Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
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
2.
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
3.
Eur J Immunol ; 42(12): 3291-301, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22936176

ABSTRACT

Malaria is a major cause of morbidity worldwide with reports of over 200-500 million infected individuals and nearly 1 million deaths each year. Antibodies have been shown to play a critical role in controlling the blood stage of this disease; however, in malaria-endemic areas antibody immunity is slow to develop despite years of exposure to Plasmodium spp. the causative parasite. Using rodent Plasmodium yoelii YM, we provide evidence that malarial infections result in a decrease in the proportion of DCs that express the B-cell survival factor, BAFF, resulting in a decreased ability of these DCs to support memory B-cell differentiation into antibody secreting cells (ASCs) and/or the survival of ASCs. Further, compared with infected WT mice, ASC numbers were significantly increased in malaria-infected transgenic mice that either overexpressed BAFF or mice with BAFF-independent B-cell survival (B-cell-restricted TRAF3 deletion). Remarkably, BAFF-overexpressing mice were protected from lethal malaria infections, indicating the significance of the role BAFF plays in determining the outcome of malaria infections. These findings describe a previously unappreciated mechanism by which Plasmodium spp. can depress the generation of protective antibody responses.


Subject(s)
Antibodies, Protozoan/immunology , B-Cell Activating Factor/immunology , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Immunologic Memory , Malaria/immunology , Plasma Cells/immunology , Plasmodium yoelii/immunology , Animals , Antibodies, Protozoan/genetics , Antibodies, Protozoan/metabolism , Antibody Formation/genetics , B-Cell Activating Factor/biosynthesis , B-Cell Activating Factor/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Cell Survival/immunology , Dendritic Cells/metabolism , Female , Gene Expression Regulation/genetics , Humans , Malaria/genetics , Malaria/metabolism , Mice , Mice, Transgenic , Plasma Cells/metabolism
4.
BMC Evol Biol ; 12: 140, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22871040

ABSTRACT

BACKGROUND: Proteins of the mammalian PYHIN (IFI200/HIN-200) family are involved in defence against infection through recognition of foreign DNA. The family member absent in melanoma 2 (AIM2) binds cytosolic DNA via its HIN domain and initiates inflammasome formation via its pyrin domain. AIM2 lies within a cluster of related genes, many of which are uncharacterised in mouse. To better understand the evolution, orthology and function of these genes, we have documented the range of PYHIN genes present in representative mammalian species, and undertaken phylogenetic and expression analyses. RESULTS: No PYHIN genes are evident in non-mammals or monotremes, with a single member found in each of three marsupial genomes. Placental mammals show variable family expansions, from one gene in cow to four in human and 14 in mouse. A single HIN domain appears to have evolved in the common ancestor of marsupials and placental mammals, and duplicated to give rise to three distinct forms (HIN-A, -B and -C) in the placental mammal ancestor. Phylogenetic analyses showed that AIM2 HIN-C and pyrin domains clearly diverge from the rest of the family, and it is the only PYHIN protein with orthology across many species. Interestingly, although AIM2 is important in defence against some bacteria and viruses in mice, AIM2 is a pseudogene in cow, sheep, llama, dolphin, dog and elephant. The other 13 mouse genes have arisen by duplication and rearrangement within the lineage, which has allowed some diversification in expression patterns. CONCLUSIONS: The role of AIM2 in forming the inflammasome is relatively well understood, but molecular interactions of other PYHIN proteins involved in defence against foreign DNA remain to be defined. The non-AIM2 PYHIN protein sequences are very distinct from AIM2, suggesting they vary in effector mechanism in response to foreign DNA, and may bind different DNA structures. The PYHIN family has highly varied gene composition between mammalian species due to lineage-specific duplication and loss, which probably indicates different adaptations for fighting infectious disease. Non-genomic DNA can indicate infection, or a mutagenic threat. We hypothesise that defence of the genome against endogenous retroelements has been an additional evolutionary driver for PYHIN proteins.


Subject(s)
Evolution, Molecular , Mammals/genetics , Nuclear Proteins/genetics , Animals , Bayes Theorem , DNA-Binding Proteins , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/chemistry , Nuclear Proteins/immunology , Phylogeny , Rats , Transcriptome
5.
Cell Rep ; 4(2): 327-39, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23850291

ABSTRACT

Mouse p202 containing two hemopoietic expression, interferon inducibility, nuclear localization (HIN) domains antagonizes AIM2 inflammasome signaling and potentially modifies lupus susceptibility. We found that only HIN1 of p202 binds double-stranded DNA (dsDNA), while HIN2 forms a homotetramer. Crystal structures of HIN1 revealed that dsDNA is bound on face opposite the site used in AIM2 and IFI16. The structure of HIN2 revealed a dimer of dimers, the face analogous to the HIN1 dsDNA binding site being a dimerization interface. Electron microscopy imaging showed that HIN1 is flexibly linked to HIN2 in p202, and tetramerization provided enhanced avidity for dsDNA. Surprisingly, HIN2 of p202 interacts with the AIM HIN domain. We propose that this results in a spatial separation of the AIM2 pyrin domains, and indeed p202 prevented the dsDNA-dependent clustering of apoptosis-associated speck-like protein containing caspase recruitment domain (ASC) and AIM2 inflammasome activation. We hypothesize that while p202 was evolutionarily selected to limit AIM2-mediated inflammation in some mouse strains, the same mechanism contributes to increased interferon production and lupus susceptibility.


Subject(s)
Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Amino Acid Sequence , Animals , DNA-Binding Proteins , Enzyme Activation , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/genetics , Protein Structure, Tertiary , Signal Transduction
6.
Immunobiology ; 217(12): 1325-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22898390

ABSTRACT

Macrophage pre-treatment with bacterial lipopolysaccharide (LPS) boosts subsequent activation of the NLRP3 inflammasome, which controls caspase-1-dependent pro-inflammatory cytokine maturation. Previous work has attributed this phenomenon (known as LPS 'priming') to LPS-dependent induction of NLRP3 expression. Whilst this plays a role, here we demonstrate that rapid LPS priming of NLRP3 inflammasome activation can occur independently of NLRP3 induction, since the priming effect of LPS is still apparent at short pre-treatment times in which NLRP3 protein expression remains unchanged. Furthermore, rapid LPS priming is still evident in Nlrp3(-/-) primary macrophages with NLRP3 expression reconstituted using a constitutive promoter. Similarly, we found that LPS potentiates AIM2 inflammasome activation to submaximal doses of cytosolic DNA without concomitant upregulation of AIM2 protein expression. Our data suggest that, in addition to augmenting NLRP3 inflammasome activity via NLRP3 induction, LPS boosts caspase-1 activation by the NLRP3 and AIM2 inflammasomes by an acute mechanism that is independent of inflammasome sensor induction.


Subject(s)
Carrier Proteins/genetics , Inflammasomes/immunology , Lipopolysaccharides/immunology , Macrophages/immunology , Animals , Carrier Proteins/agonists , Carrier Proteins/immunology , Carrier Proteins/metabolism , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , DNA/genetics , DNA-Binding Proteins , Humans , Inflammasomes/genetics , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Promoter Regions, Genetic/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Up-Regulation/genetics , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL