Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Immunol Methods ; 447: 1-13, 2017 08.
Article in English | MEDLINE | ID: mdl-28268194

ABSTRACT

Inflammasomes are protein complexes induced by diverse inflammatory stimuli that activate caspase-1, resulting in the processing and release of cytokines, IL-1ß and IL-18, and pyroptosis, an immunogenic form of cell death. To provide a homogeneous method for detecting caspase-1 activity, we developed a bioluminescent, plate-based assay that combines a substrate, Z-WEHD-aminoluciferin, with a thermostable luciferase in an optimized lytic reagent added directly to cultured cells. Assay specificity for caspase-1 is conferred by inclusion of a proteasome inhibitor in the lytic reagent and by use of a caspase-1 inhibitor to confirm activity. This approach enables a specific and rapid determination of caspase-1 activation. Caspase-1 activity is stable in the reagent thereby providing assay convenience and flexibility. Using this assay system, caspase-1 activation has been determined in THP-1 cells following treatment with α-hemolysin, LPS, nigericin, gramicidin, MSU, R848, Pam3CSK4, and flagellin. Caspase-1 activation has also been demonstrated in treated J774A.1 mouse macrophages, bone marrow-derived macrophages (BMDMs) from mice, as well as in human primary monocytes. Caspase-1 activity was not detected in treated BMDMs derived from Casp1-/- mice, further confirming the specificity of the assay. Caspase-1 activity can be measured directly in cultured cells using the lytic reagent, or caspase-1 activity released into medium can be monitored by assay of transferred supernatant. The caspase-1 assay can be multiplexed with other assays to monitor additional parameters from the same cells, such as IL-1ß release or cell death. The caspase-1 assay in combination with a sensitive real-time monitor of cell death allows one to accurately establish pyroptosis. This assay system provides a rapid, convenient, and flexible method to specifically and quantitatively monitor caspase-1 activation in cells in a plate-based format. This will allow a more efficient and effective assessment of inflammasome activation as well as enable high-throughput screening for inflammasome modulators.


Subject(s)
Caspase 1/metabolism , Inflammasomes/metabolism , Luminescent Measurements/methods , Monocytes/metabolism , Animals , Cell Line , Enzyme Activation , Humans , Luciferases/metabolism , Luminescent Measurements/instrumentation , Macrophages/drug effects , Macrophages/metabolism , Mice , Monocytes/enzymology , Pyroptosis , Sensitivity and Specificity
2.
Immunity ; 43(5): 923-32, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-26572062

ABSTRACT

The noncanonical inflammasome induced by intracellular lipopolysaccharide (LPS) leads to caspase-11-dependent pyroptosis, which is critical for induction of endotoxic shock in mice. However, the signaling pathway downstream of caspase-11 is unknown. We found that cytosolic LPS stimulation induced caspase-11-dependent cleavage of the pannexin-1 channel followed up by ATP release, which in turn activated the purinergic P2X7 receptor to mediate cytotoxicity. In the absence of P2X7 or pannexin-1, pyroptosis induced by cytosolic LPS was abrogated. Cleavage of pannexin-1 required the catalytic activity of caspase-11 and was essential for ATP release and P2X7-mediated pyroptosis. Priming the caspase-11 pathway in vivo with LPS or Toll-like receptor-3 (TLR3) agonist resulted in high mortality in wild-type mice after secondary LPS challenge, but not in Casp11(-/-), Panx1(-/-), or P2x7(-/-) mice. These results reveal a critical role for pannexin-1 and P2X7 downstream of caspase-11 for pyroptosis and susceptibility to sepsis induced by the noncanonical inflammasome.


Subject(s)
Caspases/metabolism , Connexins/metabolism , Nerve Tissue Proteins/metabolism , Pyroptosis/physiology , Receptors, Purinergic P2X7/metabolism , Shock, Septic/metabolism , Adenosine Triphosphate/metabolism , Animals , Caspases, Initiator , Cell Line , HEK293 Cells , Humans , Inflammasomes/drug effects , Inflammasomes/metabolism , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Pyroptosis/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Toll-Like Receptor 3/metabolism
3.
Immunity ; 42(4): 744-55, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862092

ABSTRACT

The microbiota stimulates inflammation, but the signaling pathways and the members of the microbiota involved remain poorly understood. We found that the microbiota induces interleukin-1ß (IL-1ß) release upon intestinal injury and that this is mediated via the NLRP3 inflammasome. Enterobacteriaceae and in particular the pathobiont Proteus mirabilis, induced robust IL-1ß release that was comparable to that induced by the pathogen Salmonella. Upon epithelial injury, production of IL-1ß in the intestine was largely mediated by intestinal Ly6C(high) monocytes, required chemokine receptor CCR2 and was abolished by deletion of IL-1ß in CCR2(+) blood monocytes. Furthermore, colonization with P. mirabilis promoted intestinal inflammation upon intestinal injury via the production of hemolysin, which required NLRP3 and IL-1 receptor signaling in vivo. Thus, upon intestinal injury, selective members of the microbiota stimulate newly recruited monocytes to induce NLRP3-dependent IL-1ß release, which promotes inflammation in the intestine.


Subject(s)
Carrier Proteins/immunology , Inflammasomes/immunology , Interleukin-1beta/immunology , Microbiota/immunology , Monocytes/immunology , Symbiosis/immunology , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Carrier Proteins/genetics , Gene Expression Regulation , Hemolysin Proteins/genetics , Hemolysin Proteins/immunology , Inflammasomes/genetics , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/genetics , Intestines/immunology , Intestines/injuries , Intestines/microbiology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/microbiology , Monocytes/pathology , NLR Family, Pyrin Domain-Containing 3 Protein , Proteus Infections/genetics , Proteus Infections/immunology , Proteus Infections/microbiology , Proteus Infections/pathology , Proteus mirabilis/immunology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Salmonella/immunology , Salmonella Infections/genetics , Salmonella Infections/immunology , Salmonella Infections/microbiology , Salmonella Infections/pathology , Signal Transduction
4.
Nat Med ; 21(3): 248-55, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25686105

ABSTRACT

The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome is a component of the inflammatory process, and its aberrant activation is pathogenic in inherited disorders such as cryopyrin-associated periodic syndrome (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer's disease and atherosclerosis. We describe the development of MCC950, a potent, selective, small-molecule inhibitor of NLRP3. MCC950 blocked canonical and noncanonical NLRP3 activation at nanomolar concentrations. MCC950 specifically inhibited activation of NLRP3 but not the AIM2, NLRC4 or NLRP1 inflammasomes. MCC950 reduced interleukin-1ß (IL-1ß) production in vivo and attenuated the severity of experimental autoimmune encephalomyelitis (EAE), a disease model of multiple sclerosis. Furthermore, MCC950 treatment rescued neonatal lethality in a mouse model of CAPS and was active in ex vivo samples from individuals with Muckle-Wells syndrome. MCC950 is thus a potential therapeutic for NLRP3-associated syndromes, including autoinflammatory and autoimmune diseases, and a tool for further study of the NLRP3 inflammasome in human health and disease.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Cryopyrin-Associated Periodic Syndromes/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Inflammasomes/antagonists & inhibitors , Interleukin-1beta/drug effects , Multiple Sclerosis , Sulfones/therapeutic use , Animals , Disease Models, Animal , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Indenes , Inflammation , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Sulfonamides , Sulfones/pharmacology
5.
Immunity ; 41(4): 620-32, 2014 Oct 16.
Article in English | MEDLINE | ID: mdl-25367575

ABSTRACT

Pathobionts play a critical role in disease development, but the immune mechanisms against pathobionts remain poorly understood. Here, we report a critical role for interleukin-22 (IL-22) in systemic protection against bacterial pathobionts that translocate into the circulation after infection with the pathogen Clostridium difficile. Infection with C. difficile induced IL-22, and infected Il22(-/-) mice harbored high numbers of pathobionts in extraintestinal organs despite comparable pathogen load and intestinal damage in mutant and wild-type mice. Pathobionts exhibited increased resistant against complement-mediated phagocytosis, and their intravenous administration resulted in high animal mortality. Selective removal of translocated commensals rescued Il22(-/-) mice, and IL-22 administration enhanced the elimination of pathobionts. Mechanistically, IL-22 augmented bacterial phagocytosis by increasing the expression and bacterial binding of complement C3. Our study demonstrates an unexpected role for IL-22 in controlling the elimination of pathobionts that enter the systemic circulation through the regulation of the complement system.


Subject(s)
Clostridioides difficile/immunology , Complement C3/immunology , Enterocolitis, Pseudomembranous/immunology , Interleukins/immunology , Intestines/microbiology , Animals , Complement C3/biosynthesis , Elapid Venoms/pharmacology , Enterobacteriaceae/growth & development , Enterocolitis, Pseudomembranous/mortality , Interleukins/genetics , Intestines/injuries , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota/immunology , Phagocytosis/immunology , Interleukin-22
6.
J Immunol ; 193(8): 4214-4222, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25225670

ABSTRACT

The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (Nlrp3) inflammasome plays an important role in inflammation by controlling the maturation and secretion of the cytokines IL-1ß and IL-18 in response to multiple stimuli including pore-forming toxins, particulate matter, and ATP. Although the pathways activated by the latter stimuli lead to a decrease in intracellular K(+) concentration, which is required for inflammasome activation, the mechanism by which microbial RNA activates Nlrp3, remains poorly understood. In this study, we found that cytosolic poly(I:C), but not total RNA from healthy macrophages, macrophages undergoing pyroptosis, or mitochondrial RNA, induces caspase-1 activation and IL-1ß release through the Nlrp3 inflammasome. Experiments with macrophages deficient in Tlr3, Myd88, or Trif, indicate that poly(I:C) induces Nlrp3 activation independently of TLR signaling. Further analyses revealed that the cytosolic sensors Rig-I and melanoma differentiation-associated gene 5 act redundantly via the common adaptor mitochondrial antiviral signaling (Mavs) to induce Nlrp3 activation in response to poly(I:C), but not ATP or nigericin. Mechanistically, Mavs triggered membrane permeabilization and K(+) efflux independently of the inflammasome which were required for poly(I:C)-induced Nlrp3 activation. We conclude that poly (I:C) activates the inflammasome through an Mavs-dependent surveillance pathway that converges into a common K(+) lowering step in the cytosol that is essential for the induction of Nlrp3 activation.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Carrier Proteins/immunology , Potassium/metabolism , RNA, Double-Stranded/immunology , Adaptor Proteins, Vesicular Transport/genetics , Animals , Caspase 1/immunology , Cytosol , DEAD Box Protein 58 , DEAD-box RNA Helicases/immunology , Inflammation/immunology , Interferon-Induced Helicase, IFIH1 , Interleukin-18/biosynthesis , Interleukin-18/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Ion Transport , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , NLR Family, Pyrin Domain-Containing 3 Protein , Poly I-C/immunology , RNA, Bacterial/immunology , RNA, Viral/immunology , Signal Transduction/immunology , Toll-Like Receptor 3/genetics
7.
Mol Immunol ; 60(1): 62-71, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24769493

ABSTRACT

Staphylococcus aureus is responsible for a large and diverse burden of human disease associated with significant morbidity and mortality. The dynamic challenge of this pathogen is exemplified by the emergence of highly virulent community-associated methicillin-resistant S. aureus strain USA300, which threatens both healthy and vulnerable individuals and constitutes a public health imperative in the United States. Though S. aureus employs many virulence factors that enable infectivity and evasion of host defenses, evidence suggests that the increased production of alpha hemolysin may be a critical contributor to the increased virulence of USA300. To enable and inform immunological targeting of alpha hemolysin, we sought to precisely map a neutralizing epitope that we hypothesized existed in the N-terminal domain. Using an in vivo mapping strategy employing peptide immunogens and an optimized in vitro toxin neutralization assay, we identified a linear neutralizing determinant in the N-terminal 19 amino acids of alpha hemolysin. Affinity purified rabbit antibody against this neutralizing epitope was shown to be highly effective at mitigating dermonecrosis in inbred and outbred mice challenged with USA300. To our knowledge, this is the first description of a linear neutralizing epitope in alpha hemolysin, and the delineation of this determinant should inform and facilitate the rational design and development of an efficacious, epitope-focused or multivalent vaccine against S. aureus.


Subject(s)
Antibodies, Neutralizing/immunology , Bacterial Toxins/immunology , Epitopes/immunology , Hemolysin Proteins/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Amino Acid Sequence , Animals , Epitopes/genetics , Female , Mice , Mice, Inbred C57BL , Rabbits , Staphylococcal Infections/immunology , Staphylococcal Vaccines/immunology , Staphylococcus aureus/pathogenicity , Virulence Factors/immunology
8.
PLoS Pathog ; 10(2): e1003926, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516390

ABSTRACT

Recognition of intracellular pathogenic bacteria by members of the nucleotide-binding domain and leucine-rich repeat containing (NLR) family triggers immune responses against bacterial infection. A major response induced by several Gram-negative bacteria is the activation of caspase-1 via the Nlrc4 inflammasome. Upon activation, caspase-1 regulates the processing of proIL-1ß and proIL-18 leading to the release of mature IL-1ß and IL-18, and induction of pyroptosis. The activation of the Nlrc4 inflammasome requires the presence of an intact type III or IV secretion system that mediates the translocation of small amounts of flagellin or PrgJ-like rod proteins into the host cytosol to induce Nlrc4 activation. Using the Salmonella system, it was shown that Naip2 and Naip5 link flagellin and the rod protein PrgJ, respectively, to Nlrc4. Furthermore, phosphorylation of Nlrc4 at Ser533 by Pkcδ was found to be critical for the activation of the Nlrc4 inflammasome. Here, we show that Naip2 recognizes the Shigella T3SS inner rod protein MxiI and induces Nlrc4 inflammasome activation. The expression of MxiI in primary macrophages was sufficient to induce pyroptosis and IL-1ß release, which were prevented in macrophages deficient in Nlrc4. In the presence of MxiI or Shigella infection, MxiI associated with Naip2, and Naip2 interacted with Nlrc4. siRNA-mediated knockdown of Naip2, but not Naip5, inhibited Shigella-induced caspase-1 activation, IL-1ß maturation and Asc pyroptosome formation. Notably, the Pkcδ kinase was dispensable for caspase-1 activation and secretion of IL-1ß induced by Shigella or Salmonella infection. These results indicate that activation of caspase-1 by Shigella is triggered by the rod protein MxiI that interacts with Naip2 to induce activation of the Nlrc4 inflammasome independently of the Pkcδ kinase.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bacterial Proteins/metabolism , Calcium-Binding Proteins/metabolism , Host-Parasite Interactions/immunology , Inflammasomes/metabolism , Neuronal Apoptosis-Inhibitory Protein/metabolism , Protein Kinase C-delta/metabolism , Animals , Caspase 1/metabolism , Immunoprecipitation , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Transfection
9.
J Biol Chem ; 289(2): 1142-50, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24265316

ABSTRACT

The NLRP3 inflammasome is a critical component of the innate immune system. NLRP3 activation is induced by diverse stimuli associated with bacterial infection or tissue damage, but its inappropriate activation is involved in the pathogenesis of inherited and acquired inflammatory diseases. However, the mechanism by which NLRP3 is activated remains poorly understood. In this study, we explored the role of kinases in NLRP3 inflammasome activation by screening a kinase inhibitor library and identified 3,4-methylenedioxy-ß-nitrostyrene (MNS) as an inhibitor for NLRP3 inflammasome activation. Notably, MNS did not affect the activation of the NLRC4 or AIM2 (absent in melanoma 2) inflammasome. Mechanistically, MNS specifically prevented NLRP3-mediated ASC speck formation and oligomerization without blocking potassium efflux induced by NLRP3 agonists. Surprisingly, Syk kinase, the reported target of MNS, did not mediate the inhibitory activity of MNS on NLRP3 inflammasome activation. We also found that the nitrovinyl group of MNS is essential for the inhibitory activity of MNS. Immunoprecipitation, mass spectrometry, and mutation studies suggest that both the nucleotide binding oligomerization domain and the leucine-rich repeat domain of NLRP3 were the intracellular targets of MNS. Administration of MNS also inhibited NLRP3 ATPase activity in vitro, suggesting that MNS blocks the NLRP3 inflammasome by directly targeting NLRP3 or NLRP3-associated complexes. These studies identified a novel chemical probe for studying the molecular mechanism of NLRP3 inflammasome activation which may advance the development of novel strategies to treat diseases associated with abnormal activation of NLRP3 inflammasome.


Subject(s)
Carrier Proteins/metabolism , Dioxolanes/pharmacology , Inflammasomes/metabolism , Macrophages/drug effects , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Blotting, Western , Carrier Proteins/genetics , Cells, Cultured , Dioxolanes/chemistry , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Inflammasomes/genetics , Interleukin-1beta/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Microscopy, Fluorescence , Molecular Structure , NLR Family, Pyrin Domain-Containing 3 Protein , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Syk Kinase , Tumor Necrosis Factor-alpha/metabolism
10.
Nature ; 503(7476): 397-401, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-24172897

ABSTRACT

Atopic dermatitis is a chronic inflammatory skin disease that affects 15-30% of children and approximately 5% of adults in industrialized countries. Although the pathogenesis of atopic dermatitis is not fully understood, the disease is mediated by an abnormal immunoglobulin-E immune response in the setting of skin barrier dysfunction. Mast cells contribute to immunoglobulin-E-mediated allergic disorders including atopic dermatitis. Upon activation, mast cells release their membrane-bound cytosolic granules leading to the release of several molecules that are important in the pathogenesis of atopic dermatitis and host defence. More than 90% of patients with atopic dermatitis are colonized with Staphylococcus aureus in the lesional skin whereas most healthy individuals do not harbour the pathogen. Several staphylococcal exotoxins can act as superantigens and/or antigens in models of atopic dermatitis. However, the role of these staphylococcal exotoxins in disease pathogenesis remains unclear. Here we report that culture supernatants of S. aureus contain potent mast-cell degranulation activity. Biochemical analysis identified δ-toxin as the mast cell degranulation-inducing factor produced by S. aureus. Mast cell degranulation induced by δ-toxin depended on phosphoinositide 3-kinase and calcium (Ca(2+)) influx; however, unlike that mediated by immunoglobulin-E crosslinking, it did not require the spleen tyrosine kinase. In addition, immunoglobulin-E enhanced δ-toxin-induced mast cell degranulation in the absence of antigen. Furthermore, S. aureus isolates recovered from patients with atopic dermatitis produced large amounts of δ-toxin. Skin colonization with S. aureus, but not a mutant deficient in δ-toxin, promoted immunoglobulin-E and interleukin-4 production, as well as inflammatory skin disease. Furthermore, enhancement of immunoglobulin-E production and dermatitis by δ-toxin was abrogated in Kit(W-sh/W-sh) mast-cell-deficient mice and restored by mast cell reconstitution. These studies identify δ-toxin as a potent inducer of mast cell degranulation and suggest a mechanistic link between S. aureus colonization and allergic skin disease.


Subject(s)
Bacterial Toxins/metabolism , Cell Degranulation , Dermatitis, Atopic/microbiology , Mast Cells/cytology , Staphylococcus aureus/pathogenicity , Animals , Bacterial Toxins/pharmacology , Calcium Signaling/drug effects , Cell Degranulation/drug effects , Culture Media, Conditioned/pharmacology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/pathology , Female , Immunoglobulin E/biosynthesis , Immunoglobulin E/immunology , Inflammation/immunology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interleukin-4/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mast Cells/drug effects , Mice , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Staphylococcus aureus/metabolism , Syk Kinase
11.
Immunity ; 38(6): 1142-53, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23809161

ABSTRACT

The NLRP3 inflammasome is an important component of the innate immune system. However, its mechanism of activation remains largely unknown. We show that NLRP3 activators including bacterial pore-forming toxins, nigericin, ATP, and particulate matter caused mitochondrial perturbation or the opening of a large membrane pore, but this was not required for NLRP3 activation. Furthermore, reactive oxygen species generation or a change in cell volume was not necessary for NLRP3 activation. Instead, the only common activity induced by all NLRP3 agonists was the permeation of the cell membrane to K⁺ and Na⁺. Notably, reduction of the intracellular K⁺ concentration was sufficient to activate NLRP3, whereas an increase in intracellular Na⁺ modulated but was not strictly required for inflammasome activation. These results provide a unifying model for the activation of the NLRP3 inflammasome in which a drop in cytosolic K⁺ is the common step that is necessary and sufficient for caspase-1 activation.


Subject(s)
Carrier Proteins/metabolism , Inflammasomes/metabolism , Macrophages/immunology , Mitochondria/metabolism , Adenosine Triphosphate/pharmacology , Animals , Carrier Proteins/drug effects , Carrier Proteins/genetics , Caspase 1/metabolism , Cell Membrane Permeability/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Immunity, Innate , Inflammasomes/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein , Nigericin/pharmacology , Particulate Matter/pharmacology , Potassium/metabolism , Potassium Channels/drug effects , Potassium Channels/metabolism , Reactive Oxygen Species/metabolism , Sodium Channels/drug effects , Sodium Channels/metabolism
12.
J Immunol ; 189(1): 328-36, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22634614

ABSTRACT

Recognition of foreign nucleic acids is important for the induction of an innate immune response against invading pathogens. Although the pathways involved in sensing bacterial DNA and viral RNA are now well established, only limited knowledge is available on mechanisms underlying recognition of bacterial RNA. It has been reported that intracellular delivery of Escherichia coli RNA activates the Nlrp3 inflammasome, but whether this is a general property of bacterial RNA remains unclear as are the pathways involved in pro-IL-1ß induction and caspase-1 activation by bacterial RNA. In this study, we report that bacterial RNA from both Gram-positive and Gram-negative bacteria induces activation of caspase-1 and secretion of IL-1ß by murine dendritic cells and bone-marrow derived macrophages. Stimulation was independent of the presence of 5'-triphosphate termini and occurred with whole RNA preparations from bacteria but not from eukaryotes. Induction of pro-IL-1ß as well as the priming for caspase-1 activation by bacterial RNA was dependent on UNC93B, an endoplasmic reticulum protein essential for delivery of TLRs to the endosome, whereas the established nucleic acid sensing endosomal TLRs 3, 7, and 9 were dispensable. Additionally, caspase-1 activation and IL-1ß production by transfected bacterial RNA were absent in MyD88-deficient cells but independent of TRIF. Thus, our data indicate the presence of a yet unidentified intracellular nucleic acid receptor involved in bacterial RNA-induced inflammasome activation and release of IL-1ß.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , Caspase 1/metabolism , Interleukin-1beta/metabolism , Membrane Glycoproteins/physiology , Membrane Transport Proteins/physiology , RNA, Bacterial/physiology , Toll-Like Receptor 3/physiology , Toll-Like Receptor 7/physiology , Toll-Like Receptor 9/physiology , Adaptor Proteins, Vesicular Transport/deficiency , Animals , Cell Line , Dendritic Cells/enzymology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Enzyme Activation/genetics , Macrophages/enzymology , Macrophages/metabolism , Macrophages/microbiology , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 9/deficiency
13.
Nat Immunol ; 13(4): 325-32, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22430785

ABSTRACT

Inflammasomes are multiprotein complexes that activate caspase-1, which leads to maturation of the proinflammatory cytokines interleukin 1ß (IL-1ß) and IL-18 and the induction of pyroptosis. Members of the Nod-like receptor (NLR) family, including NLRP1, NLRP3 and NLRC4, and the cytosolic receptor AIM2 are critical components of inflammasomes and link microbial and endogenous danger signals to the activation of caspase-1. In response to microbial infection, activation of the inflammasomes contributes to host protection by inducing immune responses that limit microbial invasion, but deregulated activation of inflammasomes is associated with autoinflammatory syndromes and other pathologies. Thus, understanding inflammasome pathways may provide insight into the mechanisms of host defense against microbes and the development of inflammatory disorders.


Subject(s)
Immunity, Innate/immunology , Inflammasomes/immunology , Signal Transduction/immunology , Animals , Humans
14.
Eur J Immunol ; 40(3): 611-5, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20201013

ABSTRACT

Members of the Nod-like receptor family and the adaptor ASC assemble into multiprotein platforms, termed inflammasomes, to mediate the activation of caspase-1 and subsequent secretion of IL-1beta and IL-18. Recent studies have identified microbial and endogenous molecules as well as possible mechanisms involved in inflammasome activation.


Subject(s)
Bacterial Infections/immunology , Inflammation/microbiology , Multiprotein Complexes/immunology , Mycoses/immunology , Virus Diseases/immunology , Animals , Apoptosis/immunology , CARD Signaling Adaptor Proteins , Cytoskeletal Proteins/immunology , Humans , Inflammation/immunology , Interleukin-1beta/immunology , Nod Signaling Adaptor Proteins/immunology , Signal Transduction/immunology
15.
J Immunol ; 183(9): 5823-9, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812205

ABSTRACT

Macrophages play a crucial role in the innate immune response against the human pathogen Streptococcus pyogenes, yet the innate immune response against the bacterium is poorly characterized. In the present study, we show that caspase-1 activation and IL-1beta secretion were induced by live, but not killed, S. pyogenes, and required expression of the pore-forming toxin streptolysin O. Using macrophages deficient in inflammasome components, we found that both NLR family pyrin domain-containing 3 (Nlrp3) and apoptosis-associated speck-like protein (Asc) were crucial for caspase-1 activation and IL-1beta secretion, but dispensable for pro-IL-1beta induction, in response to S. pyogenes infection. Conversely, macrophages deficient in the essential TLR adaptors Myd88 and Trif showed normal activation of caspase-1, but impaired induction of pro-IL-1beta and secretion of IL-1beta. Notably, activation of caspase-1 by TLR2 and TLR4 ligands in the presence of streptolysin O required Myd88/Trif, whereas that induced by S. pyogenes was blocked by inhibition of NF-kappaB. Unlike activation of the Nlrp3 inflammasome by TLR ligands, the induction of caspase-1 activation by S. pyogenes did not require exogenous ATP or the P2X7R. In vivo experiments revealed that Nlrp3 was critical for the production of IL-1beta but was not important for survival in a mouse model of S. pyogenes peritoneal infection. These results indicate that caspase-1 activation in response to S. pyogenes infection requires NF-kappaB and the virulence factor streptolysin O, but proceeds independently of P2X7R and TLR signaling.


Subject(s)
Carrier Proteins/metabolism , Inflammation Mediators/physiology , NF-kappa B/metabolism , Receptors, Purinergic P2/physiology , Signal Transduction/immunology , Streptococcus pyogenes/immunology , Streptolysins/physiology , Toll-Like Receptors/physiology , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/physiology , Carrier Proteins/genetics , Carrier Proteins/physiology , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Inflammation Mediators/metabolism , Interleukin-1beta/biosynthesis , Interleukin-1beta/metabolism , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , NF-kappa B/physiology , NLR Family, Pyrin Domain-Containing 3 Protein , Peritonitis/immunology , Peritonitis/microbiology , Peritonitis/mortality , Protein Precursors/biosynthesis , Protein Precursors/metabolism , Receptors, Purinergic P2/deficiency , Receptors, Purinergic P2/genetics , Receptors, Purinergic P2X7 , Signal Transduction/genetics , Streptococcal Infections/immunology , Streptococcal Infections/metabolism , Streptococcal Infections/mortality , Streptococcus pyogenes/genetics , Streptolysins/deficiency , Streptolysins/metabolism , Survival Analysis
16.
J Immunol ; 183(6): 3942-8, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19717510

ABSTRACT

The mechanism by which bacterial pathogens activate caspase-1 via Nlrp3 remains poorly understood. In this study, we show that the ability of Staphylococcus aureus, a leading cause of infection in humans, to activate caspase-1 and induce IL-1beta secretion resides in culture supernatants of growing bacteria. Caspase-1 activation induced by S. aureus required alpha-, beta-, and gamma-hemolysins and the host Nlrp3 inflammasome. Mechanistically, alpha- and beta-hemolysins alone did not trigger caspase-1 activation, but they did so in the presence of bacterial lipoproteins released by S. aureus. Notably, caspase-1 activation induced by S. aureus supernatant was independent of the P2X7 receptor and the essential TLR adaptors MyD88 and TIR domain-containing adapter-inducing IFN-beta, but was inhibited by extracellular K(+). These results indicate that S. aureus hemolysins circumvent the requirement of ATP and the P2X7 receptor to induce caspase-1 activation via Nlrp3. Furthermore, these studies revealed that hemolysins promote in the presence of lipoproteins the activation of the Nlrp3 inflammasome.


Subject(s)
Carrier Proteins/metabolism , Hemolysin Proteins/physiology , Lipoproteins/physiology , Staphylococcus aureus/immunology , Adenosine Triphosphate , Animals , Bacterial Proteins , Caspase 1/metabolism , Inflammation/etiology , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Potassium/pharmacology , Receptors, Purinergic P2 , Receptors, Purinergic P2X7 , Staphylococcal Infections
17.
Nat Immunol ; 10(3): 241-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19221555

ABSTRACT

The inflammasome is a multiprotein complex that mediates the activation of caspase-1, which promotes secretion of the proinflammatory cytokines interleukin 1beta (IL-1beta) and IL-18, as well as 'pyroptosis', a form of cell death induced by bacterial pathogens. Members of the Nod-like receptor family, including NLRP1, NLRP3 and NLRC4, and the adaptor ASC are critical components of the inflammasome that link microbial and endogenous 'danger' signals to caspase-1 activation. Several diseases are associated with dysregulated activation of caspase-1 and secretion of IL-1beta. Thus, understanding inflammasome pathways may provide insight into disease pathogenesis that might identify potential targets for therapeutic intervention.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Apoptosis Regulatory Proteins/immunology , CARD Signaling Adaptor Proteins/immunology , Calcium-Binding Proteins/immunology , Carrier Proteins/immunology , Caspase 1/immunology , Interleukin-1beta/immunology , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/metabolism , Carrier Proteins/metabolism , Caspase 1/metabolism , Humans , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , NLR Proteins , Protein Interaction Domains and Motifs , Uric Acid/immunology , Uric Acid/metabolism
18.
J Immunol ; 178(12): 8022-7, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17548639

ABSTRACT

Similar to Ipaf and caspase-1, the Nod-like receptor protein Naip5 restricts intracellular proliferation of Legionella pneumophila, the causative agent of a severe form of pneumonia known as Legionnaires' disease. Thus, Naip5 has been suggested to regulate Legionella replication inside macrophages through the activation of caspase-1. In this study, we show that cytosolic delivery of recombinant flagellin activated caspase-1 in A/J macrophages carrying a mutant Naip5 allele, and in C57BL/6 (B6) macrophages congenic for the mutant Naip5 allele (B6-Naip5(A/J)), but not in Ipaf(-/-) cells. In line with these results, A/J and B6-Naip5(A/J) macrophages induced high levels of caspase-1 activation and IL-1beta secretion when infected with Legionella. In addition, transgenic expression of a functional Naip5 allele in A/J macrophages did not alter Legionella-induced caspase-1 activation and IL-1beta secretion. Notably, defective Naip5 signaling renders B6-Naip5(A/J) macrophages permissive for Legionella proliferation despite normal caspase-1 activation. These results indicate that the restriction of intracellular Legionella replication is more complex than previously appreciated and requires both Ipaf-dependent caspase-1 activation as well as functional Naip5 signaling.


Subject(s)
Caspase 1/metabolism , Legionella pneumophila/growth & development , Legionnaires' Disease/immunology , Macrophages/immunology , Neuronal Apoptosis-Inhibitory Protein/physiology , Alleles , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Enzyme Activation , Flagellin/pharmacology , Interleukin-1beta/metabolism , Legionnaires' Disease/microbiology , Macrophages/drug effects , Macrophages/microbiology , Mice , Mice, Congenic , Neuronal Apoptosis-Inhibitory Protein/genetics , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL
...