Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
Add more filters

Publication year range
1.
Cell ; 140(6): 821-32, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20303873

ABSTRACT

Inflammasomes are molecular platforms activated upon cellular infection or stress that trigger the maturation of proinflammatory cytokines such as interleukin-1beta to engage innate immune defenses. Strong associations between dysregulated inflammasome activity and human heritable and acquired inflammatory diseases highlight the importance this pathway in tailoring immune responses. Here, we comprehensively review mechanisms directing normal inflammasome function and its dysregulation in disease. Agonists and activation mechanisms of the NLRP1, NLRP3, IPAF, and AIM2 inflammasomes are discussed. Regulatory mechanisms that potentiate or limit inflammasome activation are examined, as well as emerging links between the inflammasome and pyroptosis and autophagy.


Subject(s)
Inflammation/immunology , Organelles/immunology , Receptors, Pattern Recognition/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Caspases/metabolism , Humans , Inflammation/metabolism , Interleukins/metabolism
2.
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
3.
Nat Immunol ; 11(2): 136-40, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20023662

ABSTRACT

The NLRP3 inflammasome has a major role in regulating innate immunity. Deregulated inflammasome activity is associated with several inflammatory diseases, yet little is known about the signaling pathways that lead to its activation. Here we show that NLRP3 interacted with thioredoxin (TRX)-interacting protein (TXNIP), a protein linked to insulin resistance. Inflammasome activators such as uric acid crystals induced the dissociation of TXNIP from thioredoxin in a reactive oxygen species (ROS)-sensitive manner and allowed it to bind NLRP3. TXNIP deficiency impaired activation of the NLRP3 inflammasome and subsequent secretion of interleukin 1beta (IL-1beta). Akin to Txnip(-/-) mice, Nlrp3(-/-) mice showed improved glucose tolerance and insulin sensitivity. The participation of TXNIP in the NLRP3 inflammasome activation may provide a mechanistic link to the observed involvement of IL-1beta in the pathogenesis of type 2 diabetes.


Subject(s)
Carrier Proteins/immunology , Inflammation/immunology , Oxidative Stress/physiology , Signal Transduction/immunology , Thioredoxins/immunology , Animals , Carrier Proteins/metabolism , Cell Line , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/metabolism , Enzyme-Linked Immunosorbent Assay , Glucose/metabolism , Humans , Immunoblotting , Immunoprecipitation , Inflammation/metabolism , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Thioredoxins/metabolism , Transfection
4.
Immunity ; 38(6): 1154-63, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23809162

ABSTRACT

Omega-3 fatty acids (ω-3 FAs) have potential anti-inflammatory activity in a variety of inflammatory human diseases, but the mechanisms remain poorly understood. Here we show that stimulation of macrophages with ω-3 FAs, including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and other family members, abolished NLRP3 inflammasome activation and inhibited subsequent caspase-1 activation and IL-1ß secretion. In addition, G protein-coupled receptor 120 (GPR120) and GPR40 and their downstream scaffold protein ß-arrestin-2 were shown to be involved in inflammasome inhibition induced by ω-3 FAs. Importantly, ω-3 FAs also prevented NLRP3 inflammasome-dependent inflammation and metabolic disorder in a high-fat-diet-induced type 2 diabetes model. Our results reveal a mechanism through which ω-3 FAs repress inflammation and prevent inflammation-driven diseases and suggest the potential clinical use of ω-3 FAs in gout, autoinflammatory syndromes, or other NLRP3 inflammasome-driven inflammatory diseases.


Subject(s)
Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/drug therapy , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Inflammasomes/metabolism , Inflammation/prevention & control , Macrophages/drug effects , Animals , Arrestins/metabolism , Carrier Proteins/genetics , Caspase 1/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/immunology , Diet, High-Fat/adverse effects , Enzyme Activation/drug effects , Fatty Acids, Omega-3/immunology , Inflammasomes/immunology , Interleukin-1beta/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, G-Protein-Coupled/metabolism , beta-Arrestin 2 , beta-Arrestins
5.
Immunity ; 36(3): 388-400, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22444631

ABSTRACT

Through their capacity to sense danger signals and to generate active interleukin-1ß (IL-1ß), inflammasomes occupy a central role in the inflammatory response. In contrast to IL-1ß, little is known about how IL-1α is regulated. We found that all inflammasome activators also induced the secretion of IL-1α, leading to the cosecretion of both IL-1 cytokines. Depending on the type of inflammasome activator, release of IL-1α was inflammasome dependent or independent. Calcium influx induced by the opening of cation channels was sufficient for the inflammasome-independent IL-1α secretion. In both cases, IL-1α was released primarily in a processed form, resulting from intracellular cleavage by calpain-like proteases. Inflammasome-caspase-1-dependent release of IL-1α and IL-1ß was independent of caspase-1 catalytic activity, defining a mode of action for caspase-1. Because inflammasomes contribute to the pathology of numerous chronic inflammatory diseases such as gout and diabetes, IL-1α antagonists may be beneficial in the treatment of these disorders.


Subject(s)
Caspase 1/metabolism , Inflammasomes/immunology , Interleukin-1alpha/biosynthesis , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Calcium Channels/metabolism , Calcium Signaling/immunology , Calcium-Binding Proteins/metabolism , Cell Death/immunology , DNA-Binding Proteins , Female , Humans , Inflammasomes/metabolism , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/metabolism , Interleukin-1beta/biosynthesis , Male , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Peritonitis/immunology , Protein Processing, Post-Translational , Receptors, Interleukin-1/metabolism , Signal Transduction/immunology
6.
Immunity ; 36(2): 215-27, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22365665

ABSTRACT

Interleukin-1ß (IL-1ß) is a potent inflammatory cytokine that is usually cleaved and activated by inflammasome-associated caspase-1. To determine whether IL-1ß activation is regulated by inhibitor of apoptosis (IAP) proteins, we treated macrophages with an IAP-antagonist "Smac mimetic" compound or genetically deleted the genes that encode the three IAP family members cIAP1, cIAP2, and XIAP. After Toll-like receptor priming, IAP inhibition triggered cleavage of IL-1ß that was mediated not only by the NLRP3-caspase-1 inflammasome, but also by caspase-8 in a caspase-1-independent manner. In the absence of IAPs, rapid and full generation of active IL-1ß by the NLRP3-caspase-1 inflammasome, or by caspase-8, required the kinase RIP3 and reactive oxygen species production. These results demonstrate that activation of the cell death-inducing ripoptosome platform and RIP3 can generate bioactive IL-1ß and implicate them as additional targets for the treatment of pathological IL-1-driven inflammatory responses.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Interleukin-1beta/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins , Baculoviral IAP Repeat-Containing 3 Protein , Carrier Proteins/agonists , Carrier Proteins/metabolism , Caspase 1/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/deficiency , Inhibitor of Apoptosis Proteins/genetics , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/agonists , Molecular Mimicry , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases , X-Linked Inhibitor of Apoptosis Protein/deficiency , X-Linked Inhibitor of Apoptosis Protein/genetics , X-Linked Inhibitor of Apoptosis Protein/metabolism
7.
Nat Immunol ; 9(9): 1037-46, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18641654

ABSTRACT

Tumor necrosis factor receptor 1 (TNFR1) and Toll-like receptors (TLRs) regulate immune and inflammatory responses. Here we show that the TNFR1-associated death domain protein (TRADD) is critical in TNFR1, TLR3 and TLR4 signaling. TRADD deficiency abrogated TNF-induced apoptosis, prevented recruitment of the ubiquitin ligase TRAF2 and ubiquitination of the adaptor RIP1 in the TNFR1 signaling complex, and considerably inhibited but did not completely abolish activation of the transcription factor NF-kappaB and mitogen-activated protein kinases 'downstream' of TNFR1. TRIF-dependent cytokine production induced by the synthetic double-stranded RNA poly(I:C) and lipopolysaccharide was lower in TRADD-deficient mice than in wild-type mice. Moreover, TRADD deficiency inhibited poly(I:C)-mediated RIP1 ubiquitination and activation of NF-kappaB and mitogen-activated protein kinase signaling in fibroblasts but not in bone marrow macrophages. Thus, TRADD is an essential component of TNFR1 signaling and has a critical but apparently cell type-specific function in TRIF-dependent TLR responses.


Subject(s)
Signal Transduction , TNF Receptor-Associated Death Domain Protein/deficiency , TNF Receptor-Associated Factor 1/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/physiology , Animals , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , TNF Receptor-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/genetics , Ubiquitin/metabolism
8.
Immunity ; 34(2): 213-23, 2011 Feb 25.
Article in English | MEDLINE | ID: mdl-21349431

ABSTRACT

Type I interferon (IFN) is a common therapy for autoimmune and inflammatory disorders, yet the mechanisms of action are largely unknown. Here we showed that type I IFN inhibited interleukin-1 (IL-1) production through two distinct mechanisms. Type I IFN signaling, via the STAT1 transcription factor, repressed the activity of the NLRP1 and NLRP3 inflammasomes, thereby suppressing caspase-1-dependent IL-1ß maturation. In addition, type I IFN induced IL-10 in a STAT1-dependent manner; autocrine IL-10 then signaled via STAT3 to reduce the abundance of pro-IL-1α and pro-IL-1ß. In vivo, poly(I:C)-induced type I IFN diminished IL-1ß production in response to alum and Candida albicans, thus increasing susceptibility to this fungal pathogen. Importantly, monocytes from multiple sclerosis patients undergoing IFN-ß treatment produced substantially less IL-1ß than monocytes derived from healthy donors. Our findings may thus explain the effectiveness of type I IFN in the treatment of inflammatory diseases but also the observed "weakening" of the immune system after viral infection.


Subject(s)
Inflammasomes/metabolism , Interferon Type I/physiology , Interleukin-1/biosynthesis , Animals , Apoptosis Regulatory Proteins/physiology , Candida albicans/physiology , Candidiasis/etiology , Candidiasis/immunology , Carrier Proteins/physiology , Caspase 1/deficiency , Caspase 1/genetics , Caspase 1/physiology , Cells, Cultured/metabolism , Disease Susceptibility , Gene Expression Regulation/drug effects , Humans , Interferon Inducers/pharmacology , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon-beta/therapeutic use , Interleukin-1/genetics , Interleukin-10/physiology , Mice , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Multiple Sclerosis/drug therapy , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Peritonitis/etiology , Peritonitis/immunology , Poly I-C/pharmacology , STAT1 Transcription Factor/deficiency , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology
9.
Mol Cell ; 46(2): 200-11, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22464733

ABSTRACT

Caspase 1 is part of the inflammasome, which is assembled upon pathogen recognition, while caspases 3 and/or 7 are mediators of apoptotic and nonapoptotic functions. PARP1 cleavage is a hallmark of apoptosis yet not essential, suggesting it has another physiological role. Here we show that after LPS stimulation, caspase 7 is activated by caspase 1, translocates to the nucleus, and cleaves PARP1 at the promoters of a subset of NF-κB target genes negatively regulated by PARP1. Mutating the PARP1 cleavage site D214 renders PARP1 uncleavable and inhibits PARP1 release from chromatin and chromatin decondensation, thereby restraining the expression of cleavage-dependent NF-κB target genes. These findings propose an apoptosis-independent regulatory role for caspase 7-mediated PARP1 cleavage in proinflammatory gene expression and provide insight into inflammasome signaling.


Subject(s)
Caspase 7/physiology , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/physiology , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/physiology , Chromatin/metabolism , Gene Expression Regulation , Humans , Inflammation/genetics , Mice , Mutation , NLR Family, Pyrin Domain-Containing 3 Protein , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Poly(ADP-ribose) Polymerases/genetics , Signal Transduction
10.
Mol Cell ; 47(5): 681-93, 2012 Sep 14.
Article in English | MEDLINE | ID: mdl-22854598

ABSTRACT

Biochemical evidence implicates the death-domain (DD) protein PIDD as a molecular switch capable of signaling cell survival or death in response to genotoxic stress. PIDD activity is determined by binding-partner selection at its DD: whereas recruitment of RIP1 triggers prosurvival NF-κB signaling, recruitment of RAIDD activates proapoptotic caspase-2 via PIDDosome formation. However, it remains unclear how interactor selection, and thus fate decision, is regulated at the PIDD platform. We show that the PIDDosome functions in the "Chk1-suppressed" apoptotic response to DNA damage, a conserved ATM/ATR-caspase-2 pathway antagonized by Chk1. In this pathway, ATM phosphorylates PIDD on Thr788 within the DD. This phosphorylation is necessary and sufficient for RAIDD binding and caspase-2 activation. Conversely, nonphosphorylatable PIDD fails to bind RAIDD or activate caspase-2, and engages prosurvival RIP1 instead. Thus, ATM phosphorylation of the PIDD DD enables a binary switch through which cells elect to survive or die upon DNA injury.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , CRADD Signaling Adaptor Protein/metabolism , Caspase 2/metabolism , Cell Death , Cell Survival , Cells, Cultured , DNA Damage , HEK293 Cells , HeLa Cells , Humans , Phosphorylation
11.
Immunity ; 32(2): 279-89, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20171123

ABSTRACT

Dendritic cell (DC) populations consist of multiple subsets that are essential orchestrators of the immune system. Technological limitations have so far prevented systems-wide accurate proteome comparison of rare cell populations in vivo. Here, we used high-resolution mass spectrometry-based proteomics, combined with label-free quantitation algorithms, to determine the proteome of mouse splenic conventional and plasmacytoid DC subsets to a depth of 5,780 and 6,664 proteins, respectively. We found mutually exclusive expression of pattern recognition pathways not previously known to be different among conventional DC subsets. Our experiments assigned key viral recognition functions to be exclusively expressed in CD4(+) and double-negative DCs. The CD8alpha(+) DCs largely lack the receptors required to sense certain viruses in the cytoplasm. By avoiding activation via cytoplasmic receptors, including retinoic acid-inducible gene I, CD8alpha(+) DCs likely gain a window of opportunity to process and present viral antigens before activation-induced shutdown of antigen presentation pathways occurs.


Subject(s)
DEAD-box RNA Helicases/biosynthesis , Dendritic Cells/metabolism , Proteomics/methods , Respirovirus Infections/immunology , Sendai virus/immunology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , Cell Separation , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Dendritic Cells/virology , Flow Cytometry , Host-Pathogen Interactions , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Proteomics/instrumentation , Sendai virus/pathogenicity
12.
Immunity ; 28(5): 651-61, 2008 May.
Article in English | MEDLINE | ID: mdl-18439848

ABSTRACT

Upon detection of viral RNA, the helicases RIG-I and/or MDA5 trigger, via their adaptor Cardif (also known as IPS-1, MAVS, or VISA), the activation of the transcription factors NF-kappaB and IRF3, which collaborate to induce an antiviral type I interferon (IFN) response. FADD and RIP1, known as mediators of death-receptor signaling, are implicated in this antiviral pathway; however, the link between death-receptor and antiviral signaling is not known. Here we showed that TRADD, a crucial adaptor of tumor necrosis factor receptor (TNFRI), was important in RIG-like helicase (RLH)-mediated signal transduction. TRADD is recruited to Cardif and orchestrated complex formation with the E3 ubiquitin ligase TRAF3 and TANK and with FADD and RIP1, leading to the activation of IRF3 and NF-kappaB. Loss of TRADD prevented Cardif-dependent activation of IFN-beta, reduced the production of IFN-beta in response to RNA viruses, and enhanced vesicular stomatitis virus replication. Thus, TRADD is not only an essential component of proinflammatory TNFRI signaling, but is also required for RLH-Cardif-dependent antiviral immune responses.


Subject(s)
DNA Helicases/metabolism , Interferon Regulatory Factor-3/metabolism , Rhabdoviridae Infections/immunology , TNF Receptor-Associated Death Domain Protein/metabolism , Vesiculovirus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Fas-Associated Death Domain Protein/metabolism , GTPase-Activating Proteins/metabolism , Humans , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mice, Mutant Strains , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , Rhabdoviridae Infections/virology , Signal Transduction , TNF Receptor-Associated Factor 3/immunology , TNF Receptor-Associated Factor 3/metabolism , Ubiquitin-Protein Ligases/metabolism , Vesiculovirus/physiology
13.
Nature ; 469(7329): 221-5, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21124315

ABSTRACT

An inflammatory response initiated by the NLRP3 inflammasome is triggered by a variety of situations of host 'danger', including infection and metabolic dysregulation. Previous studies suggested that NLRP3 inflammasome activity is negatively regulated by autophagy and positively regulated by reactive oxygen species (ROS) derived from an uncharacterized organelle. Here we show that mitophagy/autophagy blockade leads to the accumulation of damaged, ROS-generating mitochondria, and this in turn activates the NLRP3 inflammasome. Resting NLRP3 localizes to endoplasmic reticulum structures, whereas on inflammasome activation both NLRP3 and its adaptor ASC redistribute to the perinuclear space where they co-localize with endoplasmic reticulum and mitochondria organelle clusters. Notably, both ROS generation and inflammasome activation are suppressed when mitochondrial activity is dysregulated by inhibition of the voltage-dependent anion channel. This indicates that NLRP3 inflammasome senses mitochondrial dysfunction and may explain the frequent association of mitochondrial damage with inflammatory diseases.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Mitochondria/metabolism , Animals , Apoptosis Regulatory Proteins , Autophagy/drug effects , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Cell Line , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Endoplasmic Reticulum/metabolism , Humans , Immunity, Innate , Inflammasomes/drug effects , Inflammation/metabolism , Inflammation/pathology , Interleukin-1beta/metabolism , Macrophages/cytology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Thioredoxins/genetics , Thioredoxins/metabolism , Voltage-Dependent Anion Channels/metabolism
14.
Nat Rev Immunol ; 6(3): 196-204, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16498450

ABSTRACT

Cellular caspase-8 (FLICE)-like inhibitory protein (cFLIP) was originally identified as an inhibitor of death-receptor signalling through competition with caspase-8 for recruitment to FAS-associated via death domain (FADD). More recently, it has been determined that both cFLIP and caspase-8 are required for the survival and proliferation of T cells following T-cell-receptor stimulation. This paradoxical finding launched new investigations of how these molecules might connect with signalling pathways that link to cell survival and growth following antigen-receptor activation. As discussed in this Review, insight gained from these studies indicates that cFLIP and caspase-8 form a heterodimer that ultimately links T-cell-receptor signalling to activation of nuclear factor-kappaB through a complex that includes B-cell lymphoma 10 (BCL-10), mucosa-associated-lymphoid-tissue lymphoma-translocation gene 1 (MALT1) and receptor-interacting protein 1 (RIP1).


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation , T-Lymphocytes/physiology , Animals , Autoimmunity , CASP8 and FADD-Like Apoptosis Regulating Protein , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Neoplasms/etiology , T-Lymphocytes/immunology , fas Receptor/physiology
16.
Nature ; 460(7252): 269-73, 2009 Jul 09.
Article in English | MEDLINE | ID: mdl-19494813

ABSTRACT

Inflammation is a protective attempt by the host to remove injurious stimuli and initiate the tissue healing process. The inflammatory response must be actively terminated, however, because failure to do so can result in 'bystander' damage to tissues and diseases such as arthritis or type-2 diabetes. Yet the mechanisms controlling excessive inflammatory responses are still poorly understood. Here we show that mouse effector and memory CD4(+) T cells abolish macrophage inflammasome-mediated caspase-1 activation and subsequent interleukin 1beta release in a cognate manner. Inflammasome inhibition is observed for all tested NLRP1 (commonly called NALP1) and NLRP3 (NALP3 or cryopyrin) activators, whereas NLRC4 (IPAF) inflammasome function and release of other inflammatory mediators such as CXCL2, interleukin 6 and tumour necrosis factor are not affected. Suppression of the NLRP3 inflammasome requires cell-to-cell contact and can be mimicked by macrophage stimulation with selected ligands of the tumour necrosis factor family, such as CD40L (also known as CD40LG). In a NLRP3-dependent peritonitis model, effector CD4(+) T cells are responsible for decreasing neutrophil recruitment in an antigen-dependent manner. Our findings reveal an unexpected mechanism of inflammasome inhibition, whereby effector and memory T cells suppress potentially damaging inflammation, yet leave the primary inflammatory response, crucial for the onset of immunity, intact.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Apoptosis Regulatory Proteins/antagonists & inhibitors , CD4-Positive T-Lymphocytes/immunology , Carrier Proteins/antagonists & inhibitors , Immunity, Innate/immunology , Inflammation/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antigens/immunology , Apoptosis Regulatory Proteins/metabolism , Bone Marrow Cells/cytology , Carrier Proteins/metabolism , Caspase 1/metabolism , Cells, Cultured , Immunologic Memory , Inflammation/immunology , Inflammation/pathology , Interleukin-1beta/immunology , Ligands , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Neutrophils/immunology , Peritoneal Cavity/cytology , Tumor Necrosis Factors/immunology , Tumor Necrosis Factors/metabolism
17.
Nature ; 459(7245): 433-6, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19339971

ABSTRACT

Fungal infections represent a serious threat, particularly in immunocompromised patients. Interleukin-1beta (IL-1beta) is a key pro-inflammatory factor in innate antifungal immunity. The mechanism by which the mammalian immune system regulates IL-1beta production after fungal recognition is unclear. Two signals are generally required for IL-1beta production: an NF-kappaB-dependent signal that induces the synthesis of pro-IL-1beta (p35), and a second signal that triggers proteolytic pro-IL-1beta processing to produce bioactive IL-1beta (p17) via Caspase-1-containing multiprotein complexes called inflammasomes. Here we demonstrate that the tyrosine kinase Syk, operating downstream of several immunoreceptor tyrosine-based activation motif (ITAM)-coupled fungal pattern recognition receptors, controls both pro-IL-1beta synthesis and inflammasome activation after cell stimulation with Candida albicans. Whereas Syk signalling for pro-IL-1beta synthesis selectively uses the Card9 pathway, inflammasome activation by the fungus involves reactive oxygen species production and potassium efflux. Genetic deletion or pharmalogical inhibition of Syk selectively abrogated inflammasome activation by C. albicans but not by inflammasome activators such as Salmonella typhimurium or the bacterial toxin nigericin. Nlrp3 (also known as NALP3) was identified as the critical NOD-like receptor family member that transduces the fungal recognition signal to the inflammasome adaptor Asc (Pycard) for Caspase-1 (Casp1) activation and pro-IL-1beta processing. Consistent with an essential role for Nlrp3 inflammasomes in antifungal immunity, we show that Nlrp3-deficient mice are hypersusceptible to Candida albicans infection. Thus, our results demonstrate the molecular basis for IL-1beta production after fungal infection and identify a crucial function for the Nlrp3 inflammasome in mammalian host defence in vivo.


Subject(s)
Candida albicans/immunology , Carrier Proteins/immunology , Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Animals , Candida albicans/physiology , Caspase 1/metabolism , Enzyme Activation , Humans , Inflammation/immunology , Interleukin-1beta/biosynthesis , Interleukin-1beta/immunology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Macrophages/metabolism , Mice , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nigericin/pharmacology , Potassium/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/deficiency , Protein-Tyrosine Kinases/genetics , Reactive Oxygen Species/metabolism , Syk Kinase
18.
Proc Natl Acad Sci U S A ; 109(45): 18384-9, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23090995

ABSTRACT

A chronic inflammatory microenvironment favors tumor progression through molecular mechanisms that are still incompletely defined. In inflammation-induced skin cancers, IL-1 receptor- or caspase-1-deficient mice, or mice specifically deficient for the inflammasome adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD) in myeloid cells, had reduced tumor incidence, pointing to a role for IL-1 signaling and inflammasome activation in tumor development. However, mice fully deficient for ASC were not protected, and mice specifically deficient for ASC in keratinocytes developed more tumors than controls, suggesting that, in contrast to its proinflammatory role in myeloid cells, ASC acts as a tumor-suppressor in keratinocytes. Accordingly, ASC protein expression was lost in human cutaneous squamous cell carcinoma, but not in psoriatic skin lesions. Stimulation of primary mouse keratinocytes or the human keratinocyte cell line HaCaT with UVB induced an ASC-dependent phosphorylation of p53 and expression of p53 target genes. In HaCaT cells, ASC interacted with p53 at the endogenous level upon UVB irradiation. Thus, ASC in different tissues may influence tumor growth in opposite directions: it has a proinflammatory role in infiltrating cells that favors tumor development, but it also limits keratinocyte proliferation in response to noxious stimuli, possibly through p53 activation, which helps suppressing tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytoskeletal Proteins/metabolism , Epithelium/pathology , Inflammasomes/metabolism , Skin Neoplasms/pathology , Skin/pathology , 9,10-Dimethyl-1,2-benzanthracene , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Caspase 1/deficiency , Caspase 1/metabolism , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cytokines/biosynthesis , Cytoskeletal Proteins/deficiency , Down-Regulation , Epithelium/metabolism , Humans , Inflammation/pathology , Keratinocytes/metabolism , Keratinocytes/pathology , Mice , Mice, Knockout , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms, Squamous Cell/pathology , Organ Specificity , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/prevention & control , Tetradecanoylphorbol Acetate , Tumor Microenvironment , Tumor Suppressor Protein p53/metabolism
19.
Immunol Rev ; 243(1): 136-51, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21884173

ABSTRACT

An inflammasome is a multiprotein complex that serves as a platform for caspase-1 activation and caspase-1-dependent proteolytic maturation and secretion of interleukin-1ß (IL-1ß). Though a number of inflammasomes have been described, the NLRP3 inflammasome is the most extensively studied but also the most elusive. It is unique in that it responds to numerous physically and chemically diverse stimuli. The potent proinflammatory and pyrogenic activities of IL-1ß necessitate that inflammasome activity is tightly controlled. To this end, a priming step is first required to induce the expression of both NLRP3 and proIL-1ß. This event renders the cell competent for NLRP3 inflammasome activation and IL-1ß secretion, and it is highly regulated by negative feedback loops. Despite the wide array of NLRP3 activators, the actual triggering of NLRP3 is controlled by integration a comparatively small number of signals that are common to nearly all activators. Minimally, these include potassium efflux, elevated levels of reactive oxygen species (ROS), and, for certain activators, lysosomal destabilization. Further investigation of how these and potentially other as yet uncharacterized signals are integrated by the NLRP3 inflammasome and the relevance of these biochemical events in vivo should provide new insight into the mechanisms of host defense and autoinflammatory conditions.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Inflammation Mediators/immunology , Interleukin-1beta/immunology , Receptor Cross-Talk , Animals , Feedback, Physiological , Humans , Immunity , NLR Family, Pyrin Domain-Containing 3 Protein , Receptor Cross-Talk/immunology , Signal Transduction/immunology
20.
Eur J Immunol ; 43(12): 3336-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23964013

ABSTRACT

Sterile cell death mediated inflammation is linked to several pathological disorders and involves danger recognition of intracellular molecules released by necrotic cells that activate different groups of innate pattern recognition receptors. Toll-like receptors directly interact with their extrinsic or intrinsic agonists and induce multiple proinflammatory mediators. In contrast, the NLRP3 inflammasome is rather thought to represent a downstream element integrating various indirect stimuli into proteolytic cleavage of interleukin (IL)-1ß and IL-18. Here, we report that histones released from necrotic cells induce IL-1ß secretion in an NLRP3-ASC-caspase-1-dependent manner. Genetic deletion of NLRP3 in mice significantly attenuated histone-induced IL-1ß production and neutrophil recruitment. Furthermore, necrotic cells induced neutrophil recruitment, which was significantly reduced by histone-neutralizing antibodies or depleting extracellular histones via enzymatic degradation. These results identify cytosolic uptake of necrotic cell-derived histones as a triggering mechanism of sterile inflammation, which involves NLRP3 inflammasome activation and IL-1ß secretion via oxidative stress.


Subject(s)
Carrier Proteins/immunology , Histones/immunology , Inflammasomes/immunology , Neutrophils/immunology , Oxidative Stress/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase 1/immunology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/immunology , Gene Deletion , Histones/antagonists & inhibitors , Inflammasomes/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-18/genetics , Interleukin-18/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Necrosis/genetics , Necrosis/immunology , Necrosis/pathology , Neutrophils/pathology , Oxidative Stress/drug effects , Oxidative Stress/genetics , Proteolysis/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL