Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Nat Immunol ; 15(8): 727-37, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24952505

ABSTRACT

Microbes or danger signals trigger inflammasome sensors, which induce polymerization of the adaptor ASC and the assembly of ASC specks. ASC specks recruit and activate caspase-1, which induces maturation of the cytokine interleukin 1ß (IL-1ß) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1ß. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1ß in recipient cells. ASC specks appeared in bodily fluids from inflamed tissues, and autoantibodies to ASC specks developed in patients and mice with autoimmune pathologies. Together these findings reveal extracellular functions of ASC specks and a previously unknown form of cell-to-cell communication.


Subject(s)
Apoptosis/immunology , Caspase 1/immunology , Cytoskeletal Proteins/immunology , Inflammation/immunology , Interleukin-1beta/immunology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Antibodies/immunology , Apoptosis Regulatory Proteins , Autoantibodies/immunology , Autoimmune Diseases/immunology , CARD Signaling Adaptor Proteins , Carrier Proteins/genetics , Caspase 1/genetics , Caspase Inhibitors/pharmacology , Cell Communication/immunology , Cytoskeletal Proteins/genetics , Humans , Inflammasomes/immunology , Lysosomes/pathology , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Phagocytosis/immunology , Prions/chemistry , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology
2.
Nat Immunol ; 11(5): 395-402, 2010 May.
Article in English | MEDLINE | ID: mdl-20351692

ABSTRACT

Inflammasomes regulate the activity of caspase-1 and the maturation of interleukin 1beta (IL-1beta) and IL-18. AIM2 has been shown to bind DNA and engage the caspase-1-activating adaptor protein ASC to form a caspase-1-activating inflammasome. Using Aim2-deficient mice, we identify a central role for AIM2 in regulating caspase-1-dependent maturation of IL-1beta and IL-18, as well as pyroptosis, in response to synthetic double-stranded DNA. AIM2 was essential for inflammasome activation in response to Francisella tularensis, vaccinia virus and mouse cytomegalovirus and had a partial role in the sensing of Listeria monocytogenes. Moreover, production of IL-18 and natural killer cell-dependent production of interferon-gamma, events critical in the early control of virus replication, were dependent on AIM2 during mouse cytomegalovirus infection in vivo. Collectively, our observations demonstrate the importance of AIM2 in the sensing of both bacterial and viral pathogens and in triggering innate immunity.


Subject(s)
DNA Virus Infections/immunology , DNA Viruses/immunology , Francisella tularensis/immunology , Killer Cells, Natural/metabolism , Listeriosis/immunology , Macrophages/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Tularemia/immunology , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Caspase 1/genetics , Caspase 1/immunology , Caspase 1/metabolism , Cell Line , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Cytoskeletal Proteins/genetics , DNA/immunology , DNA Virus Infections/genetics , DNA Virus Infections/metabolism , DNA Viruses/growth & development , DNA Viruses/pathogenicity , DNA-Binding Proteins , Francisella tularensis/pathogenicity , Humans , Immunity, Innate , Killer Cells, Natural/immunology , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Killer Cells, Natural/virology , Listeriosis/genetics , Listeriosis/metabolism , Lymphocyte Activation/genetics , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/immunology , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Transcription Factors/genetics , Transcription Factors/immunology , Transcription Factors/metabolism , Tularemia/genetics , Tularemia/metabolism , Viral Load/genetics , Viral Load/immunology
3.
Nat Immunol ; 9(8): 857-65, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18604209

ABSTRACT

The fibrillar peptide amyloid-beta (A beta) has a chief function in the pathogenesis of Alzheimer's disease. Interleukin 1 beta (IL-1 beta) is a key cytokine in the inflammatory response to A beta. Insoluble materials such as crystals activate the inflammasome formed by the cytoplasmic receptor NALP3, which results in the release of IL-1 beta. Here we identify the NALP3 inflammasome as a sensor of A beta in a process involving the phagocytosis of A beta and subsequent lysosomal damage and release of cathepsin B. Furthermore, the IL-1 beta pathway was essential for the microglial synthesis of proinflammatory and neurotoxic factors, and the inflammasome, caspase-1 and IL-1 beta were critical for the recruitment of microglia to exogenous A beta in the brain. Our findings suggest that activation of the NALP3 inflammasome is important for inflammation and tissue damage in Alzheimer's disease.


Subject(s)
Alzheimer Disease/immunology , Amyloid beta-Peptides/immunology , Immunity, Innate/immunology , Inflammation/metabolism , Carrier Proteins/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation Mediators/physiology , NLR Family, Pyrin Domain-Containing 3 Protein
4.
J Immunol ; 197(6): 2421-33, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27521339

ABSTRACT

A critical component of innate immune response to infection and tissue damage is the NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome, and this pathway and its activation products have been implicated in the pathophysiology of a variety of diseases. NLRP3 inflammasome activation leads to the cleavage of pro-IL-1ß and pro-IL-18, as well as the subsequent release of biologically active IL-1ß, IL-18, and other soluble mediators of inflammation. In this study, we further define the pharmacology of the previously reported NLRP3 inflammasome-selective, IL-1ß processing inhibitor CP-456,773 (also known as MCC950), and we demonstrate its efficacy in two in vivo models of inflammation. Specifically, we show that in human and mouse innate immune cells CP-456,773 is an inhibitor of the cellular release of IL-1ß, IL-1α, and IL-18, that CP-456,773 prevents inflammasome activation induced by disease-relevant soluble and crystalline NLRP3 stimuli, and that CP-456,773 inhibits R848- and imiquimod-induced IL-1ß release. In mice, CP-456,773 demonstrates potent inhibition of the release of proinflammatory cytokines following acute i.p. challenge with LPS plus ATP in a manner that is proportional to the free/unbound concentrations of the drug, thereby establishing an in vivo pharmacokinetic/pharmacodynamic model for CP-456,773. Furthermore, CP-456,773 reduces ear swelling in an imiquimod cream-induced mouse model of skin inflammation, and it reduces airway inflammation in mice following acute challenge with house dust mite extract. These data implicate the NLRP3 inflammasome in the pathogenesis of dermal and airway inflammation, and they highlight the utility of CP-456,773 for interrogating the contribution of the NLRP3 inflammasome and its outputs in preclinical models of inflammation and disease.


Subject(s)
Dermatitis/drug therapy , Heterocyclic Compounds, 4 or More Rings/pharmacology , Inflammasomes/antagonists & inhibitors , Inflammation/physiopathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Pneumonia/drug therapy , Pneumonia/immunology , Sulfones/pharmacology , Animals , Cytokines/antagonists & inhibitors , Cytokines/immunology , Dermatitis/immunology , Dermatitis/physiopathology , Disease Models, Animal , Furans , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Heterocyclic Compounds, 4 or More Rings/therapeutic use , Humans , Immunity, Innate/drug effects , Indenes , Inflammation/drug therapy , Inflammation/immunology , Interleukin-18/antagonists & inhibitors , Interleukin-18/metabolism , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/metabolism , Interleukin-1beta/antagonists & inhibitors , Interleukin-1beta/immunology , Mice , Pneumonia/physiopathology , Signal Transduction , Sulfonamides , Sulfones/administration & dosage , Sulfones/therapeutic use
5.
J Immunol ; 196(4): 1732-40, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773149

ABSTRACT

Neisseria gonorrhoeae, the causative agent of the sexually transmitted infection gonorrhea, has developed resistance to almost every conventional antibiotic. There is an urgent need to develop novel therapies against gonorrhea. Many pathogens, including N. gonorrhoeae, bind the complement inhibitor factor H (FH) to evade complement-dependent killing. Sialylation of gonococcal lipooligosaccharide, as occurs in vivo, augments binding of human FH through its domains 18-20 (FH18-20). We explored the use of fusing FH18-20 with IgG Fc (FH18-20/Fc) to create a novel anti-infective immunotherapeutic. FH18-20 also binds to select host glycosaminoglycans to limit unwanted complement activation on host cells. To identify mutation(s) in FH18-20 that eliminated complement activation on host cells, yet maintained binding to N. gonorrhoeae, we created four mutations in domains 19 or 20 described in atypical hemolytic uremic syndrome that prevented binding of mutated fH to human erythrocytes. One of the mutant proteins (D to G at position 1119 in domain 19; FHD1119G/Fc) facilitated complement-dependent killing of gonococci similar to unmodified FH18-20/Fc but, unlike FH18-20/Fc, did not lyse human erythrocytes. FHD1119G/Fc bound to all (100%) of 15 sialylated clinical N. gonorrhoeae isolates tested (including three contemporary ceftriaxone-resistant strains), mediated complement-dependent killing of 10 of 15 (67%) strains, and enhanced C3 deposition (≥10-fold above baseline levels) on each of the five isolates not directly killed by complement. FHD1119G/Fc facilitated opsonophagocytic killing of a serum-resistant strain by human polymorphonuclear neutrophils. FHD1119G/Fc administered intravaginally significantly reduced the duration and burden of gonococcal infection in the mouse vaginal colonization model. FHD1119G/Fc represents a novel immunotherapeutic against multidrug-resistant N. gonorrhoeae.


Subject(s)
Complement Factor H/immunology , Gonorrhea/immunology , Immunoglobulin Fc Fragments/immunology , Immunotherapy/methods , Recombinant Fusion Proteins/immunology , Animals , Complement Factor H/pharmacology , Disease Models, Animal , Female , Flow Cytometry , Humans , Immunoglobulin Fc Fragments/pharmacology , Mice , Mice, Inbred BALB C , Neisseria gonorrhoeae/immunology , Recombinant Fusion Proteins/pharmacology
6.
Nat Methods ; 10(2): 147-154, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23291722

ABSTRACT

Measurement of protease activity in living cells or organisms remains a challenging task. We here present a transgene-encoded biosensor that reports the proteolytic activity of caspase-1 in the course of inflammasome activation and that of other proteases in a highly sensitive and specific manner. This protease reporter is based on the biological activity of a pro-interleukin (IL)-1ß-Gaussia luciferase (iGLuc) fusion construct, in which pro-IL-1ß-dependent formation of protein aggregates renders GLuc enzyme inactive. Cleavage leads to monomerization of this biosensor protein, resulting in a strong gain in luciferase activity. Exchange of the canonical caspase-1 cleavage site in this reporter construct allows the generation of protease biosensors with additional specificities. The high sensitivity, signal-to-background ratio and specificity of the iGLuc system renders it a useful tool to study proteolytic events in mouse and human cells at high throughput and to monitor protease activity in mice in vivo.


Subject(s)
Biosensing Techniques/methods , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Luciferases/metabolism , Animals , Caspase 1/metabolism , Cell Line , Enzyme Activation , Humans , Mice , Transfection
7.
J Biol Chem ; 289(20): 13701-5, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24692555

ABSTRACT

The inflammatory cytokine IL-1ß is critical for host responses against many human pathogens. Here, we define Group B Streptococcus (GBS)-mediated activation of the Nod-like receptor-P3 (NLRP3) inflammasome in macrophages. NLRP3 activation requires GBS expression of the cytolytic toxin, ß-hemolysin, lysosomal acidification, and leakage. These processes allow the interaction of GBS RNA with cytosolic NLRP3. The present study supports a model in which GBS RNA, along with lysosomal components including cathepsins, leaks out of lysosomes and interacts with NLRP3 to induce IL-1ß production.


Subject(s)
Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Hemolysin Proteins/metabolism , Inflammasomes/metabolism , Interleukin-1beta/biosynthesis , Macrophages/metabolism , RNA, Bacterial/metabolism , Streptococcus agalactiae/physiology , Animals , Humans , Interleukin-1beta/metabolism , Lysosomes/metabolism , Lysosomes/microbiology , Macrophages/cytology , Macrophages/microbiology , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Phagosomes/metabolism , Phagosomes/microbiology , Streptococcus agalactiae/metabolism
8.
Nat Chem Biol ; 9(6): 398-405, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23603659

ABSTRACT

Formation of the inflammasome, a scaffolding complex that activates caspase-1, is important in numerous diseases. Pyroptotic cell death induced by anthrax lethal toxin (LT) is a model for inflammasome-mediated caspase-1 activation. We discovered 7-desacetoxy-6,7-dehydrogedunin (7DG) in a phenotypic screen as a small molecule that protects macrophages from LT-induced death. Using chemical proteomics, we identified protein kinase R (PKR) as the target of 7DG and show that RNAi knockdown of PKR phenocopies treatment with 7DG. Further, we show that PKR's role in ASC assembly and caspase-1 activation induced by several different inflammasome stimuli is independent of PKR's kinase activity, demonstrating that PKR has a previously uncharacterized role in caspase-1 activation and pyroptosis that is distinct from its reported kinase-dependent roles in apoptosis and inflammasome formation in lipopolysaccharide-primed cells. Remarkably, PKR has different roles in two distinct cell death pathways and has a broad role in inflammasome function relevant in other diseases.


Subject(s)
Cell Death , eIF-2 Kinase/chemistry , Animals , Bacillus anthracis/enzymology , Caspase 1/metabolism , Catalytic Domain , Cell Line , Enzyme-Linked Immunosorbent Assay , HSP90 Heat-Shock Proteins/metabolism , Hydrogen-Ion Concentration , Inflammation , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Models, Biological , Peptide Hydrolases/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Conformation
9.
J Immunol ; 190(10): 5216-25, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23585680

ABSTRACT

Vascular disrupting agents such as 5,6-dimethylxanthenone-4-acetic acid (DMXAA) represent a novel approach for cancer treatment. DMXAA has potent antitumor activity in mice and, despite significant preclinical promise, failed human clinical trials. The antitumor activity of DMXAA has been linked to its ability to induce type I IFNs in macrophages, although the molecular mechanisms involved are poorly understood. In this study, we identify stimulator of IFN gene (STING) as a direct receptor for DMXAA leading to TANK-binding kinase 1 and IFN regulatory factor 3 signaling. Remarkably, the ability to sense DMXAA was restricted to murine STING. Human STING failed to bind to or signal in response to DMXAA. Human STING also failed to signal in response to cyclic dinucleotides, conserved bacterial second messengers known to bind and activate murine STING signaling. Collectively, these findings detail an unexpected species-specific role for STING as a receptor for an anticancer drug and uncover important insights that may explain the failure of DMXAA in clinical trials for human cancer.


Subject(s)
Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Membrane Proteins/metabolism , Xanthones/metabolism , Xanthones/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/drug effects , Interferon-beta/metabolism , Leukocytes, Mononuclear/immunology , Macrophages/immunology , Mice , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects
10.
J Biol Chem ; 286(13): 11875-82, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21325272

ABSTRACT

Myeloid differentiation protein 88 (MyD88) is a key signaling adapter in Toll-like receptor (TLR) signaling. MyD88 is also one of the most polymorphic adapter proteins. We screened the reported nonsynonymous coding mutations in MyD88 to identify variants with altered function. In reporter assays, a death domain variant, S34Y, was found to be inactive. Importantly, in reconstituted macrophage-like cell lines derived from knock-out mice, MyD88 S34Y was severely compromised in its ability to respond to all MyD88-dependent TLR ligands. Unlike wild-type MyD88, S34Y is unable to form distinct foci in the cells but is present diffused in the cytoplasm. We observed that IRAK4 co-localizes with MyD88 in these aggregates, and thus these foci appear to be "Myddosomes." The MyD88 S34Y loss-of-function mutant demonstrates how proper cellular localization of MyD88 to the Myddosome is a feature required for MyD88 function.


Subject(s)
Amino Acid Substitution , Cytoplasm/metabolism , Mutation, Missense , Myeloid Differentiation Factor 88/metabolism , Animals , Cytoplasm/genetics , HEK293 Cells , Humans , Mice , Myeloid Differentiation Factor 88/genetics , Protein Transport/physiology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
11.
J Biol Chem ; 286(25): 22235-42, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21531728

ABSTRACT

Human factor H (HufH), a key inhibitor of the alternative pathway of complement, binds to Neisseria gonorrhoeae and constitutes an important mechanism of human-specific complement evasion. The C-terminal domain 20 of HufH contains the binding site for sialylated gonococci. We exploited differences in amino acid sequences between human and non-binding chimpanzee fH domain 20 to create cross-species mutations to define amino acids important for binding to sialylated gonococci. We used fH/Fc fusion constructs that contained contiguous fH domains 18-20 fused to Fc fragments of murine IgG2a. The Fc region was used both as a tag for detection of each fusion molecule on the bacterial surface and as an indicator for complement-dependent killing. Arg-1203 was critical for binding to both porin (Por) B.1A and PorB.1B strains. Modeling of the R1203N human-to-chimpanzee mutation using the crystal structure of HufH19-20 as a template showed a loss of positive charge that protrudes at the C terminus of domain 20. We tested the functional importance of Arg-1203 by incubating sialylated gonococci with normal human serum, in the presence of wild-type HufH18-20/Fc or its R1203A mutant. Gonococci bound and were killed by wild-type HufH18-20/Fc but not by the R1203A mutant. A recombinant fH/Fc molecule that contained chimpanzee domain 20, humanized only at amino acid 1203 (N1203R) also bound to sialylated gonococci and restored killing. These findings provide further insights into the species specificity of gonococcal infections and proof-of-concept of a novel therapeutic approach against gonorrhea, a disease rapidly becoming resistant to conventional antibiotics.


Subject(s)
Complement Factor H/metabolism , N-Acetylneuraminic Acid/metabolism , Neisseria gonorrhoeae/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Arginine , Binding Sites , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/pharmacology , Heparin/pharmacology , Humans , Immunoglobulin Fc Fragments/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Neisseria gonorrhoeae/drug effects , Pan troglodytes , Protein Binding/drug effects , Protein Structure, Tertiary , Species Specificity
12.
J Immunol ; 184(3): 1168-79, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-20042593

ABSTRACT

Human metapneumoviruses (HMPVs) are recently identified Paramyxoviridae that contribute to respiratory tract infections in children. No effective treatments or vaccines are available. Successful defense against virus infection relies on early detection by germ line-encoded pattern recognition receptors and activation of cytokine and type I IFN genes. Recently, the RNA helicase retinoic acid-inducible gene I (RIG-I) has been shown to sense HMPV. In this study, we investigated the abilities of two prototype strains of HMPV (A1 [NL\1\00] and B1 [NL\1\99]) to activate RIG-I and induce type I IFNs. Despite the abilities of both HMPV-A1 and HMPV-B1 to infect and replicate in cell lines and primary cells, only the HMPV-A1 strain triggered RIG-I to induce IFNA/B gene transcription. The failure of the HMPV-B1 strain to elicit type I IFN production was dependent on the B1 phosphoprotein, which specifically prevented RIG-I-mediated sensing of HMPV viral 5' triphosphate RNA. In contrast to most cell types, plasmacytoid dendritic cells displayed a unique ability to sense both HMPV-A1 and HMPV-B1 and in this case sensing was via TLR7 rather than RIG-I. Collectively, these data reveal differential mechanisms of sensing for two closely related viruses, which operate in cell type-specific manners.


Subject(s)
DEAD-box RNA Helicases/metabolism , Metapneumovirus/immunology , Phosphoproteins/metabolism , Toll-Like Receptor 7/metabolism , Viral Interference/immunology , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , DEAD Box Protein 58 , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/physiology , Gene Expression Regulation, Viral/immunology , Humans , Immunity, Innate , Interferon-alpha/biosynthesis , Interferon-alpha/genetics , Interferon-beta/biosynthesis , Interferon-beta/genetics , Ligands , Metapneumovirus/genetics , Metapneumovirus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/metabolism , Paramyxoviridae Infections/virology , Phosphoproteins/genetics , RNA, Viral/genetics , Receptors, Immunologic , Species Specificity , Toll-Like Receptor 7/deficiency , Toll-Like Receptor 7/physiology , Vero Cells
13.
J Biol Chem ; 285(36): 27935-43, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20592019

ABSTRACT

Lipopolysaccharide (LPS) activates the innate immune response through the Toll-like receptor 4 (TLR4).MD-2 complex. A synthetic lipid A precursor, lipid IV(A), induces an innate immune response in mice but not in humans. Both TLR4 and MD-2 are required for the agonist activity of lipid IV(A) in mice, with TLR4 interacting through specific surface charges at the dimerization interface. In this study, we used site-directed mutagenesis to identify the MD-2 residues that determine lipid IV(A) species specificity. A single mutation of murine MD-2 at the hydrophobic pocket entrance, E122K, substantially reduced the response to lipid IV(A). Combining the murine MD-2 E122K with the murine TLR4 K367E/S386K/R434Q mutations completely abolished the response to lipid IV(A), effectively converting the murine cellular response to a human-like response. In human cells, however, simultaneous mutations of K122E, K125L, Y41F, and R69G on human MD-2 were required to promote a response to lipid IV(A). Combining the human MD-2 quadruple mutations with the human TLR4 E369K/Q436R mutations completely converted the human MD-2/human TLR4 receptor to a murine-like receptor. Because MD-2 residues 122 and 125 reside at the dimerization interface near the pocket entrance, surface charge differences here directly affect receptor dimerization. In comparison, residues 42 and 69 reside at the MD-2/TLR4 interaction surface opposite the dimerization interface. Surface charge differences there likely affect the binding angle and/or rigidity between MD-2 and TLR4, exerting an indirect influence on receptor dimerization and activation. Thus, surface charge differences at the two MD-2/TLR4 interfaces determine the species-specific activation of lipid IV(A).


Subject(s)
Glycolipids/metabolism , Lipid A/analogs & derivatives , Lymphocyte Antigen 96/chemistry , Lymphocyte Antigen 96/metabolism , Animals , Arginine , Aspartic Acid , Cell Line , Humans , Hydrophobic and Hydrophilic Interactions , Leucine , Lipid A/metabolism , Lymphocyte Antigen 96/genetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , Mutation , Phenotype , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Species Specificity , Static Electricity , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tyrosine
14.
J Immunol ; 183(2): 787-91, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19570822

ABSTRACT

The IL-1 family cytokines are regulated on transcriptional and posttranscriptional levels. Pattern recognition and cytokine receptors control pro-IL-1beta transcription whereas inflammasomes regulate the proteolytic processing of pro-IL-1beta. The NLRP3 inflammasome, however, assembles in response to extracellular ATP, pore-forming toxins, or crystals only in the presence of proinflammatory stimuli. How the activation of gene transcription by signaling receptors enables NLRP3 activation remains elusive and controversial. In this study, we show that cell priming through multiple signaling receptors induces NLRP3 expression, which we identified to be a critical checkpoint for NLRP3 activation. Signals provided by NF-kappaB activators are necessary but not sufficient for NLRP3 activation, and a second stimulus such as ATP or crystal-induced damage is required for NLRP3 activation.


Subject(s)
Carrier Proteins/genetics , Gene Expression Regulation/immunology , Inflammation/metabolism , NF-kappa B/physiology , Receptors, Cytokine/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Antigen Presentation , Carrier Proteins/metabolism , Cells, Cultured , Humans , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Knockout , Monocytes/cytology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Transcriptional Activation
15.
J Biol Chem ; 284(38): 25742-8, 2009 Sep 18.
Article in English | MEDLINE | ID: mdl-19509286

ABSTRACT

The adapter protein MyD88 adapter-like (Mal), encoded by TIR-domain containing adapter protein (Tirap) (MIM 606252), is the most polymorphic of the five adapter proteins involved in Toll-like receptor signaling, harboring eight non-synonymous single nucleotide polymorphisms in its coding region. We screened reported mutations of Mal for activity in reporter assays to test the hypothesis that variants of Mal existed with altered signaling potential. A TIR domain variant, Mal D96N (rs8177400), was found to be inactive. In reconstituted cell lines, Mal D96N acted as a hypomorphic mutation, with impaired cytokine production and NF-kappaB activation upon lipopolysaccharide or PAM2CSK4 stimulation. Moreover, co-immunoprecipitation studies revealed that Mal D96N is unable to interact with MyD88, a prerequisite for downstream signaling to occur. Computer modeling data suggested that residue 96 resides in the MyD88 binding site, further supporting these findings. Genotyping of Mal D96N in three different cohorts suggested that it is a rare mutation. We, thus, describe a rare variant in Mal that exerts its effect via its inability to bind MyD88.


Subject(s)
Membrane Glycoproteins/metabolism , Myeloid Differentiation Factor 88/metabolism , Receptors, Interleukin-1/metabolism , Signal Transduction/physiology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Amino Acid Substitution , Binding Sites/physiology , Cell Line , Cohort Studies , Computer Simulation , Female , Humans , Lipopeptides/pharmacology , Lipopolysaccharides/pharmacology , Male , Membrane Glycoproteins/genetics , Models, Molecular , Mutation, Missense , Myeloid Differentiation Factor 88/genetics , Polymorphism, Single Nucleotide , Protein Binding/drug effects , Protein Binding/physiology , Protein Structure, Tertiary , Receptors, Interleukin-1/genetics , Signal Transduction/drug effects , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics
16.
J Exp Med ; 198(7): 1043-55, 2003 Oct 06.
Article in English | MEDLINE | ID: mdl-14517278

ABSTRACT

Toll-IL-1-resistance (TIR) domain-containing adaptor-inducing IFN-beta (TRIF)-related adaptor molecule (TRAM) is the fourth TIR domain-containing adaptor protein to be described that participates in Toll receptor signaling. Like TRIF, TRAM activates interferon regulatory factor (IRF)-3, IRF-7, and NF-kappaB-dependent signaling pathways. Toll-like receptor (TLR)3 and 4 activate these pathways to induce IFN-alpha/beta, regulated on activation, normal T cell expressed and secreted (RANTES), and gamma interferon-inducible protein 10 (IP-10) expression independently of the adaptor protein myeloid differentiation factor 88 (MyD88). Dominant negative and siRNA studies performed here demonstrate that TRIF functions downstream of both the TLR3 (dsRNA) and TLR4 (LPS) signaling pathways, whereas the function of TRAM is restricted to the TLR4 pathway. TRAM interacts with TRIF, MyD88 adaptor-like protein (Mal)/TIRAP, and TLR4 but not with TLR3. These studies suggest that TRIF and TRAM both function in LPS-TLR4 signaling to regulate the MyD88-independent pathway during the innate immune response to LPS.


Subject(s)
Adaptor Proteins, Vesicular Transport/physiology , DNA-Binding Proteins/physiology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/physiology , NF-kappa B/physiology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Transcription Factors/physiology , Amino Acid Sequence , Animals , Cell Line , Chemokine CCL5/metabolism , Humans , Interferon Regulatory Factor-3 , Interferon Regulatory Factor-7 , Mice , Mice, Inbred C57BL , Molecular Sequence Data , RNA, Double-Stranded/pharmacology , RNA, Small Interfering/physiology , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Toll-Like Receptors
17.
J Leukoc Biol ; 80(6): 1584-92, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16946018

ABSTRACT

The cell surface receptor complex formed by TLR4 and myeloid differentiation 2 (MD-2) is engaged when cells are exposed to LPS. Recent studies suggested that surface localization of functional mouse TLR4 (mTLR4) depends on the simultaneous expression of MD-2. As we did not observe a similar requirement, we conducted a comparative study of human TLR4 and mTLR4 surface expression in immune cells derived from the MD-2 knockout mouse and LPS-responsive cell lines and in cells that ectopically express TLR4. Our results indicate that in the human and mouse models, neither TLR4 function nor TLR4 surface targeting requires MD-2 coexpression. Accordingly, we report on one human cell line, which constitutively expresses functional TLR4 on the cell surface in the absence of MD-2 expression.


Subject(s)
Gene Expression Regulation , Lymphocyte Antigen 96/biosynthesis , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96/deficiency , Lymphocyte Antigen 96/immunology , Mice , Mice, Knockout , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/immunology
18.
AJP Rep ; 7(4): e211-e214, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29177107

ABSTRACT

Objective Morbidly adherent placentation is associated with increased maternal morbidity and mortality. Recently, there has been mounting evidence supporting the benefits of a standardized multidisciplinary approach at tertiary teaching hospitals. Our objective was to estimate the impact of the implementation of a similar program at a high-volume private community hospital. Study Design In this retrospective cohort study, we evaluated maternal outcomes in all cases of histopathologically confirmed morbidly adherent placentation since the initiation of our multidisciplinary program (2012-2016). Our data were compared with the previously published outcomes of two large cohorts from tertiary teaching hospitals in Utah and Texas. Results In the 28 cases included for evaluation, our group's median estimated blood loss, median packed red blood cells transfused, median anesthesia time, median length of stay, or rates of maternal morbidity did not statistically differ from the published data in Utah or Texas. Conclusion Our data demonstrate the feasibility and utility of a multidisciplinary morbidly adherent placentation program in the private practice/community hospital setting with outcomes similar to those at tertiary teaching hospitals. Implementation of such program may prove beneficial in remote centers, where various factors may prohibit patient travel to a larger center.

19.
Immunobiology ; 211(6-8): 437-47, 2006.
Article in English | MEDLINE | ID: mdl-16920483

ABSTRACT

Toll-like receptors (TLRs) are a small family of type-I glycoproteins that bind to and are activated by conserved non-self molecular signatures carried by microorganisms. Toll-like receptor 4 is triggered by most lipopolysaccharides (LPS). LPS is a complex amphipathic saccharolipidic glycan derived from Gram-negative bacteria. Unique among TLRs, TLR4 activity and interaction with its natural ligand(s) strictly depends on the presence of the extracellular adaptor MD-2. MD-2 is a small secreted glycoprotein that binds with cytokine-like affinities to both the hydrophobic portion of LPS and to the extracellular domain of TLR4. The interaction between MD-2 and LPS induces a triggering event on TLR4, which involves the molecular rearrangement of the receptor complex and its homotypic aggregation. In silico analysis suggests that MD-2 and MD-1 are paralogs derived from a common predecessor at the level of early vertebrates. In this review, we summarize the current state of knowledge concerning MD-2.


Subject(s)
Lymphocyte Antigen 96/physiology , Amino Acid Sequence , Animals , Bacteria/immunology , Humans , Immunity, Innate/physiology , Inflammation/immunology , Lipopolysaccharides/immunology , Lymphocyte Antigen 96/genetics , Molecular Sequence Data , Toll-Like Receptor 4/physiology
20.
Cell Rep ; 16(2): 571-582, 2016 07 12.
Article in English | MEDLINE | ID: mdl-27346360

ABSTRACT

Inflammasome activation is associated with numerous diseases. However, in vivo detection of the activated inflammasome complex has been limited by a dearth of tools. We have developed transgenic mice that ectopically express the fluorescent adaptor protein, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) and characterized the formation of assembled inflammasome complexes ("specks") in primary cells and tissues. In addition to hematopoietic cells, we have found that a stromal population in the lung tissues formed specks during the early phase of influenza infection, whereas myeloid cells showed speck formation after 2 days. In a peritonitis and group B streptococcus infection model, a higher percentage of neutrophils formed specks at early phases of infection, while dendritic cells formed specks at later time points. Furthermore, speck-forming cells underwent pyroptosis and extensive release of specks to the extracellular milieu in vivo. These data underscore the importance of free specks during inflammatory processes in vivo.


Subject(s)
CARD Signaling Adaptor Proteins/genetics , Inflammasomes/metabolism , Animals , CARD Signaling Adaptor Proteins/biosynthesis , Female , Gene Expression , Genes, Reporter , Male , Mice, Inbred C57BL , Mice, Transgenic , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/metabolism , Peritonitis/immunology , Peritonitis/metabolism , Protein Multimerization , Streptococcal Infections/immunology , Streptococcal Infections/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL