Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.111
Filter
Add more filters

Publication year range
1.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Article in English | MEDLINE | ID: mdl-31923399

ABSTRACT

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Subject(s)
Immunity, Mucosal/immunology , Interleukin-18/immunology , Intestinal Mucosa/immunology , Animals , Cytokines/immunology , Enteric Nervous System/immunology , Enteric Nervous System/metabolism , Epithelial Cells/immunology , Female , Goblet Cells/immunology , Interleukin-18/biosynthesis , Intestinal Mucosa/metabolism , Intestine, Small/immunology , Male , Mice , Mice, Inbred C57BL , Neurons/immunology , Rats , Rats, Sprague-Dawley , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology
2.
Nat Immunol ; 22(3): 322-335, 2021 03.
Article in English | MEDLINE | ID: mdl-33531712

ABSTRACT

Immune system dysfunction is paramount in coronavirus disease 2019 (COVID-19) severity and fatality rate. Mucosal-associated invariant T (MAIT) cells are innate-like T cells involved in mucosal immunity and protection against viral infections. Here, we studied the immune cell landscape, with emphasis on MAIT cells, in cohorts totaling 208 patients with various stages of disease. MAIT cell frequency is strongly reduced in blood. They display a strong activated and cytotoxic phenotype that is more pronounced in lungs. Blood MAIT cell alterations positively correlate with the activation of other innate cells, proinflammatory cytokines, notably interleukin (IL)-18, and with the severity and mortality of severe acute respiratory syndrome coronavirus 2 infection. We also identified a monocyte/macrophage interferon (IFN)-α-IL-18 cytokine shift and the ability of infected macrophages to induce the cytotoxicity of MAIT cells in an MR1-dependent manner. Together, our results suggest that altered MAIT cell functions due to IFN-α-IL-18 imbalance contribute to disease severity, and their therapeutic manipulation may prevent deleterious inflammation in COVID-19 aggravation.


Subject(s)
COVID-19/immunology , Interferon-alpha/immunology , Interleukin-18/immunology , Macrophages/immunology , Monocytes/immunology , Mucosal-Associated Invariant T Cells/immunology , Adult , Aged , Aged, 80 and over , Animals , Bronchoalveolar Lavage , Case-Control Studies , Chlorocebus aethiops , Cohort Studies , Female , France , Humans , Immunophenotyping , Interleukin-10/immunology , Interleukin-15/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Interleukin-8/immunology , Male , Middle Aged , RNA-Seq , SARS-CoV-2 , Severity of Illness Index , Single-Cell Analysis , Vero Cells , Young Adult
3.
Immunity ; 57(7): 1445-1448, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38986437

ABSTRACT

Interleukin-1 (IL-1) family cytokines are key immunological regulators that achieve their signaling prowess after post-translational proteolytic processing. In this issue of Immunity, Dong et al. reveal the structural consequences of this process on proinflammatory IL-18, demonstrating that pro-IL-18 and mature IL-18 are structurally distinct.


Subject(s)
Interleukin-18 , Signal Transduction , Interleukin-18/metabolism , Interleukin-18/immunology , Humans , Signal Transduction/immunology , Animals , Protein Processing, Post-Translational
4.
Immunity ; 57(9): 2077-2094.e12, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38906145

ABSTRACT

Tissues are exposed to diverse inflammatory challenges that shape future inflammatory responses. While cellular metabolism regulates immune function, how metabolism programs and stabilizes immune states within tissues and tunes susceptibility to inflammation is poorly understood. Here, we describe an innate immune metabolic switch that programs long-term intestinal tolerance. Intestinal interleukin-18 (IL-18) stimulation elicited tolerogenic macrophages by preventing their proinflammatory glycolytic polarization via metabolic reprogramming to fatty acid oxidation (FAO). FAO reprogramming was triggered by IL-18 activation of SLC12A3 (NCC), leading to sodium influx, release of mitochondrial DNA, and activation of stimulator of interferon genes (STING). FAO was maintained in macrophages by a bistable switch that encoded memory of IL-18 stimulation and by intercellular positive feedback that sustained the production of macrophage-derived 2'3'-cyclic GMP-AMP (cGAMP) and epithelial-derived IL-18. Thus, a tissue-reinforced metabolic switch encodes durable immune tolerance in the gut and may enable reconstructing compromised immune tolerance in chronic inflammation.


Subject(s)
Immune Tolerance , Interleukin-18 , Macrophages , Nucleotides, Cyclic , Interleukin-18/metabolism , Interleukin-18/immunology , Animals , Mice , Nucleotides, Cyclic/metabolism , Macrophages/immunology , Macrophages/metabolism , Humans , Mice, Inbred C57BL , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Knockout , Fatty Acids/metabolism , Intestines/immunology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Glycolysis , Oxidation-Reduction
5.
Cell ; 175(6): 1651-1664.e14, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30392956

ABSTRACT

The activator and composition of the NLRP6 inflammasome remain poorly understood. We find that lipoteichoic acid (LTA), a molecule produced by Gram-positive bacteria, binds and activates NLRP6. In response to cytosolic LTA or infection with Listeria monocytogenes, NLRP6 recruited caspase-11 and caspase-1 via the adaptor ASC. NLRP6 activation by LTA induced processing of caspase-11, which promoted caspase-1 activation and interleukin-1ß (IL-1ß)/IL-18 maturation in macrophages. Nlrp6-/- and Casp11-/- mice were less susceptible to L. monocytogenes infection, which was associated with reduced pathogen loads and impaired IL-18 production. Administration of IL-18 to Nlrp6-/- or Casp11-/- mice restored the susceptibility of mutant mice to L. monocytogenes infection. These results reveal a previously unrecognized innate immunity pathway triggered by cytosolic LTA that is sensed by NLRP6 and exacerbates systemic Gram-positive pathogen infection via the production of IL-18.


Subject(s)
Immunity, Innate , Inflammasomes/immunology , Lipopolysaccharides/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Receptors, Cell Surface/immunology , Teichoic Acids/immunology , Animals , Caspase 1/genetics , Caspase 1/immunology , Caspases/genetics , Caspases/immunology , Caspases, Initiator , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Listeriosis/genetics , Listeriosis/pathology , Mice , Mice, Knockout , Receptors, Cell Surface/genetics
6.
Cell ; 167(2): 444-456.e14, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27716507

ABSTRACT

While conventional pathogenic protists have been extensively studied, there is an underappreciated constitutive protist microbiota that is an integral part of the vertebrate microbiome. The impact of these species on the host and their potential contributions to mucosal immune homeostasis remain poorly studied. Here, we show that the protozoan Tritrichomonas musculis activates the host epithelial inflammasome to induce IL-18 release. Epithelial-derived IL-18 promotes dendritic cell-driven Th1 and Th17 immunity and confers dramatic protection from mucosal bacterial infections. Along with its role as a "protistic" antibiotic, colonization with T. musculis exacerbates the development of T-cell-driven colitis and sporadic colorectal tumors. Our findings demonstrate a novel mutualistic host-protozoan interaction that increases mucosal host defenses at the cost of an increased risk of inflammatory disease.


Subject(s)
Colitis/immunology , Colitis/parasitology , Host-Parasite Interactions , Inflammasomes/immunology , Intestinal Mucosa/parasitology , Microbiota/immunology , Trichomonas Infections/immunology , Trichomonas/immunology , Animals , Colitis/microbiology , Dientamoeba/immunology , Immunity, Mucosal , Interleukin-18/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Mice , Mice, Inbred C57BL , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Symbiosis , Th1 Cells/immunology , Th17 Cells/immunology
7.
Cell ; 163(6): 1310-2, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638066

ABSTRACT

The fidelity of the intestinal barrier is critical to maintaining a healthy relationship between the immune system and the microbiota. Levy et al. and Nowarski et al. reveal how microbiota-derived metabolites modulate the activation of the inflammasome to influence the expression of the cytokine IL-18, intestinal barrier function, and intestinal inflammation.


Subject(s)
Colitis, Ulcerative/pathology , Colitis, Ulcerative/physiopathology , Colon/immunology , Colon/microbiology , Inflammasomes/immunology , Interleukin-18/immunology , Microbiota , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Female , Male
8.
Cell ; 163(6): 1428-43, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638072

ABSTRACT

Host-microbiome co-evolution drives homeostasis and disease susceptibility, yet regulatory principles governing the integrated intestinal host-commensal microenvironment remain obscure. While inflammasome signaling participates in these interactions, its activators and microbiome-modulating mechanisms are unknown. Here, we demonstrate that the microbiota-associated metabolites taurine, histamine, and spermine shape the host-microbiome interface by co-modulating NLRP6 inflammasome signaling, epithelial IL-18 secretion, and downstream anti-microbial peptide (AMP) profiles. Distortion of this balanced AMP landscape by inflammasome deficiency drives dysbiosis development. Upon fecal transfer, colitis-inducing microbiota hijacks this microenvironment-orchestrating machinery through metabolite-mediated inflammasome suppression, leading to distorted AMP balance favoring its preferential colonization. Restoration of the metabolite-inflammasome-AMP axis reinstates a normal microbiota and ameliorates colitis. Together, we identify microbial modulators of the NLRP6 inflammasome and highlight mechanisms by which microbiome-host interactions cooperatively drive microbial community stability through metabolite-mediated innate immune modulation. Therefore, targeted "postbiotic" metabolomic intervention may restore a normal microenvironment as treatment or prevention of dysbiosis-driven diseases.


Subject(s)
Colon/immunology , Colon/microbiology , Inflammasomes/immunology , Microbiota , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Antimicrobial Cationic Peptides , Colitis/chemically induced , Colitis/drug therapy , Colon/metabolism , Dysbiosis/metabolism , Germ-Free Life , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/drug therapy , Interleukin-18/immunology , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/genetics , Taurine/administration & dosage
9.
Cell ; 163(6): 1444-56, 2015 Dec 03.
Article in English | MEDLINE | ID: mdl-26638073

ABSTRACT

The intestinal mucosal barrier controlling the resident microbiome is dependent on a protective mucus layer generated by goblet cells, impairment of which is a hallmark of the inflammatory bowel disease, ulcerative colitis. Here, we show that IL-18 is critical in driving the pathologic breakdown of barrier integrity in a model of colitis. Deletion of Il18 or its receptor Il18r1 in intestinal epithelial cells (Δ/EC) conferred protection from colitis and mucosal damage in mice. In contrast, deletion of the IL-18 negative regulator Il18bp resulted in severe colitis associated with loss of mature goblet cells. Colitis and goblet cell loss were rescued in Il18bp(-/-);Il18r(Δ/EC) mice, demonstrating that colitis severity is controlled at the level of IL-18 signaling in intestinal epithelial cells. IL-18 inhibited goblet cell maturation by regulating the transcriptional program instructing goblet cell development. These results inform on the mechanism of goblet cell dysfunction that underlies the pathology of ulcerative colitis.


Subject(s)
Colitis, Ulcerative/pathology , Colitis, Ulcerative/physiopathology , Interleukin-18/immunology , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Dextran Sulfate , Endothelial Cells/metabolism , Epithelial Cells/cytology , Female , Goblet Cells/metabolism , Goblet Cells/pathology , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18 Receptor alpha Subunit/genetics , Interleukin-18 Receptor alpha Subunit/metabolism , Intestinal Mucosa/physiopathology , Male , Mice , Signal Transduction
10.
Nature ; 625(7994): 377-384, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38057668

ABSTRACT

Cytokines mediate cell-cell communication in the immune system and represent important therapeutic targets1-3. A myriad of studies have highlighted their central role in immune function4-13, yet we lack a global view of the cellular responses of each immune cell type to each cytokine. To address this gap, we created the Immune Dictionary, a compendium of single-cell transcriptomic profiles of more than 17 immune cell types in response to each of 86 cytokines (>1,400 cytokine-cell type combinations) in mouse lymph nodes in vivo. A cytokine-centric view of the dictionary revealed that most cytokines induce highly cell-type-specific responses. For example, the inflammatory cytokine interleukin-1ß induces distinct gene programmes in almost every cell type. A cell-type-centric view of the dictionary identified more than 66 cytokine-driven cellular polarization states across immune cell types, including previously uncharacterized states such as an interleukin-18-induced polyfunctional natural killer cell state. Based on this dictionary, we developed companion software, Immune Response Enrichment Analysis, for assessing cytokine activities and immune cell polarization from gene expression data, and applied it to reveal cytokine networks in tumours following immune checkpoint blockade therapy. Our dictionary generates new hypotheses for cytokine functions, illuminates pleiotropic effects of cytokines, expands our knowledge of activation states of each immune cell type, and provides a framework to deduce the roles of specific cytokines and cell-cell communication networks in any immune response.


Subject(s)
Cytokines , Immunity , Single-Cell Analysis , Animals , Mice , Cell Communication/drug effects , Cytokines/immunology , Gene Expression Profiling , Gene Expression Regulation , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity/drug effects , Interleukin-18/immunology , Interleukin-1beta/immunology , Killer Cells, Natural/immunology , Lymph Nodes/cytology , Lymph Nodes/immunology , Neoplasms/immunology , Neoplasms/therapy , Signal Transduction/drug effects , Software
11.
Cell ; 150(6): 1235-48, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22980983

ABSTRACT

The lymphatic network that transports interstitial fluid and antigens to lymph nodes constitutes a conduit system that can be hijacked by invading pathogens to achieve systemic spread unless dissemination is blocked in the lymph node itself. Here, we show that a network of diverse lymphoid cells (natural killer cells, γδ T cells, natural killer T cells, and innate-like CD8+ T cells) are spatially prepositioned close to lymphatic sinus-lining sentinel macrophages where they can rapidly and efficiently receive inflammasome-generated IL-18 and additional cytokine signals from the pathogen-sensing phagocytes. This leads to rapid IFNγ secretion by the strategically positioned innate lymphocytes, fostering antimicrobial resistance in the macrophage population. Interference with this innate immune response loop allows systemic spread of lymph-borne bacteria. These findings extend our understanding of the functional significance of cellular positioning and local intercellular communication within lymph nodes while emphasizing the role of these organs as highly active locations of innate host defense.


Subject(s)
Bacterial Infections/immunology , Immunity, Innate , Lymph Nodes/cytology , Lymph Nodes/immunology , Virus Diseases/immunology , Animals , Host-Pathogen Interactions , Inflammasomes/metabolism , Interferon-gamma/immunology , Interleukin-18/immunology , Lymph/microbiology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin Diseases, Infectious/immunology , Specific Pathogen-Free Organisms , T-Lymphocytes/immunology
12.
Nature ; 595(7865): 101-106, 2021 07.
Article in English | MEDLINE | ID: mdl-34108686

ABSTRACT

T cell immunoglobulin and mucin-containing molecule 3 (TIM-3), first identified as a molecule expressed on interferon-γ producing T cells1, is emerging as an important immune-checkpoint molecule, with therapeutic blockade of TIM-3 being investigated in multiple human malignancies. Expression of TIM-3 on CD8+ T cells in the tumour microenvironment is considered a cardinal sign of T cell dysfunction; however, TIM-3 is also expressed on several other types of immune cell, confounding interpretation of results following blockade using anti-TIM-3 monoclonal antibodies. Here, using conditional knockouts of TIM-3 together with single-cell RNA sequencing, we demonstrate the singular importance of TIM-3 on dendritic cells (DCs), whereby loss of TIM-3 on DCs-but not on CD4+ or CD8+ T cells-promotes strong anti-tumour immunity. Loss of TIM-3 prevented DCs from expressing a regulatory program and facilitated the maintenance of CD8+ effector and stem-like T cells. Conditional deletion of TIM-3 in DCs led to increased accumulation of reactive oxygen species resulting in NLRP3 inflammasome activation. Inhibition of inflammasome activation, or downstream effector cytokines interleukin-1ß (IL-1ß) and IL-18, completely abrogated the protective anti-tumour immunity observed with TIM-3 deletion in DCs. Together, our findings reveal an important role for TIM-3 in regulating DC function and underscore the potential of TIM-3 blockade in promoting anti-tumour immunity by regulating inflammasome activation.


Subject(s)
Hepatitis A Virus Cellular Receptor 2/metabolism , Inflammasomes/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , Dendritic Cells , Female , Hepatitis A Virus Cellular Receptor 2/deficiency , Hepatitis A Virus Cellular Receptor 2/genetics , Interleukin-18/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neoplasms/pathology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
13.
Blood ; 144(2): 171-186, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38579288

ABSTRACT

ABSTRACT: Multiple myeloma is a plasma cell malignancy that is currently incurable with conventional therapies. Following the success of CD19-targeted chimeric antigen receptor (CAR) T cells in leukemia and lymphoma, CAR T cells targeting B-cell maturation antigen (BCMA) more recently demonstrated impressive activity in relapsed and refractory myeloma patients. However, BCMA-directed therapy can fail due to weak expression of BCMA on myeloma cells, suggesting that novel approaches to better address this antigen-low disease may improve patient outcomes. We hypothesized that engineered secretion of the proinflammatory cytokine interleukin-18 (IL-18) and multiantigen targeting could improve CAR T-cell activity against BCMA-low myeloma. In a syngeneic murine model of myeloma, CAR T cells targeting the myeloma-associated antigens BCMA and B-cell activating factor receptor (BAFF-R) failed to eliminate myeloma when these antigens were weakly expressed, whereas IL-18-secreting CAR T cells targeting these antigens promoted myeloma clearance. IL-18-secreting CAR T cells developed an effector-like T-cell phenotype, promoted interferon-gamma production, reprogrammed the myeloma bone marrow microenvironment through type-I/II interferon signaling, and activated macrophages to mediate antimyeloma activity. Simultaneous targeting of weakly-expressed BCMA and BAFF-R with dual-CAR T cells enhanced T-cell:target-cell avidity, increased overall CAR signal strength, and stimulated antimyeloma activity. Dual-antigen targeting augmented CAR T-cell secretion of engineered IL-18 and facilitated elimination of larger myeloma burdens in vivo. Our results demonstrate that combination of engineered IL-18 secretion and multiantigen targeting can eliminate myeloma with weak antigen expression through distinct mechanisms.


Subject(s)
B-Cell Maturation Antigen , Immunotherapy, Adoptive , Interleukin-18 , Multiple Myeloma , Animals , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Multiple Myeloma/pathology , Mice , Interleukin-18/immunology , Immunotherapy, Adoptive/methods , B-Cell Maturation Antigen/immunology , Humans , Receptors, Chimeric Antigen/immunology , Disease Models, Animal , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Line, Tumor
14.
Cell ; 145(5): 745-57, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21565393

ABSTRACT

Inflammasomes are multiprotein complexes that function as sensors of endogenous or exogenous damage-associated molecular patterns. Here, we show that deficiency of NLRP6 in mouse colonic epithelial cells results in reduced IL-18 levels and altered fecal microbiota characterized by expanded representation of the bacterial phyla Bacteroidetes (Prevotellaceae) and TM7. NLRP6 inflammasome-deficient mice were characterized by spontaneous intestinal hyperplasia, inflammatory cell recruitment, and exacerbation of chemical colitis induced by exposure to dextran sodium sulfate (DSS). Cross-fostering and cohousing experiments revealed that the colitogenic activity of this microbiota is transferable to neonatal or adult wild-type mice, leading to exacerbation of DSS colitis via induction of the cytokine, CCL5. Antibiotic treatment and electron microscopy studies further supported the role of Prevotellaceae as a key representative of this microbiota-associated phenotype. Altogether, perturbations in this inflammasome pathway, including NLRP6, ASC, caspase-1, and IL-18, may constitute a predisposing or initiating event in some cases of human IBD.


Subject(s)
Colitis/immunology , Colitis/microbiology , Colon/microbiology , Inflammasomes/immunology , Receptors, Cell Surface/metabolism , Animals , Bacteria/classification , Bacteroidetes , Chemokine CCL5/metabolism , Colitis/chemically induced , Colitis/physiopathology , Colon/immunology , Dextran Sulfate , Disease Susceptibility , Interleukin-18/immunology , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Receptors, Cell Surface/genetics
15.
Nature ; 583(7817): 609-614, 2020 07.
Article in English | MEDLINE | ID: mdl-32581358

ABSTRACT

Cytokines were the first modern immunotherapies to produce durable responses in patients with advanced cancer, but they have only modest efficacy and limited tolerability1,2. In an effort to identify alternative cytokine pathways for immunotherapy, we found that components of the interleukin-18 (IL-18) pathway are upregulated on tumour-infiltrating lymphocytes, suggesting that IL-18 therapy could enhance anti-tumour immunity. However, recombinant IL-18 previously did not demonstrate efficacy in clinical trials3. Here we show that IL-18BP, a high-affinity IL-18 decoy receptor, is frequently upregulated in diverse human and mouse tumours and limits the anti-tumour activity of IL-18 in mice. Using directed evolution, we engineered a 'decoy-resistant' IL-18 (DR-18) that maintains signalling potential but is impervious to inhibition by IL-18BP. Unlike wild-type IL-18, DR-18 exerted potent anti-tumour effects in mouse tumour models by promoting the development of poly-functional effector CD8+ T cells, decreasing the prevalence of exhausted CD8+ T cells that express the transcriptional regulator of exhaustion TOX, and expanding the pool of stem-like TCF1+ precursor CD8+ T cells. DR-18 also enhanced the activity and maturation of natural killer cells to effectively treat anti-PD-1 resistant tumours that have lost surface expression of major histocompatibility complex class I molecules. These results highlight the potential of the IL-18 pathway for immunotherapeutic intervention and implicate IL-18BP as a major therapeutic barrier.


Subject(s)
Immunotherapy , Intercellular Signaling Peptides and Proteins/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-18/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , Histocompatibility Antigens Class I/immunology , Humans , Kaplan-Meier Estimate , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Receptors, Interleukin-18/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
16.
Nat Immunol ; 14(12): 1247-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24185614

ABSTRACT

The inflammasome adaptor ASC contributes to innate immunity through the activation of caspase-1. Here we found that signaling pathways dependent on the kinases Syk and Jnk were required for the activation of caspase-1 via the ASC-dependent inflammasomes NLRP3 and AIM2. Inhibition of Syk or Jnk abolished the formation of ASC specks without affecting the interaction of ASC with NLRP3. ASC was phosphorylated during inflammasome activation in a Syk- and Jnk-dependent manner, which suggested that Syk and Jnk are upstream of ASC phosphorylation. Moreover, phosphorylation of Tyr144 in mouse ASC was critical for speck formation and caspase-1 activation. Our results suggest that phosphorylation of ASC controls inflammasome activity through the formation of ASC specks.


Subject(s)
Cytoskeletal Proteins/immunology , Inflammasomes/immunology , Intracellular Signaling Peptides and Proteins/immunology , JNK Mitogen-Activated Protein Kinases/immunology , Protein-Tyrosine Kinases/immunology , Animals , Apoptosis Regulatory Proteins , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 1/immunology , Caspase 1/metabolism , Cells, Cultured , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , HEK293 Cells , Humans , Immunoblotting , Inflammasomes/genetics , Inflammasomes/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nigericin/pharmacology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Phosphorylation/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA Interference , Syk Kinase , Tyrosine/genetics , Tyrosine/immunology , Tyrosine/metabolism
17.
Clin Exp Immunol ; 217(1): 31-44, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38587448

ABSTRACT

Allergic asthma (AA) is closely associated with the polarization of T helper (Th)2 and Th17 cells. Interleukin (IL)-18 acts as an inducer of Th2 and Th17 cell responses. However, expressions of IL-18 and IL-18 receptor alpha (IL-18Rα) in blood Th2 and Th17 cells of patients with AA remain unclear. We therefore investigated their expressions in Th2 and Th17 cells using flow cytometric analysis, quantitative real-time PCR (qPCR), and murine AA model. We observed increased proportions of Th2, Th17, IL-18+, IL-18+ Th2, and IL-18+ Th17 cells in blood CD4+ T cells of patients with AA. Additionally, house dust mite seemed to upregulate further IL-18 expression in Th2 and Th17, and upregulate IL-18Rα expression in CD4+ T, Th2, and Th17 cells of AA patients. It was also found that the plasma levels of IL-4, IL-17A, and IL-18 in AA patients were elevated, and they were correlated between each other. In ovalbumin (OVA)-induced asthma mouse (AM), we observed that the percentages of blood CD4+ T, Th2, and Th17 cells were increased. Moreover, OVA-induced AM expressed higher level of IL-18Rα in blood Th2 cells, which was downregulated by IL-18. Increased IL-18Rα expression was also observed in blood Th2 cells of OVA-induced FcεRIα-/- mice. Collectively, our findings suggest the involvement of Th2 cells in AA by expressing excessive IL-18 and IL-18Rα in response to allergen, and that IL-18 and IL-18Rα expressing Th2 cells are likely to be the potential targets for AA therapy.


Subject(s)
Allergens , Asthma , Interleukin-18 , Th17 Cells , Th2 Cells , Humans , Interleukin-18/immunology , Interleukin-18/blood , Asthma/immunology , Asthma/blood , Animals , Th2 Cells/immunology , Mice , Female , Th17 Cells/immunology , Male , Adult , Allergens/immunology , Middle Aged , Up-Regulation/immunology , Interleukin-18 Receptor alpha Subunit/immunology , Interleukin-18 Receptor alpha Subunit/genetics , Ovalbumin/immunology , Receptors, Interleukin-18/immunology , Mice, Inbred BALB C , Disease Models, Animal , Pyroglyphidae/immunology , Young Adult
18.
Nat Immunol ; 13(2): 162-9, 2012 Jan 08.
Article in English | MEDLINE | ID: mdl-22231517

ABSTRACT

Memory T cells exert antigen-independent effector functions, but how these responses are regulated is unclear. We discovered an in vivo link between flagellin-induced NLRC4 inflammasome activation in splenic dendritic cells (DCs) and host protective interferon-γ (IFN-γ) secretion by noncognate memory CD8(+) T cells, which could be activated by Salmonella enterica serovar Typhimurium, Yersinia pseudotuberculosis and Pseudomonas aeruginosa. We show that CD8α(+) DCs were particularly efficient at sensing bacterial flagellin through NLRC4 inflammasomes. Although this activation released interleukin 18 (IL-18) and IL-1ß, only IL-18 was required for IFN-γ production by memory CD8(+) T cells. Conversely, only the release of IL-1ß, but not IL-18, depended on priming signals mediated by Toll-like receptors. These findings provide a comprehensive mechanistic framework for the regulation of noncognate memory T cell responses during bacterial immunity.


Subject(s)
Apoptosis Regulatory Proteins/immunology , CD8-Positive T-Lymphocytes/immunology , Calcium-Binding Proteins/immunology , Dendritic Cells/immunology , Immunologic Memory , Inflammasomes/immunology , Interferon-gamma/immunology , Animals , Flagellin/immunology , Interleukin-18/immunology , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Salmonella Infections, Animal/immunology , Salmonella typhimurium/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptors/immunology , Yersinia pseudotuberculosis Infections/immunology
19.
Immunity ; 43(5): 987-97, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572063

ABSTRACT

Defective neutrophils in patients with chronic granulomatous disease (CGD) cause susceptibility to extracellular and intracellular infections. Microbes must first be ejected from intracellular niches to expose them to neutrophil attack, so we hypothesized that inflammasomes detect certain CGD pathogens upstream of neutrophil killing. Here, we identified one such ubiquitous environmental bacterium, Chromobacterium violaceum, whose extreme virulence was fully counteracted by the NLRC4 inflammasome. Caspase-1 protected via two parallel pathways that eliminated intracellular replication niches. Pyroptosis was the primary bacterial clearance mechanism in the spleen, but both pyroptosis and interleukin-18 (IL-18)-driven natural killer (NK) cell responses were required for liver defense. NK cells cleared hepatocyte replication niches via perforin-dependent cytotoxicity, whereas interferon-γ was not required. These insights suggested a therapeutic approach: exogenous IL-18 restored perforin-dependent cytotoxicity during infection by the inflammasome-evasive bacterium Listeria monocytogenes. Therefore, inflammasomes can trigger complementary programmed cell death mechanisms, directing sterilizing immunity against intracellular bacterial pathogens.


Subject(s)
Bacterial Infections/immunology , Inflammasomes/immunology , Killer Cells, Natural/immunology , Pyroptosis/immunology , Animals , Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Caspase 1/immunology , Cell Death/immunology , Chromobacterium/immunology , Granulomatous Disease, Chronic/immunology , Interferon-gamma/immunology , Interleukin-18/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Liver/immunology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Spleen/immunology
20.
Immunity ; 42(2): 321-331, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680273

ABSTRACT

T helper 1 (Th1) cell-associated immunity exacerbates ileitis induced by oral Toxoplasma gondii infection. We show here that attenuated ileitis observed in interleukin-22 (IL-22)-deficient mice was associated with reduced production of Th1-cell-promoting IL-18. IL-22 not only augmented the expression of Il18 mRNA and inactive precursor protein (proIL-18) in intestinal epithelial cells after T. gondii or Citrobacter rodentium infection, but also maintained the homeostatic amount of proIL-18 in the ileum. IL-22, however, did not induce the processing to active IL-18, suggesting a two-step regulation of IL-18 in these cells. Although IL-18 exerted pathogenic functions during ileitis triggered by T. gondii, it was required for host defense against C. rodentium. Conversely, IL-18 was required for the expression of IL-22 in innate lymphoid cells (ILCs) upon T. gondii infection. Our results define IL-18 as an IL-22 target gene in epithelial cells and describe a complex mutual regulation of both cytokines during intestinal infection.


Subject(s)
Enterobacteriaceae Infections/immunology , Interleukin-18/immunology , Interleukins/immunology , Intestinal Mucosa/immunology , Toxoplasmosis/immunology , Animals , Cells, Cultured , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/microbiology , Epithelial Cells/immunology , Ileitis/immunology , Ileitis/microbiology , Ileitis/parasitology , Ileum/immunology , Ileum/microbiology , Ileum/parasitology , Inflammation/immunology , Interferon-gamma/biosynthesis , Interleukin-18/biosynthesis , Interleukins/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/parasitology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration/immunology , Neutrophils/immunology , Organ Culture Techniques , RNA, Messenger/biosynthesis , Th1 Cells/immunology , Toxoplasma/immunology , Toxoplasmosis/parasitology , Up-Regulation , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL