Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Sci Signal ; 17(820): eabg8145, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38261657

ABSTRACT

Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome-nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form "specks," and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant-associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1ß in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.


Subject(s)
Hereditary Autoinflammatory Diseases , Inflammasomes , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing , Intracellular Signaling Peptides and Proteins , Leukocytes, Mononuclear , NLR Family, Pyrin Domain-Containing 3 Protein , Syndrome
2.
Thorax ; 79(3): 227-235, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38148147

ABSTRACT

BACKGROUND: Inflammatory subphenotypes have been identified in acute respiratory distress syndrome (ARDS). Hyperferritinaemia in sepsis is associated with hyperinflammation, worse clinical outcomes, and may predict benefit with immunomodulation. Our aim was to determine if raised ferritin identified a subphenotype in patients with ARDS. METHODS: Baseline plasma ferritin concentrations were measured in patients with ARDS from two randomised controlled trials of simvastatin (Hydroxymethylglutaryl-CoA Reductase Inhibition with Simvastatin in Acute Lung Injury to Reduce Pulmonary Dysfunction-2 (HARP-2); discovery cohort, UK) and neuromuscular blockade (ROSE; validation cohort, USA). Results were analysed using a logistic regression model with restricted cubic splines, to determine the ferritin threshold associated with 28-day mortality. RESULTS: Ferritin was measured in 511 patients from HARP-2 (95% of patients enrolled) and 847 patients (84% of patients enrolled) from ROSE. Ferritin was consistently associated with 28-day mortality in both studies and following a meta-analysis, a log-fold increase in ferritin was associated with an OR 1.71 (95% CI 1.01 to 2.90) for 28-day mortality. Patients with ferritin >1380 ng/mL (HARP-2 28%, ROSE 24%) had a significantly higher 28-day mortality and fewer ventilator-free days in both studies. Mediation analysis, including confounders (acute physiology and chronic health evaluation-II score and ARDS aetiology) demonstrated a statistically significant contribution of interleukin (IL)-18 as an intermediate pathway between ferritin and mortality. CONCLUSIONS: Ferritin is a clinically useful biomarker in ARDS and is associated with worse patient outcomes. These results provide support for prospective interventional trials of immunomodulatory agents targeting IL-18 in this hyperferritinaemic subgroup of patients with ARDS.


Subject(s)
Interleukin-18 , Respiratory Distress Syndrome , Humans , Prospective Studies , Simvastatin , Respiratory Distress Syndrome/etiology , Inflammation
3.
Cell Rep ; 42(8): 113012, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37598340

ABSTRACT

How the opportunistic Gram-negative pathogens of the genus Achromobacter interact with the innate immune system is poorly understood. Using three Achromobacter clinical isolates from two species, we show that the type 3 secretion system (T3SS) is required to induce cell death in human macrophages by inflammasome-dependent pyroptosis. Macrophages deficient in the inflammasome sensors NLRC4 or NLRP3 undergo pyroptosis upon bacterial internalization, but those deficient in both NLRC4 and NLRP3 do not, suggesting either sensor mediates pyroptosis in a T3SS-dependent manner. Detailed analysis of the intracellular trafficking of one isolate indicates that the intracellular bacteria reside in a late phagolysosome. Using an intranasal mouse infection model, we observe that Achromobacter damages lung structure and causes severe illness, contingent on a functional T3SS. Together, we demonstrate that Achromobacter species can survive phagocytosis by promoting macrophage cell death and inflammation by redundant mechanisms of pyroptosis induction in a T3SS-dependent manner.


Subject(s)
Achromobacter , Pyroptosis , Humans , Animals , Mice , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Type III Secretion Systems , Disease Models, Animal , Calcium-Binding Proteins , CARD Signaling Adaptor Proteins
4.
Trends Pharmacol Sci ; 43(8): 653-668, 2022 08.
Article in English | MEDLINE | ID: mdl-35513901

ABSTRACT

The nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has emerged as a key mediator of pathological inflammation in many diseases and is an exciting drug target. Here, we review the molecular basis of NLRP3 inhibition by drug-like small molecules under development as novel therapeutics. We also summarize recent strategies to block pyroptosis as a novel approach to suppress chronic inflammation. Major recent developments in this area include the elucidation of mechanisms of action (MoAs) by which small molecules block NLRP3 inflammasome assembly and gasdermin D (GSDMD)-induced pyroptosis. We also discuss the status of clinical trials using agents that block specific components of the NLRP3 pathway, including their potential clinical applications for the treatment of many diseases.


Subject(s)
Inflammasomes , Pyroptosis , Humans , Inflammasomes/metabolism , Inflammation/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis/physiology
5.
Methods Mol Biol ; 2459: 51-63, 2022.
Article in English | MEDLINE | ID: mdl-35212953

ABSTRACT

The non-canonical inflammasome is a signaling platform that allows for the detection of cytoplasmic lipopolysaccharides (LPS) in immune and non-immune cells. Upon detection of LPS, this inflammasome activates the signaling proteases caspase-4 and -5 (in humans) and caspase-11 (in mice). Inflammatory caspases activation leads to caspase self-processing and the cleavage of the pore-forming protein Gasdermin D (GSDMD). GSDMD N-terminal fragments oligomerize and form pores at the plasma membranes, leading to an inflammatory form of cell death called pyroptosis. Here, we describe a simple method to activate the non-canonical inflammasome in myeloid and epithelial cells and to measure its activity using cell death assay and immunoblotting.


Subject(s)
Inflammasomes , Intracellular Signaling Peptides and Proteins , Animals , Humans , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasm Proteins/metabolism , Phosphate-Binding Proteins , Pyroptosis
6.
Biochem Soc Trans ; 49(6): 2495-2507, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34854899

ABSTRACT

Inflammasomes are protein complexes in the innate immune system that regulate the production of pro-inflammatory cytokines and inflammatory cell death. Inflammasome activation and subsequent cell death often occur within minutes to an hour, so the pathway must be dynamically controlled to prevent excessive inflammation and the development of inflammatory diseases. Phosphorylation is a fundamental post-translational modification that allows rapid control over protein function and the phosphorylation of inflammasome proteins has emerged as a key regulatory step in inflammasome activation. Phosphorylation of inflammasome sensor and adapter proteins regulates their inter- and intra-molecular interactions, subcellular localisation, and function. The control of inflammasome phosphorylation may thus provide a new strategy for the development of anti-inflammatory therapeutics. Herein we describe the current knowledge of how phosphorylation operates as a critical switch for inflammasome signalling.


Subject(s)
Inflammasomes/metabolism , Signal Transduction , Animals , Humans , Phosphorylation , Protein Processing, Post-Translational , Subcellular Fractions/metabolism
7.
Cell ; 184(26): 6224-6226, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34942097

ABSTRACT

How the danger sensor NLRP3 is activated is intensively debated. Using cryo-electron microscopy (EM) approaches, Andreeva and colleagues made the remarkable discovery that inactive NLRP3 forms a double ring of 12-16 monomers that shield its pyrin domains from the cytosol. We discuss this surprising new mechanism of inflammasome regulation.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Cryoelectron Microscopy , Cytosol
8.
J Leukoc Biol ; 108(3): 937-952, 2020 09.
Article in English | MEDLINE | ID: mdl-32745339

ABSTRACT

The NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome is an immunological sensor that detects a wide range of microbial- and host-derived signals. Inflammasome activation results in the release of the potent pro-inflammatory cytokines IL-1ß and IL-18 and triggers a form of inflammatory cell death known as pyroptosis. Excessive NLRP3 activity is associated with the pathogenesis of a wide range of inflammatory diseases, thus NLRP3 activation mechanisms are an area of intensive research. NLRP3 inflammasome activation is a tightly regulated process that requires both priming and activation signals. In particular, recent research has highlighted the highly complex nature of the priming step, which involves transcriptional and posttranslational mechanisms, and numerous protein binding partners. This review will describe the current understanding of NLRP3 priming and will discuss the potential opportunities for targeting this process therapeutically to treat NLRP3-associated diseases.


Subject(s)
Inflammasomes/immunology , Inflammation/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , CARD Signaling Adaptor Proteins/immunology , CARD Signaling Adaptor Proteins/metabolism , Caspase Activation and Recruitment Domain , Gene Expression Regulation/immunology , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Inflammation/therapy , Mice , Molecular Targeted Therapy , NIMA-Related Kinases/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , Pyrin Domain , Pyroptosis/immunology , Transcription, Genetic , Ubiquitination
9.
EMBO J ; 38(21): e103533, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31617599

ABSTRACT

Signalling by innate immune cells is critical to shaping the adaptive immune response to microbial infection. In this issue of The EMBO Journal, Labzin et al reveal that the adaptive immune system can instruct the innate response to adenovirus infection. In human macrophages, antibody-coated adenovirus triggers a novel TRIM21-dependent pathway that activates the NLRP3 inflammasome and the secretion of IL-1ß.


Subject(s)
Infections , Inflammasomes , Adaptive Immunity , DNA , Humans , Interleukin-1beta , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein
10.
Nat Chem Biol ; 15(6): 556-559, 2019 06.
Article in English | MEDLINE | ID: mdl-31086327

ABSTRACT

Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and specific small-molecule inhibitor of the NLRP3 pathway, but its molecular target is not defined. Here, we show that MCC950 directly interacts with the Walker B motif within the NLRP3 NACHT domain, thereby blocking ATP hydrolysis and inhibiting NLRP3 activation and inflammasome formation.


Subject(s)
Adenosine Triphosphate/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Sulfones/pharmacology , Adenosine Triphosphate/metabolism , Binding Sites/drug effects , Furans , Heterocyclic Compounds, 4 or More Rings/chemistry , Humans , Hydrolysis/drug effects , Indenes , Inflammasomes/biosynthesis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sulfonamides , Sulfones/chemistry
11.
Immunity ; 49(6): 989-991, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566886

ABSTRACT

The NOD-like receptor protein NLRC3 attenuates myeloid cell inflammatory responses. In this issue of Immunity, Uchimura et al. (2018) reveal additional T-cell-intrinsic functions for NLRC3 in restricting T cell metabolism, T helper 1 and T helper 17 cell responses, and antiviral and autoimmune responses.


Subject(s)
Autoimmunity , Intercellular Signaling Peptides and Proteins , Carrier Proteins , Immunity, Innate , T-Lymphocytes
12.
Cell Res ; 28(12): 1202, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30467413

ABSTRACT

In the initial published version of this article, there was a mistake in the title. The correct title should be "Mitochondrial DNA synthesis fuels NLRP3 activation". This correction does not affect the description of the results or the conclusions of this work.

14.
J Exp Med ; 215(3): 827-840, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29432122

ABSTRACT

Host-protective caspase-1 activity must be tightly regulated to prevent pathology, but mechanisms controlling the duration of cellular caspase-1 activity are unknown. Caspase-1 is activated on inflammasomes, signaling platforms that facilitate caspase-1 dimerization and autoprocessing. Previous studies with recombinant protein identified a caspase-1 tetramer composed of two p20 and two p10 subunits (p20/p10) as an active species. In this study, we report that in the cell, the dominant species of active caspase-1 dimers elicited by inflammasomes are in fact full-length p46 and a transient species, p33/p10. Further p33/p10 autoprocessing occurs with kinetics specified by inflammasome size and cell type, and this releases p20/p10 from the inflammasome, whereupon the tetramer becomes unstable in cells and protease activity is terminated. The inflammasome-caspase-1 complex thus functions as a holoenzyme that directs the location of caspase-1 activity but also incorporates an intrinsic self-limiting mechanism that ensures timely caspase-1 deactivation. This intrinsic mechanism of inflammasome signal shutdown offers a molecular basis for the transient nature, and coordinated timing, of inflammasome-dependent inflammatory responses.


Subject(s)
Caspase 1/metabolism , Inflammasomes/metabolism , Animals , Kinetics , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Models, Biological , Nigericin/pharmacology , Protein Multimerization
15.
ChemMedChem ; 12(17): 1449-1457, 2017 09 07.
Article in English | MEDLINE | ID: mdl-28703484

ABSTRACT

Insulin-secretory sulfonylureas are widely used, cost-effective treatments for type 2 diabetes (T2D). However, pancreatic ß-cells are continually depleted as T2D progresses, thereby rendering the sulfonylurea drug class ineffective in controlling glycaemia. Dysregulation of the innate immune system via activation of the NLRP3 inflammasome, and the consequent production of interleukin-1ß, has been linked to pancreatic ß-cell death and multiple inflammatory complications of T2D disease. One proposed strategy for treating T2D is the use of sulfonylurea insulin secretagogues that are also NLRP3 inhibitors. We report the synthesis and biological evaluation of nine sulfonylureas that inhibit NLRP3 activation in murine bone-marrow- derived macrophages in a potent, dose-dependent manner. Six of these compounds inhibited NLRP3 at nanomolar concentrations and can also stimulate insulin secretion from a murine pancreatic cell line (MIN6). These novel compounds possess unprecedented dual modes of action, paving the way for a new generation of sulfonylureas that may be useful as therapeutic candidates and/or tool compounds in T2D and its associated inflammatory complications.


Subject(s)
Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Inflammasomes/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pancreas/drug effects , Sulfonylurea Compounds/chemistry , Sulfonylurea Compounds/pharmacology , Animals , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/immunology , HEK293 Cells , Humans , Inflammasomes/immunology , Insulin/immunology , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Pancreas/cytology , Pancreas/immunology
16.
Proc Natl Acad Sci U S A ; 114(32): E6480-E6489, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28739909

ABSTRACT

MyD88 adaptor-like (MAL) is a critical protein in innate immunity, involved in signaling by several Toll-like receptors (TLRs), key pattern recognition receptors (PRRs). Crystal structures of MAL revealed a nontypical Toll/interleukin-1 receptor (TIR)-domain fold stabilized by two disulfide bridges. We therefore undertook a structural and functional analysis of the role of reactive cysteine residues in the protein. Under reducing conditions, the cysteines do not form disulfides, but under oxidizing conditions they are highly amenable to modification. The solution structure of the reduced form of the MAL TIR domain, determined by NMR spectroscopy, reveals a remarkable structural rearrangement compared with the disulfide-bonded structure, which includes the relocation of a ß-strand and repositioning of the functionally important "BB-loop" region to a location more typical for TIR domains. Redox measurements by NMR further reveal that C91 has the highest redox potential of all cysteines in MAL. Indeed, mass spectrometry revealed that C91 undergoes glutathionylation in macrophages activated with the TLR4 ligand lipopolysaccharide (LPS). The C91A mutation limits MAL glutathionylation and acts as a dominant negative, blocking the interaction of MAL with its downstream target MyD88. The H92P mutation mimics the dominant-negative effects of the C91A mutation, presumably by preventing C91 glutathionylation. The MAL C91A and H92P mutants also display diminished degradation and interaction with interleukin-1 receptor-associated kinase 4 (IRAK4). We conclude that in the cell, MAL is not disulfide-bonded and requires glutathionylation of C91 for signaling.


Subject(s)
Glutathione/metabolism , Membrane Glycoproteins , Protein Processing, Post-Translational , Receptors, Interleukin-1 , Signal Transduction , Amino Acid Substitution , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Glutathione/chemistry , Glutathione/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mutation, Missense , Nuclear Magnetic Resonance, Biomolecular , Protein Domains , Protein Structure, Secondary , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/metabolism , Structure-Activity Relationship
17.
J Biol Chem ; 292(29): 12077-12087, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28576828

ABSTRACT

The NLRP3 inflammasome is a multiprotein complex that regulates the activation of caspase-1 leading to the maturation of the proinflammatory cytokines IL-1ß and IL-18 and promoting pyroptosis. Classically, the NLRP3 inflammasome in murine macrophages is activated by the recognition of pathogen-associated molecular patterns and by many structurally unrelated factors. Understanding the precise mechanism of NLRP3 activation by such a wide array of stimuli remains elusive, but several signaling events, including cytosolic efflux and influx of select ions, have been suggested. Accordingly, several studies have indicated a role of anion channels in NLRP3 inflammasome assembly, but their direct involvement has not been shown. Here, we report that the chloride intracellular channel proteins CLIC1 and CLIC4 participate in the regulation of the NLRP3 inflammasome. Confocal microscopy and cell fractionation experiments revealed that upon LPS stimulation of macrophages, CLIC1 and CLIC4 translocated into the nucleus and cellular membrane. In LPS/ATP-stimulated bone marrow-derived macrophages (BMDMs), CLIC1 or CLIC4 siRNA transfection impaired transcription of IL-1ß, ASC speck formation, and secretion of mature IL-1ß. Collectively, our results demonstrate that CLIC1 and CLIC4 participate both in the priming signal for IL-1ß and in NLRP3 activation.


Subject(s)
Chloride Channels/metabolism , Inflammasomes/drug effects , Interleukin-1beta/agonists , Macrophage Activation/drug effects , Macrophages/drug effects , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Active Transport, Cell Nucleus/drug effects , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Line , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Protein Transport/drug effects , Pyroptosis/drug effects , RAW 264.7 Cells , RNA Interference , Signal Transduction/drug effects
18.
Cell Mol Immunol ; 14(1): 118-126, 2017 01.
Article in English | MEDLINE | ID: mdl-26996064

ABSTRACT

Inflammation is the host response to microbial infection or sterile injury that aims to eliminate the insult, repair the tissue and restore homeostasis. Macrophages and the NLRP3 inflammasome are key sentinels for both types of insult. Although it is well established that the NLRP3 inflammasome is activated by microbial products and molecules released during sterile injury, it is unclear whether the responses elicited by these different types of signals are distinct. In this study, we used lipopolysaccharide and tumor necrosis factor as prototypical microbial and sterile signal 1 stimuli, respectively, to prime the NLRP3 inflammasome. We then used the bacterial toxin nigericin and a common product released from necrotic cells, ATP, as prototypical microbial and sterile signal 2 stimuli, respectively, to trigger the assembly of the NLRP3 inflammasome complex in mouse and human macrophages. We found that NLRP3 inflammasome responses were weakest when both signal 1 and signal 2 were sterile, but responses were faster and stronger when at least one of the two signals was microbial. Ultimately, the most rapid and potent responses were elicited when both signals were microbial. Together, these data suggest that microbial versus sterile signals are distinct, both kinetically and in magnitude, in their ability to generate inflammasome-dependent responses. This hierarchy of NLRP3 responses to sterile versus microbial stimuli likely reflects the urgent need for the immune system to respond rapidly to the presence of infection to halt pathogen dissemination.


Subject(s)
Inflammasomes/metabolism , Macrophages/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Animals , Cross-Priming/drug effects , Cross-Priming/immunology , Humans , Interleukin-1beta/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice, Inbred C57BL , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
19.
Blood ; 128(25): 2960-2975, 2016 12 22.
Article in English | MEDLINE | ID: mdl-27737891

ABSTRACT

Despite genetic heterogeneity, myelodysplastic syndromes (MDSs) share features of cytological dysplasia and ineffective hematopoiesis. We report that a hallmark of MDSs is activation of the NLRP3 inflammasome, which drives clonal expansion and pyroptotic cell death. Independent of genotype, MDS hematopoietic stem and progenitor cells (HSPCs) overexpress inflammasome proteins and manifest activated NLRP3 complexes that direct activation of caspase-1, generation of interleukin-1ß (IL-1ß) and IL-18, and pyroptotic cell death. Mechanistically, pyroptosis is triggered by the alarmin S100A9 that is found in excess in MDS HSPCs and bone marrow plasma. Further, like somatic gene mutations, S100A9-induced signaling activates NADPH oxidase (NOX), increasing levels of reactive oxygen species (ROS) that initiate cation influx, cell swelling, and ß-catenin activation. Notably, knockdown of NLRP3 or caspase-1, neutralization of S100A9, and pharmacologic inhibition of NLRP3 or NOX suppress pyroptosis, ROS generation, and nuclear ß-catenin in MDSs and are sufficient to restore effective hematopoiesis. Thus, alarmins and founder gene mutations in MDSs license a common redox-sensitive inflammasome circuit, which suggests new avenues for therapeutic intervention.


Subject(s)
Inflammasomes/metabolism , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Calgranulin B/metabolism , Cell Size , Colony-Forming Units Assay , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Ion Channel Gating , Ion Channels/metabolism , Mice, Transgenic , Mutation/genetics , NADPH Oxidases/metabolism , Phenotype , Pyroptosis , Reactive Oxygen Species/metabolism , beta Catenin/metabolism
20.
Science ; 352(6292): aad1210, 2016 Jun 17.
Article in English | MEDLINE | ID: mdl-27313051

ABSTRACT

The NLRP3 inflammasome controls interleukin-1ß maturation in antigen-presenting cells, but a direct role for NLRP3 in human adaptive immune cells has not been described. We found that the NLRP3 inflammasome assembles in human CD4(+) T cells and initiates caspase-1-dependent interleukin-1ß secretion, thereby promoting interferon-γ production and T helper 1 (T(H)1) differentiation in an autocrine fashion. NLRP3 assembly requires intracellular C5 activation and stimulation of C5a receptor 1 (C5aR1), which is negatively regulated by surface-expressed C5aR2. Aberrant NLRP3 activity in T cells affects inflammatory responses in human autoinflammatory disease and in mouse models of inflammation and infection. Our results demonstrate that NLRP3 inflammasome activity is not confined to "innate immune cells" but is an integral component of normal adaptive T(H)1 responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Carrier Proteins/metabolism , Complement C5a/immunology , Inflammasomes/immunology , Interferon-gamma/biosynthesis , Th1 Cells/immunology , Adaptive Immunity , Animals , Autocrine Communication , Carrier Proteins/genetics , Complement Activation , Cryopyrin-Associated Periodic Syndromes/immunology , Disease Models, Animal , HEK293 Cells , Humans , Immunity, Innate , Inflammation/immunology , Membrane Cofactor Protein/immunology , Mice , Mice, Mutant Strains , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species/metabolism , Receptor, Anaphylatoxin C5a/agonists , Receptor, Anaphylatoxin C5a/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Antigen, T-Cell/agonists , Receptors, Antigen, T-Cell/metabolism , Receptors, Chemokine/agonists , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...