Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
Add more filters










Publication year range
1.
Parasite Immunol ; 33(4): 217-25, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21204848

ABSTRACT

Enterocytozoon bieneusi is clinically the most significant microsporidian parasite associated with persistent diarrhoea, wasting and cholangitis in 30-50% of individuals with HIV/AIDS, as well as in malnutritional children and in the recipients of immunosuppressive therapy. However, the host immune responses to E. bieneusi have not been investigated until recently because of lack of sources of spores, cell culture system and animal models. In this study, we purified spores from heavily infected human or monkey faeces by serial salt-Percoll-sucrose-iodixanol centrifugation, and the purity of spores was confirmed by FACS and scanning electron microscopy. Exposure of dendritic cells to E. bieneusi spores induced the upregulation of the surface markers and production of pro-inflammatory cytokines. The cytokine production was independent of toll-like receptor 4, but MyD88 dependent, because dendritic cells from MyD88 knockout mice failed to secrete these pro-inflammatory cytokines, whereas dendritic cells from C3H/HeJ (a toll-like receptor 4 mutant) were activated by E. bieneusi and secreted these cytokines. Furthermore, MyD88-deficient mice were susceptible to E. bieneusi infection, in contrast to wild-type mice that resisted the infection. Collectively, the data demonstrate innate recognition of E. bieneusi by dendritic cells and the importance of MyD88-dependent signalling in resisting infection in a murine challenge model.


Subject(s)
Cytokines/biosynthesis , Enterocytozoon/immunology , Immunity, Innate , Microsporidiosis/immunology , Myeloid Differentiation Factor 88/immunology , Animals , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disease Models, Animal , Enterocytozoon/isolation & purification , Feces/microbiology , Haplorhini , Humans , Mice , Mice, Inbred C3H , Mice, Knockout , Myeloid Differentiation Factor 88/genetics
2.
J Immunol ; 167(12): 7069-76, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11739528

ABSTRACT

Group B streptococcus (GBS) imposes a major health threat to newborn infants. Little is known about the molecular basis of GBS-induced sepsis. Both heat-inactivated whole GBS bacteria and a heat-labile soluble factor released by GBS during growth (GBS-F) induce nuclear translocation of NF-kappaB, the secretion of TNF-alpha, and the formation of NO in mouse macrophages. Macrophages from mice with a targeted disruption of MyD88 failed to secrete TNF-alpha in response to both heat-inactivated whole bacteria and GBS-F, suggesting that Toll-like receptors (TLRs) are involved in different aspects of GBS recognition. Immune cell activation by whole bacteria differed profoundly from that by secreted GBS-F. Whole GBS activated macrophages independently of TLR2 and TLR6, whereas a response to the secreted GBS-F was not observed in macrophages from TLR2-deficient animals. In addition to TLR2, TLR6 and CD14 expression were essential for GBS-F responses, whereas TLR1 and TLR4 or MD-2 did not appear to be involved. Heat lability distinguished GBS-F from peptidoglycan and lipoproteins. GBS mutants deficient in capsular polysaccharide or beta-hemolysin had GBS-F activity comparable to that of wild-type streptococci. We suggest that CD14 and TLR2 and TLR6 function as coreceptors for secreted microbial products derived from GBS and that cell wall components of GBS are recognized by TLRs distinct from TLR1, 2, 4, or 6.


Subject(s)
Drosophila Proteins , Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Interleukin-1 , Streptococcus agalactiae/physiology , Animals , Antigens, Surface/physiology , Biological Factors/metabolism , CHO Cells , Carrier Proteins/genetics , Carrier Proteins/physiology , Cells, Cultured , Cricetinae , Humans , Inflammation Mediators/metabolism , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Models, Immunological , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Sepsis/immunology , Streptococcal Infections/immunology , Toll-Like Receptor 1 , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptor 6 , Toll-Like Receptors , Transfection , Tumor Necrosis Factor-alpha/biosynthesis
4.
J Immunol ; 167(4): 2257-67, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11490013

ABSTRACT

Down-regulation of cell surface expression of Toll-like receptor (TLR) 4 following LPS stimulation has been suggested to underlie endotoxin tolerance. In this study, we examined whether overexpression of TLR2 or TLR4 would affect the ability of cells to become tolerant to LPS or the mycobacterial components, arabinose-capped lipoarabinomannan (LAM) and soluble tuberculosis factor (STF). To this end, Chinese hamster ovary/CD14 cells stably transfected with a NF-kappaB-dependent reporter construct, endothelial leukocyte adhesion molecule CD25 (the 3E10 clone), were engineered to overexpress either human TLR2 or TLR4. Transfected TLRs exhibited proper signaling functions, as evidenced by increased LPS responsiveness of 3E10/TLR4 cells and acquisition of sensitivity to TLR2-specific ligands upon transfection of TLR2 into TLR2-negative 3E10 cells. Pretreatment of cells with LPS, LAM, or STF did not modulate TLR2 or TLR4 cell surface expression. Following LPS exposure, 3E10, 3E10/TLR2, and 3E10/TLR4 cells exhibited comparable decreases in LPS-mediated NF-kappaB activation and mitogen-activated protein (MAP) kinase phosphorylation. Likewise, LPS pretreatment profoundly inhibited LPS-induced NF-kappaB translocation in Chinese hamster ovary cells that concomitantly overexpressed human TLR4 and myeloid differentiation protein-2 (MD-2), but failed to modulate TLR4 or MD-2 cell surface expression. Pretreatment of 3E10/TLR2 cells with LAM or STF decreased their NF-kappaB responses induced by subsequent stimulation with these substances or LPS. Conversely, prior exposure of 3E10/TLR2 cells to LPS led to hyporesponsiveness to LPS, LAM, and STF, indicating that LPS and mycobacterial products induce cross-tolerance. Thus, tolerance to LPS and mycobacterial components cannot be attributed solely to a decrease in TLR/MD-2 expression levels, suggesting inhibition of expression or function of other signaling intermediates.


Subject(s)
Antigens, Bacterial/immunology , CHO Cells/immunology , Drosophila Proteins , Immune Tolerance , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharides/immunology , Membrane Glycoproteins/biosynthesis , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/biosynthesis , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , CHO Cells/metabolism , Cell Line , Clone Cells , Cricetinae , Enzyme Activation/genetics , Enzyme Activation/immunology , Humans , Immune Tolerance/genetics , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/antagonists & inhibitors , Lymphocyte Antigen 96 , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Receptors, Cell Surface/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection
5.
Circ Res ; 89(3): 244-50, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11485974

ABSTRACT

An early component of atherogenesis is abnormal vascular smooth muscle cell (VSMC) proliferation. The presence of Chlamydia pneumoniae in many atherosclerotic lesions raises the possibility that this organism plays a causal role in atherogenesis. In this study, C pneumoniae elementary bodies (EBs) rapidly activated p44/p42 mitogen-activated protein kinases (MAPKs) and stimulated proliferation of VSMCs in vitro. Exposure of VSMCs derived from human saphenous vein to C pneumoniae EBs (3x10(7) inclusion forming units/mL) enhanced bromodeoxyuridine (BrdU) incorporation 12+/-3-fold. UV- and heat-inactivated C pneumoniae EBs also stimulated VSMC proliferation, indicating a role of direct stimulation by chlamydial antigens. However, the mitogenic activity of C pneumoniae was heat-labile, thus excluding a role of lipopolysaccharide. Chlamydial hsp60 (25 microg/mL) replicated the effect of C pneumoniae, stimulating BrdU incorporation 7+/-3-fold. Exposure to C pneumoniae or chlamydial hsp60 rapidly activated p44/p42 MAPK, within 5 to 10 minutes of exposure. In addition, PD98059 and U0126, which are two distinct inhibitors of upstream MAPK kinase 1/2 (MEK1/2), abolished the mitogenic effect of C pneumoniae and chlamydial hsp60. Toll-like receptors (TLRs) act as sensors for microbial antigens and can signal via the p44/p42 MAPK pathway. Human VSMCs were shown to express TLR4 mRNA and protein, and a TLR4 antagonist abolished chlamydial hsp60-induced VSMC proliferation and attenuated C pneumoniae-induced MAPK activation and VSMC proliferation. Together these results indicate that C pneumoniae and chlamydial hsp60 are potent inducers of human VSMC proliferation and that these effects are mediated, at least in part, by rapid TLR4-mediated activation of p44/p42 MAPK.


Subject(s)
Chaperonin 60/metabolism , Drosophila Proteins , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Cell Surface/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Bromodeoxyuridine , Butadienes/pharmacology , Cell Division/drug effects , Cells, Cultured , Chaperonin 60/antagonists & inhibitors , Chaperonin 60/pharmacology , Chlamydophila Infections/metabolism , Chlamydophila pneumoniae/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Hot Temperature , Humans , Lipid A/analogs & derivatives , Lipid A/pharmacology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Mitogen-Activated Protein Kinase 3 , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/microbiology , Nitriles/pharmacology , RNA, Messenger/biosynthesis , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Reverse Transcriptase Polymerase Chain Reaction , Saphenous Vein , Toll-Like Receptor 4 , Toll-Like Receptors
6.
J Immunol ; 167(2): 910-8, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11441098

ABSTRACT

Mycobacterium tuberculosis (MTB) induces vigorous immune responses, yet persists inside macrophages, evading host immunity. MTB bacilli or lysate was found to inhibit macrophage expression of class II MHC (MHC-II) molecules and MHC-II Ag processing. This report characterizes and identifies a specific component of MTB that mediates these inhibitory effects. The inhibitor was extracted from MTB lysate with Triton X-114, isolated by gel electroelution, and identified with Abs to be MTB 19-kDa lipoprotein. Electroelution- or immunoaffinity-purified MTB 19-kDa lipoprotein inhibited MHC-II expression and processing of both soluble Ags and Ag 85B from intact MTB bacilli. Inhibition of MHC-II Ag processing by either MTB bacilli or purified MTB 19-kDa lipoprotein was dependent on Toll-like receptor (TLR) 2 and independent of TLR 4. Synthetic analogs of lipopeptides from Treponema pallidum also inhibited Ag processing. Despite the ability of MTB 19-kDa lipoprotein to activate microbicidal and innate immune functions early in infection, TLR 2-dependent inhibition of MHC-II expression and Ag processing by MTB 19-kDa lipoprotein during later phases of macrophage infection may prevent presentation of MTB Ags and decrease recognition by T cells. This mechanism may allow intracellular MTB to evade immune surveillance and maintain chronic infection.


Subject(s)
Acyltransferases , Antigen Presentation/immunology , Antigens, Bacterial , Bacterial Proteins/pharmacology , Drosophila Proteins , Histocompatibility Antigens Class II/biosynthesis , Immunosuppressive Agents/pharmacology , Lipoproteins/pharmacology , Macrophages/immunology , Membrane Glycoproteins/physiology , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/physiology , Animals , Antigen Presentation/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/isolation & purification , Bacterial Proteins/metabolism , Detergents , Epitopes/metabolism , Female , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Macrophages/metabolism , Macrophages/microbiology , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Octoxynol , Polyethylene Glycols , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors
7.
J Exp Med ; 194(1): 79-88, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11435474

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.


Subject(s)
Antigens, Surface/genetics , Drosophila Proteins , Endotoxins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , 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
8.
J Immunol ; 167(1): 416-23, 2001 Jul 01.
Article in English | MEDLINE | ID: mdl-11418678

ABSTRACT

Glycosylphosphatidylinositol (GPI) anchors and glycoinositolphospholipids (GIPLs) from parasitic protozoa have been shown to exert a wide variety of effects on cells of the host innate immune system. However, the receptor(s) that are triggered by these protozoan glycolipids has not been identified. Here we present evidence that Trypanosoma cruzi-derived GPI anchors and GIPLs trigger CD25 expression on Chinese hamster ovary-K1 cells transfected with CD14 and Toll-like receptor-2 (TLR-2), but not wild-type (TLR-2-deficient) Chinese hamster ovary cells. The protozoan-derived GPI anchors and GIPLs containing alkylacylglycerol and saturated fatty acid chains or ceramide were found to be active in a concentration range of 100 nM to 1 microM. More importantly, the GPI anchors purified from T. cruzi trypomastigotes, which contain a longer glycan core and unsaturated fatty acids in the sn-2 position of the alkylacylglycerolipid component, triggered TLR-2 at subnanomolar concentrations. We performed experiments with macrophages from TLR-2 knockout and TLR-4 knockout mice, and found that TLR-2 expression appears to be essential for induction of IL-12, TNF-alpha, and NO by GPI anchors derived from T. cruzi trypomastigotes. Thus, highly purified GPI anchors from T. cruzi parasites are potent activators of TLR-2 from both mouse and human origin. The activation of TLR-2 may initiate host innate defense mechanisms and inflammatory response during protozoan infection, and may provide new strategies for immune intervention during protozoan infections.


Subject(s)
Drosophila Proteins , Glycosylphosphatidylinositols/physiology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Trypanosoma cruzi/immunology , Animals , CHO Cells , Cell Line , Cricetinae , Dose-Response Relationship, Immunologic , Glycolipids/physiology , Glycosylphosphatidylinositols/isolation & purification , Inflammation/immunology , Inflammation/parasitology , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , NF-kappa B/physiology , Phospholipids/physiology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Receptors, Interleukin-2/biosynthesis , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection , Trypanosoma cruzi/chemistry , Trypanosoma cruzi/growth & development
9.
Infect Immun ; 69(4): 2230-6, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11254578

ABSTRACT

The recent isolation of a lipopolysaccharide (LPS)-deficient mutant of Neisseria meningitidis has allowed us to explore the roles of other gram-negative cell wall components in the host response to infection. The experiments in this study were designed to examine the ability of this mutant strain to activate cells. Although it was clearly less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in monocytic cells, inducing a response similar to that seen with Staphylococcus aureus. Cellular activation by the LPS mutant was related to expression of CD14, a high-affinity receptor for LPS and other microbial products, as well as Toll-like receptor 2, a member of the Toll family of receptors recently implicated in host responses to gram-positive bacteria. In contrast to the parental strain, the synthetic LPS antagonist E5564 did not inhibit the LPS-deficient mutant. We conclude that even in the absence of LPS, the gram-negative cell wall remains a potent inflammatory stimulant, utilizing signaling pathways independent of those involved in LPS signaling.


Subject(s)
Bacterial Proteins/physiology , Drosophila Proteins , Lipid A/analogs & derivatives , Lipopolysaccharides/toxicity , Membrane Glycoproteins/physiology , Membrane Proteins/physiology , Neisseria meningitidis/immunology , Receptors, Cell Surface/physiology , Animals , CHO Cells , Cell Wall/physiology , Cricetinae , Humans , Lipid A/pharmacology , Lipopolysaccharide Receptors/physiology , Mutation , Toll-Like Receptor 2 , Toll-Like Receptors , Tumor Necrosis Factor-alpha/metabolism
10.
J Immunol ; 166(7): 4620-6, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11254720

ABSTRACT

Toll-like receptors (TLR) 2 and 4 are cell surface receptors that in association with CD14 enable phagocytic inflammatory responses to a variety of microbial products. Activation via these receptors triggers signaling cascades, resulting in nuclear translocation of NF-kappa B and a proinflammatory response including TNF-alpha production. We investigated whether TLRs participate in the host response to Cryptococcus neoformans glucuronoxylomannan (GXM), the major capsular polysaccharide of this fungus. Chinese hamster ovary fibroblasts transfected with human TLR2, TLR4, and/or CD14 bound fluorescently labeled GXM. The transfected Chinese hamster ovary cells were challenged with GXM, and activation of an NF-kappa B-dependent reporter construct was evaluated. Activation was observed in cells transfected with both CD14 and TLR4. GXM also stimulated nuclear NF-kappa B translocation in PBMC and RAW 264.7 cells. However, stimulation of these cells with GXM resulted in neither TNF-alpha secretion nor activation of the extracellular signal-regulated kinase 1/2, p38, and stress-activated protein kinase/c-Jun N-terminal kinase mitogen-activated protein kinase pathways. These findings suggest that TLRs, in conjunction with CD14, function as pattern recognition receptors for GXM. Furthermore, whereas GXM stimulates cells to translocate NF-kappa B to the nucleus, it does not induce activation of mitogen-activated protein kinase pathways or release of TNF-alpha. Taken together, these observations suggest a novel scenario whereby GXM stimulates cells via CD14 and TLR4, resulting in an incomplete activation of pathways necessary for TNF-alpha production.


Subject(s)
Cryptococcus neoformans/immunology , Drosophila Proteins , Membrane Glycoproteins/physiology , Polysaccharides/immunology , Receptors, Cell Surface/physiology , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism , Active Transport, Cell Nucleus/immunology , Animals , Binding Sites/genetics , Binding Sites/immunology , CHO Cells , Cell Line , Cricetinae , Gene Expression Regulation/immunology , Humans , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lipopolysaccharide Receptors/physiology , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Membrane Glycoproteins/genetics , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Polysaccharides/metabolism , Polysaccharides/physiology , Receptors, Cell Surface/genetics , Signal Transduction/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
12.
J Immunol ; 166(6): 4074-82, 2001 Mar 15.
Article in English | MEDLINE | ID: mdl-11238656

ABSTRACT

We previously showed that viable Mycobacterium tuberculosis (Mtb) bacilli contain distinct ligands that activate cells via the mammalian Toll-like receptor (TLR) proteins TLR2 and TLR4. We now demonstrate that expression of a dominant negative TLR2 or TLR4 proteins in RAW 264.7 macrophages partially blocked Mtb-induced NF-kappa B activation. Coexpression of both dominant negative proteins blocked virtually all Mtb-induced NF-kappa B activation. The role of the TLR4 coreceptor MD-2 was also examined. Unlike LPS, Mtb-induced macrophage activation was not augmented by overexpression of ectopic MD-2. Moreover, cells expressing an LPS-unresponsive MD-2 mutant responded normally to Mtb. We also observed that the lipid A-like antagonist E5531 specifically inhibited TLR4-dependent Mtb-induced cellular responses. E5531 could substantially block LPS- and Mtb-induced TNF-alpha production in both RAW 264.7 cells and primary human alveolar macrophages (AM phi). E5531 inhibited Mtb-induced AM phi apoptosis in vitro, an effect that was a consequence of the inhibition of TNF-alpha production by E5531. In contrast, E5531 did not inhibit Mtb-induced NO production in RAW 264.7 cells and AM phi. Mtb-stimulated peritoneal macrophages from TLR2- and TLR4-deficient animals produced similar amounts of NO compared with control animals, demonstrating that these TLR proteins are not required for Mtb-induced NO production. Lastly, we demonstrated that a dominant negative MyD88 mutant could block Mtb-induced activation of the TNF-alpha promoter, but not the inducible NO synthase promoter, in murine macrophages. Together, these data suggest that Mtb-induced TNF-alpha production is largely dependent on TLR signaling. In contrast, Mtb-induced NO production may be either TLR independent or mediated by TLR proteins in a MyD88-independent manner.


Subject(s)
Drosophila Proteins , Lipid A/pharmacology , Macrophages/microbiology , Membrane Glycoproteins/antagonists & inhibitors , Mycobacterium tuberculosis/physiology , Receptors, Cell Surface/antagonists & inhibitors , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/physiology , Antitubercular Agents/pharmacology , Apoptosis/drug effects , CHO Cells , Cell Line , Cricetinae , Cricetulus , Female , Gene Expression Regulation , Lipid A/analogs & derivatives , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/pharmacology , Lymphocyte Antigen 96 , Macrophages/drug effects , Macrophages/metabolism , Macrophages, Alveolar/cytology , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/microbiology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/pharmacology , Membrane Glycoproteins/physiology , Mesocricetus , Mice , Mice, Inbred C3H , Mutation , Mycobacterium tuberculosis/drug effects , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Promoter Regions, Genetic/immunology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/physiology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Tuberculosis/mortality , Tuberculosis/prevention & control , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
13.
J Biol Chem ; 276(3): 1873-80, 2001 Jan 19.
Article in English | MEDLINE | ID: mdl-11032843

ABSTRACT

ER-112022 is a novel acyclic synthetic lipid A analog that contains six symmetrically organized fatty acids on a noncarbohydrate backbone. Chinese hamster ovary (CHO)-K1 fibroblasts and U373 human astrocytoma cells do not respond to lipopolysaccharide (LPS) in the absence of CD14. In contrast, exposure to ER-112022 effectively induced activation of CHO and U373 cells under serum-free conditions. Expression of CD14 was not necessary for cells to respond to ER-112022, although the presence of soluble CD14 enhanced the sensitivity of the response. Several lines of evidence suggested that ER-112022 stimulates cells via the LPS signal transduction pathway. First, the diglucosamine-based LPS antagonists E5564 and E5531 blocked ER-112022-induced stimulation of CHO-K1, U373, and RAW264.7 cells. Second, ER-112022 was unable to activate C3H/HeJ mouse peritoneal macrophages, containing a mutation in Toll-like receptor (TLR) 4, as well as HEK293 cells, an epithelial cell line that does not express TLR4. Third, ER-112022 activated NF-kappaB in HEK293 cells transfected with TLR4/MD-2. Finally, tumor necrosis factor release from primary human monocytes exposed to ER-112022 was blocked by TLR4 antibodies but not by TLR2 antibodies. Our results suggest that ER-112022 and the family of lipid A-like LPS antagonists can functionally associate with TLR4 in the absence of CD14. Synthetic molecules like ER-112022 may prove to be valuable tools to characterize elements in the LPS receptor complex, as well as to activate or inhibit the TLR4 signaling pathway for therapeutic purposes.


Subject(s)
Drosophila Proteins , Lipid A/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Carbohydrate Conformation , Cell Line , Lipid A/chemistry , Lipopolysaccharides/chemistry , Phospholipids/pharmacology , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection
14.
Infect Immun ; 68(12): 6770-6, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11083794

ABSTRACT

Lipopolysaccharide (LPS) and related bacterial products can be recognized by host inflammatory cells in a particulate, bacterium-bound form, as well as in various soluble, released forms. In the present study we have compared the mechanisms used by LPS, detoxified LPS (DLPS), and mannuronic acid polymers (M-polymers), in solution or covalently linked to particles, in stimulating monocytes to tumor necrosis factor (TNF) production. The addition of recombinant LPS binding protein (LBP) and/or soluble CD14 (sCD14) enhanced the production of TNF from monocytes stimulated with soluble LPS, DLPS, or M-polymer, but did not affect the response to M-polymer or DLPS attached to particles. Treatment of monocytes with antibody to CD14, CD18, or CD11b showed that CD14, but not CR3 (CD11b/CD18), mediated monocyte TNF production in response to the soluble antigens. In contrast, anti-CD14, anti-CD11b and anti-CD18 monoclonal antibodies all inhibited the response to the particulate stimuli. On the other hand, B975, a synthetic analog of Rhodobacter capsulatus lipid A, completely abrogated the monocyte TNF response induced by LPS but did not affect the TNF induction by DLPS or M-polymer, either in soluble or particulate forms. These data demonstrate that the engagement of immune receptors by bacterial products such as LPS, DLPS, and M-polymer is dependent upon the presentation form of their constituent carbohydrates, and that factors such as aggregation state, acylation, carbohydrate chain length, and solid versus liquid phase of bacterial ligands influence the mechanisms used by cells in mediating proinflammatory responses.


Subject(s)
Acute-Phase Proteins , Alginates/pharmacology , CD18 Antigens/physiology , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins , Animals , CHO Cells , Carrier Proteins/physiology , Cricetinae , Glucuronic Acid , Hexuronic Acids , Macrophage-1 Antigen/physiology , Monocytes/drug effects , Tumor Necrosis Factor-alpha/biosynthesis
15.
J Immunol ; 165(8): 4272-80, 2000 Oct 15.
Article in English | MEDLINE | ID: mdl-11035061

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.


Subject(s)
Escherichia coli/immunology , Gene Deletion , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , 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
16.
Cytokine Growth Factor Rev ; 11(3): 219-32, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10817965

ABSTRACT

In 1997, a human homologue of the Drosophila Toll protein was described, a protein later to be designated Toll-like receptor 4 (TLR4). Since that time, additional human and murine TLR proteins have been identified. Mammalian TLR proteins appear to represent a conserved family of innate immune recognition receptors. These receptors are coupled to a signaling pathway that is conserved in mammals, insects, and plants, resulting in the activation of genes that mediate innate immune defenses. Numerous studies have now identified a wide variety of chemically-diverse bacterial products that serve as putative ligands for TLR proteins. More recent studies have identified the first endogenous protein ligands for TLR proteins. TLR signaling represents a key feature of innate immune response to pathogen invasion.


Subject(s)
Drosophila Proteins , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Drosophila/genetics , Drosophila/metabolism , Humans , Insect Proteins/physiology , Ligands , Lipopolysaccharides/chemistry , Mammals , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , Mice, Knockout , Molecular Sequence Data , Receptors, Cell Surface/genetics , Sequence Homology, Amino Acid , Toll-Like Receptor 4 , Toll-Like Receptor 5 , Toll-Like Receptor 6 , Toll-Like Receptors
17.
J Immunol ; 164(4): 2064-9, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657659

ABSTRACT

Human Toll like receptor (TLR) 2 has been implicated as a signaling receptor for LPS from Gram-negative bacteria and cell wall components from Gram-positive organisms. In this study, we investigated whether TLR2 can signal cell activation by the heat-killed group B streptococci type III (GBS) and Listeria monocytogenes (HKLM). HKLM, but not GBS, showed a time- and dose-dependent activation of Chinese hamster ovary cells transfected with human TLR2, as measured by translocation of NF-kappaB and induction of IL-6 production. A mAb recognizing a TLR2-associated epitope (TL2.1) was generated that inhibited IL-6 production from Chinese hamster ovary-TLR2 cells stimulated with HKLM or LPS. The TL2.1 mAb reduced HKLM-induced TNF production from human monocytes by 60%, whereas a CD14 mAb (3C10) reduced the TNF production by 30%. However, coadministrating TL2.1 and 3C10 inhibited the TNF response by 80%. In contrast to this, anti-CD14 blocked LPS-induced TNF production from monocytes, whereas anti-TLR2 showed no inhibition. Neither TL2.1 nor 3C10 affected GBS-induced TNF production. These results show that TLR2 can function as a signaling receptor for HKLM, possibly together with CD14, but that TLR2 is unlikely to be involved in cell activation by GBS. Furthermore, although LPS can activate transfected cell lines through TLR2, this receptor does not seem to be the main transducer of LPS activation of human monocytes. Thus, our data demonstrate the ability of TLR2 to distinguish between different pathogens.


Subject(s)
Drosophila Proteins , Lipopolysaccharides/immunology , Listeria monocytogenes/immunology , Membrane Glycoproteins/physiology , Monocytes/immunology , Monocytes/microbiology , Receptors, Cell Surface/physiology , Streptococcus agalactiae/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antigen-Antibody Reactions , CHO Cells , Cricetinae , Dose-Response Relationship, Immunologic , Epitopes/immunology , Hot Temperature , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/antagonists & inhibitors , Macrophage Activation , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , NF-kappa B/metabolism , Receptors, Cell Surface/immunology , Toll-Like Receptor 2 , Toll-Like Receptors , Transfection
18.
J Clin Invest ; 105(4): 497-504, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10683379

ABSTRACT

Lipopolysaccharide (LPS) is the main inducer of shock and death in Gram-negative sepsis. Recent evidence suggests that LPS-induced signal transduction begins with CD14-mediated activation of 1 or more Toll-like receptors (TLRs). The lipid A analogues lipid IVa and Rhodobacter sphaeroides lipid A (RSLA) exhibit an uncommon species-specific pharmacology. Both compounds inhibit the effects of LPS in human cells but display LPS-mimetic activity in hamster cells. We transfected human TLR4 or human TLR2 into hamster fibroblasts to determine if either of these LPS signal transducers is responsible for the species-specific pharmacology. RSLA and lipid IVa strongly induced NF-kappaB activity and IL-6 release in Chinese hamster ovary fibroblasts expressing CD14 (CHO/CD14), but these compounds antagonized LPS antagonists in CHO/CD14 fibroblasts that overexpressed human TLR4. No such antagonism occurred in cells overexpressing human TLR2. We cloned TLR4 from hamster macrophages and found that human THP-1 cells expressing the hamster TLR4 responded to lipid IVa as an LPS mimetic, as if they were hamster in origin. Hence, cells heterologously overexpressing TLR4 from different species acquired a pharmacological phenotype with respect to recognition of lipid A substructures that corresponded to the species from which the TLR4 transgene originated. These data suggest that TLR4 is the central lipid A-recognition protein in the LPS receptor complex.


Subject(s)
Drosophila Proteins , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/metabolism , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Animals , CHO Cells , Cricetinae , Glycolipids/metabolism , Humans , Ligands , Lipid A/analogs & derivatives , Lipid A/metabolism , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/antagonists & inhibitors , Macrophages/cytology , Macrophages/metabolism , Membrane Glycoproteins/genetics , Molecular Mimicry , Receptors, Cell Surface/genetics , Recombinant Proteins/metabolism , Rhodobacter sphaeroides , Signal Transduction , Species Specificity , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors
19.
Life Sci ; 68(3): 241-58, 2000 Dec 08.
Article in English | MEDLINE | ID: mdl-11191641

ABSTRACT

Beginning in 1997 with the identification of the first human homologue of the Drosophila protein Toll, a family of related molecules have been identified in both humans and other mammals. These Toll-like receptor (TLR) proteins appear to represent a conserved family of innate immune recognition receptors. TLR proteins share extended homology with receptors for the cytokines interleukin 1 (IL-1) and interleukin 18 (IL-18). These receptors are coupled to a signaling pathway that is conserved in mammals, insects, and plants, resulting in cellular activation, thereby stimulating innate immune defenses. A variety of bacterial and fungal products have been identified that serve as TLR ligands, and more recent studies have identified the first endogenous protein ligands for TLR proteins. While TLR signaling is likely to be a key feature of innate immune responses, these proteins may also regulate homeostasis via interaction with endogenous protein ligands.


Subject(s)
Drosophila Proteins , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/physiology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/physiology , Amino Acid Sequence , Animals , Drosophila , Humans , Interleukin-18 Receptor alpha Subunit , Mice , Molecular Sequence Data , Receptors, Interleukin/chemistry , Receptors, Interleukin/physiology , Receptors, Interleukin-1/chemistry , Receptors, Interleukin-1/physiology , Receptors, Interleukin-18 , Sequence Homology, Amino Acid , Signal Transduction , Toll-Like Receptor 5 , Toll-Like Receptors
20.
J Endotoxin Res ; 6(5): 411-5, 2000.
Article in English | MEDLINE | ID: mdl-11521065

ABSTRACT

The inflammatory response to bacterial infections plays an important role in the detection and elimination of invading micro-organisms. Various components of the bacterial cell wall are capable of activating this pro-inflammatory response. In the case of Gram-negative bacteria, lipopolysaccharide (LPS) is the dominant trigger, although other bacterial factors are also capable of activating this systemic inflammatory response. Recently, Toll-like receptors (TLRs) have been implicated in host responses to bacterial pathogens. Specifically, TLR4 mediates LPS responses while TLR2 plays a broader role in the recognition of a variety of bacteria and bacterial antigens. The experiments in this study were designed to examine the role of Gram-negative cell wall components, other than LPS, and their cellular receptors in the host response to infection using an LPS-deficient mutant of Neisseria meningitidis. Although less potent than the parental strain, we found the LPS-deficient mutant to be a capable inducer of the inflammatory response in a variety of cell types. Moreover, cellular activation by this mutant required expression of CD14 and TLR2.


Subject(s)
Drosophila Proteins , Lipopolysaccharides/immunology , Membrane Glycoproteins/immunology , Neisseria meningitidis/immunology , Receptors, Cell Surface/immunology , Animals , CHO Cells , Cell Wall/immunology , Cricetinae , Humans , In Vitro Techniques , Inflammation Mediators/immunology , Leukocytes, Mononuclear/immunology , Lipopolysaccharide Receptors/genetics , Membrane Glycoproteins/genetics , Mutation , Neisseria meningitidis/genetics , Receptors, Cell Surface/genetics , Toll-Like Receptor 2 , Toll-Like Receptor 4 , Toll-Like Receptors , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...