39 results on '"Monks BG"'
Search Results
2. Stable serum resistance of Neisseria gonorrhoeaeis mediated by binding of C4B-binding protein to gonococcal porin
- Author
-
Ram, S, Cullinane, M, Gulati, S, Blom, A, Dahlback, B, Elkins, C, Pangburn, MK, McQuillen, DP, Monks, BG, and Rice, PA
- Published
- 2000
- Full Text
- View/download PDF
3. Efficacy and Pharmacology of the NLRP3 Inflammasome Inhibitor CP-456,773 (CRID3) in Murine Models of Dermal and Pulmonary Inflammation.
- Author
-
Primiano MJ, Lefker BA, Bowman MR, Bree AG, Hubeau C, Bonin PD, Mangan M, Dower K, Monks BG, Cushing L, Wang S, Guzova J, Jiao A, Lin LL, Latz E, Hepworth D, and Hall JP
- Subjects
- 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, 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
- 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-1β and pro-IL-18, as well as the subsequent release of biologically active IL-1β, IL-18, and other soluble mediators of inflammation. In this study, we further define the pharmacology of the previously reported NLRP3 inflammasome-selective, IL-1β 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-1β, 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-1β 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., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
4. A Novel Factor H-Fc Chimeric Immunotherapeutic Molecule against Neisseria gonorrhoeae.
- Author
-
Shaughnessy J, Gulati S, Agarwal S, Unemo M, Ohnishi M, Su XH, Monks BG, Visintin A, Madico G, Lewis LA, Golenbock DT, Reed GW, Rice PA, and Ram S
- Subjects
- 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, Complement Factor H immunology, Gonorrhea immunology, Immunoglobulin Fc Fragments immunology, Immunotherapy methods, Recombinant Fusion Proteins immunology
- 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., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
5. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases.
- Author
-
Coll RC, Robertson AA, Chae JJ, Higgins SC, Muñoz-Planillo R, Inserra MC, Vetter I, Dungan LS, Monks BG, Stutz A, Croker DE, Butler MS, Haneklaus M, Sutton CE, Núñez G, Latz E, Kastner DL, Mills KH, Masters SL, Schroder K, Cooper MA, and O'Neill LA
- Subjects
- 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, 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
- 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-1β (IL-1β) 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.
- Published
- 2015
- Full Text
- View/download PDF
6. The adaptor ASC has extracellular and 'prionoid' activities that propagate inflammation.
- Author
-
Franklin BS, Bossaller L, De Nardo D, Ratter JM, Stutz A, Engels G, Brenker C, Nordhoff M, Mirandola SR, Al-Amoudi A, Mangan MS, Zimmer S, Monks BG, Fricke M, Schmidt RE, Espevik T, Jones B, Jarnicki AG, Hansbro PM, Busto P, Marshak-Rothstein A, Hornemann S, Aguzzi A, Kastenmüller W, and Latz E
- Subjects
- 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, Apoptosis immunology, Caspase 1 immunology, Cytoskeletal Proteins immunology, Inflammation immunology, Interleukin-1beta immunology
- 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 1β (IL-1β) and pyroptotic cell death. Here we found that after pyroptosis, ASC specks accumulated in the extracellular space, where they promoted further maturation of IL-1β. In addition, phagocytosis of ASC specks by macrophages induced lysosomal damage and nucleation of soluble ASC, as well as activation of IL-1β 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.
- Published
- 2014
- Full Text
- View/download PDF
7. Chemical genetics reveals a kinase-independent role for protein kinase R in pyroptosis.
- Author
-
Hett EC, Slater LH, Mark KG, Kawate T, Monks BG, Stutz A, Latz E, and Hung DT
- Subjects
- 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, Cell Death, eIF-2 Kinase chemistry
- 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.
- Published
- 2013
- Full Text
- View/download PDF
8. ASC speck formation as a readout for inflammasome activation.
- Author
-
Stutz A, Horvath GL, Monks BG, and Latz E
- Subjects
- Animals, Apoptosis Regulatory Proteins, CARD Signaling Adaptor Proteins, Cell Line, Cytoskeletal Proteins genetics, Gene Expression, Genetic Vectors genetics, Humans, Inflammasomes immunology, Macrophages immunology, Macrophages metabolism, Mice, Microscopy, Confocal, Retroviridae genetics, Transduction, Genetic, Cytoskeletal Proteins metabolism, Inflammasomes metabolism
- Abstract
All inflammasomes require the adapter protein apoptosis associated speck-like protein containing a CARD (ASC) for the activation of caspase-1. After inflammasome activation, ASC assembles into a large protein complex, which is termed "speck". ASC specks can be observed as they reach a size of around 1 μm and in most cells only one speck forms upon inflammasome activation. Hence, ASC speck formation can be used as a simple upstream readout for inflammasome activation. Here, we describe a method for analyzing inflammasome activation by ASC speck visualization. First, we describe the generation of a clonal inflammasome reporter macrophage cell line overexpressing fluorescently tagged ASC. We then discuss stimulation conditions and the microscopic evaluation of ASC speck formation.
- Published
- 2013
- Full Text
- View/download PDF
9. Natural loss-of-function mutation of myeloid differentiation protein 88 disrupts its ability to form Myddosomes.
- Author
-
Nagpal K, Plantinga TS, Sirois CM, Monks BG, Latz E, Netea MG, and Golenbock DT
- Subjects
- Animals, Cytoplasm genetics, HEK293 Cells, Humans, Mice, Myeloid Differentiation Factor 88 genetics, Protein Transport physiology, Toll-Like Receptors genetics, Toll-Like Receptors metabolism, Amino Acid Substitution, Cytoplasm metabolism, Mutation, Missense, Myeloid Differentiation Factor 88 metabolism
- 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.
- Published
- 2011
- Full Text
- View/download PDF
10. MD-2 residues tyrosine 42, arginine 69, aspartic acid 122, and leucine 125 provide species specificity for lipid IVA.
- Author
-
Meng J, Drolet JR, Monks BG, and Golenbock DT
- Subjects
- 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, Glycolipids metabolism, Lipid A analogs & derivatives, Lymphocyte Antigen 96 chemistry, Lymphocyte Antigen 96 metabolism
- 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).
- Published
- 2010
- Full Text
- View/download PDF
11. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses.
- Author
-
Rathinam VA, Jiang Z, Waggoner SN, Sharma S, Cole LE, Waggoner L, Vanaja SK, Monks BG, Ganesan S, Latz E, Hornung V, Vogel SN, Szomolanyi-Tsuda E, and Fitzgerald KA
- Subjects
- 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, 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
- 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.
- Published
- 2010
- Full Text
- View/download PDF
12. Cell type-specific recognition of human metapneumoviruses (HMPVs) by retinoic acid-inducible gene I (RIG-I) and TLR7 and viral interference of RIG-I ligand recognition by HMPV-B1 phosphoprotein.
- Author
-
Goutagny N, Jiang Z, Tian J, Parroche P, Schickli J, Monks BG, Ulbrandt N, Ji H, Kiener PA, Coyle AJ, and Fitzgerald KA
- Subjects
- 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, DEAD-box RNA Helicases metabolism, Metapneumovirus immunology, Phosphoproteins metabolism, Toll-Like Receptor 7 metabolism, Viral Interference immunology
- 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.
- Published
- 2010
- Full Text
- View/download PDF
13. A TIR domain variant of MyD88 adapter-like (Mal)/TIRAP results in loss of MyD88 binding and reduced TLR2/TLR4 signaling.
- Author
-
Nagpal K, Plantinga TS, Wong J, Monks BG, Gay NJ, Netea MG, Fitzgerald KA, and Golenbock DT
- Subjects
- 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, 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
- 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.
- Published
- 2009
- Full Text
- View/download PDF
14. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression.
- Author
-
Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, Hornung V, and Latz E
- Subjects
- 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, Carrier Proteins genetics, Gene Expression Regulation immunology, Inflammation metabolism, NF-kappa B physiology, Receptors, Cytokine metabolism, Receptors, Pattern Recognition metabolism
- 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.
- Published
- 2009
- Full Text
- View/download PDF
15. The NALP3 inflammasome is involved in the innate immune response to amyloid-beta.
- Author
-
Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, Fitzgerald KA, Latz E, Moore KJ, and Golenbock DT
- Subjects
- Carrier Proteins metabolism, Inflammation genetics, Inflammation immunology, Inflammation Mediators physiology, NLR Family, Pyrin Domain-Containing 3 Protein, Alzheimer Disease immunology, Amyloid beta-Peptides immunology, Immunity, Innate immunology, Inflammation metabolism
- 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.
- Published
- 2008
- Full Text
- View/download PDF
16. Ligand-induced conformational changes allosterically activate Toll-like receptor 9.
- Author
-
Latz E, Verma A, Visintin A, Gong M, Sirois CM, Klein DC, Monks BG, McKnight CJ, Lamphier MS, Duprex WP, Espevik T, and Golenbock DT
- Subjects
- Allosteric Regulation, Cell Line, CpG Islands immunology, Humans, Ligands, Oligodeoxyribonucleotides metabolism, Protein Binding, Protein Conformation, Toll-Like Receptor 9 chemistry, Toll-Like Receptor 9 metabolism
- Abstract
Microbial and synthetic DNA rich in CpG dinucleotides stimulates Toll-like receptor 9 (TLR9), whereas DNA lacking CpG either is inert or can inhibit TLR9 activation. The molecular mechanisms by which TLR9 becomes activated or is inhibited are not well understood. Here we show that TLR9 bound to stimulatory and inhibitory DNA; however, only stimulatory DNA led to substantial conformational changes in the TLR9 ectodomain. In the steady state, 'inactive' TLR9 homodimers formed in an inactivated conformation. Binding of DNA containing CpG, but not of DNA lacking CpG, to TLR9 dimers resulted in allosteric changes in the TLR9 cytoplasmic signaling domains. In endosomes, conformational changes induced by DNA containing CpG resulted in close apposition of the cytoplasmic signaling domains, a change that is probably required for the recruitment of signaling adaptor molecules. Our results indicate that the formation of TLR9 dimers is not sufficient for its activation but instead that TLR9 activation is regulated by conformational changes induced by DNA containing CpG.
- Published
- 2007
- Full Text
- View/download PDF
17. Malaria hemozoin is immunologically inert but radically enhances innate responses by presenting malaria DNA to Toll-like receptor 9.
- Author
-
Parroche P, Lauw FN, Goutagny N, Latz E, Monks BG, Visintin A, Halmen KA, Lamphier M, Olivier M, Bartholomeu DC, Gazzinelli RT, and Golenbock DT
- Subjects
- Animals, DNA, Protozoan immunology, Humans, Lymphocyte Activation immunology, Melanoma, Experimental, Mice, Plasmodium falciparum immunology, Toll-Like Receptor 9 immunology, Antigen Presentation, DNA, Protozoan metabolism, Hemeproteins physiology, Immunity, Innate, Plasmodium falciparum genetics, Toll-Like Receptor 9 metabolism
- Abstract
Hemozoin (HZ) is an insoluble crystal formed in the food vacuole of malaria parasites. HZ has been reported to induce inflammation by directly engaging Toll-like receptor (TLR) 9, an endosomal receptor. "Synthetic" HZ (beta-hematin), typically generated from partially purified extracts of bovine hemin, is structurally identical to natural HZ. When HPLC-purified hemin was used to synthesize the crystal, beta-hematin had no inflammatory activity. In contrast, natural HZ from Plasmodium falciparum cultures was a potent TLR9 inducer. Natural HZ bound recombinant TLR9 ectodomain, but not TLR2. Both TLR9 stimulation and TLR9 binding of HZ were abolished by nuclease treatment. PCR analysis demonstrated that natural HZ is coated with malarial but not human DNA. Purified malarial DNA activated TLR9 but only when DNA was targeted directly to the endosome with a transfection reagent. Stimulatory quantities of natural HZ contain <1 microg of malarial DNA; its potency in activating immune responses was even greater than transfecting malarial DNA. Thus, although the malarial genome is extremely AT-rich, its DNA is highly proinflammatory, with the potential to induce cytokinemia and fever during disease. However, its activity depends on being bound to HZ, which we propose amplifies the biological responses to malaria DNA by targeting it to a TLR9(+) intracellular compartment.
- Published
- 2007
- Full Text
- View/download PDF
18. MD-2 expression is not required for cell surface targeting of Toll-like receptor 4 (TLR4).
- Author
-
Visintin A, Halmen KA, Khan N, Monks BG, Golenbock DT, and Lien E
- Subjects
- Animals, Cell Line, Humans, Lipopolysaccharides pharmacology, Lymphocyte Antigen 96 deficiency, Lymphocyte Antigen 96 immunology, Mice, Mice, Knockout, Toll-Like Receptor 4 deficiency, Toll-Like Receptor 4 immunology, Gene Expression Regulation drug effects, Gene Expression Regulation genetics, Gene Expression Regulation immunology, Lymphocyte Antigen 96 biosynthesis, Toll-Like Receptor 4 biosynthesis, Toll-Like Receptor 4 metabolism
- 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.
- Published
- 2006
- Full Text
- View/download PDF
19. The myristoylation of TRIF-related adaptor molecule is essential for Toll-like receptor 4 signal transduction.
- Author
-
Rowe DC, McGettrick AF, Latz E, Monks BG, Gay NJ, Yamamoto M, Akira S, O'Neill LA, Fitzgerald KA, and Golenbock DT
- Subjects
- Adaptor Proteins, Vesicular Transport analysis, Adaptor Proteins, Vesicular Transport genetics, Amino Acid Sequence, Animals, Bone Marrow Cells drug effects, Bone Marrow Cells immunology, Cell Membrane chemistry, Cell Membrane metabolism, Cells, Cultured, Humans, Lipopolysaccharides immunology, Lipopolysaccharides pharmacology, Macrophages drug effects, Mice, Mice, Mutant Strains, Molecular Sequence Data, Mutation, Myristic Acid metabolism, Signal Transduction, Toll-Like Receptor 4 analysis, Adaptor Proteins, Vesicular Transport metabolism, Macrophages immunology, Toll-Like Receptor 4 metabolism
- Abstract
TRIF-related adaptor molecule (TRAM) is the fourth Toll/IL-1 resistance domain-containing adaptor to be described that participates in Toll-like receptor (TLR) signaling. TRAM functions exclusively in the TLR4 pathway. Here we show by confocal microscopy that TRAM is localized in the plasma membrane and the Golgi apparatus, where it colocalizes with TLR4. Membrane localization of TRAM is the result of myristoylation because mutation of a predicted myristoylation site in TRAM (TRAM-G2A) brought about dissociation of TRAM from the membrane and its relocation to the cytosol. Further, TRAM, but not TRAM-G2A, was radiolabeled with [3H]myristate in vivo. Unlike wild-type TRAM, overexpression of TRAM-G2A failed to elicit either IFN regulatory factor 3 or NF-kappaB signaling. Moreover, TRAM-G2A was unable to reconstitute LPS responses in bone marrow-derived macrophages from TRAM-deficient mice. These observations provide clear evidence that the myristoylation of TRAM targets it to the plasma membrane, where it is essential for LPS responses through the TLR4 signal transduction pathway, and suggest a hitherto unappreciated manner in which LPS responses can be regulated.
- Published
- 2006
- Full Text
- View/download PDF
20. MD-2.
- Author
-
Visintin A, Iliev DB, Monks BG, Halmen KA, and Golenbock DT
- Subjects
- 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, Lymphocyte Antigen 96 physiology
- 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.
- Published
- 2006
- Full Text
- View/download PDF
21. Pharmacological inhibition of endotoxin responses is achieved by targeting the TLR4 coreceptor, MD-2.
- Author
-
Visintin A, Halmen KA, Latz E, Monks BG, and Golenbock DT
- Subjects
- Cell Line, Humans, Kinetics, Lipid A analogs & derivatives, Lipid A pharmacology, Lipopolysaccharides antagonists & inhibitors, Lipopolysaccharides metabolism, Lymphocyte Antigen 96 blood, Lymphocyte Antigen 96 chemistry, Protein Binding drug effects, Protein Structure, Tertiary, Recombinant Fusion Proteins metabolism, Recombinant Fusion Proteins pharmacology, Signal Transduction, Solubility, Toll-Like Receptor 4 chemistry, Lipopolysaccharides toxicity, Lymphocyte Antigen 96 metabolism, Toll-Like Receptor 4 metabolism
- Abstract
The detection of Gram-negative LPS depends upon the proper function of the TLR4-MD-2 receptor complex in immune cells. TLR4 is the signal transduction component of the LPS receptor, whereas MD-2 is the endotoxin-binding unit. MD-2 appears to activate TLR4 when bound to TLR4 and ligated by LPS. Only the monomeric form of MD-2 was found to bind LPS and only monomeric MD-2 interacts with TLR4. Monomeric MD-2 binds TLR4 with an apparent Kd of 12 nM; this binding avidity was unaltered in the presence of endotoxin. E5564, an LPS antagonist, appears to inhibit cellular activation by competitively preventing the binding of LPS to MD-2. Depletion of endogenous soluble MD-2 from human serum, with an immobilized TLR4 fusion protein, abrogated TLR4-mediated LPS responses. By determining the concentration of added-back MD-2 that restored normal LPS responsiveness, the concentration of MD-2 was estimated to be approximately 50 nM. Similarly, purified TLR4-Fc fusion protein, when added to the supernatants of TLR4-expressing cells in culture, inhibited the interaction of MD-2 with TLR4, thus preventing LPS stimulation. The ability to inhibit the effects of LPS as a result of the binding of TLR4-Fc or E5564 to MD-2 highlights MD-2 as the logical target for drug therapies designed to pharmacologically intervene against endotoxin-induced disease.
- Published
- 2005
- Full Text
- View/download PDF
22. The RNA helicase Lgp2 inhibits TLR-independent sensing of viral replication by retinoic acid-inducible gene-I.
- Author
-
Rothenfusser S, Goutagny N, DiPerna G, Gong M, Monks BG, Schoenemeyer A, Yamamoto M, Akira S, and Fitzgerald KA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Adaptor Proteins, Vesicular Transport genetics, Animals, Antigens, Differentiation genetics, Cell Line, Humans, Membrane Transport Proteins genetics, Mice, Mice, Knockout, Myelin Proteins genetics, Myelin and Lymphocyte-Associated Proteolipid Proteins, Myeloid Differentiation Factor 88, Proteolipids genetics, RNA, Double-Stranded metabolism, Receptors, Immunologic genetics, Sendai virus physiology, Signal Transduction physiology, TNF Receptor-Associated Factor 6 genetics, Toll-Like Receptor 3 genetics, Toll-Like Receptor 3 metabolism, Trans-Activators genetics, RNA Helicases physiology, Toll-Like Receptors physiology, Trans-Activators physiology, Virus Replication physiology
- Abstract
The paramyxovirus Sendai (SV), is a well-established inducer of IFN-alphabeta gene expression. In this study we show that SV induces IFN-alphabeta gene expression normally in cells from mice with targeted deletions of the Toll-IL-1 resistance domain containing adapters MyD88, Mal, Toll/IL-1R domain-containing adaptor inducing IFN-beta (TRIF), and TRIF-related adaptor molecule TLR3, or the E3 ubiquitin ligase, TNFR-associated factor 6. This TLR-independent induction of IFN-alphabeta after SV infection is replication dependent and mediated by the RNA helicase, retinoic acid-inducible gene-I (RIG-I) and not the related family member, melanoma differentiation-associated gene 5. Furthermore, we characterize a RIG-I-like RNA helicase, Lgp2. In contrast to RIG-I or melanoma differentiation-associated gene 5, Lgp2 lacks signaling caspase recruitment and activation domains. Overexpression of Lgp2 inhibits SV and Newcastle disease virus signaling to IFN-stimulated regulatory element- and NF-kappaB-dependent pathways. Importantly, Lgp2 does not prevent TLR3 signaling. Like RIG-I, Lgp2 binds double-stranded, but not single-stranded, RNA. Quantitative PCR analysis demonstrates that Lgp2 is present in unstimulated cells at a lower level than RIG-I, although both helicases are induced to similar levels after virus infection. We propose that Lgp2 acts as a negative feedback regulator of antiviral signaling by sequestering dsRNA from RIG-I.
- Published
- 2005
- Full Text
- View/download PDF
23. TLR9 signals after translocating from the ER to CpG DNA in the lysosome.
- Author
-
Latz E, Schoenemeyer A, Visintin A, Fitzgerald KA, Monks BG, Knetter CF, Lien E, Nilsen NJ, Espevik T, and Golenbock DT
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Antigens, Differentiation genetics, Antigens, Differentiation immunology, Biological Transport, Active, Cell Line, DNA-Binding Proteins genetics, Dendritic Cells immunology, Humans, In Vitro Techniques, Luminescent Proteins genetics, Luminescent Proteins immunology, Membrane Glycoproteins genetics, Mice, Mice, Inbred C57BL, Mice, Knockout, Myeloid Differentiation Factor 88, Receptors, Cell Surface genetics, Receptors, Immunologic deficiency, Receptors, Immunologic genetics, Receptors, Immunologic immunology, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Signal Transduction, Toll-Like Receptor 9, Toll-Like Receptors, CpG Islands immunology, DNA-Binding Proteins immunology, Endoplasmic Reticulum immunology, Lysosomes immunology, Membrane Glycoproteins immunology, Receptors, Cell Surface immunology
- Abstract
Microbial DNA sequences containing unmethylated CpG dinucleotides activate Toll-like receptor 9 (TLR9). We have found that TLR9 is localized to the endoplasmic reticulum (ER) of dendritic cells (DCs) and macrophages. Because there is no precedent for immune receptor signaling in the ER, we investigated how TLR9 is activated. We show that CpG DNA binds directly to TLR9 in ligand-binding studies. CpG DNA moves into early endosomes and is subsequently transported to a tubular lysosomal compartment. Concurrent with the movement of CpG DNA in cells, TLR9 redistributes from the ER to CpG DNA-containing structures, which also accumulate MyD88. Our data indicate a previously unknown mechanism of cellular activation involving the recruitment of TLR9 from the ER to sites of CpG DNA uptake, where signal transduction is initiated.
- Published
- 2004
- Full Text
- View/download PDF
24. Lysines 128 and 132 enable lipopolysaccharide binding to MD-2, leading to Toll-like receptor-4 aggregation and signal transduction.
- Author
-
Visintin A, Latz E, Monks BG, Espevik T, and Golenbock DT
- Subjects
- Amino Acid Sequence, Biotinylation, Blotting, Western, Cell Line, Cell Membrane metabolism, Cell Membrane ultrastructure, Cysteine chemistry, Humans, Lipopolysaccharide Receptors biosynthesis, Lipopolysaccharide Receptors metabolism, Lymphocyte Antigen 96, Microscopy, Electron, Scanning, Microscopy, Fluorescence, Molecular Sequence Data, Precipitin Tests, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins metabolism, Sequence Homology, Amino Acid, Toll-Like Receptor 4, Toll-Like Receptors, Transfection, Tyrosine chemistry, Antigens, Surface metabolism, Lipopolysaccharides metabolism, Lysine chemistry, Membrane Glycoproteins metabolism, Receptors, Cell Surface metabolism, Signal Transduction
- Abstract
Three cell-surface proteins have been recognized as components of the mammalian signaling receptor for bacterial lipopolysaccharide (LPS): CD14, Toll-like receptor-4 (TLR4), and MD-2. Biochemical and visual studies shown here demonstrate that the role of CD14 in signal transduction is to enhance LPS binding to MD-2, although its expression is not essential for cellular activation. These studies clarify how MD-2 functions: we found that MD-2 enables TLR4 binding to LPS and allows the formation of stable receptor complexes. MD-2 must be bound to TLR4 on the cell surface before binding can occur. Consequently, TLR4 clusters into receptosomes (many of which are massive) that recruit intracellular toll/IL-1/resistance domain-containing adapter proteins within minutes, thus initiating signal transduction. TLR4 activation correlates with the ability of MD-2 to bind LPS, as MD-2 mutants that still bind TLR4, but are impaired in the ability to bind LPS, conferred a greatly blunted LPS response. These findings help clarify the earliest events of TLR4 triggering by LPS and identify MD-2 as an attractive target for pharmacological intervention in endotoxin-mediated diseases.
- Published
- 2003
- Full Text
- View/download PDF
25. Lipopolysaccharide rapidly traffics to and from the Golgi apparatus with the toll-like receptor 4-MD-2-CD14 complex in a process that is distinct from the initiation of signal transduction.
- Author
-
Latz E, Visintin A, Lien E, Fitzgerald KA, Monks BG, Kurt-Jones EA, Golenbock DT, and Espevik T
- Subjects
- Adaptor Proteins, Signal Transducing, Antigens, Differentiation metabolism, Blotting, Western, Brefeldin A pharmacology, Cell Line, Cell Separation, Dose-Response Relationship, Drug, Escherichia coli metabolism, Flow Cytometry, Fluorescent Dyes pharmacology, Genes, Reporter, Green Fluorescent Proteins, Humans, Luciferases metabolism, Luminescent Proteins metabolism, Lymphocyte Antigen 96, Microscopy, Confocal, Microscopy, Fluorescence, Myeloid Differentiation Factor 88, Plasmids metabolism, Precipitin Tests, Protein Binding, Protein Structure, Tertiary, Protein Transport, Receptors, Immunologic metabolism, Recombinant Fusion Proteins metabolism, Time Factors, Toll-Like Receptor 4, Toll-Like Receptors, Transfection, Antigens, Surface metabolism, Drosophila Proteins, Golgi Apparatus metabolism, Lipopolysaccharide Receptors metabolism, Lipopolysaccharides metabolism, Membrane Glycoproteins metabolism, Receptors, Cell Surface metabolism, Signal Transduction
- Abstract
Mammalian responses to LPS require the expression of Toll-like receptor 4 (TLR4), CD14, and MD-2. We expressed fluorescent TLR4 in cell lines and found that TLR4 densely localized to the surface and the Golgi. Similar distributions were observed in human monocytes. Confocal imaging revealed rapid recycling of TLR4-CD14-MD-2 complexes between the Golgi and the plasma membrane. Fluorescent LPS followed these trafficking pathways in CD14-positive cells. The TLR4- adapter protein, MyD88, translocated to the cell surface upon LPS exposure, and cross-linking of surface TLR4 with antibody induced signaling. Golgi-associated TLR4 expression was disrupted by brefeldin A, yet LPS signaling was preserved. We conclude that LPS signaling may be initiated by surface aggregation of TLR4 and is not dependent upon LPS trafficking to the Golgi.
- Published
- 2002
- Full Text
- View/download PDF
26. Involvement of toll-like receptor (TLR) 2 and TLR4 in cell activation by mannuronic acid polymers.
- Author
-
Flo TH, Ryan L, Latz E, Takeuchi O, Monks BG, Lien E, Halaas Ø, Akira S, Skjåk-Braek G, Golenbock DT, and Espevik T
- Subjects
- Animals, Cell Line, Cell Membrane metabolism, Humans, Interleukin-6 biosynthesis, Mice, Mice, Inbred C3H, Signal Transduction, Toll-Like Receptor 2, Toll-Like Receptor 4, Toll-Like Receptors, Transfection, Tumor Necrosis Factor-alpha biosynthesis, Biopolymers physiology, Drosophila Proteins, Hexuronic Acids metabolism, Membrane Glycoproteins physiology, Protein Isoforms physiology, Receptors, Cell Surface physiology
- Abstract
The alginate capsule produced by the human pathogen Pseudomonas aeruginosa is composed mainly of mannuronic acid polymers (poly-M) that have immunostimulating properties. Poly-M shares with lipopolysaccharide the ability to stimulate cytokine production from human monocytes in a CD14-dependent manner. In the present study we examined the role of Toll-like receptor (TLR) 2 and TLR4 in responses to poly-M. Blocking antibodies to TLR2 and TLR4 partly inhibited tumor necrosis factor production induced by poly-M in human monocytes, and further inhibition was obtained by combining the antibodies. By transiently transfecting HEK293 cells, we found that membrane CD14 together with either TLR2 or TLR4/MD-2 could mediate activation by poly-M. Transfection of HEK293 cells with TLR2 and fluorescently labeled TLR4 followed by co-patching of TLR2 with an antibody revealed no association of these molecules on the plasma membrane. However, macrophages from the Tlr4 mutant C3H/HeJ mice and TLR4 knockout mice were completely non-responsive to poly-M, whereas the tumor necrosis factor release from TLR2 knockout macrophages was half of that seen with wild type cells. Taken together the results suggest that both TLR2 and TLR4 are involved in cell activation by poly-M and that TLR4 may be required in primary murine macrophages.
- Published
- 2002
- Full Text
- View/download PDF
27. Molecular genetic analysis of an endotoxin nonresponder mutant cell line: a point mutation in a conserved region of MD-2 abolishes endotoxin-induced signaling.
- Author
-
Schromm AB, Lien E, Henneke P, Chow JC, Yoshimura A, Heine H, Latz E, Monks BG, Schwartz DA, Miyake K, and Golenbock DT
- Subjects
- Animals, Antigens, Surface metabolism, CHO Cells drug effects, Cell Line, Clone Cells, Cloning, Molecular, Cricetinae, DNA Mutational Analysis, Genetic Complementation Test, Humans, Interleukin-1 pharmacology, Interleukin-6 metabolism, Lipopolysaccharide Receptors genetics, Lipopolysaccharide Receptors metabolism, Lipopolysaccharides metabolism, Lipopolysaccharides toxicity, Lymphocyte Antigen 96, Membrane Glycoproteins genetics, Mutation, Receptors, Cell Surface genetics, Signal Transduction, Toll-Like Receptor 4, Toll-Like Receptors, Tumor Necrosis Factor-alpha pharmacology, Antigens, Surface genetics, Drosophila Proteins, Endotoxins metabolism, Membrane Glycoproteins metabolism, Receptors, Cell Surface metabolism
- Abstract
Somatic cell mutagenesis is a powerful tool for characterizing receptor systems. We reported previously two complementation groups of mutant cell lines derived from CD14-transfected Chinese hamster ovary--K1 fibroblasts defective in responses to bacterial endotoxin. Both classes of mutants expressed a normal gene product for Toll-like receptor (TLR)4, and fully responded to stimulation by tumor necrosis factor (TNF)-alpha or interleukin (IL)-1 beta. We identified the lesion in one of the complementation groups in the gene for MD-2, a putative TLR4 coreceptor. The nonresponder phenotype of this mutant was reversed by transfection with MD-2. Cloning of MD-2 from the nonresponder cell line revealed a point mutation in a highly conserved region resulting in a C95Y amino acid exchange. Both forms of MD-2 colocalized with TLR4 on the cell surface after transfection, but only the wild-type cDNA reverted the lipopolysaccharide (LPS) nonresponder phenotype. Furthermore, soluble MD-2, but not soluble MD-2(C95Y), functioned to enable LPS responses in cells that expressed TLR4. Thus, MD-2 is a required component of the LPS signaling complex and can function as a soluble receptor for cells that do not otherwise express it. We hypothesize that MD-2 conformationally affects the extracellular domain of TLR4, perhaps resulting in a change in affinity for LPS or functioning as a portion of the true ligand for TLR4.
- Published
- 2001
- Full Text
- View/download PDF
28. C4bp binding to porin mediates stable serum resistance of Neisseria gonorrhoeae.
- Author
-
Ram S, Cullinane M, Blom AM, Gulati S, McQuillen DP, Boden R, Monks BG, O'Connell C, Elkins C, Pangburn MK, Dahlbäck B, and Rice PA
- Subjects
- Amino Acid Sequence, Binding Sites, Blood Bactericidal Activity immunology, Complement C4 metabolism, Humans, Immunoglobulin M metabolism, In Vitro Techniques, Molecular Sequence Data, Neisseria gonorrhoeae genetics, Neisseria gonorrhoeae pathogenicity, Phenotype, Porins chemistry, Porins genetics, Porins immunology, Sequence Homology, Amino Acid, Complement Inactivator Proteins, Glycoproteins, Neisseria gonorrhoeae immunology, Porins metabolism, Receptors, Complement metabolism
- Abstract
Screening of 29 strains of Neisseria gonorrhoeae revealed that 16/21 serum resistant strains and 0/8 serum sensitive strains bound C4bp, suggesting that C4bp binding to gonococci could contribute to serum resistance. C4bp bound to gonococci retained cofactor (C4b-degrading) function. Using allelic exchange to construct strains with hybrid Por1A/B molecules, we demonstrate that the N-terminal loop (loop 1) of Por1A is required for C4bp binding. Serum resistant Por1B gonococcal strains also bind C4bp via their Por molecule. Using allelic exchange and site-directed mutagenesis, we have shown that loops 5 and 7 together form a negatively charged C4bp binding domain. C4bp-Por1B interactions are ionic in nature (inhibited by high salt as well as by heparin), while the C4bp-Por1A bond is hydrophobic. mAbs directed against SCR1 of the alpha-chain of C4bp inhibit C4bp binding to both Por1A and Por1B. Furthermore, only recombinant C4bp mutant molecules that contain alpha-chain SCR1 bind both Por1A and Por1B gonococci, confirming that SCR1 contains Por binding sites. C4bp alpha-chain monomers do not bind strains with either Por molecule, suggesting that the polymeric form of C4bp is required for binding to gonococci. Inhibition of C4bp binding to serum resistant Por1A and Por1B strains in a serum bactericidal assay using fAb fragments against C4bp SCR1 results in complete killing at 30 min of otherwise fully serum resistant strains in only 10% normal serum, underscoring the role of C4bp in mediating gonococcal serum resistance.
- Published
- 2001
- Full Text
- View/download PDF
29. Binding of C4b-binding protein to porin: a molecular mechanism of serum resistance of Neisseria gonorrhoeae.
- Author
-
Ram S, Cullinane M, Blom AM, Gulati S, McQuillen DP, Monks BG, O'Connell C, Boden R, Elkins C, Pangburn MK, Dahlbäck B, and Rice PA
- Subjects
- Amino Acid Sequence, Base Sequence, Cell Line, Complement C4 immunology, Humans, Molecular Sequence Data, Peptide Fragments immunology, Porins genetics, Protein S immunology, Receptors, Complement genetics, Complement C4b immunology, Complement Inactivator Proteins, Glycoproteins, Neisseria gonorrhoeae immunology, Porins immunology, Receptors, Complement immunology
- Abstract
We screened 29 strains of Neisseria gonorrhoeae and found 16/21 strains that resisted killing by normal human serum and 0/8 serum sensitive strains that bound the complement regulator, C4b-binding protein (C4bp). Microbial surface-bound C4bp demonstrated cofactor activity. We constructed gonococcal strains with hybrid porin (Por) molecules derived from each of the major serogroups (Por1A and Por1B) of N. gonorrhoeae, and showed that the loop 1 of Por1A is required for C4bp binding. Por1B loops 5 and 7 of serum-resistant gonococci together formed a negatively charged C4bp-binding domain. C4bp-Por1B interactions were ionic in nature (inhibited by high salt or by heparin), whereas the C4bp-Por1A bond was hydrophobic. Only recombinant C4bp mutant molecules containing the NH2-terminal alpha-chain short consensus repeat (SCR1) bound to both Por1A and Por1B gonococci, suggesting that SCR1 contained Por binding sites. C4bp alpha-chain monomers did not bind gonococci, indicating that the polymeric form of C4bp was required for binding. Using fAb fragments against C4bp SCR1, C4bp binding to Por1A and Por1B strains was inhibited in a complement-dependent serum bactericidal assay. This resulted in complete killing of these otherwise fully serum resistant strains in only 10% normal serum, underscoring the importance of C4bp in mediating gonococcal serum resistance.
- Published
- 2001
- Full Text
- View/download PDF
30. Divergent response to LPS and bacteria in CD14-deficient murine macrophages.
- Author
-
Moore KJ, Andersson LP, Ingalls RR, Monks BG, Li R, Arnaout MA, Golenbock DT, and Freeman MW
- Subjects
- Animals, Bacterial Adhesion genetics, Bacterial Adhesion immunology, Bacterial Outer Membrane Proteins physiology, CD18 Antigens physiology, Cell Line, Cytokines biosynthesis, Escherichia coli physiology, Female, Lipopolysaccharide Receptors blood, Macrophage Activation genetics, Macrophage-1 Antigen physiology, Macrophages, Peritoneal metabolism, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutagenesis, Insertional, Phagocytosis immunology, Signal Transduction immunology, Escherichia coli immunology, Gene Deletion, Lipopolysaccharide Receptors genetics, Lipopolysaccharides immunology, Macrophages, Peritoneal immunology, Macrophages, Peritoneal microbiology
- Abstract
Gram-negative bacteria and the LPS constituent of their outer membranes stimulate the release of inflammatory mediators believed to be responsible for the clinical manifestations of septic shock. The GPI-linked membrane protein, CD14, initiates the signaling cascade responsible for the induction of this inflammatory response by LPS. In this paper, we report the generation and characterization of CD14-null mice in which the entire coding region of CD14 was deleted. As expected, LPS failed to elicit TNF-alpha and IL-6 production in macrophages taken from these animals, and this loss in responsiveness is associated with impaired activation of both the NF-kappaB and the c-Jun N-terminal mitogen-activated protein kinase pathways. The binding and uptake of heat-killed Escherichia coli, measured by FACS analysis, did not differ between CD14-null and wild-type macrophages. However, in contrast to the findings with LPS, whole E. coli stimulated similar levels of TNF-alpha release from CD14-null and wild-type macrophages at a dose of 10 bioparticles per cell. This effect was dose dependent, and at lower bacterial concentrations CD14-deficient macrophages produced significantly less TNF-alpha than wild type. Approximately half of this CD14-independent response appeared to be mediated by CD11b/CD18, as demonstrated by receptor blockade using neutrophil inhibitory factor. An inhibitor of phagocytosis, cytochalasin B, abrogated the induction of TNF-alpha in CD14-deficient macrophages by E. coli. These data indicate that CD14 is essential for macrophage responses to free LPS, whereas other receptors, including CD11b/CD18, can compensate for the loss of CD14 in response to whole bacteria.
- Published
- 2000
- Full Text
- View/download PDF
31. Bacterial lipopolysaccharide induces expression of the stress response genes hop and H411.
- Author
-
Heine H, Delude RL, Monks BG, Espevik T, and Golenbock DT
- Subjects
- Amino Acid Sequence, Animals, CHO Cells, Cricetinae, Fungal Proteins metabolism, Growth Substances metabolism, Heat-Shock Proteins metabolism, Humans, Mice, Molecular Sequence Data, Rats, Sequence Alignment, Fungal Proteins genetics, Gene Expression Regulation drug effects, Growth Substances genetics, Heat-Shock Proteins genetics, Lipopolysaccharide Receptors genetics, Lipopolysaccharides pharmacology, Molecular Chaperones
- Abstract
CD14-transfected Chinese hamster ovary K1 fibroblasts (CHO/CD14) respond to lipopolysaccharide (LPS) by metabolizing arachidonic acid and with translocation of NF-kappaB to the nucleus. Although previous experiments failed to identify the production of tumor necrosis factor-alpha and interleukin (IL)-1beta by CHO/CD14 cells, LPS did induce the expression of IL-6 mRNA and the subsequent release of the IL-6 protein. To identify additional LPS-inducible genes, a cDNA library derived from LPS-stimulated CHO/CD14 cells was screened by subtractive hybridization. Fourteen genes were found to be expressed differentially, and two were analyzed in detail: hop (Hsp70/Hsp90-organizing protein), which is the hamster homologue of the stress-inducible yeast gene, STI1, and clone H411, which encodes a novel LPS-inducible growth factor. In response to LPS, the expression of Hop mRNA was also increased in both the murine macrophage cell line, RAW 264.7, as well as in primary hamster macrophages. This suggested that the up-regulation of Hop expression is part of the macrophage stress response to LPS. Clone H411 encodes a protein in the epidermal growth factor-like repeat protein family. Overexpression of H411 cDNA in the RAW 264.7 macrophage cell line promoted an increased growth rate, suggesting that expression of H411 is part of the proliferative cell response to LPS. Both Hop and H411 represent novel gene products not previously recognized as part of the complex biological response to endotoxin.
- Published
- 1999
- Full Text
- View/download PDF
32. Cutting edge: cells that carry A null allele for toll-like receptor 2 are capable of responding to endotoxin.
- Author
-
Heine H, Kirschning CJ, Lien E, Monks BG, Rothe M, and Golenbock DT
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, CHO Cells immunology, Cloning, Molecular, Cricetinae, Female, Humans, Macrophages, Peritoneal immunology, Macrophages, Peritoneal metabolism, Membrane Glycoproteins chemistry, Membrane Glycoproteins deficiency, Mice, Molecular Sequence Data, Receptors, Cell Surface chemistry, Receptors, Cell Surface deficiency, Sequence Deletion, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Signal Transduction genetics, Signal Transduction immunology, Toll-Like Receptor 2, Toll-Like Receptor 4, Toll-Like Receptors, Alleles, CHO Cells metabolism, Drosophila Proteins, Lipopolysaccharides pharmacology, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism
- Abstract
Toll-like receptor (TLR) 2 and TLR4 have been implicated in the responses of cells to LPS (endotoxin). CD14-transfected Chinese hamster ovary (CHO)-K1 fibroblasts (CHO/CD14) are exquisitely sensitive to endotoxin. Sequence analysis of CHO-TLR2, compared with human and mouse TLR2, revealed a single base pair deletion. This frameshift mutation resulted in an alternative stop codon, encoding a protein devoid of transmembrane and intracellular domains. CHO-TLR2 cDNA failed to enable LPS signaling upon transient transfection into human epithelial kidney 293 cells. Site-directed mutagenesis of CHO-TLR2 enabled expression of a presumed full-length hamster TLR2 that conferred LPS responsiveness in human epithelial kidney 293 cells. Genomic TLR2 DNA from primary hamster macrophages also contained the frameshift mutation found in CHO fibroblasts. Nevertheless, hamster peritoneal macrophages were found to respond normally to LPS, as evidenced by the induction of cytokines. These results imply that expression of TLR2 is sufficient but not essential for mammalian responses to endotoxin.
- Published
- 1999
33. Membrane expression of soluble endotoxin-binding proteins permits lipopolysaccharide signaling in Chinese hamster ovary fibroblasts independently of CD14.
- Author
-
Ingalls RR, Monks BG, and Golenbock DT
- Subjects
- Amino Acid Sequence, Animals, Antimicrobial Cationic Peptides, Base Sequence, Blood Bactericidal Activity, Blood Proteins metabolism, CD11 Antigens metabolism, CHO Cells, Cricetinae, Fibroblasts drug effects, Fibroblasts metabolism, Gram-Negative Bacteria metabolism, Humans, Molecular Sequence Data, Lipopolysaccharide Receptors metabolism, Lipopolysaccharides pharmacology, Membrane Proteins metabolism, Signal Transduction
- Abstract
The activation of phagocytes by lipopolysaccharide (LPS) has been implicated in the pathogenesis of Gram-negative sepsis. Although the interaction between CD14 and LPS is a key event in the signaling cascade, the molecular mechanism by which cellular activation occurs remains obscure. We hypothesized that the main function of CD14 was to bind LPS and transfer it to a second receptor, which then initiates the subsequent signal for cellular activation. Thus, surface binding of LPS to the cell membrane would be the critical step that CD14 carries out. To test this hypothesis, we examined the activity of two other proteins known to bind LPS, lipopolysaccharide-binding protein and bactericidal/permeability-increasing protein. We found that when these normally soluble proteins were expressed in Chinese hamster ovary-K1 fibroblasts as glycosylphosphatidylinositol-anchored proteins, both could substitute for CD14 in initiating LPS signaling. Pharmacological studies with synthetic lipid A analogues demonstrated that these surface expressed LPS-binding proteins had characteristics that were qualitatively identical to membrane CD14. These data support the hypothesis that a receptor distinct from CD14 functions as the actual signal transducer and suggest that surface binding of LPS to the cell membrane is the crucial first step for initiating downstream signaling events.
- Published
- 1999
- Full Text
- View/download PDF
34. CD11/CD18 and CD14 share a common lipid A signaling pathway.
- Author
-
Ingalls RR, Monks BG, Savedra R Jr, Christ WJ, Delude RL, Medvedev AE, Espevik T, and Golenbock DT
- Subjects
- Animals, CD11 Antigens genetics, CHO Cells, Carrier Proteins physiology, Cell Line, Cricetinae, Gram-Negative Bacteria immunology, Humans, Lipid A analogs & derivatives, Lipopolysaccharides antagonists & inhibitors, Lipopolysaccharides chemistry, Lipopolysaccharides metabolism, Lipopolysaccharides pharmacology, Macrophage Activation drug effects, Mice, Mycobacterium immunology, Peptidoglycan pharmacology, Transfection drug effects, Transfection immunology, Tumor Cells, Cultured, Acute-Phase Proteins, CD11 Antigens physiology, CD18 Antigens physiology, Lipid A physiology, Lipopolysaccharide Receptors physiology, Membrane Glycoproteins, Signal Transduction immunology
- Abstract
The activation of phagocytes by the lipid A moiety of LPS has been implicated in the pathogenesis of Gram-negative sepsis. While two LPS receptors, CD14 and CD11/CD18, have been associated with cell signaling, details of the LPS signal transduction cascade remain obscure. CD14, which exists as a GPI-anchored and a soluble protein, lacks cytoplasmic-signaling domains, suggesting that an ancillary molecule is required to activate cells. The CD11/CD18 integrins are transmembrane proteins. Like CD14, they are capable of mediating LPS-induced cellular activation when expressed on the surface of hamster fibroblasts Chinese hamster ovary (CHO)-K1. The observation that a cytoplasmic deletion mutant is still capable of activating transfected CHO-K1 argues that CD11/CD18 also utilizes an associated signal transducer. We sought to identify further similarities between the signaling systems utilized by CD14 and CD11/CD18. LPS-binding protein, which transfers LPS to CD14, enhanced both LPS-induced cellular activation and binding of Gram-negative bacteria in CD11/CD18-transfected CHO-K1, thus implying that LPS-binding protein can also transfer LPS to CD11/CD18. When synthetic lipid A analogues were analyzed for their ability to function as LPS agonists, or antagonists, in the CHO transfectants, we found the effects were identical regardless of which LPS receptor was expressed. This supports the hypothesis that a receptor distinct from CD14 and CD11/CD18 is responsible for discriminating between the lipid A of LPS and the LPS antagonists. We propose that this receptor, which is the target of the LPS antagonists, functions as the true signal transducer in LPS-induced cellular activation for both CD14 and CD11/CD18.
- Published
- 1998
35. Targeted deletion of the lipopolysaccharide (LPS)-binding protein gene leads to profound suppression of LPS responses ex vivo, whereas in vivo responses remain intact.
- Author
-
Wurfel MM, Monks BG, Ingalls RR, Dedrick RL, Delude R, Zhou D, Lamping N, Schumann RR, Thieringer R, Fenton MJ, Wright SD, and Golenbock D
- Subjects
- Animals, Carrier Proteins genetics, Chimera, In Vitro Techniques, Kidney chemistry, Kidney drug effects, Lipopolysaccharide Receptors metabolism, Liver chemistry, Liver drug effects, Mice, Mice, Knockout, Mice, Mutant Strains, Tumor Necrosis Factor-alpha metabolism, Acute-Phase Proteins genetics, Carrier Proteins physiology, Gene Deletion, Lipopolysaccharides pharmacology, Membrane Glycoproteins
- Abstract
Gram-negative bacterial lipopolysaccharide (LPS) stimulates phagocytic leukocytes by interacting with the cell surface protein CD14. Cellular responses to LPS are markedly potentiated by the LPS-binding protein (LBP), a lipid-transfer protein that binds LPS aggregates and transfers LPS monomers to CD14. LBP also transfers LPS to lipoproteins, thereby promoting the neutralization of LPS. LBP present in normal plasma has been shown to enhance the LPS responsiveness of cells in vitro. The role of LBP in promoting LPS responsiveness in vivo was tested in LBP-deficient mice produced by gene targeting in embryonic stem cells. Whole blood from LBP-deficient animals was 1,000-fold less responsive to LPS as assessed by the release of tumor necrosis factor (TNF)-alpha. Blood from gene-targeted mice was devoid of immunoreactive LBP, essentially incapable of transferring LPS to CD14 in vitro, and failed to support cellular responses to LPS. These activities were restored by the addition of exogenous recombinant murine LBP to the plasma. Despite these striking in vitro findings, no significant differences in TNF-alpha levels were observed in plasma from wild-type and LBP-deficient mice injected with LPS. These data suggest the presence of an LBP-independent mechanism for responding to LPS. These LBP knockout mice may provide a tool for discovering the nature of the presumed second mechanism for transferring LPS to responsive cells.
- Published
- 1997
- Full Text
- View/download PDF
36. Dual effects of LPS antibodies on cellular uptake of LPS and LPS-induced proinflammatory functions.
- Author
-
Pollack M, Ohl CA, Golenbock DT, Di Padova F, Wahl LM, Koles NL, Guelde G, and Monks BG
- Subjects
- Adult, Animals, Antibodies, Bacterial chemistry, Antibodies, Monoclonal chemistry, Antigens, Bacterial chemistry, Antigens, Bacterial immunology, Biological Transport immunology, CHO Cells, Cricetinae, Fibroblasts immunology, Fibroblasts metabolism, Humans, Inflammation blood, Inflammation etiology, Inflammation immunology, Lipopolysaccharide Receptors immunology, Lipopolysaccharides blood, Lipopolysaccharides chemistry, Monocytes metabolism, NF-kappa B immunology, NF-kappa B metabolism, Protein Conformation, Receptors, Complement 3b immunology, Antibodies, Bacterial pharmacology, Antibodies, Monoclonal pharmacology, Lipopolysaccharides immunology, Lipopolysaccharides metabolism, Monocytes immunology, Monocytes pathology
- Abstract
Human phagocytes recognize bacterial LPS (endotoxin) through membrane CD14 (mCD14), a proinflammatory LPS receptor. This study tested the hypothesis that anti-LPS Abs neutralize endotoxin by blocking cellular uptake through mCD14. Ab-associated changes in the uptake and cellular distribution of FITC-LPS were assessed by flow cytometry and laser scanning confocal microscopy in human CD14-transfected Chinese hamster ovary fibroblasts (CHO-CD14 cells) and human peripheral blood monocytes. LPS core- and O-side chain-specific mAbs inhibited mCD14-mediated LPS uptake by both cell types in the presence of serum. O-side chain-specific mAb concurrently enhanced complement-dependent LPS uptake by monocytes through complement receptor-1 (CR1) and uptake by CHO-CD14 cells involving another heat-labile serum factor(s) and cell-associated recognition molecule(s). Core-specific mAb inhibited mCD14-mediated uptake of homologous and heterologous LPS, while producing less concurrent enhancement of non-mCD14-mediated LPS uptake. The modulation by anti-LPS mAbs of mCD14-mediated LPS uptake was associated with inhibition of LPS-induced nuclear factor-kappaB (NF-kappaB) translocation and TNF-alpha secretion in CHO-CD14 cells and monocytes, respectively, while mAb enhancement of non-mCD14-mediated LPS uptake stimulated these activities. LPS-specific Abs thus mediate anti-inflammatory and proinflammatory functions, respectively, by preventing target cell uptake of LPS through mCD14 and augmenting uptake through CR1 or other cell receptors.
- Published
- 1997
37. The NF-beta A-binding element, not an overlapping NF-IL-6-binding element, is required for maximal IL-1 beta gene expression.
- Author
-
Buras JA, Monks BG, and Fenton MJ
- Subjects
- Animals, Base Sequence, CCAAT-Enhancer-Binding Proteins, Cell Line, DNA genetics, DNA Probes genetics, DNA-Binding Proteins genetics, Enhancer Elements, Genetic drug effects, Gene Expression, HeLa Cells, Humans, Lipopolysaccharides pharmacology, Mice, Molecular Sequence Data, Nuclear Proteins genetics, Promoter Regions, Genetic, Transcription Factors genetics, Transcription, Genetic, DNA-Binding Proteins metabolism, Interleukin-1 genetics, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
NF-beta A is a monocyte, neutrophil, and B cell-specific nuclear protein that is involved in regulation of the IL-1 beta gene. These studies further define the functional role of NF-beta A in RAW264.7 monocytic cells by using transient transfection analysis. We showed that NF-beta A was able to activate transcription from a heterologous promoter in a distance-independent and dose-dependent manner. NF-beta A also appeared to function in a positionally independent manner within the IL-1 beta cap-site proximal (CSP) promoter. NF-beta A was required for maximal IL-1 beta gene expression directed by the upstream LPS-inducible enhancer element. Deletion of the NF-beta A-binding sequence resulted in an 80% reduction in basal reporter gene activity and an 86% reduction in LPS-inducible reporter gene activity in constructs containing only the enhancer and CSP promoter. Other regulatory elements located between the enhancer and the cap site were not able to substitute functionally for the absence of NF-beta A. Recently, other investigators have reported that IL-1 beta CSP promoter function was decreased by introducing multiple mutations within both the NF-beta A-binding sequence, and a putative overlapping NF-IL-6-binding sequence. We have found that these mutations predominantly affect NF-beta A binding. Furthermore NF-beta A, and not NF-IL-6, was required for supporting basal and LPS-inducible transcription from a minimal IL-1 beta CSP promoter (positions -58 to +11). This promoter region did not appear to direct monocyte-specific IL-1 beta gene expression because reporter constructs containing the IL-1 beta CSP promoter were also active in transiently transfected HeLa cells.
- Published
- 1994
38. An upstream protein interacts with a distinct protein that binds to the cap site of the human interleukin 1 beta gene.
- Author
-
Monks BG, Martell BA, Buras JA, and Fenton MJ
- Subjects
- Animals, Base Sequence, Binding, Competitive, Cells, Cultured, DNA genetics, DNA metabolism, Humans, Mice, Molecular Sequence Data, NF-kappa B metabolism, Protein Binding, DNA-Binding Proteins metabolism, Interleukin-1 genetics, Nuclear Proteins metabolism, Promoter Regions, Genetic
- Abstract
Interleukin 1 beta (IL-1 beta) is a proinflammatory cytokine that exhibits a wide variety of biological activities. Genomic sequences that mediate the induction of human IL-1 beta gene transcription by lipopolysaccharide and phorbol esters are located more than 2,700 bp upstream of the transcriptional start site (cap site). These upstream elements require additional cap site-proximal (CSP) sequences which are necessary for basal transcription of the human IL-1 beta gene. In addition, these CSP sequences have been shown to mediate both cell type-specific expression of this gene, and trans-activation by some viral proteins. In this study, we report the identification of a novel nuclear protein, termed NF beta C, that binds to a DNA sequence which spans the cap site of the human IL-1 beta gene (positions -12 to +8). We have also identified a second region (positions -305 to -280) containing a putative NF-kappa B binding site. We show here that this region can bind three distinct nuclear proteins. One protein is similar or identical to NF-kappa B, a second protein (termed NF beta B) binds a distinct sequence that substantially overlaps the 5' half of the NF kappa B binding sequence, and a third protein (termed NF beta D) binds a distinct sequence that substantially overlaps the 3' half of the NF kappa B binding sequence. Unlike NF kappa B, NF1 beta B and NF beta D are present in nuclear extracts prepared from unstimulated monocytic cells. Although the NF beta D and NF beta C binding sequences share no significant similarity, each sequence can specifically compete for the binding of either protein to DNA, whereas oligonucleotides containing only the NF kappa B or NF beta B motifs do not compete for the binding of NF beta C or NF beta D. This suggests that NF beta C and NF beta D can specifically interact in vitro, possibly through a common subunit.
- Published
- 1994
- Full Text
- View/download PDF
39. The functional importance of a cap site-proximal region of the human prointerleukin 1 beta gene is defined by viral protein trans-activation.
- Author
-
Hunninghake GW, Monks BG, Geist LJ, Monick MM, Monroy MA, Stinski MF, Webb AC, Dayer JM, Auron PE, and Fenton MJ
- Subjects
- Animals, Base Sequence, Cell Line, Chloramphenicol O-Acetyltransferase genetics, Chloramphenicol O-Acetyltransferase metabolism, DNA Probes, Exons, Humans, Methylation, Mice, Molecular Sequence Data, Regulatory Sequences, Nucleic Acid, Restriction Mapping, Retroviridae Proteins, Oncogenic, Sequence Homology, Nucleic Acid, TATA Box, Transfection, DNA-Binding Proteins metabolism, Interleukin-1 genetics, Promoter Regions, Genetic, Protein Precursors genetics, Transcription, Genetic, Transcriptional Activation
- Abstract
Prointerleukin 1 beta (IL-1 beta) is a cytokine that mediates a broad range of biological activities. Genomic sequences that regulate IL-1 beta transcription include both inducible regulatory elements located more than 2,700 bp upstream of the transcriptional start site (cap site) and proximal elements located near the TATA box of this gene. In this study, we focused on the identification and characterization of trans-acting nuclear regulatory proteins that bind to the cap site-proximal region of the human IL-1 beta gene. We identified a protein, termed NFIL-1 beta A (NF beta A), that binds to a highly conserved 12-bp DNA sequence (-49 to -38) located upstream of the TATA box motif in both the human and murine IL-1 beta genes. The IL-1 alpha gene, which lacks a TATA motif, does not possess an NF beta A-binding sequence within the promoter region, suggesting that NF beta A may selectively regulate IL-1 beta expression. Using electrophoretic mobility shift assays, we identified several distinct DNA-protein complexes that are expressed in a cell-type-specific manner. In monocytic cell lines, the relative abundance of these complexes varies rapidly following stimulation of the cells with phorbol esters or lipopolysaccharide. UV cross-linking analysis identified two distinct DNA-binding polypeptides that comprise distinct complexes. The functional role of NF beta A was assessed in transient transfection assays. These data indicate that NF beta A is required for both basal and inducible promoter activity in monocytic cells. Furthermore, the human cytomegalovirus immediate-early 1 gene product requires the presence of NF beta A in order to trans-activate the proximal IL-1 beta promoter in a monocytic cell line. We propose that NF beta A is a factor that mediates either direct or indirect activation by the immediate-early 1 gene product. The proximity of this essential factor to the TATA motif suggests a possible role in transcriptional initiation.
- Published
- 1992
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.