Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Brain ; 144(8): 2361-2374, 2021 09 04.
Article in English | MEDLINE | ID: mdl-34145876

ABSTRACT

Autoantibodies are a hallmark of numerous neurological disorders, including multiple sclerosis, autoimmune encephalitides and neuromyelitis optica. Whilst well understood in peripheral myeloid cells, the pathophysiological significance of autoantibody-induced Fc receptor signalling in microglia remains unknown, in part due to the lack of a robust in vivo model. Moreover, the application of therapeutic antibodies for neurodegenerative disease also highlights the importance of understanding Fc receptor signalling in microglia. Here, we describe a novel in vivo experimental paradigm that allows for selective engagement of Fc receptors within the CNS by peripherally injecting anti-myelin oligodendrocyte glycoprotein (MOG) monoclonal antibodies into normal wild-type mice. MOG antigen-bound immunoglobulins were detected throughout the CNS and triggered a rapid and tightly regulated proliferative response in both brain and spinal cord microglia. This microglial response was abrogated when anti-MOG antibodies were deprived of Fc receptor effector function or injected into Fcγ receptor knockout mice and was associated with the downregulation of Fc receptors in microglia, but not peripheral myeloid cells, establishing that this response was dependent on central Fc receptor engagement. Downstream of the Fc receptors, BTK was a required signalling node for this response, as microglia proliferation was amplified in BtkE41K knock-in mice expressing a constitutively active form of the enzyme and blunted in mice treated with a CNS-penetrant small molecule inhibitor of BTK. Finally, this response was associated with transient and stringently regulated changes in gene expression predominantly related to cellular proliferation, which markedly differed from transcriptional programs typically associated with Fc receptor engagement in peripheral myeloid cells. Together, these results establish a physiologically-meaningful functional response to Fc receptor and BTK signalling in microglia, while providing a novel in vivo tool to further dissect the roles of microglia-specific Fc receptor and BTK-driven responses to both pathogenic and therapeutic antibodies in CNS homeostasis and disease.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Autoantibodies/immunology , Brain/pathology , Microglia/pathology , Myelin-Oligodendrocyte Glycoprotein/immunology , Receptors, Fc/metabolism , Spinal Cord/pathology , Animals , Brain/immunology , Brain/metabolism , Cell Proliferation/physiology , Mice , Microglia/immunology , Microglia/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
2.
Elife ; 92020 06 24.
Article in English | MEDLINE | ID: mdl-32579115

ABSTRACT

To date, microglia subsets in the healthy CNS have not been identified. Utilizing autofluorescence (AF) as a discriminating parameter, we identified two novel microglia subsets in both mice and non-human primates, termed autofluorescence-positive (AF+) and negative (AF-). While their proportion remained constant throughout most adult life, the AF signal linearly and specifically increased in AF+ microglia with age and correlated with a commensurate increase in size and complexity of lysosomal storage bodies, as detected by transmission electron microscopy and LAMP1 levels. Post-depletion repopulation kinetics revealed AF- cells as likely precursors of AF+ microglia. At the molecular level, the proteome of AF+ microglia showed overrepresentation of endolysosomal, autophagic, catabolic, and mTOR-related proteins. Mimicking the effect of advanced aging, genetic disruption of lysosomal function accelerated the accumulation of storage bodies in AF+ cells and led to impaired microglia physiology and cell death, suggestive of a mechanistic convergence between aging and lysosomal storage disorders.


Microglia are a unique type of immune cell found in the brain and spinal cord. Their job is to support neurons, defend against invading microbes, clear debris and remove dying neurons by engulfing them. Despite these diverse roles, scientists have long believed that there is only a single type of microglial cell, which adapts to perform whatever task is required. But more recent evidence suggests that this is not the whole story. Burns et al. now show that we can distinguish two subtypes of microglia based on a property called autofluorescence. This is the tendency of cells and tissues to emit light of one color after they have absorbed light of another. Burns et al. show that about 70% of microglia in healthy mouse and monkey brains display autofluorescence. However, about 30% of microglia show no autofluorescence at all. This suggests that there are two subtypes of microglia: autofluorescence-positive and autofluorescence-negative. But does this difference have any implications for how the microglia behave? Autofluorescence occurs because specific substances inside the cells absorb light. In the case of microglia, electron microscopy revealed that autofluorescence was caused by structures within the cell called lysosomal storage bodies accumulating certain materials. The stored material included fat molecules, cholesterol crystals and other substances that are typical of disorders that affect these compartments. Burns et al. show that autofluorescent microglia contain larger amounts of proteins involved in storing and digesting waste materials than their non-autofluorescent counterparts. Moreover, as the brain ages, lysosomal storage material builds up inside autofluorescent microglia, which increase their autofluorescence as a result. Unfortunately, this accumulation of cellular debris also makes it harder for the microglia to perform their tasks. Increasing evidence suggests that the accumulation of waste materials inside the brain contributes to diseases of aging. Future work should examine how autofluorescent microglia behave in animal models of neurodegenerative diseases. If these cells do help protect the brain from the effects of aging, targeting them could be a new strategy for treating aging-related diseases.


Subject(s)
Aging , Brain/metabolism , Microglia/metabolism , Animals , Autophagy , Disease Models, Animal , Endosomes/metabolism , Female , Lysosomal Membrane Proteins/metabolism , Lysosomes/metabolism , Macaca mulatta , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Myelin Sheath/chemistry , Neurons/metabolism , Phagocytosis , Proteomics , Reactive Oxygen Species/metabolism , Receptors, Fc/metabolism , Receptors, Immunologic/metabolism
3.
Proc Natl Acad Sci U S A ; 117(20): 10946-10957, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32350141

ABSTRACT

Four decades ago, it was identified that muramyl dipeptide (MDP), a peptidoglycan-derived bacterial cell wall component, could display immunosuppressive functions in animals through mechanisms that remain unexplored. We sought to revisit these pioneering observations because mutations in NOD2, the gene encoding the host sensor of MDP, are associated with increased risk of developing the inflammatory bowel disease Crohn's disease, thus suggesting that the loss of the immunomodulatory functions of NOD2 could contribute to the development of inflammatory disease. Here, we demonstrate that intraperitoneal (i.p.) administration of MDP triggered regulatory T cells and the accumulation of a population of tolerogenic CD103+ dendritic cells (DCs) in the spleen. This was found to occur not through direct sensing of MDP by DCs themselves, but rather via the production of the cytokine GM-CSF, another factor with an established regulatory role in Crohn's disease pathogenesis. Moreover, we demonstrate that populations of CD103-expressing DCs in the gut lamina propria are enhanced by the activation of NOD2, indicating that MDP sensing plays a critical role in shaping the immune response to intestinal antigens by promoting a tolerogenic environment via manipulation of DC populations.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immune Tolerance , Integrin alpha Chains/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/genetics , Acetylmuramyl-Alanyl-Isoglutamine/metabolism , Animals , Crohn Disease , Cytokines , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Nod2 Signaling Adaptor Protein/genetics , T-Lymphocytes, Regulatory/metabolism
4.
Nat Commun ; 9(1): 4578, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30385785

ABSTRACT

Previous studies have reported that microglia depletion leads to impairment of synapse formation and these cells rapidly repopulate from CNS progenitors. However, the impact of microglia depletion and repopulation in the long-term state of the CNS environment has not been characterized. Here, we report that acute and synchronous microglia depletion and subsequent repopulation induces gray matter microgliosis, neuronal death in the somatosensory cortex and ataxia-like behavior. We find a type 1 interferon inflammatory signature in degenerating somatosensory cortex from microglia-depleted mice. Transcriptomic and mass cytometry analysis of repopulated microglia demonstrates an interferon regulatory factor 7-driven activation state. Minocycline and anti-IFNAR1 antibody treatment attenuate the CNS type 1 interferon-driven inflammation, restore microglia homeostasis and reduce ataxic behavior. Neither microglia depletion nor repopulation impact neuropathology or T-cell responses during experimental autoimmune encephalomyelitis. Together, we found that acute microglia ablation induces a type 1 interferon activation state of gray matter microglia associated with acute neurodegeneration.


Subject(s)
Cell Death/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Gray Matter/immunology , Interferon Type I/immunology , Microglia/immunology , Neurons/immunology , Somatosensory Cortex/immunology , Animals , Anti-Bacterial Agents/pharmacology , Ataxia/immunology , Ataxia/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Flow Cytometry , Gene Expression Profiling , Gray Matter/pathology , Homeostasis , Immunohistochemistry , Interferon Regulatory Factor-7/genetics , Interferon Regulatory Factor-7/immunology , Mice , Microscopy, Confocal , Minocycline/pharmacology , Neurons/pathology , Receptor, Interferon alpha-beta/antagonists & inhibitors , Rotarod Performance Test , Somatosensory Cortex/pathology
5.
Sci Rep ; 8(1): 6351, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29679021

ABSTRACT

Formalin-fixed paraffin-embedded (FFPE) tissues are valuable resources commonly used in pathology. However, formalin fixation modifies nucleic acids challenging the isolation of high-quality RNA for genetic profiling. Here, we assessed feasibility and reliability of microarray studies analysing transcriptome data from fresh, fresh-frozen (FF) and FFPE tissues. We show that reproducible microarray data can be generated from only 2 ng FFPE-derived RNA. For RNA quality assessment, fragment size distribution (DV200) and qPCR proved most suitable. During RNA isolation, extending tissue lysis time to 10 hours reduced high-molecular-weight species, while additional incubation at 70 °C markedly increased RNA yields. Since FF- and FFPE-derived microarrays constitute different data entities, we used indirect measures to investigate gene signal variation and relative gene expression. Whole-genome analyses revealed high concordance rates, while reviewing on single-genes basis showed higher data variation in FFPE than FF arrays. Using an experimental model, gene set enrichment analysis (GSEA) of FFPE-derived microarrays and fresh tissue-derived RNA-Seq datasets yielded similarly affected pathways confirming the applicability of FFPE tissue in global gene expression analysis. Our study provides a workflow comprising RNA isolation, quality assessment and microarray profiling using minimal RNA input, thus enabling hypothesis-generating pathway analyses from limited amounts of precious, pathologically significant FFPE tissues.


Subject(s)
RNA/analysis , Reproducibility of Results , Sequence Analysis, RNA/methods , Animals , Formaldehyde/adverse effects , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Hippocampus , Humans , Microarray Analysis/methods , Paraffin Embedding/methods , RNA/genetics , Rats , Real-Time Polymerase Chain Reaction , Tissue Fixation/methods , Transcriptome , Visual Cortex
6.
Eur J Immunol ; 43(11): 2896-906, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23881368

ABSTRACT

The enteric pathogen Citrobacter rodentium induces a mucosal IL-17 response in CD4(+) T helper (Th17) cells that is dependent on the Nod-like receptors Nod1 and Nod2. Here, we sought to determine whether this early Th17 response required antigen presentation by major histocompatibility complex class II (MHCII) for full induction. At early phases of C. rodentium infection, we observed that the intestinal mucosal Th17 response was fully blunted in irradiated mice reconstituted with MHCII-deficient (MHCII(-/-) →WT) hematopoietic cells. Surprisingly, we also observed a substantial increase in the relative frequency of IL-17(+) CD8(+) CD4(-) TCR-ß(+) cells (Tc17 cells) and FOXP3(+) CD8(+) CD4(-) TCR-ß(+) cells in the lamina propria and intraepithelial lymphocyte compartment of MHCII(-/-) →WT mice compared with that in WT→WT counterparts. Moreover, MHCII(-/-) →WT mice displayed increased susceptibility, increased bacterial translocation to deeper organs, and more severe colonic histopathology after infection with C. rodentium. Finally, a similar phenotype was observed in mice deficient for CIITA, a transcriptional regulator of MHCII expression. Together, these results indicate that MHCII is required to mount early mucosal Th17 responses to an enteric pathogen, and that MHCII regulates the induction of atypical CD8(+) T-cell subsets, such as Tc17 cells and FOXP3(+) CD8(+) cells, in vivo.


Subject(s)
Antigen Presentation/immunology , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/immunology , Hematopoietic Stem Cells/immunology , Histocompatibility Antigens Class II/immunology , Th17 Cells/immunology , Animals , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Genes, MHC Class II/genetics , Histocompatibility Antigens Class II/genetics , Interleukin-17/metabolism , Intestines/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics
7.
Gut Microbes ; 4(3): 222-31, 2013.
Article in English | MEDLINE | ID: mdl-23549220

ABSTRACT

Patients with inflammatory bowel diseases (IBD) harbour intestinal bacterial communities with altered composition compared with healthy counterparts; however, it is unknown whether changes in the microbiota are associated with genetic susceptibility of individuals for developing disease or instead reflect other changes in the intestinal environment related to the disease itself. Since deficiencies in the innate immune receptors Nod1 and Nod2 are linked to IBD, we tested the hypothesis that Nod-signaling alters intestinal immune profiles and subsequently alters bacterial community structure. We used qPCR to analyze expression patterns of selected immune mediators in the ileum and cecum of Nod-deficient mice compared with their Nod-sufficient littermates and assessed the relative abundance of major bacterial groups sampled from the ileum, cecum and colon. The Nod1-deficient ileum exhibited significantly lower expression of Nod2, Muc2, α- and ß-defensins and keratinocyte-derived chemokine (KC), suggesting a weakened epithelial barrier compared with WT littermates; however, there were no significant differences in the relative abundance of targeted bacterial groups, indicating that Nod1-associated immune differences alone do not promote dysbiosis. Furthermore, Nod2-deficient mice did not display any changes in the expression of immune markers or bacterial communities. Shifts in bacterial communities that were observed in this study correlated with housing conditions and were independent of genotype. These findings emphasize the importance of using F2 littermate controls to minimize environmental sources of variation in microbial analyses, to establish baseline conditions for host-microbe homeostasis in Nod-deficient mice and to strengthen models for testing factors contributing to microbial dysbiosis associated with IBD.


Subject(s)
Biota , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Homeostasis , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Signal Transduction , Animals , Biomarkers/analysis , Female , Gene Expression Profiling , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nod1 Signaling Adaptor Protein/deficiency , Nod2 Signaling Adaptor Protein/deficiency , Real-Time Polymerase Chain Reaction
8.
Innate Immun ; 19(5): 493-503, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23339926

ABSTRACT

Muramyl peptides (MPs) represent the building blocks of bacterial peptidoglycan, a critical component of bacterial cell walls. MPs are well characterized for their immunomodulatory properties, and numerous studies have delineated the role of MPs or synthetic MP analogs in host defense, adjuvanticity and inflammation. More recently, Nod1 and Nod2 have been identified as the host sensors for specific MPs, and, in particular, Nod2 was shown to detect muramyl dipeptide (MDP), a MP found in both Gram-positive and Gram-negative bacterial cell walls. Because mutations in Nod2 are associated with the etiology of Crohn's disease, there is a need to identify synthetic MP analogs that could potentiate Nod2-dependent immunity. Here, we analyzed the Nod2-activating property of 36 MP analogs that had been tested previously for their adjuvanticity and anti-infectious activity. Using a luciferase-based screen, we demonstrate that addition of a methyl group to the second amino acid of MDP generates a MDP derivative with enhanced Nod2-activating capacity. We further validated these results in murine macrophages, human dendritic cells and in vivo. These results offer a basis for the rational development of synthetic MPs that could be used in the treatment of inflammatory disorders that have been associated with Nod2 dysfunction, such as Crohn's disease.


Subject(s)
Acetylmuramyl-Alanyl-Isoglutamine/immunology , Crohn Disease/therapy , Dendritic Cells/immunology , Macrophages/immunology , Nod2 Signaling Adaptor Protein/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/analogs & derivatives , Acetylmuramyl-Alanyl-Isoglutamine/chemical synthesis , Animals , Crohn Disease/genetics , Crohn Disease/immunology , Enzyme Activation/genetics , Genetic Predisposition to Disease , HEK293 Cells , Humans , Interleukin-6/blood , Methylation , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/immunology , Polymorphism, Genetic , Protein Engineering , Signal Transduction/genetics , Transgenes/genetics
9.
Biol Open ; 1(12): 1239-47, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23259058

ABSTRACT

It is indispensable to thoroughly characterize each animal model in order to distinguish between primary and secondary effects of genetic changes. The present study analyzed Nod1 and Nod2 double deficient (Nod1/2 DKO) mice under physiological and inflammatory conditions. Nod1 and Nod2 are members of the Nucleotide-binding domain and Leucine-rich repeat containing Receptor (NLR) family. Several inflammatory disorders, such as Crohn's disease and asthma, are linked to genetic changes in either Nod1 or Nod2. These associations suggest that Nod1 and Nod2 play important roles in regulating the immune system.Three-month-old wildtype (Wt) and Nod1/2 DKO mice were sacrificed, body and organ weight were determined, and blood was drawn. Except for lower liver weight in Nod1/2 DKO mice, no differences were found in body/organ weight between both strains. Leukocyte count and composition was comparable. No significant changes in analyzed plasma biochemical markers were found. Additionally, intestinal and vascular permeability was determined. Nod1/2 DKO mice show increased susceptibility for intestinal permeability while vascular permeability was not affected. Next we induced septic shock and organ damage by administering LPS+PGN intraperitoneally to Wt and Nod1/2 DKO mice and sacrificed animals after 2 and 24 hours. The systemic inflammatory and metabolic response was comparable between both strains. However, renal response was different as indicated by partly preserved kidney function and tubular epithelial cell damage in Nod1/2 DKO at 24 hours. Remarkably, renal inflammatory mediators Tnfα, KC and Il-10 were significantly increased in Nod1/2 DKO compared with Wt mice at 2 hours.Systematic analysis of Nod1/2 DKO mice revealed a possible role of Nod1/2 in the development of renal disease during systemic inflammation.

10.
Curr Opin Immunol ; 24(4): 398-404, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22677577

ABSTRACT

The Nod-like receptor (NLR) family of intracellular pattern recognition molecules plays critical roles in the control of inflammation through the modulation of different signalling pathways, including those dependent on NF-κB and caspase-1-mediated cleavage of interleukin (IL)-1ß and IL-18. A number of NLRs or NLR-associated proteins have been genetically associated with susceptibility to inflammatory bowel disease (IBD), either Crohn's disease or ulcerative colitis. Accordingly, recent studies have examined the role of NLR proteins in chemical-induced or bacteria-induced murine models of colitis. In this review, we will discuss the genetic associations of NLRs with IBD and the research using NLR-deficient mice in different colitis models.


Subject(s)
Colitis, Ulcerative/immunology , Crohn Disease/immunology , Nod1 Signaling Adaptor Protein/physiology , Nod2 Signaling Adaptor Protein/physiology , Receptors, Cytoplasmic and Nuclear/physiology , Animals , Humans , Mice
11.
Semin Immunol ; 24(1): 9-16, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22296734

ABSTRACT

Nod-like receptors (NLRs) for detecting microbial invaders are features of many plant and animal families. Although broadly similar in form and function, intimate co-evolutionary events with environmental microbes have shaped specific classes of NLRs in different types of hosts. Details of the roles of different NLRs in signaling cellular immune responses to invading microbes are only beginning to emerge. This review will discuss the current understanding of NLRs in plants, invertebrates, and mammals, with emphasis on their role in regulating NF-κB and inflammasome activity in mammals.


Subject(s)
Nod Signaling Adaptor Proteins/metabolism , Signal Transduction , Animals , Gene Dosage , Humans , Inflammation/metabolism , Plants/metabolism , Plants/microbiology
12.
Trends Immunol ; 33(3): 112-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22342740

ABSTRACT

With the identification of T helper (Th)17 cells, a specific subset of CD4 T cells expressing interleukin (IL)-17 and IL-22, research on the function of these cytokines initially largely focused on traditional adaptive immune responses. However, IL-17 and IL-22 enhance basic innate barrier defenses at mucosal surfaces, such as antimicrobial peptide production and neutrophil recruitment; both events that occur rapidly and precede adaptive phase immunity. At the intestinal mucosal surface, it is now clear that innate lymphoid cells are also important sources of IL-17 and IL-22 during early phases of infection. Here, we discuss the function of innate IL-17- and IL-22-producing lymphocytes during enteric bacterial infection and their regulation by the intestinal microbiota, Toll-like receptors (TLRs) and Nod-like receptors (NLRs).


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Interleukin-17/immunology , Interleukins/immunology , Animals , Bacterial Infections/microbiology , Homeostasis , Humans , Intestines/immunology , Intestines/microbiology , Interleukin-22
13.
Proc Natl Acad Sci U S A ; 108(36): 14896-901, 2011 Sep 06.
Article in English | MEDLINE | ID: mdl-21856952

ABSTRACT

Although a number of studies have examined the development of T-helper cell type 2 (Th2) immunity in different settings, the mechanisms underlying the initiation of this arm of adaptive immunity are not well understood. We exploited the fact that immunization with antigen plus either nucleotide-binding oligomerization domain-containing proteins 1 (Nod1) or 2 (Nod2) agonists drives Th2 induction to understand how these pattern-recognition receptors mediate the development of systemic Th2 immune responses. Here, we show in bone-marrow chimeric mice that Nod1 and Nod2 expression within the stromal compartment is necessary for priming of effector CD4(+) Th2 responses and specific IgG1 antibodies. In contrast, sensing of these ligands by dendritic cells was not sufficient to induce Th2 immunity, although these cells contribute to the response. Moreover, we determined that CD11c(+) cells were the critical antigen-presenting cells, whereas basophils and B cells did not affect the capacity of Nod ligands to induce CD4(+) Th2 effector function. Finally, we found that full Th2 induction upon Nod1 and Nod2 activation was dependent on both thymic stromal lymphopoietin production by the stromal cells and the up-regulation of the costimulatory molecule, OX40 ligand, on dendritic cells. This study provides in vivo evidence of how systemic Th2 immunity is induced in the context of Nod stimulation. Such understanding will influence the rational design of therapeutics that could reprogram the immune system during an active Th1-mediated disease, such as Crohn's disease.


Subject(s)
Cytokines/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Th2 Cells/immunology , Animals , B-Lymphocytes/immunology , Basophils/immunology , Crohn Disease/genetics , Crohn Disease/immunology , Crohn Disease/therapy , Cytokines/genetics , Dendritic Cells/immunology , Immunity, Cellular/physiology , Immunization , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Nod1 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/genetics , OX40 Ligand , Protein Structure, Tertiary , Th1 Cells/immunology , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/immunology , Thymic Stromal Lymphopoietin
14.
Nat Med ; 17(7): 837-44, 2011 Jun 12.
Article in English | MEDLINE | ID: mdl-21666695

ABSTRACT

Interleukin 17 (IL-17) is a central cytokine implicated in inflammation and antimicrobial defense. After infection, both innate and adaptive IL-17 responses have been reported, but the type of cells involved in innate IL-17 induction, as well as their contribution to in vivo responses, are poorly understood. Here we found that Citrobacter and Salmonella infection triggered early IL-17 production, which was crucial for host defense and was mediated by CD4(+) T helper cells. Enteric innate T helper type 17 (iT(H)17) responses occurred principally in the cecum, were dependent on the Nod-like receptors Nod1 and Nod2, required IL-6 induction and were associated with a decrease in mucosal CD103(+) dendritic cells. Moreover, imprinting by the intestinal microbiota was fully required for the generation of iT(H)17 responses. Together, these results identify the Nod-iT(H)17 axis as a central element in controlling enteric pathogens, which may implicate Nod-driven iT(H)17 responses in the development of inflammatory bowel diseases.


Subject(s)
Intestines/microbiology , Th17 Cells/immunology , Animals , Citrobacter rodentium/immunology , Colitis/immunology , Colitis/microbiology , Enterobacteriaceae Infections/immunology , Female , Immunity, Innate/immunology , Interleukin-17/immunology , Interleukin-6/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestines/immunology , Male , Mice , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology
SELECTION OF CITATIONS
SEARCH DETAIL