Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 316
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 167(5): 1339-1353.e21, 2016 11 17.
Article in English | MEDLINE | ID: mdl-27863247

ABSTRACT

Despite the accepted health benefits of consuming dietary fiber, little is known about the mechanisms by which fiber deprivation impacts the gut microbiota and alters disease risk. Using a gnotobiotic mouse model, in which animals were colonized with a synthetic human gut microbiota composed of fully sequenced commensal bacteria, we elucidated the functional interactions between dietary fiber, the gut microbiota, and the colonic mucus barrier, which serves as a primary defense against enteric pathogens. We show that during chronic or intermittent dietary fiber deficiency, the gut microbiota resorts to host-secreted mucus glycoproteins as a nutrient source, leading to erosion of the colonic mucus barrier. Dietary fiber deprivation, together with a fiber-deprived, mucus-eroding microbiota, promotes greater epithelial access and lethal colitis by the mucosal pathogen, Citrobacter rodentium. Our work reveals intricate pathways linking diet, the gut microbiome, and intestinal barrier dysfunction, which could be exploited to improve health using dietary therapeutics.


Subject(s)
Dietary Fiber/administration & dosage , Gastrointestinal Microbiome , Intestinal Mucosa/microbiology , Animals , Citrobacter rodentium/physiology , Colitis/microbiology , Colon/microbiology , Disease Susceptibility , Enterobacteriaceae Infections/microbiology , Escherichia coli , Female , Germ-Free Life , Humans , Male , Mice , Mucin-2/genetics
3.
Cell ; 167(7): 1719-1733.e12, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27984723

ABSTRACT

Aging is the major risk factor for many human diseases. In vitro studies have demonstrated that cellular reprogramming to pluripotency reverses cellular age, but alteration of the aging process through reprogramming has not been directly demonstrated in vivo. Here, we report that partial reprogramming by short-term cyclic expression of Oct4, Sox2, Klf4, and c-Myc (OSKM) ameliorates cellular and physiological hallmarks of aging and prolongs lifespan in a mouse model of premature aging. Similarly, expression of OSKM in vivo improves recovery from metabolic disease and muscle injury in older wild-type mice. The amelioration of age-associated phenotypes by epigenetic remodeling during cellular reprogramming highlights the role of epigenetic dysregulation as a driver of mammalian aging. Establishing in vivo platforms to modulate age-associated epigenetic marks may provide further insights into the biology of aging.


Subject(s)
Aging/genetics , Cellular Reprogramming , Epigenesis, Genetic , Metabolic Diseases/genetics , Transcription Factors/metabolism , Aging, Premature/genetics , Aging, Premature/metabolism , Animals , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Lamin Type A/genetics , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control , Mice , Models, Animal , Pancreas/metabolism , Sarcopenia/metabolism
4.
Cell ; 163(2): 367-80, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26411289

ABSTRACT

Intestinal Th17 cells are induced and accumulate in response to colonization with a subgroup of intestinal microbes such as segmented filamentous bacteria (SFB) and certain extracellular pathogens. Here, we show that adhesion of microbes to intestinal epithelial cells (ECs) is a critical cue for Th17 induction. Upon monocolonization of germ-free mice or rats with SFB indigenous to mice (M-SFB) or rats (R-SFB), M-SFB and R-SFB showed host-specific adhesion to small intestinal ECs, accompanied by host-specific induction of Th17 cells. Citrobacter rodentium and Escherichia coli O157 triggered similar Th17 responses, whereas adhesion-defective mutants of these microbes failed to do so. Moreover, a mixture of 20 bacterial strains, which were selected and isolated from fecal samples of a patient with ulcerative colitis on the basis of their ability to cause a robust induction of Th17 cells in the mouse colon, also exhibited EC-adhesive characteristics.


Subject(s)
Bacterial Adhesion , Citrobacter rodentium/physiology , Enterobacteriaceae Infections/immunology , Escherichia coli Infections/immunology , Escherichia coli O157/physiology , Intestinal Mucosa/immunology , Th17 Cells/immunology , Animals , Bacterial Infections/immunology , Epithelial Cells/immunology , Epithelial Cells/microbiology , Epithelial Cells/ultrastructure , Feces/microbiology , Humans , Immunoglobulin A/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Microscopy, Electron, Scanning , Rats , Rats, Inbred F344 , Species Specificity
5.
Trends Biochem Sci ; 48(4): 331-344, 2023 04.
Article in English | MEDLINE | ID: mdl-36336552

ABSTRACT

The NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome is a cytoplasmic supramolecular complex that is activated in response to cellular perturbations triggered by infection and sterile injury. Assembly of the NLRP3 inflammasome leads to activation of caspase-1, which induces the maturation and release of interleukin-1ß (IL-1ß) and IL-18, as well as cleavage of gasdermin D (GSDMD), which promotes a lytic form of cell death. Production of IL-1ß via NLRP3 can contribute to the pathogenesis of inflammatory disease, whereas aberrant IL-1ß secretion through inherited NLRP3 mutations causes autoinflammatory disorders. In this review, we discuss recent developments in the structure of the NLRP3 inflammasome, and the cellular processes and signaling events controlling its assembly and activation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Caspase 1/metabolism , Gene Expression , Interleukin-1beta/genetics , Interleukin-1beta/metabolism
6.
Cell ; 149(4): 847-59, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22541070

ABSTRACT

Alu RNA accumulation due to DICER1 deficiency in the retinal pigmented epithelium (RPE) is implicated in geographic atrophy (GA), an advanced form of age-related macular degeneration that causes blindness in millions of individuals. The mechanism of Alu RNA-induced cytotoxicity is unknown. Here we show that DICER1 deficit or Alu RNA exposure activates the NLRP3 inflammasome and triggers TLR-independent MyD88 signaling via IL18 in the RPE. Genetic or pharmacological inhibition of inflammasome components (NLRP3, Pycard, Caspase-1), MyD88, or IL18 prevents RPE degeneration induced by DICER1 loss or Alu RNA exposure. These findings, coupled with our observation that human GA RPE contains elevated amounts of NLRP3, PYCARD, and IL18 and evidence of increased Caspase-1 and MyD88 activation, provide a rationale for targeting this pathway in GA. Our findings also reveal a function of the inflammasome outside the immune system and an immunomodulatory action of mobile elements.


Subject(s)
Alu Elements , DEAD-box RNA Helicases/metabolism , Geographic Atrophy/immunology , Geographic Atrophy/pathology , Inflammasomes/immunology , Myeloid Differentiation Factor 88/metabolism , Retinal Pigment Epithelium/metabolism , Ribonuclease III/metabolism , Animals , Carrier Proteins/metabolism , Geographic Atrophy/metabolism , Humans , Inflammasomes/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Retinal Pigment Epithelium/pathology , Toll-Like Receptors/metabolism
7.
Nat Immunol ; 14(7): 685-90, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23778796

ABSTRACT

A dense resident microbial community in the gut, referred as the commensal microbiota, coevolved with the host and is essential for many host physiological processes that include enhancement of the intestinal epithelial barrier, development of the immune system and acquisition of nutrients. A major function of the microbiota is protection against colonization by pathogens and overgrowth of indigenous pathobionts that can result from the disruption of the healthy microbial community. The mechanisms that regulate the ability of the microbiota to restrain pathogen growth are complex and include competitive metabolic interactions, localization to intestinal niches and induction of host immune responses. Pathogens, in turn, have evolved strategies to escape from commensal-mediated resistance to colonization. Thus, the interplay between commensals and pathogens or indigenous pathobionts is critical for controlling infection and disease. Understanding pathogen-commensal interactions may lead to new therapeutic approaches to treating infectious diseases.


Subject(s)
Bacterial Infections/immunology , Bacterial Infections/microbiology , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Metagenome/immunology , Animals , Gastrointestinal Tract/metabolism , Host-Pathogen Interactions , Humans
8.
Nat Immunol ; 14(2): 101-5, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23334821

ABSTRACT

Leading scientists working on the microbiome gathered in an October 2012 meeting in Baeza, Spain, to discuss recent advances in the understanding of the role of the microbiota in immunity, pathogen colonization, metabolism and disease.


Subject(s)
Immunity , Metagenome/immunology , Animals , Bacteria/growth & development , Host-Pathogen Interactions , Humans , Spain
9.
Immunity ; 44(3): 647-658, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26944199

ABSTRACT

The gut microbiota is compartmentalized in the intestinal lumen and induces local immune responses, but it remains unknown whether the gut microbiota can induce systemic response and contribute to systemic immunity. We report that selective gut symbiotic gram-negative bacteria were able to disseminate systemically to induce immunoglobulin G (IgG) response, which primarily targeted gram-negative bacterial antigens and conferred protection against systemic infections by E. coli and Salmonella by directly coating bacteria to promote killing by phagocytes. T cells and Toll-like receptor 4 on B cells were important in the generation of microbiota-specific IgG. We identified murein lipoprotein (MLP), a highly conserved gram-negative outer membrane protein, as a major antigen that induced systemic IgG homeostatically in both mice and humans. Administration of anti-MLP IgG conferred crucial protection against systemic Salmonella infection. Thus, our findings reveal an important function for the gut microbiota in combating systemic infection through the induction of protective IgG.


Subject(s)
Gram-Negative Bacteria/immunology , Gram-Negative Bacterial Infections/immunology , Immunoglobulin G/metabolism , Intestines/immunology , Peptidoglycan/immunology , Animals , Bacterial Load/genetics , Homeostasis/genetics , Host-Pathogen Interactions , Immunoglobulin G/genetics , Intestines/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
10.
Nature ; 570(7761): 338-343, 2019 06.
Article in English | MEDLINE | ID: mdl-31189953

ABSTRACT

The NLRP3 inflammasome can be activated by stimuli that include nigericin, uric acid crystals, amyloid-ß fibrils and extracellular ATP. The mitotic kinase NEK7 licenses the assembly and activation of the NLRP3 inflammasome in interphase. Here we report a cryo-electron microscopy structure of inactive human NLRP3 in complex with NEK7, at a resolution of 3.8 Å. The earring-shaped NLRP3 consists of curved leucine-rich-repeat and globular NACHT domains, and the C-terminal lobe of NEK7 nestles against both NLRP3 domains. Structural recognition between NLRP3 and NEK7 is confirmed by mutagenesis both in vitro and in cells. Modelling of an active NLRP3-NEK7 conformation based on the NLRC4 inflammasome predicts an additional contact between an NLRP3-bound NEK7 and a neighbouring NLRP3. Mutations to this interface abolish the ability of NEK7 or NLRP3 to rescue NLRP3 activation in NEK7-knockout or NLRP3-knockout cells. These data suggest that NEK7 bridges adjacent NLRP3 subunits with bipartite interactions to mediate the activation of the NLRP3 inflammasome.


Subject(s)
Cryoelectron Microscopy , Inflammasomes/metabolism , Inflammasomes/ultrastructure , NIMA-Related Kinases/metabolism , NIMA-Related Kinases/ultrastructure , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/ultrastructure , Binding, Competitive , Humans , Inflammasomes/chemistry , Inflammasomes/genetics , Models, Molecular , Mutation , NIMA-Related Kinases/chemistry , NIMA-Related Kinases/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Protein Binding , Protein Domains , Protein Multimerization , Protein Structure, Quaternary
11.
Nat Immunol ; 13(4): 325-32, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22430785

ABSTRACT

Inflammasomes are multiprotein complexes that activate caspase-1, which leads to maturation of the proinflammatory cytokines interleukin 1ß (IL-1ß) and IL-18 and the induction of pyroptosis. Members of the Nod-like receptor (NLR) family, including NLRP1, NLRP3 and NLRC4, and the cytosolic receptor AIM2 are critical components of inflammasomes and link microbial and endogenous danger signals to the activation of caspase-1. In response to microbial infection, activation of the inflammasomes contributes to host protection by inducing immune responses that limit microbial invasion, but deregulated activation of inflammasomes is associated with autoinflammatory syndromes and other pathologies. Thus, understanding inflammasome pathways may provide insight into the mechanisms of host defense against microbes and the development of inflammatory disorders.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/immunology , Signal Transduction/immunology , Animals , Humans
12.
Nat Immunol ; 13(5): 449-56, 2012 May.
Article in English | MEDLINE | ID: mdl-22484733

ABSTRACT

Intestinal phagocytes transport oral antigens and promote immune tolerance, but their role in innate immune responses remains unclear. Here we found that intestinal phagocytes were anergic to ligands for Toll-like receptors (TLRs) or commensals but constitutively expressed the precursor to interleukin 1ß (pro-IL-1ß). After infection with pathogenic Salmonella or Pseudomonas, intestinal phagocytes produced mature IL-1ß through the NLRC4 inflammasome but did not produce tumor necrosis factor (TNF) or IL-6. BALB/c mice deficient in NLRC4 or the IL-1 receptor were highly susceptible to orogastric but not intraperitoneal infection with Salmonella. That enhanced lethality was preceded by impaired expression of endothelial adhesion molecules, lower neutrophil recruitment and poor intestinal pathogen clearance. Thus, NLRC4-dependent production of IL-1ß by intestinal phagocytes represents a specific response that discriminates pathogenic bacteria from commensal bacteria and contributes to host defense in the intestine.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Calcium-Binding Proteins/immunology , Clonal Anergy , Host-Pathogen Interactions/immunology , Interleukin-1beta/metabolism , Intestines/immunology , Intestines/microbiology , Phagocytes/immunology , Animals , Apoptosis Regulatory Proteins/genetics , Calcium-Binding Proteins/genetics , Caspase 1/metabolism , Flagellin/immunology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-6/biosynthesis , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Neutrophil Infiltration/genetics , Neutrophil Infiltration/immunology , Neutrophils/immunology , Neutrophils/pathology , Phagocytes/microbiology , Pseudomonas/immunology , Pseudomonas Infections/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Salmonella/genetics , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptors/immunology , Toll-Like Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/immunology
13.
Immunity ; 43(5): 923-32, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572062

ABSTRACT

The noncanonical inflammasome induced by intracellular lipopolysaccharide (LPS) leads to caspase-11-dependent pyroptosis, which is critical for induction of endotoxic shock in mice. However, the signaling pathway downstream of caspase-11 is unknown. We found that cytosolic LPS stimulation induced caspase-11-dependent cleavage of the pannexin-1 channel followed up by ATP release, which in turn activated the purinergic P2X7 receptor to mediate cytotoxicity. In the absence of P2X7 or pannexin-1, pyroptosis induced by cytosolic LPS was abrogated. Cleavage of pannexin-1 required the catalytic activity of caspase-11 and was essential for ATP release and P2X7-mediated pyroptosis. Priming the caspase-11 pathway in vivo with LPS or Toll-like receptor-3 (TLR3) agonist resulted in high mortality in wild-type mice after secondary LPS challenge, but not in Casp11(-/-), Panx1(-/-), or P2x7(-/-) mice. These results reveal a critical role for pannexin-1 and P2X7 downstream of caspase-11 for pyroptosis and susceptibility to sepsis induced by the noncanonical inflammasome.


Subject(s)
Caspases/metabolism , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Pyroptosis/physiology , Receptors, Purinergic P2X7/metabolism , Shock, Septic/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspases, Initiator , Cell Line , HEK293 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Pyroptosis/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 3/metabolism
14.
Immunity ; 43(3): 451-62, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26341399

ABSTRACT

Endoplasmic reticulum (ER) stress is observed in many human diseases, often associated with inflammation. ER stress can trigger inflammation through nucleotide-binding domain and leucine-rich repeat containing (NLRP3) inflammasome, which might stimulate inflammasome formation by association with damaged mitochondria. How ER stress triggers mitochondrial dysfunction and inflammasome activation is ill defined. Here we have used an infection model to show that the IRE1α ER stress sensor regulates regulated mitochondrial dysfunction through an NLRP3-mediated feed-forward loop, independently of ASC. IRE1α activation increased mitochondrial reactive oxygen species, promoting NLRP3 association with mitochondria. NLRP3 was required for ER stress-induced cleavage of caspase-2 and the pro-apoptotic factor, Bid, leading to subsequent release of mitochondrial contents. Caspase-2 and Bid were necessary for activation of the canonical inflammasome by infection-associated or general ER stress. These data identify an NLRP3-caspase-2-dependent mechanism that relays ER stress to the mitochondria to promote inflammation, integrating cellular stress and innate immunity.


Subject(s)
Carrier Proteins/immunology , Caspase 2/immunology , Endoplasmic Reticulum Stress/immunology , Inflammasomes/immunology , Mitochondria/immunology , Animals , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/immunology , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Brucella abortus/immunology , Brucella abortus/physiology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Caspase 2/genetics , Caspase 2/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress/genetics , Endoribonucleases/immunology , Endoribonucleases/metabolism , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Inflammasomes/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , RNA Interference/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism
15.
Immunity ; 42(4): 744-55, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862092

ABSTRACT

The microbiota stimulates inflammation, but the signaling pathways and the members of the microbiota involved remain poorly understood. We found that the microbiota induces interleukin-1ß (IL-1ß) release upon intestinal injury and that this is mediated via the NLRP3 inflammasome. Enterobacteriaceae and in particular the pathobiont Proteus mirabilis, induced robust IL-1ß release that was comparable to that induced by the pathogen Salmonella. Upon epithelial injury, production of IL-1ß in the intestine was largely mediated by intestinal Ly6C(high) monocytes, required chemokine receptor CCR2 and was abolished by deletion of IL-1ß in CCR2(+) blood monocytes. Furthermore, colonization with P. mirabilis promoted intestinal inflammation upon intestinal injury via the production of hemolysin, which required NLRP3 and IL-1 receptor signaling in vivo. Thus, upon intestinal injury, selective members of the microbiota stimulate newly recruited monocytes to induce NLRP3-dependent IL-1ß release, which promotes inflammation in the intestine.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Microbiota/immunology , Monocytes/immunology , Symbiosis/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Carrier Proteins/genetics , Gene Expression Regulation , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Inflammasomes/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Intestines/immunology , Intestines/injuries , Intestines/microbiology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/microbiology , Monocytes/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Proteus Infections/genetics , Proteus Infections/immunology , Proteus Infections/microbiology , Proteus Infections/pathology , Proteus mirabilis/immunology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/pathology , Signal Transduction
16.
EMBO J ; 38(20): e101266, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31544965

ABSTRACT

Inflammasomes are cytosolic protein complexes, which orchestrate the maturation of active IL-1ß by proteolytic cleavage via caspase-1. Although many principles of inflammasome activation have been described, mechanisms that limit inflammasome-dependent immune responses remain poorly defined. Here, we show that the thiol-specific peroxidase peroxiredoxin-4 (Prdx4) directly regulates IL-1ß generation by interfering with caspase-1 activity. We demonstrate that caspase-1 and Prdx4 form a redox-sensitive regulatory complex via caspase-1 cysteine 397 that leads to caspase-1 sequestration and inactivation. Mice lacking Prdx4 show an increased susceptibility to LPS-induced septic shock. This effect was phenocopied in mice carrying a conditional deletion of Prdx4 in the myeloid lineage (Prdx4-ΔLysMCre). Strikingly, we demonstrate that Prdx4 co-localizes with inflammasome components in extracellular vesicles (EVs) from inflammasome-activated macrophages. Purified EVs are able to transmit a robust IL-1ß-dependent inflammatory response in vitro and also in recipient mice in vivo. Loss of Prdx4 boosts the pro-inflammatory potential of EVs. These findings identify Prdx4 as a critical regulator of inflammasome activity and provide new insights into remote cell-to-cell communication function of inflammasomes via macrophage-derived EVs.


Subject(s)
Caspase 1/metabolism , Extracellular Vesicles/metabolism , Inflammasomes/immunology , Macrophages/immunology , Peroxiredoxins/physiology , Shock, Septic/prevention & control , Animals , Caspase 1/genetics , Cytokines/metabolism , Female , Inflammasomes/metabolism , Lipopolysaccharides/toxicity , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Shock, Septic/chemically induced , Shock, Septic/immunology , Shock, Septic/pathology , Signal Transduction
17.
Int Immunol ; 34(8): 409-420, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35641096

ABSTRACT

IL-17 plays important roles in host defense against Candida albicans at barrier surfaces and during invasive infection. However, the role of IL-17 in host defense after colonization of the epidermis, a main site of C. albicans infection, remains poorly understood. Using a murine model of epicutaneous candidiasis without skin abrasion, we found that skin inflammation triggered by epidermal C. albicans colonization was self-limiting with fungal clearance completed by day 7 after inoculation in wild-type mice or animals deficient in IL-17A or IL-17F. In contrast, marked neutrophilic inflammation in the epidermis and impaired fungal clearance were observed in mice lacking both IL-17A and IL-17F. Clearance of C. albicans was independent of Dectin-1, Dectin-2, CARD9 (caspase-recruitment domain family, member 9), TLR2 (Toll-like receptor 2) and MyD88 in the epidermal colonization model. We found that group 3 innate lymphoid cells (ILC3s) and γδT cells were the major IL-17 producers in the epicutaneous candidiasis model. Analyses of Rag2-/- mice and Rag2-/-Il2rg-/- mice revealed that production of IL-17A and IL-17F by ILC3s was sufficient for C. albicans clearance. Finally, we found that depletion of neutrophils impaired C. albicans clearance in the epidermal colonization model. Taken together, these findings indicate a critical and redundant function of IL-17A and IL-17F produced by ILC3s in host defense against C. albicans in the epidermis. The results also suggest that epidermal C. albicans clearance is independent of innate immune receptors or that these receptors act redundantly in fungal recognition and clearance.


Subject(s)
Candida albicans , Candidiasis , Interleukin-17/immunology , Animals , CARD Signaling Adaptor Proteins , Epidermis/metabolism , Immunity, Innate , Inflammation , Lymphocytes , Mice , Mice, Inbred C57BL , Mice, Knockout
18.
Immunity ; 41(6): 898-908, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25526305

ABSTRACT

The nucleotide-binding oligomerization domain (NOD) proteins NOD1 and NOD2, the founding members of the intracellular NOD-like receptor family, sense conserved motifs in bacterial peptidoglycan and induce proinflammatory and antimicrobial responses. Here, we discuss recent developments about the mechanisms by which NOD1 and NOD2 are activated by bacterial ligands, the regulation of their signaling pathways, and their role in host defense and inflammatory disease. Several routes for the entry of peptidoglycan ligands to the host cytosol to trigger activation of NOD1 and NOD2 have been elucidated. Furthermore, genetic screens and biochemical analyses have revealed mechanisms that regulate NOD1 and NOD2 signaling. Finally, recent studies have suggested several mechanisms to account for the link between NOD2 variants and susceptibility to Crohn's disease. Further understanding of NOD1 and NOD2 should provide new insight into the pathogenesis of disease and the development of new strategies to treat inflammatory and infectious disorders.


Subject(s)
Bacterial Infections/immunology , Inflammation/immunology , Nod1 Signaling Adaptor Protein/immunology , Nod2 Signaling Adaptor Protein/immunology , Receptors, Pattern Recognition/immunology , Animals , Antigens, Bacterial/immunology , Host-Pathogen Interactions , Humans , Immunity, Innate , Peptidoglycan/immunology , Signal Transduction
19.
Immunity ; 41(4): 620-32, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25367575

ABSTRACT

Pathobionts play a critical role in disease development, but the immune mechanisms against pathobionts remain poorly understood. Here, we report a critical role for interleukin-22 (IL-22) in systemic protection against bacterial pathobionts that translocate into the circulation after infection with the pathogen Clostridium difficile. Infection with C. difficile induced IL-22, and infected Il22(-/-) mice harbored high numbers of pathobionts in extraintestinal organs despite comparable pathogen load and intestinal damage in mutant and wild-type mice. Pathobionts exhibited increased resistant against complement-mediated phagocytosis, and their intravenous administration resulted in high animal mortality. Selective removal of translocated commensals rescued Il22(-/-) mice, and IL-22 administration enhanced the elimination of pathobionts. Mechanistically, IL-22 augmented bacterial phagocytosis by increasing the expression and bacterial binding of complement C3. Our study demonstrates an unexpected role for IL-22 in controlling the elimination of pathobionts that enter the systemic circulation through the regulation of the complement system.


Subject(s)
Clostridioides difficile/immunology , Complement C3/immunology , Enterocolitis, Pseudomembranous/immunology , Interleukins/immunology , Intestines/microbiology , Animals , Complement C3/biosynthesis , Elapid Venoms/pharmacology , Enterobacteriaceae/growth & development , Enterocolitis, Pseudomembranous/mortality , Interleukins/genetics , Intestines/injuries , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Phagocytosis/immunology , Interleukin-22
20.
Chem Biodivers ; 20(10): e202301015, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37624683

ABSTRACT

Bee bread (BB) is a beehive product generated upon fermentation of pollen combined with flower nectar and glandular secretions. The potential application of BB is related to its nutritional and functional components, including phenolic compounds. This is the first prospective study on palynological parameters, phenolics, antioxidant, and antibacterial activity of Chilean bee bread in vitro. The tested material exhibited high levels of phenolics (1340±186 mg GAE/100 g BB) and showed antioxidant capacity as determined by the FRAP (51±2 µmol Trolox equivalent/g BB) and ORAC-FL (643±64 µmol Trolox equivalent/g BB) and antibacterial activity against Streptococcus pyogenes. Furthermore, the phenolic acids and flavonoids was determined using liquid chromatography-mass spectrometry, and the concentration was determined using liquid chromatography with diode array detection. Kaempferol, quercetin, ferulic acid, and rutin were the main phenolics found. This study demonstrates the bioactive potential of Chilean BB and supports the evidence that this bee product is a promising source of antioxidants and antimicrobial compounds.

SELECTION OF CITATIONS
SEARCH DETAIL