uncontrolled colonic inflammation are at a higher risk for developing colorectal cancer (CRC). Intestinal microbes can impact many immune functions, and here we asked if they could be used to improve intestinal inflammation. By utilizing an intestinal adherent E. coli that we find increases IL-10 producing macrophages, we were able to limit intestinal inflammation and restrict tumor formation. Macrophage IL-10 along with IL-10 signaling to the intestinal epithelium were required for protection in both inflammation and tumor development. Our work highlights that administration of immune modulating microbes can improve intestinal outcomes by altering tissue inflammation.


Subject(s)
Colitis-Associated Neoplasms , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Microbiota , Animals , Disease Models, Animal , Escherichia coli , Humans , Inflammation , Inflammatory Bowel Diseases/therapy , Interleukin-10 , Macrophages
13.
Gut Microbes ; 13(1): 1-14, 2021.
Article in English | MEDLINE | ID: mdl-33929291

ABSTRACT

During both health and disease, a coordinated response between the epithelium, immune system, and enteric nervous system is required for proper intestinal function. While each system responds to a number of common stimuli, their coordinated responses support digestion as well as responses and recovery following injury or pathogenic infections. In this review, we discuss how individual responses to common signals work together to support these critical functions.


Subject(s)
Enteric Nervous System/physiology , Epithelium/physiology , Gastrointestinal Microbiome , Host Microbial Interactions , Immunity , Intestines/immunology , Intestines/microbiology , Humans , Intestinal Mucosa/physiology
14.
Cell Host Microbe ; 29(4): 607-619.e8, 2021 04 14.
Article in English | MEDLINE | ID: mdl-33539767

ABSTRACT

Adherent-invasive E. coli (AIEC) are enriched in the intestinal microbiota of patients with Crohn's disease (CD) and promote intestinal inflammation. Yet, how AIEC metabolism of nutrients impacts intestinal homeostasis is poorly defined. Here, we show that AIEC encoding the large subunit of propanediol dehydratase (PduC), which facilitates the utilization of fucose fermentation product 1,2-propanediol, are increased in the microbiome of CD patients and drive AIEC-induced intestinal T cell inflammation. In murine models, CX3CR1+ mononuclear phagocytes (MNP) are required for PduC-dependent induction of T helper 17 (Th17) cells and interleukin-1ß (IL-1ß) production that leads to AIEC-induced inflammatory colitis. Activation of this inflammatory cascade requires the catalytic activity of PduC to generate propionate, which synergizes with lipopolysaccharide (LPS) to induce IL-1ß by MNPs. Disrupting fucose availability limits AIEC-induced propionate production and intestinal inflammation. These findings identify MNPs as metabolic sensors linking AIEC metabolism with intestinal inflammation and identify microbial metabolism as a potential therapeutic target in Crohn's disease treatment.


Subject(s)
Crohn Disease/metabolism , Escherichia coli Infections/metabolism , Escherichia coli/metabolism , Inflammation/metabolism , Intestines/immunology , Phagocytes/metabolism , Propylene Glycols/metabolism , Animals , Bacterial Adhesion , Crohn Disease/microbiology , Escherichia coli Infections/microbiology , Female , Host-Pathogen Interactions , Humans , Immunity , Interleukin-1beta , Intestinal Mucosa/metabolism , Intestines/microbiology , Male , Mice , Phagocytes/immunology , Th17 Cells
15.
Front Immunol ; 11: 597966, 2020.
Article in English | MEDLINE | ID: mdl-33424846

ABSTRACT

Autoimmune diseases and chronic inflammatory disorders are characterized by dysregulated immune responses resulting in excessive and uncontrolled tissue inflammation. Multiple factors including genetic variation, environmental stimuli, and infection are all thought to contribute to continued inflammation and pathology. Current evidence supports the microbiota as one such factor with emerging data linking commensal organisms to the onset and progression of disease. In this review, we will discuss links between the microbiota and specific diseases as well as highlight common pathways that link intestinal microbes with multiple autoimmune and inflammatory diseases.


Subject(s)
Autoimmune Diseases/etiology , Autoimmunity , Disease Susceptibility , Gastrointestinal Microbiome , Inflammation/etiology , Animals , Autoantibodies/immunology , Autoimmune Diseases/diagnosis , Autoimmune Diseases/metabolism , Disease Susceptibility/immunology , Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Humans , Inflammation/diagnosis , Inflammation/metabolism , Organ Specificity/immunology
16.
Cell Microbiol ; 10(8): 1646-61, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18397382

ABSTRACT

Host resistance against Salmonella enterica serovar Typhimurium (S. Typhimurium) is mediated by natural resistance-associated macrophage protein 1 (Nramp1/Slc11a1). Nramp1 is critical to host defence, as mice lacking Nramp1 fail to control bacterial replication and succumb to low doses of S. Typhimurium. Despite this crucial role, the mechanisms underlying Nramp1's protective effects are unclear. Dendritic cells (DCs) that sample the intestinal lumen are among the first cells encountered by S. Typhimurium following oral infection and act as a conduit for S. Typhimurium to cross the intestinal epithelial barrier. We report that DCs, including intestinal, splenic and bone marrow-derived DCs (BMDCs), express Nramp1 protein. In the small intestine, Nramp1 expression is greater in a subset of DCs (CD11c(+)CD103(-)) characterized by the elevated expression of pro-inflammatory cytokines in response to bacterial products. While Nramp1 expression did not affect S. Typhimurium replication in BMDCs, infected Nramp1+/+ BMDCs and intestinal CD11c(+)CD103(-) DCs secreted more inflammatory cytokines (IL-6, IL-12 and TNF-alpha) than Nramp1-/-, suggesting that Nramp1 expression may promote a more rapid inflammatory response following infection. Collectively, these findings reveal a new role for DCs and Nramp1 in modulating the host inflammatory response to S. Typhimurium.


Subject(s)
Cation Transport Proteins/immunology , Dendritic Cells/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Animals , Cytokines/immunology , Intestine, Small/microbiology , Mice , Mucous Membrane/immunology
17.
Gut Microbes ; 10(4): 540-546, 2019 Jul 04.
Article in English | MEDLINE | ID: mdl-33970784

ABSTRACT

Intestinal damage driven by unrestricted immune responses against the intestinal microbiota can lead to the development of inflammatory diseases including inflammatory bowel disease. How such breakdown in tolerance occurs alongside the mechanisms to reinforce homeostasis with the microbiota are a focus of many studies. Our recent work demonstrates coordinated interactions between intact microbiota and CX3CR1 expressing intestinal antigen presenting cells (APCs) that limits T helper 1 cell responses and promotes differentiation of regulatory T cells (Treg) against intestinal antigens including pathogens, soluble proteins and the microbiota itself. We find a microbial attachment to intestinal epithelial cells is necessary to support these anti-inflammatory immune functions. In this addendum, we discuss how our findings enhance understanding of microbiota-directed homeostatic functions of the intestinal immune system and implications of modulating this interaction in ameliorating inflammatory disease.

18.
Front Immunol ; 10: 75, 2019.
Article in English | MEDLINE | ID: mdl-30761142

ABSTRACT

Epidemiological evidence finds cigarette smoking is a common risk factor for a number of diseases, not only in the lung but also in other tissues, such as the gastrointestinal tract. While it is well-documented that smoking directly drives lung inflammatory disease, how it promotes disease in peripheral tissues is incompletely understood. In this study, we utilized a mouse model of short-term smoke exposure and found increased Th17 cells and neutrophilia in the lung as well as in the circulation. Following intestinal inflammatory challenge, smoke exposed mice showed increased pathology which corresponds to enhanced intestinal Th17 cells, ILC3 and neutrophils within intestinal tissue. Using cellular depletion and genetic deficiencies, we define a cellular loop by which IL-17A and downstream neutrophils drive cigarette smoke-enhanced intestinal inflammation. Collectively, cigarette smoke induced local lung Th17 responses lead to increased systemic susceptibility to inflammatory insult through enhanced circulating neutrophils. These data demonstrate a cellular pathway by which inflammatory challenge in the lung can sensitize the intestine to enhanced pathological innate and adaptive immune responses.


Subject(s)
Intestines/drug effects , Lung/drug effects , Neutrophils/drug effects , Smoke/adverse effects , Th17 Cells/drug effects , Tobacco Products , Animals , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Cytokines/genetics , Cytokines/immunology , Female , Intestines/immunology , Intestines/pathology , Lung/immunology , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/immunology , Th17 Cells/immunology
19.
Sci Transl Med ; 9(376)2017 02 08.
Article in English | MEDLINE | ID: mdl-28179509

ABSTRACT

Peripheral spondyloarthritis (SpA) is a common extraintestinal manifestation in patients with active inflammatory bowel disease (IBD) characterized by inflammatory enthesitis, dactylitis, or synovitis of nonaxial joints. However, a mechanistic understanding of the link between intestinal inflammation and SpA has yet to emerge. We evaluated and functionally characterized the fecal microbiome of IBD patients with or without peripheral SpA. Coupling the sorting of immunoglobulin A (IgA)-coated microbiota with 16S ribosomal RNA-based analysis (IgA-seq) revealed a selective enrichment in IgA-coated Escherichia coli in patients with Crohn's disease-associated SpA (CD-SpA) compared to CD alone. E. coli isolates from CD-SpA-derived IgA-coated bacteria were similar in genotype and phenotype to an adherent-invasive E. coli (AIEC) pathotype. In comparison to non-AIEC E. coli, colonization of germ-free mice with CD-SpA E. coli isolates induced T helper 17 cell (TH17) mucosal immunity, which required the virulence-associated metabolic enzyme propanediol dehydratase (pduC). Modeling the increase in mucosal and systemic TH17 immunity we observed in CD-SpA patients, colonization of interleukin-10-deficient or K/BxN mice with CD-SpA-derived E. coli lead to more severe colitis or inflammatory arthritis, respectively. Collectively, these data reveal the power of IgA-seq to identify immunoreactive resident pathosymbionts that link mucosal and systemic TH17-dependent inflammation and offer microbial and immunophenotype stratification of CD-SpA that may guide medical and biologic therapy.


Subject(s)
Crohn Disease/immunology , Crohn Disease/microbiology , Escherichia coli/metabolism , Immunoglobulin A/metabolism , Inflammation/pathology , Spondylarthritis/immunology , Spondylarthritis/microbiology , Th17 Cells/immunology , Animals , Biomarkers/metabolism , Colitis/chemically induced , Colitis/immunology , Colitis/microbiology , Crohn Disease/complications , Dextran Sulfate , Epithelium/immunology , Escherichia coli/isolation & purification , Humans , Immunity, Mucosal , Immunophenotyping , Inflammation/complications , Interleukin-10/metabolism , Interleukin-23/metabolism , Intestines/microbiology , Joints/pathology , Mice, Inbred C57BL , Spondylarthritis/complications
20.
Cell Host Microbe ; 18(4): 383-5, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468738

ABSTRACT

While acute infections cause short-term tissue damage, their long-term impact remains unknown. In a recent publication in Cell, Morais da Fonseca et al. (2015) demonstrate disruption of mesenteric lymph nodes and associated lymphatics after Yersinia pseudotuberculosis infection and clearance. This leads to chronic inflammation and an inability to initiate subsequent intestinal immune responses.


Subject(s)
Gastrointestinal Microbiome , Immune System Diseases/microbiology , Immune System Diseases/pathology , Lymphatic Diseases/pathology , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis/physiology , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL