Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Eur J Immunol ; 54(5): e2350515, 2024 May.
Article in English | MEDLINE | ID: mdl-38361219

ABSTRACT

Caspase-1 location in cells has been studied with fluorochrome-labeled inhibitors of caspase-1 (FLICA reagents). We report that FLICA reagents have limited cell-membrane permeability. This impacts experimental design as cells with intact membranes, including caspase-1 knockout cells, are not appropriate controls for cells with inflammasome-induced gasdermin D membrane pores.


Subject(s)
Caspase 1 , Caspase Inhibitors , Cell Membrane Permeability , Fluorescent Dyes , Inflammasomes , Macrophages , Caspase 1/metabolism , Animals , Macrophages/immunology , Macrophages/metabolism , Cell Membrane Permeability/drug effects , Mice , Inflammasomes/metabolism , Caspase Inhibitors/pharmacology , Mice, Knockout , Phosphate-Binding Proteins/metabolism , Humans
2.
Immunity ; 45(2): 333-45, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27533014

ABSTRACT

Many pathogens, including Plasmodium spp., exploit the interaction of programmed death-1 (PD-1) with PD-1-ligand-1 (PD-L1) to "deactivate" T cell functions, but the role of PD-L2 remains unclear. We studied malarial infections to understand the contribution of PD-L2 to immunity. Here we have shown that higher PD-L2 expression on blood dendritic cells, from Plasmodium falciparum-infected individuals, correlated with lower parasitemia. Mechanistic studies in mice showed that PD-L2 was indispensable for establishing effective CD4(+) T cell immunity against malaria, because it not only inhibited PD-L1 to PD-1 activity but also increased CD3 and inducible co-stimulator (ICOS) expression on T cells. Importantly, administration of soluble multimeric PD-L2 to mice with lethal malaria was sufficient to dramatically improve immunity and survival. These studies show immuno-regulation by PD-L2, which has the potential to be translated into an effective treatment for malaria and other diseases where T cell immunity is ineffective or short-lived due to PD-1-mediated signaling.


Subject(s)
B7-H1 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adamantane/analogs & derivatives , Adamantane/therapeutic use , Adult , Animals , Antimalarials/therapeutic use , B7-H1 Antigen/genetics , Cells, Cultured , Clinical Trials as Topic , Dendritic Cells/parasitology , Female , Humans , Immunity, Cellular , Lymphocyte Activation , Malaria, Falciparum/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Parasitemia/immunology , Peroxides/therapeutic use , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Receptor/genetics , Pyrimidines/therapeutic use , Triazoles/therapeutic use , Young Adult
3.
EMBO J ; 39(17): e106202, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32869315

ABSTRACT

Shigella, a major cause of bacterial dysentery, knows when it is not wanted. To generate and maintain its niche within host cells, this unwelcome guest injects several dozen virulence factors via a type 3 secretion system (T3SS). In this issue, Ashida et al (2020) have elucidated the role of two such factors from Shigella flexneri-OspC1 and OspD3-that together counteract apoptotic and necroptotic death pathways in colonised epithelial cells. As a result, Shigella can replicate to high levels within the colonic epithelium, leading to the substantial epithelial damage in shigellosis and efficient bacterial release for faecal transmission.


Subject(s)
Dysentery, Bacillary , Shigella , Caspase 8 , Cell Death , Epithelial Cells , Humans , Shigella/genetics , Shigella flexneri/genetics
4.
Eur J Immunol ; 53(7): e2250056, 2023 07.
Article in English | MEDLINE | ID: mdl-37058370

ABSTRACT

TLRs engage numerous adaptor proteins and signaling molecules, enabling a complex series of post-translational modifications (PTMs) to mount inflammatory responses. TLRs themselves are post-translationally modified following ligand-induced activation, with this being required to relay the full spectrum of proinflammatory signaling responses. Here, we reveal indispensable roles for TLR4 Y672 and Y749 phosphorylation in mounting optimal LPS-inducible inflammatory responses in primary mouse macrophages. LPS promotes phosphorylation at both tyrosine residues, with Y749 phosphorylation being required for maintenance of total TLR4 protein levels and Y672 phosphorylation exerting its pro-inflammatory effects more selectively by initiating ERK1/2 and c-FOS phosphorylation. Our data also support a role for the TLR4-interacting membrane proteins SCIMP and the SYK kinase axis in mediating TLR4 Y672 phosphorylation to permit downstream inflammatory responses in murine macrophages. The corresponding residue in human TLR4 (Y674) is also required for optimal LPS signaling responses. Our study, thus, reveals how a single PTM on one of the most widely studied innate immune receptors orchestrates downstream inflammatory responses.


Subject(s)
Cytokines , Lipopolysaccharides , Humans , Animals , Mice , Phosphorylation , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Toll-Like Receptor 4 , Tyrosine/metabolism , Tyrosine/pharmacology , Macrophages
5.
J Virol ; 97(11): e0125123, 2023 Nov 30.
Article in English | MEDLINE | ID: mdl-37850747

ABSTRACT

IMPORTANCE: Dengue virus, an arbovirus, causes an estimated 100 million symptomatic infections annually and is an increasing threat as the mosquito range expands with climate change. Dengue epidemics are a substantial strain on local economies and health infrastructure, and an understanding of what drives severe disease may enable treatments to help reduce hospitalizations. Factors exacerbating dengue disease are debated, but gut-related symptoms are much more frequent in severe than mild cases. Using mouse models of dengue infection, we have shown that inflammation and damage are earlier and more severe in the gut than in other tissues. Additionally, we observed impairment of the gut mucus layer and propose that breakdown of the barrier function exacerbates inflammation and promotes severe dengue disease. This idea is supported by recent data from human patients showing elevated bacteria-derived molecules in dengue patient serum. Therapies aiming to maintain gut integrity may help to abrogate severe dengue disease.


Subject(s)
Dengue Virus , Severe Dengue , Animals , Humans , Mice , Culicidae , Dengue Virus/physiology , Inflammation/virology , Severe Dengue/pathology , Kinetics
6.
Mol Psychiatry ; 28(7): 2878-2893, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36316366

ABSTRACT

Coronavirus disease-2019 (COVID-19) is primarily a respiratory disease, however, an increasing number of reports indicate that SARS-CoV-2 infection can also cause severe neurological manifestations, including precipitating cases of probable Parkinson's disease. As microglial NLRP3 inflammasome activation is a major driver of neurodegeneration, here we interrogated whether SARS-CoV-2 can promote microglial NLRP3 inflammasome activation. Using SARS-CoV-2 infection of transgenic mice expressing human angiotensin-converting enzyme 2 (hACE2) as a COVID-19 pre-clinical model, we established the presence of virus in the brain together with microglial activation and NLRP3 inflammasome upregulation in comparison to uninfected mice. Next, utilising a model of human monocyte-derived microglia, we identified that SARS-CoV-2 isolates can bind and enter human microglia in the absence of viral replication. This interaction of virus and microglia directly induced robust inflammasome activation, even in the absence of another priming signal. Mechanistically, we demonstrated that purified SARS-CoV-2 spike glycoprotein activated the NLRP3 inflammasome in LPS-primed microglia, in a ACE2-dependent manner. Spike protein also could prime the inflammasome in microglia through NF-κB signalling, allowing for activation through either ATP, nigericin or α-synuclein. Notably, SARS-CoV-2 and spike protein-mediated microglial inflammasome activation was significantly enhanced in the presence of α-synuclein fibrils and was entirely ablated by NLRP3-inhibition. Finally, we demonstrate SARS-CoV-2 infected hACE2 mice treated orally post-infection with the NLRP3 inhibitory drug MCC950, have significantly reduced microglial inflammasome activation, and increased survival in comparison with untreated SARS-CoV-2 infected mice. These results support a possible mechanism of microglial innate immune activation by SARS-CoV-2, which could explain the increased vulnerability to developing neurological symptoms akin to Parkinson's disease in COVID-19 infected individuals, and a potential therapeutic avenue for intervention.


Subject(s)
COVID-19 , Parkinson Disease , Humans , Mice , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Microglia/metabolism , alpha-Synuclein/metabolism , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/metabolism , Mice, Transgenic
7.
Mol Cell ; 64(2): 236-250, 2016 10 20.
Article in English | MEDLINE | ID: mdl-27746017

ABSTRACT

Caspase-8 activation can be triggered by death receptor-mediated formation of the death-inducing signaling complex (DISC) and by the inflammasome adaptor ASC. Caspase-8 assembles with FADD at the DISC and with ASC at the inflammasome through its tandem death effector domain (tDED), which is regulated by the tDED-containing cellular inhibitor cFLIP and the viral inhibitor MC159. Here we present the caspase-8 tDED filament structure determined by cryoelectron microscopy. Extensive assembly interfaces not predicted by the previously proposed linear DED chain model were uncovered, and were further confirmed by structure-based mutagenesis in filament formation in vitro and Fas-induced apoptosis and ASC-mediated caspase-8 recruitment in cells. Structurally, the two DEDs in caspase-8 use quasi-equivalent contacts to enable assembly. Using the tDED filament structure as a template, structural analyses reveal the interaction surfaces between FADD and caspase-8 and the distinct mechanisms of regulation by cFLIP and MC159 through comingling and capping, respectively.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Fas-Associated Death Domain Protein/chemistry , Viral Proteins/chemistry , Amino Acid Sequence , Apoptosis/drug effects , Binding Sites , CARD Signaling Adaptor Proteins , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Cryoelectron Microscopy , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Death Effector Domain , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Humans , Jurkat Cells , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection , Viral Proteins/genetics , Viral Proteins/metabolism , fas Receptor/pharmacology
8.
J Enzyme Inhib Med Chem ; 39(1): 2313055, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38416868

ABSTRACT

Toll-like receptor (TLR) innate immunity signalling protects against pathogens, but excessive or prolonged signalling contributes to a range of inflammatory conditions. Structural information on the TLR cytoplasmic TIR (Toll/interleukin-1 receptor) domains and the downstream adaptor proteins can help us develop inhibitors targeting this pathway. The small molecule o-vanillin has previously been reported as an inhibitor of TLR2 signalling. To study its mechanism of action, we tested its binding to the TIR domain of the TLR adaptor MAL/TIRAP (MALTIR). We show that o-vanillin binds to MALTIR and inhibits its higher-order assembly in vitro. Using NMR approaches, we show that o-vanillin forms a covalent bond with lysine 210 of MAL. We confirm in mouse and human cells that o-vanillin inhibits TLR2 but not TLR4 signalling, independently of MAL, suggesting it may covalently modify TLR2 signalling complexes directly. Reactive aldehyde-containing small molecules such as o-vanillin may target multiple proteins in the cell.


Subject(s)
Benzaldehydes , Lysine , Toll-Like Receptor 2 , Humans , Animals , Mice , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptors/metabolism , Membrane Glycoproteins/metabolism , Receptors, Interleukin-1/metabolism
9.
J Biol Chem ; 298(12): 102666, 2022 12.
Article in English | MEDLINE | ID: mdl-36334634

ABSTRACT

Conventional assays to probe signaling protein interactions and function involve measurement of luciferase reporter expression within the bulk cell population, with lack of control over target-protein expression level. To address this issue, we have developed a rapid and robust flow cytometric assay for analysis of signaling protein function. A fluorescent reporter and fluorescent tagging of the target protein enables simultaneous assessment of protein expression and signaling within individual cells. We have applied our technique to the analysis of variants of the lipopolysaccharide receptor Toll-like receptor 4 (TLR4) and its adapter protein MyD88, using a NF-кB-responsive promoter driving mScarlet-I expression. The assay enables exclusion of nontransfected cells and overexpressing cells that signal spontaneously. Additionally, our assay allows the identification of protein variants that fail to express. We found that the assays were highly sensitive, with cells expressing an appropriate level of GFP-MyD88 showing approximately 200-fold induction of mScarlet-I by lipopolysaccharide, and we can detect subtle protein concentration-dependent effects of mutations. Importantly, the assay is adaptable to various signaling pathways.


Subject(s)
Lipopolysaccharides , Myeloid Differentiation Factor 88 , Adaptor Proteins, Signal Transducing/metabolism , Lipopolysaccharides/pharmacology , Lipopolysaccharides/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Signal Transduction , Humans
10.
EMBO Rep ; 20(9): e48891, 2019 09.
Article in English | MEDLINE | ID: mdl-31379068

ABSTRACT

The non-canonical inflammasome mediates pyroptotic cell death in response to bacterial lipopolysaccharide (LPS) found in the cytosol. Understanding the mechanism and regulation of this system is of great interest, given its central role in mouse models of bacterial septic shock. In this issue of EMBO Reports, Benaoudia and colleagues sought to discover extra players in the human non-canonical inflammasome using a CRISPR library screen; the only strongly positive hit apart from the known components caspase-4 and gasdermin D was interferon regulatory factor-2 (IRF2) [1 ]. IRF2 was found to be a transcriptional activator of caspase-4, and in its absence, induction of IRF1 could substitute to maintain caspase-4 expression.


Subject(s)
Inflammasomes , Lipopolysaccharides , Animals , Humans , Interferon Regulatory Factor-1 , Interferon Regulatory Factor-2 , Mice
11.
Immunol Cell Biol ; 97(1): 17-28, 2019 01.
Article in English | MEDLINE | ID: mdl-30052286

ABSTRACT

Inflammasomes are protein complexes activated by infection and cellular stress that promote caspase-1 activation and subsequent inflammatory cytokine processing and cell death. It has been anticipated that inflammasome activity contributes to autoimmunity. However, we previously showed that macrophages from autoimmune New Zealand Black (NZB) mice lack NLRP3 inflammasome function, and their absent in melanoma 2 (AIM2) inflammasome responses are compromised by high expression of the AIM2 antagonist protein p202. Here we found that the point mutation leading to lack of NLRP3 expression occurred early in the NZB strain establishment, as it is shared with the related obese strain New Zealand Obese, but not with the unrelated New Zealand White (NZW) strain. The first cross progeny of NZB and NZW mice develop more severe lupus nephritis than the NZB strain. We have compared AIM2 and NLRP3 inflammasome function in macrophages from NZB, NZW, and NZB/W F1 mice. The NZW parental strain showed strong inflammasome function, whereas the NZB/W F1 have haploinsufficient expression of NLRP3 and show reduced NLRP3 and AIM2 inflammasome responses, particularly at low stimulus strength. It remains to be established whether the low inflammasome function could contribute to loss of tolerance and the onset of autoimmunity in NZB and NZB/W F1. However, with amplifying inflammatory stimuli through the course of disease, the NLRP3 response in the NZB/W F1 may be sufficient to contribute to kidney damage at later stages of disease.


Subject(s)
Autoimmunity , DNA-Binding Proteins/deficiency , Inflammasomes , Macrophages , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Animals , Autoimmunity/genetics , DNA-Binding Proteins/immunology , Female , Inflammasomes/genetics , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred NZB , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Point Mutation
12.
Immunol Cell Biol ; 96(10): 1120-1130, 2018 11.
Article in English | MEDLINE | ID: mdl-30003588

ABSTRACT

Outer membrane vesicles (OMVs) are constitutively produced by Gram-negative bacteria both in vivo and in vitro. These lipid-bound structures carry a range of immunogenic components derived from the parent cell, which are transported into host target cells and activate the innate immune system. Recent advances in the field have shed light on some of the multifaceted roles of OMVs in host-pathogen interactions. In this study, we investigated the ability of OMVs from two clinically important pathogens, Pseudomonas aeruginosa and Helicobacter pylori, to activate canonical and noncanonical inflammasomes. P. aeruginosa OMVs induced inflammasome activation in mouse macrophages, as evidenced by "speck" formation, as well as the cleavage and secretion of interleukin-1ß and caspase-1. These responses were independent of AIM2 and NLRC4 canonical inflammasomes, but dependent on the noncanonical caspase-11 pathway. Moreover, P. aeruginosa OMVs alone were able to activate the inflammasome in a TLR-dependent manner, without requiring an exogenous priming signal. In contrast, H. pylori OMVs were not able to induce inflammasome activation in macrophages. Using CRISPR/Cas9 knockout THP-1 cells lacking the human caspase-11 homologs, caspase-4 and -5,we demonstrated that caspase-5 but not caspase-4 is required for inflammasome activation by P. aeruginosa OMVs in human monocytes. In contrast, free P. aeruginosa lipopolysaccharide (LPS) transfected into cells induced inflammasome responses via caspase-4. This suggests that caspase-4 and caspase-5 differentially recognize LPS depending on its physical form or route of delivery into the cell. These findings have relevance to Gram-negative infections in humans and the use of OMVs as novel vaccines.


Subject(s)
Caspases/metabolism , Extracellular Vesicles/metabolism , Inflammasomes/metabolism , Monocytes/immunology , Monocytes/metabolism , Pseudomonas Infections/immunology , Pseudomonas Infections/metabolism , Pseudomonas aeruginosa/physiology , Caspase 1/metabolism , Cell Line , Humans , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Pseudomonas Infections/microbiology , Signal Transduction
13.
Adv Exp Med Biol ; 1062: 89-106, 2018.
Article in English | MEDLINE | ID: mdl-29845527

ABSTRACT

Recent structural and functional advances provide fresh insight into the biology of the dengue virus non-structural protein, NS1 and suggest new avenues of research. The work of our lab and others have shown that the secreted, hexameric form of NS1 has a systemic toxic effect, inducing inflammatory cytokines and acting directly on endothelial cells to produce the hallmark of dengue disease, vascular leak. We also demonstrated that NS1 exerts its toxic activity through recognition by the innate immune receptor TLR4, mimicking the bacterial endotoxin LPS. This monograph covers the background underpinning these new findings and discusses new avenues for antiviral and vaccine intervention.


Subject(s)
Dengue Virus/immunology , Severe Dengue/virology , Viral Nonstructural Proteins/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dengue Virus/chemistry , Dengue Virus/genetics , Endothelial Cells/immunology , Endothelial Cells/virology , Humans , Receptors, Virus/genetics , Receptors, Virus/immunology , Severe Dengue/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/genetics
14.
Immunol Cell Biol ; 95(5): 491-495, 2017 05.
Article in English | MEDLINE | ID: mdl-28220810

ABSTRACT

The secreted hexameric form of the dengue virus (DENV) non-structural protein 1 (NS1) has recently been shown to elicit inflammatory cytokine release and disrupt endothelial cell monolayer integrity. This suggests that circulating NS1 contributes to the vascular leak that plays a major role in the pathology of dengue haemorrhagic fever and shock. Pathways activated by NS1 are thus of great interest as potential therapeutic targets. Recent works have separately implicated both toll-like receptor 4 (TLR4) and the TLR2/6 heterodimer in immune cell activation by NS1. Here we have used mouse gene knockout macrophages and antibodies blocking TLR function in human peripheral blood mononuclear cells to show that recombinant NS1, expressed and purified from eukaryotic cells, induces cytokine production via TLR4 but not TLR2/6. Furthermore, the commercial Escherichia coli-derived recombinant NS1 preparation used in other work to implicate TLR2/6 in the response is not correctly folded and appears to be contaminated by several microbial TLR ligands. Thus TLR4 remains a therapeutic target for DENV infections, with TLR4 antagonists holding promise for the treatment of dengue disease.


Subject(s)
Dengue Virus/immunology , Leukocytes/virology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 6/metabolism , Viral Nonstructural Proteins/immunology , Animals , Dengue Virus/drug effects , Escherichia coli/metabolism , Humans , Leukocytes/drug effects , Leukocytes/pathology , Lipopolysaccharides , Mice, Inbred C57BL , Polymyxin B/pharmacology , Protein Multimerization/drug effects
15.
FASEB J ; 30(5): 1901-12, 2016 05.
Article in English | MEDLINE | ID: mdl-26839376

ABSTRACT

We aimed to characterize antimicrobial zinc trafficking within macrophages and to determine whether the professional intramacrophage pathogen Salmonella enterica serovar Typhimurium (S Typhimurium) subverts this pathway. Using both Escherichia coli and S Typhimurium, we show that TLR signaling promotes the accumulation of vesicular zinc within primary human macrophages. Vesicular zinc is delivered to E. coli to promote microbial clearance, whereas S. Typhimurium evades this response via Salmonella pathogenicity island (SPI)-1. Even in the absence of SPI-1 and the zinc exporter ZntA, S Typhimurium resists the innate immune zinc stress response, implying the existence of additional host subversion mechanisms. We also demonstrate the combinatorial antimicrobial effects of zinc and copper, a pathway that S. Typhimurium again evades. Our use of complementary tools and approaches, including confocal microscopy, direct assessment of intramacrophage bacterial zinc stress responses, specific E. coli and S Typhimurium mutants, and inductively coupled plasma mass spectroscopy, has enabled carefully controlled characterization of this novel innate immune antimicrobial pathway. In summary, our study provides new insights at the cellular level into the well-documented effects of zinc in promoting host defense against infectious disease, as well as the complex host subversion strategies employed by S Typhimurium to combat this pathway.-Kapetanovic, R., Bokil, N. J., Achard, M. E. S., Ong, C.-L. Y., Peters, K. M., Stocks, C. J., Phan, M.-D., Monteleone, M., Schroder, K., Irvine, K. M., Saunders, B. M., Walker, M. J., Stacey, K. J., McEwan, A. G., Schembri, M. A., Sweet, M. J. Salmonella employs multiple mechanisms to subvert the TLR-inducible zinc-mediated antimicrobial response of human macrophages.


Subject(s)
Macrophages/immunology , Macrophages/metabolism , Salmonella typhimurium/physiology , Salmonella/physiology , Toll-Like Receptors/metabolism , Zinc/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Cells, Cultured , Copper , Cytoplasmic Vesicles/chemistry , Cytoplasmic Vesicles/metabolism , Gene Expression Regulation, Bacterial , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Toll-Like Receptors/genetics
16.
J Immunol ; 194(1): 455-62, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25404358

ABSTRACT

Inflammasomes are large protein complexes induced by a wide range of microbial, stress, and environmental stimuli that function to induce cell death and inflammatory cytokine processing. Formation of an inflammasome involves dramatic relocalization of the inflammasome adapter protein apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) into a single speck. We have developed a flow cytometric assay for inflammasome formation, time of flight inflammasome evaluation, which detects the change in ASC distribution within the cell. The transit of ASC into the speck is detected by a decreased width or increased height of the pulse of emitted fluorescence. This assay can be used to quantify native inflammasome formation in subsets of mixed cell populations ex vivo. It can also provide a rapid and sensitive technique for investigating molecular interactions in inflammasome formation, by comparison of wild-type and mutant proteins in inflammasome reconstitution experiments.


Subject(s)
Apoptosis Regulatory Proteins/immunology , Flow Cytometry/methods , Inflammasomes/immunology , Animals , Apoptosis/immunology , Apoptosis Regulatory Proteins/genetics , Bone Marrow Cells/immunology , CARD Signaling Adaptor Proteins/immunology , Caspase 1/genetics , Cell Line , HEK293 Cells , Humans , Inflammasomes/analysis , Inflammation Mediators/immunology , Macrophages/immunology , Mice , Mice, Knockout
17.
J Immunol ; 195(3): 1233-41, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26116505

ABSTRACT

Inflammasomes are protein complexes that promote caspase activation, resulting in processing of IL-1ß and cell death, in response to infection and cellular stresses. Inflammasomes have been anticipated to contribute to autoimmunity. The New Zealand Black (NZB) mouse develops anti-erythrocyte Abs and is a model of autoimmune hemolytic anemia. These mice also develop anti-nuclear Abs typical of lupus. In this article, we show that NZB macrophages have deficient inflammasome responses to a DNA virus and fungal infection. Absent in melanoma 2 (AIM2) inflammasome responses are compromised in NZB by high expression of the AIM 2 antagonist protein p202, and consequently NZB cells had low IL-1ß output in response to both transfected DNA and mouse CMV infection. Surprisingly, we also found that a second inflammasome system, mediated by the NLR family, pyrin domain containing 3 (NLRP3) initiating protein, was completely lacking in NZB cells. This was due to a point mutation in an intron of the Nlrp3 gene in NZB mice, which generates a novel splice acceptor site. This leads to incorporation of a pseudoexon with a premature stop codon. The lack of full-length NLRP3 protein results in NZB being effectively null for Nlrp3, with no production of bioactive IL-1ß in response to NLRP3 stimuli, including infection with Candida albicans. Thus, this autoimmune strain harbors two inflammasome deficiencies, mediated through quite distinct mechanisms. We hypothesize that the inflammasome deficiencies in NZB alter the interaction of the host with both microflora and pathogens, promoting prolonged production of cytokines that contribute to development of autoantibodies.


Subject(s)
Anemia, Hemolytic, Autoimmune/genetics , Carrier Proteins/genetics , DNA-Binding Proteins/genetics , Inflammasomes/genetics , Macrophages/immunology , Anemia, Hemolytic, Autoimmune/immunology , Animals , Antibodies, Antinuclear/immunology , Autoimmunity/genetics , Autoimmunity/immunology , Candida albicans/immunology , Candidiasis/immunology , Candidiasis/microbiology , Carrier Proteins/immunology , Caspase 1/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/virology , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/biosynthesis , Intracellular Signaling Peptides and Proteins/biosynthesis , Mice , Mice, Inbred NZB , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction/genetics , Signal Transduction/immunology
18.
J Biol Chem ; 290(49): 29217-30, 2015 Dec 04.
Article in English | MEDLINE | ID: mdl-26468282

ABSTRACT

Inflammasomes mediate inflammatory and cell death responses to pathogens and cellular stress signals via activation of procaspases-1 and -8. During inflammasome assembly, activated receptors of the NLR or PYHIN family recruit the adaptor protein ASC and initiate polymerization of its pyrin domain (PYD) into filaments. We show that ASC filaments in turn nucleate procaspase-8 death effector domain (DED) filaments in vitro and in vivo. Interaction between ASC PYD and procaspase-8 tandem DEDs optimally required both DEDs and represents an unusual heterotypic interaction between domains of the death fold superfamily. Analysis of ASC PYD mutants showed that interaction surfaces that mediate procaspase-8 interaction overlap with those required for ASC self-association and interaction with the PYDs of inflammasome initiators. Our data indicate that multiple types of death fold domain filaments form at inflammasomes and that PYD/DED and homotypic PYD interaction modes are similar. Interestingly, we observed condensation of procaspase-8 filaments containing the catalytic domain, suggesting that procaspase-8 interactions within and/or between filaments may be involved in caspase-8 activation. Procaspase-8 filaments may also be relevant to apoptosis induced by death receptors.


Subject(s)
Caspase 8/metabolism , Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Apoptosis , CARD Signaling Adaptor Proteins , Caspase 1/metabolism , Catalytic Domain , Cell Death , HEK293 Cells , Humans , Inflammation , Microscopy, Fluorescence , Mutation , Protein Binding , Signal Transduction
19.
Immunol Cell Biol ; 94(5): 520-4, 2016 05.
Article in English | MEDLINE | ID: mdl-26833024

ABSTRACT

Inflammasomes are molecular complexes activated by infection and cellular stress, leading to caspase-1 activation and subsequent interleukin-1ß (IL-1ß) processing and cell death. The autoimmune NZB mouse strain does not express NLRP3, a key inflammasome initiator mediating responses to a wide variety of stimuli including endogenous danger signals, environmental irritants and a range of bacterial, fungal and viral pathogens. We have previously identified an intronic point mutation in the Nlrp3 gene from NZB mice that generates a splice acceptor site. This leads to inclusion of a pseudoexon that introduces an early termination codon and is proposed to be the cause of NLRP3 inflammasome deficiency in NZB cells. Here we have used exon skipping antisense oligonucleotides (AONs) to prevent aberrant splicing of Nlrp3 in NZB macrophages, and this restored both NLRP3 protein expression and NLRP3 inflammasome activity. Thus, the single point mutation leading to aberrant splicing is the sole cause of NLRP3 inflammasome deficiency in NZB macrophages. The NZB mouse provides a model for addressing a splicing defect in macrophages and could be used to further investigate AON design and delivery of AONs to macrophages in vivo.


Subject(s)
Autoimmunity/drug effects , Exons/genetics , Inflammasomes/metabolism , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Oligonucleotides, Antisense/pharmacology , Alternative Splicing/genetics , Animals , Base Sequence , Macrophages/drug effects , Mice, Inbred C57BL , Mice, Inbred NZB , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
20.
EMBO Rep ; 15(9): 982-90, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24990442

ABSTRACT

A current paradigm proposes that mitochondrial damage is a critical determinant of NLRP3 inflammasome activation. Here, we genetically assess whether mitochondrial signalling represents a unified mechanism to explain how NLRP3 is activated by divergent stimuli. Neither co-deletion of the essential executioners of mitochondrial apoptosis BAK and BAX, nor removal of the mitochondrial permeability transition pore component cyclophilin D, nor loss of the mitophagy regulator Parkin, nor deficiency in MAVS affects NLRP3 inflammasome function. In contrast, caspase-8, a caspase essential for death-receptor-mediated apoptosis, is required for efficient Toll-like-receptor-induced inflammasome priming and cytokine production. Collectively, these results demonstrate that mitochondrial apoptosis is not required for NLRP3 activation, and highlight an important non-apoptotic role for caspase-8 in regulating inflammasome activation and pro-inflammatory cytokine levels.


Subject(s)
Carrier Proteins/biosynthesis , Caspase 8/biosynthesis , Inflammasomes/metabolism , Mitochondria/metabolism , Apoptosis/genetics , Autophagy/genetics , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carrier Proteins/genetics , Caspase 8/genetics , Cells, Cultured , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Cyclophilins/genetics , Humans , Interleukin-1beta/biosynthesis , Mitochondria/pathology , Mitophagy/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Toll-Like Receptors/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL