Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Respir Cell Mol Biol ; 66(6): 601-611, 2022 06.
Article in English | MEDLINE | ID: mdl-35290170

ABSTRACT

Silicosis is a multifaceted lung disease, characterized by persistent inflammation and structural remodeling. Despite its poor prognosis, there are no treatments currently available for patients with silicosis. Recent preclinical findings in models of lung fibrosis have suggested a major role for the NLRP3 (nucleotide-binding domain and leucine-rich repeat pyrin domain containing 3) inflammasome in silica-driven inflammation and fibrosis. This review outlines the beneficial effects of targeting the NLRP3 inflammasome in in vitro cell experiments and in in vivo animal models, whereby inflammation and fibrosis are abrogated after NLRP3 inflammasome inhibition. Although preclinical evidence is promising, studies that explore NLRP3 inflammasomes in the clinical setting are warranted. In particular, there is still a need to identify biomarkers that may be helpful for the early detection of silicosis and to fully elucidate mechanisms underlying these beneficial effects to further develop or repurpose existing anti-NLRP3 drugs as novel treatments that limit disease progression.


Subject(s)
Inflammasomes , Silicosis , Animals , Dust , Fibrosis , Humans , Inflammation , NLR Family, Pyrin Domain-Containing 3 Protein , Silicosis/drug therapy
2.
Immunol Cell Biol ; 99(7): 737-748, 2021 08.
Article in English | MEDLINE | ID: mdl-33834544

ABSTRACT

Hyperinflammatory responses including the production of NLRP3-dependent interleukin (IL)-1ß is a characteristic feature of severe and fatal influenza A virus (IAV) infections. The NLRP3 inflammasome has been shown to play a temporal role during severe IAV immune responses, with early protective and later detrimental responses. However, the specific contribution of IL-1ß in modulating IAV disease in vivo is currently not well defined. Here, we identified that activation of NLRP3-dependent IL-1ß responses occurs rapidly following HKx31 H3N2 infection, prior to the onset of severe IAV disease. Mature IL-1ß was detectable in vivo in both hemopoietic and nonhemopoietic cells. Significantly, therapeutic inhibition of IL-1ß in the airways with intranasal anti-IL-1ß antibody treatment from day 3 postinfection, corresponding to the onset of clinical signs of disease, significantly prolonged survival and reduced inflammation in the airways. Importantly, early targeting of IL-1ß from day 1 postinfection also improved survival. Together, these studies specifically define a role for IL-1ß in contributing to the development of hyperinflammation and disease and indicate that targeting IL-1ß is a potential therapeutic strategy for severe IAV infections.


Subject(s)
Influenza A virus , Pneumonia , Humans , Inflammasomes , Influenza A Virus, H3N2 Subtype , Interleukin-1beta , NLR Family, Pyrin Domain-Containing 3 Protein
3.
J Immunol ; 202(8): 2384-2396, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30787108

ABSTRACT

MyD88 adaptor-like (Mal) protein is the most polymorphic of the four key adaptor proteins involved in TLR signaling. TLRs play a critical role in the recognition and immune response to pathogens through activation of the prototypic inflammatory transcription factor NF-κB. The study of single nucleotide polymorphisms in TLRs, adaptors, and signaling mediators has provided key insights into the function of the corresponding genes but also into the susceptibility to infectious diseases in humans. In this study, we have analyzed the immune response of mice carrying the human Mal-D96N genetic variation that has previously been proposed to confer protection against septic shock. We have found that Mal-D96N macrophages display reduced cytokine expression in response to TLR4 and TLR2 ligand challenge. Mal-D96N macrophages also display reduced MAPK activation, NF-κB transactivation, and delayed NF-κB nuclear translocation, presumably via delayed kinetics of Mal interaction with MyD88 following LPS stimulation. Importantly, Mal-D96N genetic variation confers a physiological protective phenotype to in vivo models of LPS-, Escherichia coli-, and influenza A virus-induced hyperinflammatory disease in a gene dosage-dependent manner. Together, these results highlight the critical role Mal plays in regulating optimal TLR-induced inflammatory signaling pathways and suggest the potential therapeutic advantages of targeting the Mal D96 signaling nexus.


Subject(s)
Lipopolysaccharides/toxicity , MAP Kinase Signaling System , Macrophages/immunology , Mutation, Missense , Myeloid Differentiation Factor 88 , Polymorphism, Single Nucleotide , Toll-Like Receptors , Amino Acid Substitution , Animals , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Mice , Mice, Mutant Strains , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
4.
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
5.
Nature ; 501(7466): 247-51, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24025841

ABSTRACT

Successful infection by enteric bacterial pathogens depends on the ability of the bacteria to colonize the gut, replicate in host tissues and disseminate to other hosts. Pathogens such as Salmonella, Shigella and enteropathogenic and enterohaemorrhagic (EPEC and EHEC, respectively) Escherichia coli use a type III secretion system (T3SS) to deliver virulence effector proteins into host cells during infection that promote colonization and interfere with antimicrobial host responses. Here we report that the T3SS effector NleB1 from EPEC binds to host cell death-domain-containing proteins and thereby inhibits death receptor signalling. Protein interaction studies identified FADD, TRADD and RIPK1 as binding partners of NleB1. NleB1 expressed ectopically or injected by the bacterial T3SS prevented Fas ligand or TNF-induced formation of the canonical death-inducing signalling complex (DISC) and proteolytic activation of caspase-8, an essential step in death-receptor-induced apoptosis. This inhibition depended on the N-acetylglucosamine transferase activity of NleB1, which specifically modified Arg 117 in the death domain of FADD. The importance of the death receptor apoptotic pathway to host defence was demonstrated using mice deficient in the FAS signalling pathway, which showed delayed clearance of the EPEC-like mouse pathogen Citrobacter rodentium and reversion to virulence of an nleB mutant. The activity of NleB suggests that EPEC and other attaching and effacing pathogens antagonize death-receptor-induced apoptosis of infected cells, thereby blocking a major antimicrobial host response.


Subject(s)
Enteropathogenic Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Escherichia coli Proteins/metabolism , Gastrointestinal Tract/microbiology , Signal Transduction , Virulence Factors/metabolism , Animals , Caspase 8/metabolism , Cell Death , Citrobacter rodentium/pathogenicity , Citrobacter rodentium/physiology , Enteropathogenic Escherichia coli/pathogenicity , Enzyme Activation , Escherichia coli Infections/pathology , Fas Ligand Protein/antagonists & inhibitors , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/chemistry , Fas-Associated Death Domain Protein/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , N-Acetylglucosaminyltransferases/metabolism , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/metabolism , fas Receptor/deficiency , fas Receptor/metabolism
6.
J Biol Chem ; 292(3): 826-836, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27913620

ABSTRACT

The emergence of avian H7N9 influenza A virus in humans with associated high mortality has highlighted the threat of a potential pandemic. Fatal H7N9 infections are characterized by hyperinflammation and increased cellular infiltrates in the lung. Currently there are limited therapies to address the pathologies associated with H7N9 infection and the virulence factors that contribute to these pathologies. We have found that PB1-F2 derived from H7N9 activates the NLRP3 inflammasome and induces lung inflammation and cellular recruitment that is NLRP3-dependent. We have also shown that H7N9 and A/Puerto Rico/H1N1 (PR8)PB1-F2 peptide treatment induces significant mitochondrial reactive oxygen production, which contributes to NLRP3 activation. Importantly, treatment of cells or mice with the specific NLRP3 inhibitor MCC950 significantly reduces IL-1ß maturation, lung cellular recruitment, and cytokine production. Together, these results suggest that PB1-F2 from H7N9 avian influenza A virus may be a major contributory factor to disease pathophysiology and excessive inflammation characteristic of clinical infections and that targeting the NLRP3 inflammasome may be an effective means to reduce the inflammatory burden associated with H7N9 infections.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Orthomyxoviridae Infections/immunology , Peptides/immunology , Viral Proteins/immunology , Animals , Cell Line, Transformed , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Indenes , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mitochondria/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Reactive Oxygen Species/immunology , Sulfonamides , Sulfones/pharmacology
7.
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
9.
FASEB J ; 31(1): 85-95, 2017 01.
Article in English | MEDLINE | ID: mdl-27682204

ABSTRACT

The NLRP3 inflammasome is a multimeric protein complex that controls the production of IL-1ß, a cytokine that influences the development of both innate and adaptive immune responses. Helminth parasites secrete molecules that interact with innate immune cells, modulating their activity to ultimately determine the phenotype of differentiated T cells, thus creating an immune environment that is conducive to sustaining chronic infection. We show that one of these molecules, FhHDM-1, a cathelicidin-like peptide secreted by the helminth parasite, Fasciola hepatica, inhibits the activation of the NLRP3 inflammasome resulting in reduced secretion of IL-1ß by macrophages. FhHDM-1 had no effect on the synthesis of pro-IL-1ß. Rather, the inhibitory effect was associated with the capacity of the peptide to prevent acidification of the endolysosome. The activation of cathepsin B protease by lysosomal destabilization was prevented in FhHDM-1-treated macrophages. By contrast, peptide derivatives of FhHDM-1 that did not alter the lysosomal pH did not inhibit secretion of IL-1ß. We propose a novel immune modulatory strategy used by F. hepatica, whereby secretion of the FhHDM-1 peptide impairs the activation of NLRP3 by lysosomal cathepsin B protease, which prevents the downstream production of IL-1ß and the development of protective T helper 1 type immune responses that are detrimental to parasite survival.-Alvarado, R., To, J., Lund, M. E., Pinar, A., Mansell, A., Robinson, M. W., O'Brien, B. A., Dalton, J. P., Donnelly, S. The immune modulatory peptide FhHDM-1 secreted by the helminth Fasciola hepatica prevents NLRP3 inflammasome activation by inhibiting endolysosomal acidification in macrophages.


Subject(s)
Fasciola hepatica/metabolism , Helminth Proteins/metabolism , Macrophages/physiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Cathepsin B/genetics , Cathepsin B/metabolism , Cytokines/genetics , Cytokines/metabolism , Fasciola hepatica/genetics , Gene Expression Regulation/physiology , Helminth Proteins/genetics , Hydrogen-Ion Concentration , Lysosomes/metabolism , Macrophages/drug effects , Mice , Mice, Inbred BALB C , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Silicon Dioxide/toxicity
10.
Proc Natl Acad Sci U S A ; 112(5): 1535-40, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25605927

ABSTRACT

Inflammation is critical for host defense, but without appropriate control, it can cause chronic disease or even provoke fatal responses. Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like receptors, we identify that the Broad-complex, Tramtrack and Bric-a-brac/poxvirus and zinc finger (BTB/POZ), transcriptional regulator promyelocytic leukemia zinc finger (PLZF) limits the expression of inflammatory gene products. In accord with this finding, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated inflammatory responses to infectious stimuli. Temporal quantitation of inflammatory gene transcripts shows increased gene induction in the absence of PLZF. Genome-wide analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune homeostasis and limit damaging inflammation. We show that PLZF stabilizes a corepressor complex that encompasses histone deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while curbing damaging inflammation.


Subject(s)
Chromatin/metabolism , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , Signal Transduction , Animals , Bacterial Infections/metabolism , Chromatin Immunoprecipitation , Fluorescence Resonance Energy Transfer , Histone Deacetylases/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Promyelocytic Leukemia Zinc Finger Protein , Real-Time Polymerase Chain Reaction
11.
Infect Immun ; 85(9)2017 09.
Article in English | MEDLINE | ID: mdl-28630066

ABSTRACT

Porphyromonas gulae is an anaerobic, Gram-negative coccobacillus that has been associated with periodontal disease in companion animals. The aims of this study were to analyze the ligation of pattern recognition receptors by P. gulae and the subsequent activation of macrophages. Exposure of HEK cells transfected with Toll-like receptors (TLRs) or NOD-like receptors to P. gulae resulted in the ligation of TLR2, TLR4, and NOD2. The effects of this engagement of receptors were investigated by measuring the synthesis of nitric oxide (NO), CD86 expression, and inflammatory cytokine production by wild-type, TLR2-/-, and TLR4-/- macrophages. The addition of P. gulae to unprimed and gamma interferon (IFN-γ)-primed (M1 phenotype) macrophages significantly increased the surface expression of CD86, but only M1 macrophages produced nitric oxide. P. gulae-induced expression of CD86 on unprimed macrophages was dependent on both TLR2 and TLR4, but CD86 expression and NO production in M1 macrophages were only TLR2 dependent. P. gulae induced an increase in secretion of interleukin-1α (IL-1α), IL-1ß, IL-6, IL-12p70, IL-13, tumor necrosis factor alpha (TNF-α), granulocyte colony-stimulating factor (G-CSF), monocyte chemoattractant protein 1 (MCP-1), and macrophage inflammatory protein 1α (MIP-1α) by M1 macrophages compared to that by unprimed controls. Among these cytokines, secretion of IL-6 and TNF-α by M1 macrophages was dependent on either TLR2 or TLR4. Our data indicate that TLR2 and TLR4 are important for P. gulae activation of unprimed macrophages and that activation and effector functions induced in M1 macrophages by P. gulae are mainly dependent on TLR2. In conclusion, P. gulae induces a strong TLR2-dependent inflammatory M1 macrophage response which may be important in establishing the chronic inflammation associated with periodontal disease in companion animals.


Subject(s)
Macrophage Activation , Macrophages/immunology , Nod2 Signaling Adaptor Protein/metabolism , Porphyromonas/immunology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cells, Cultured , Cytokines/metabolism , Interferon-gamma/metabolism , Macrophages/microbiology , Mice, Inbred C57BL , Nitric Oxide/metabolism , Receptors, Pattern Recognition/metabolism
12.
Pharmacol Res ; 116: 77-86, 2017 02.
Article in English | MEDLINE | ID: mdl-27986554

ABSTRACT

OBJECTIVE: To determine whether a clinically-utilised IL-1 receptor antagonist, anakinra, reduces renal inflammation, structural damage and blood pressure (BP) in mice with established hypertension. METHODS: Hypertension was induced in male mice by uninephrectomy, deoxycorticosterone acetate (2.4mg/d,s.c.) and replacement of drinking water with saline (1K/DOCA/salt). Control mice received uninephrectomy, a placebo pellet and normal drinking water. 10days post-surgery, mice commenced treatment with anakinra (75mg/kg/d, i.p.) or vehicle (0.9% saline, i.p.) for 11days. Systolic BP was measured by tail cuff while qPCR, immunohistochemistry and flow cytometry were used to measure inflammatory markers, collagen and immune cell infiltration in the kidneys. RESULTS: By 10days post-surgery, 1K/DOCA/salt-treated mice displayed elevated systolic BP (148.3±2.4mmHg) compared to control mice (121.7±2.7mmHg; n=18, P<0.0001). The intervention with anakinra reduced BP in 1K/DOCA/salt-treated mice by ∼20mmHg (n=16, P<0.05), but had no effect in controls. In 1K/DOCA/salt-treated mice, anakinra modestly reduced (∼30%) renal expression of some (CCL5, CCL2; n=7-8; P<0.05) but not all (ICAM-1, IL-6) inflammatory markers, and had no effect on immune cell infiltration (n=7-8, P>0.05). Anakinra reduced renal collagen content (n=6, P<0.01) but paradoxically appeared to exacerbate the renal and glomerular hypertrophy (n=8-9, P<0.001) that accompanied 1K/DOCA/salt-induced hypertension. CONCLUSION: Despite its anti-hypertensive and renal anti-fibrotic actions, anakinra had minimal effects on inflammation and leukocyte infiltration in mice with 1K/DOCA/salt-induced hypertension. Future studies will assess whether the anti-hypertensive actions of anakinra are mediated by protective actions in other BP-regulating or salt-handling organs such as the arteries, skin and brain.


Subject(s)
Blood Pressure/drug effects , Fibrosis/drug therapy , Hypertension, Renal/drug therapy , Interleukin 1 Receptor Antagonist Protein/pharmacology , Kidney Diseases/drug therapy , Kidney/drug effects , Animals , Antihypertensive Agents/pharmacology , Biomarkers/metabolism , Desoxycorticosterone Acetate/pharmacology , Fibrosis/metabolism , Hypertension, Renal/chemically induced , Hypertension, Renal/metabolism , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Kidney/metabolism , Kidney Diseases/metabolism , Male , Mice , Mice, Inbred C57BL , Sodium Chloride, Dietary/pharmacology
13.
Proc Natl Acad Sci U S A ; 111(7): 2650-5, 2014 Feb 18.
Article in English | MEDLINE | ID: mdl-24481253

ABSTRACT

IL-37 is a fundamental inhibitor of innate immunity. Human IL-37 has a caspase-1 cleavage site and translocates to the nucleus upon LPS stimulation. Here, we investigated whether caspase-1 processing affects IL-37-mediated suppression of LPS-induced cytokines and the release from cells by analyzing a caspase-1 cleavage site mutant IL-37 (IL-37D20A). Nuclear translocation of IL-37D20A is significantly impaired compared with WT IL-37 in transfected cells. LPS-induced IL-6 was decreased in cells expressing WT IL-37 but not IL-37D20A. The function of IL-37 in transfected bone marrow-derived macrophages is nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-dependent, because IL-37 transfection in apoptosis-associated speck-like protein containing a carboxyl-terminal caspase recruitment domain- and NLRP3-deficient cells does not reduce levels of IL-6 and IL-1ß upon LPS stimulation. IL-37-expressing macrophages release both precursor and mature IL-37, but only the externalization of mature IL-37 was dependent on ATP. Precursor and mature IL-37 was also secreted from human dendritic cells and peripheral blood mononuclear cells. To determine whether IL-37 is active in the extracellular compartment, we pretreated IL-37 transgenic mice with IL-37-neutralizing antibodies before LPS challenge. In IL-37-expressing mice, neutralizing IL-37 antibodies reversed the suppression of LPS-induced serum IL-6. In contrast, the addition of neutralizing antibody did not reverse suppression of LPS-induced IL-6 in mouse macrophages transfected with IL-37. Although caspase-1 is required for nuclear translocation of intracellular IL-37 and for secretion of mature IL-37, the release of the IL-37 precursor is independent of caspase-1 activation. IL-37 now emerges as a dual-function cytokine with intra- and extracellular properties for suppressing innate inflammation.


Subject(s)
Antibodies, Neutralizing/immunology , Caspase 1/metabolism , Cell Nucleus/metabolism , Immunity, Innate/immunology , Interleukin-1/metabolism , Active Transport, Cell Nucleus/immunology , Animals , Blotting, Western , Caspase 1/genetics , Cell Line , Escherichia coli , Fluorescent Antibody Technique , Humans , Interleukin-6/blood , Lipopolysaccharides , Mice , Mice, Transgenic , Microscopy, Confocal , Mutagenesis, Site-Directed
14.
Immunol Cell Biol ; 94(5): 439-46, 2016 05.
Article in English | MEDLINE | ID: mdl-26667784

ABSTRACT

Expression of the viral virulence protein PB1-F2 during infection has been linked to NLRP3 inflammasome complex activation in macrophages and induction of early inflammatory events enhancing immunopathology during influenza disease. We sought to determine whether PB1-F2-specific NLRP3 inflammasome activation influenced the magnitude and/or robustness of the CD8(+) T-cell responses specific for conserved viral antigens and subsequent virus elimination. Using murine heterosubtypic viral infection models, we showed that mice infected with virus unable to produce PB1-F2 protein showed no deficit in the overall magnitude and functional memory responses of CD8(+) T cells established during the effector phase compared with those infected with wild-type PB1-F2-expressing virus and were equally capable of mounting robust recall responses. These data indicate that while expression of PB1-F2 protein can induce inflammatory events, the capacity to generate memory CD8(+) T cells specific for immunodominant viral epitopes remains uncompromised.


Subject(s)
Immunologic Memory , Inflammasomes/metabolism , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , T-Lymphocytes, Cytotoxic/immunology , Viral Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , Female , Inflammation/pathology , Interleukin-1beta/metabolism , Mice, Inbred C57BL , Orthomyxoviridae Infections/pathology , Species Specificity
15.
J Virol ; 89(13): 6575-84, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25855743

ABSTRACT

UNLABELLED: Dendritic cells (DCs) and macrophages are present in the tissues of the anogenital tract, where HIV-1 transmission occurs in almost all cases. These cells are both target cells for HIV-1 and represent the first opportunity for the virus to interfere with innate recognition. Previously we have shown that both cell types fail to produce type I interferons (IFNs) in response to HIV-1 but that, unlike T cells, the virus does not block IFN induction by targeting IFN regulatory factor 3 (IRF3) for cellular degradation. Thus, either HIV-1 inhibits IFN induction by an alternate mechanism or, less likely, these cells fail to sense HIV-1. Here we show that HIV-1 (but not herpes simplex virus 2 [HSV-2] or Sendai virus)-exposed DCs and macrophages fail to induce the expression of all known type I and III IFN genes. These cells do sense the virus, and pattern recognition receptor (PRR)-induced signaling pathways are triggered. The precise stage in the IFN-inducing signaling pathway that HIV-1 targets to block IFN induction was identified; phosphorylation but not K63 polyubiquitination of TANK-binding kinase 1 (TBK1) was completely inhibited. Two HIV-1 accessory proteins, Vpr and Vif, were shown to bind to TBK1, and their individual deletion partly restored IFN-ß expression. Thus, the inhibition of TBK1 autophosphorylation by binding of these proteins appears to be the principal mechanism by which HIV-1 blocks type I and III IFN induction in myeloid cells. IMPORTANCE: Dendritic cells (DCs) and macrophages are key HIV target cells. Therefore, definition of how HIV impairs innate immune responses to initially establish infection is essential to design preventative interventions, especially by restoring initial interferon production. Here we demonstrate how HIV-1 blocks interferon induction by inhibiting the function of a key kinase in the interferon signaling pathway, TBK1, via two different viral accessory proteins. Other viral proteins have been shown to target the general effects of TBK1, but this precise targeting between ubiquitination and phosphorylation of TBK1 is novel.


Subject(s)
Dendritic Cells/immunology , HIV-1/immunology , Host-Pathogen Interactions , Macrophages/immunology , Protein Serine-Threonine Kinases/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , vpr Gene Products, Human Immunodeficiency Virus/metabolism , Cells, Cultured , Dendritic Cells/virology , Humans , Immune Evasion , Interferons/antagonists & inhibitors , Macrophages/virology , Phosphorylation , Protein Processing, Post-Translational , Signal Transduction , Ubiquitination
16.
J Biol Chem ; 289(2): 654-68, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24265315

ABSTRACT

Upon activation of Toll-like receptors (TLRs), cytoplasmic Toll/interleukin-1 receptor (TIR) domains of the receptors undergo homo- or heterodimerization. This in turn leads to the recruitment of adaptor proteins, activation of transcription factors, and the secretion of pro-inflammatory cytokines. Recent studies have described the TIR domain-containing protein from Brucella melitensis, TcpB (BtpA/Btp1), to be involved in virulence and suppression of host innate immune responses. TcpB interferes with TLR4 and TLR2 signaling pathways by a mechanism that remains controversial. In this study, we show using co-immunoprecipitation analyses that TcpB interacts with MAL, MyD88, and TLR4 but interferes only with the MAL-TLR4 interaction. We present the crystal structure of the TcpB TIR domain, which reveals significant structural differences in the loop regions compared with other TIR domain structures. We demonstrate that TcpB forms a dimer in solution, and the crystal structure reveals the dimerization interface, which we validate by mutagenesis and biophysical studies. Our study advances the understanding of the molecular mechanisms of host immunosuppression by bacterial pathogens.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Protein Structure, Tertiary , Toll-Like Receptor 4/metabolism , Virulence Factors/chemistry , Virulence Factors/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Binding Sites/genetics , Brucella melitensis/genetics , Brucella melitensis/metabolism , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Models, Molecular , Molecular Sequence Data , Mutation , Myelin and Lymphocyte-Associated Proteolipid Proteins/genetics , Myelin and Lymphocyte-Associated Proteolipid Proteins/metabolism , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Receptors, Interleukin-1/metabolism , Scattering, Small Angle , Sequence Homology, Amino Acid , Signal Transduction , Toll-Like Receptor 4/genetics , Virulence Factors/genetics , X-Ray Diffraction
17.
J Biol Chem ; 289(10): 6429-6437, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24407287

ABSTRACT

The apoptosis-associated speck-like protein containing a caspase-activating recruitment domain (ASC) is an essential component of several inflammasomes, multiprotein complexes that regulate caspase-1 activation and inflammation. We report here an interaction between promyelocytic leukemia protein (PML) and ASC. We observed enhanced formation of ASC dimers in PML-deficient macrophages. These macrophages also display enhanced levels of ASC in the cytosol. Furthermore, IL-1ß production was markedly enhanced in these macrophages in response to both NLRP3 and AIM2 inflammasome activation and following bone marrow-derived macrophage infection with herpes simplex virus-1 (HSV-1) and Salmonella typhimurium. Collectively, our data indicate that PML limits ASC function, retaining ASC in the nucleus.


Subject(s)
Cytoskeletal Proteins/metabolism , Inflammasomes/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , CARD Signaling Adaptor Proteins , Carrier Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoskeletal Proteins/genetics , Cytosol/metabolism , DNA-Binding Proteins , HEK293 Cells , Humans , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein , Protein Multimerization , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
18.
PLoS Pathog ; 9(5): e1003392, 2013.
Article in English | MEDLINE | ID: mdl-23737748

ABSTRACT

The ability for a host to recognize infection is critical for virus clearance and often begins with induction of inflammation. The PB1-F2 of pathogenic influenza A viruses (IAV) contributes to the pathophysiology of infection, although the mechanism for this is unclear. The NLRP3-inflammasome has been implicated in IAV pathogenesis, but whether IAV virulence proteins can be activators of the complex is unknown. We investigated whether PB1-F2-mediated activation of the NLRP3-inflammasome is a mechanism contributing to overt inflammatory responses to IAV infection. We show PB1-F2 induces secretion of pyrogenic cytokine IL-1ß by activating the NLRP3-inflammasome, contributing to inflammation triggered by pathogenic IAV. Compared to infection with wild-type virus, mice infected with reverse engineered PB1-F2-deficient IAV resulted in decreased IL-1ß secretion and cellular recruitment to the airways. Moreover, mice exposed to PB1-F2 peptide derived from pathogenic IAV had enhanced IL-1ß secretion compared to mice exposed to peptide derived from seasonal IAV. Implicating the NLRP3-inflammasome complex specifically, we show PB1-F2 derived from pathogenic IAV induced IL-1ß secretion was Caspase-1-dependent in human PBMCs and NLRP3-dependent in mice. Importantly, we demonstrate PB1-F2 is incorporated into the phagolysosomal compartment, and upon acidification, induces ASC speck formation. We also show that high molecular weight aggregated PB1-F2, rather than soluble PB1-F2, induces IL-1ß secretion. Furthermore, NLRP3-deficient mice exposed to PB1-F2 peptide or infected with PB1-F2 expressing IAV were unable to efficiently induce the robust inflammatory response as observed in wild-type mice. In addition to viral pore forming toxins, ion channel proteins and RNA, we demonstrate inducers of NLRP3-inflammasome activation may include disordered viral proteins, as exemplified by PB1-F2, acting as host pathogen 'danger' signals. Elucidating immunostimulatory PB1-F2 mediation of NLRP3-inflammasome activation is a major step forward in our understanding of the aetiology of disease attributable to exuberant inflammatory responses to IAV infection.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Influenza A virus/metabolism , Influenza, Human/metabolism , Viral Proteins/immunology , Virulence Factors/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Line, Transformed , Female , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Inflammation/virology , Influenza A virus/genetics , Influenza A virus/immunology , Influenza A virus/pathogenicity , Influenza, Human/genetics , Influenza, Human/immunology , Influenza, Human/physiopathology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-1beta/metabolism , Male , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence Factors/genetics , Virulence Factors/immunology
19.
Am J Pathol ; 184(5): 1411-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24631024

ABSTRACT

In acute kidney injury, which is a significant cause of morbidity and mortality, cytokines and leukocytes promote inflammation and injury. We examined the pathogenic role of IL-17A in cisplatin-induced acute kidney injury. Intrarenal IL-17A mRNA transcription and protein expression were increased in wild-type mice after cisplatin-induced renal injury. An important role for IL-17A in the nephrotoxicity of cisplatin was demonstrated by observing protection from cisplatin-induced functional and histological renal injury in Il17a(-/-) and Rorγt(-/-) mice, as well as in mice treated pre-emptively with anti-IL-17A antibodies. Both renal injury and renal IL-1ß and IL-17A production were attenuated in Asc(-/-) and Tlr2(-/-) mice, suggesting that cisplatin induces endogenous TLR2 ligand production and activates the ASC-dependent inflammasome complex, resulting in IL-1ß and injurious IL-17A production. Neutrophils and natural killer cells are the likely targets of these pathways, because combined depletion of these cells was strongly protective; anti-IL-17A antibodies had no additional effect in this setting. Although IL-17A can also be produced by CD4(+) and γδ T cells, IL-17A from those cells does not contribute to renal injury. Cisplatin-induced injury was unchanged in γδ T-cell-deficient mice, whereas Il17a(-/-) CD4(+) T cells induced similar injury as did wild-type CD4(+) T cells on transfer to cisplatin-injected Rag1(-/-) mice. These studies demonstrate an important role for TLR2, the ASC inflammasome, and IL-17A in innate leukocytes in cisplatin-induced renal injury.


Subject(s)
Acute Kidney Injury/pathology , Inflammasomes/metabolism , Interleukin-17/biosynthesis , Leukocytes/metabolism , Toll-Like Receptor 2/metabolism , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Animals , Antibodies/pharmacology , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Chemokines/genetics , Chemokines/metabolism , Cisplatin/adverse effects , Gene Expression Regulation/drug effects , Inflammasomes/drug effects , Interleukin-17/genetics , Interleukin-17/metabolism , Kidney/metabolism , Kidney/pathology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukocytes/drug effects , Male , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Protein Expr Purif ; 106: 31-40, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25306876

ABSTRACT

TRAM/TICAM-2 is used by Toll-like receptor 4 (TLR4) as a bridging adaptor during the mammalian innate immune response. It recruits TRIF, another TIR domain-containing adaptor protein, to TLR4 via TIR domain interactions, which leads to the activation of transcription factors responsible for the production of type-1 interferon and cytokines. The molecular mechanisms of these dual interactions mediated by the TRAM TIR domain are not clear. To understand the molecular basis of TIR:TIR domain interactions, structural and biochemical studies of TRAM TIR domain are necessary, and require a functional soluble protein. In this paper, we report a successful purification and characterization of full-length TRAM. Because full-length TRAM likely contains unstructured regions that may be disadvantageous for structural studies, we also carried out a systematic construct design to determine the boundaries of the TRAM TIR domain. The truncated TRAM constructs were designed based on secondary structure predictions and screened by small-scale expression. Selected constructs were subjected to biophysical analyses. We show that the expressed TRAM TIR domain is functional using in vitro GST pull-down assays that demonstrate a physical interaction with the TLR4 TIR domain. We further show, by site-directed mutagenesis, that the "BB loop" regions of both the TRAM TIR domain and the TLR4 TIR domain are crucial for this physical interaction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Biochemistry/methods , Interferons/metabolism , Recombinant Proteins/biosynthesis , Signal Transduction , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/isolation & purification , Chromatography, Gel , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , HEK293 Cells , Humans , Protein Binding , Protein Structure, Tertiary , Proton Magnetic Resonance Spectroscopy , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Toll-Like Receptor 4/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL