Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Immunol ; 372: 104483, 2022 02.
Article in English | MEDLINE | ID: mdl-35085880

ABSTRACT

The occurring in SR-A/CD204- or CD36-deficient mice increased susceptibility to infections with Staphylococcus aureus (Sa) had traditionally been ascribed to the impairment of macrophage-mediated phagocytosis, which is, however, inconsistent with low effectiveness of unopsonized Sa killing within macrophages and redundant roles of both receptors in this process. We have found that Sa-stimulated cytokine production in mouse macrophages seems to be exclusively mediated by TLR2, mainly from within endosomes in response to Sa-derived lipoteichoic acid. By driving endocytic trafficking of TLR2 and its ligands through the clathrin-dependent pathway, CD36 and SR-A sensitize macrophages to activation by Sa as well as regulate the type and amount of cytokines produced. Additionally, upon direct Sa binding, both receptors autonomously generate anti-inflammatory signaling. Consequently, the delayed induction of acute inflammation in knockout mice may allow for the initial, uncontrolled multiplication of bacteria, stimulating excessive, septic shock-inducing production of inflammatory cytokines in later stages of infection.


Subject(s)
CD36 Antigens/immunology , Cytokines/biosynthesis , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Scavenger Receptors, Class A/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Animals , CD36 Antigens/deficiency , CD36 Antigens/genetics , Endocytosis/immunology , Ligands , Lipopolysaccharide Receptors/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Pattern Recognition/immunology , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Signal Transduction/immunology , Toll-Like Receptor 2/immunology
2.
Cell Immunol ; 312: 42-50, 2017 02.
Article in English | MEDLINE | ID: mdl-27908440

ABSTRACT

Acting through CD14 and TLR4/MD-2, lipopolysaccharide (LPS) triggers strong pro-inflammatory activation of macrophages, which, if not appropriately controlled, may lead to lethal septic shock. Therefore, numerous mechanisms of negative regulation of responses to LPS exist, but whether they include down-regulation of LPS receptors is not clear. We have found that in J774 cells, the clathrin-dependent endocytic pathway enables activation of TRIF-dependent TLR4 signaling within endosomes, but is not associated with the down-regulation of TLR4 or CD14 surface expression. In contrast, lipid raft-dependent endocytosis negatively regulates the basal cell surface expression of LPS receptors and, consequently, responsiveness to LPS. Together with observations that treatments, known to selectively disrupt lipid rafts, do not inhibit LPS-stimulated cytokine production, our results suggest that lipid rafts may serve as sites in which LPS receptors are sorted for endocytosis, rather than being platforms for the assembly of TLR4-centered signaling complexes, as suggested previously.


Subject(s)
Lipopolysaccharide Receptors/metabolism , Macrophages/immunology , Membrane Microdomains/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Cell Line , Clathrin/metabolism , Cytokines/metabolism , Endocytosis , Gene Expression Regulation , Humans , Lipopolysaccharide Receptors/genetics , Lipopolysaccharides/immunology , Mice , Signal Transduction , Toll-Like Receptor 4/metabolism
3.
Immunol Cell Biol ; 95(5): 424-435, 2017 05.
Article in English | MEDLINE | ID: mdl-27826145

ABSTRACT

Scavenger receptor (SR)-mediated opsonin-independent phagocytosis of bacteria by macrophages has been suggested to represent an important, early mechanism of anti-bacterial host defense. However, although the ability to bind bacteria has been demonstrated to be a shared feature of all types of SRs, in many cases the evidence is limited to the demonstration of increased binding of killed, fluorescently labeled bacteria to non-phagocytic cells transfected with these receptors. We sought to verify the ability of SRs to mediate non-opsonic phagocytosis of live Escherichia coli (Ec) and Staphylococcus aureus (Sa), model species of Gram-negative and -positive bacteria, respectively, and to assess the relative contributions of different SRs expressed on murine macrophages in this process. We found that the class A SR SR-A/CD204 was the major receptor mediating phagocytosis of fluorescently labeled Sa, whereas different SRs had highly redundant roles in the phagocytosis of live Sa. Conversely, different SRs contributed to the phagocytosis of fluorescently labeled Ec. In comparison, phagocytosis of live Ec was of much lower magnitude and was selectively mediated by SR-A. These results question the use of fluorescently labeled bacteria as valid replacements for live bacteria. The low magnitude of opsonin-independent phagocytosis of Ec and unimpaired phagocytosis of Sa in SR-A- or CD36-deficient macrophages indicate that the defect in this process might not be responsible for the reported impaired bacteria clearance in mice deficient in these receptors. We postulate that this impairment might result to a larger extent from inhibition of intracellular bacteria killing caused by pro-inflammatory cytokines, produced in excessive amounts by SR-deficient cells in response to bacterial products.


Subject(s)
CD36 Antigens/metabolism , Fluorescent Dyes/metabolism , Macrophages/metabolism , Macrophages/microbiology , Microbial Viability , Phagocytosis , Receptors, Scavenger/metabolism , Animals , Escherichia coli/physiology , Mice, Inbred C57BL , Opsonin Proteins/metabolism , Recombinant Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcal Infections/pathology , Staphylococcus aureus/physiology
4.
Immunol Cell Biol ; 94(5): 525, 2016 05.
Article in English | MEDLINE | ID: mdl-27216566
5.
PLoS One ; 11(4): e0153558, 2016.
Article in English | MEDLINE | ID: mdl-27073833

ABSTRACT

Lipopolysaccharide (LPS) is the major pathogen-associated molecular pattern of Gram-negative bacterial infections, and includes smooth (S-LPS) and rough (R-LPS) chemotypes. Upon activation by LPS through CD14, TLR4/MD-2 heterodimers sequentially induce two waves of intracellular signaling for macrophage activation: the MyD88-dependent pathway from the plasma membrane and, following internalization, the TRIF-dependent pathway from endosomes. We sought to better define the role of scavenger receptors CD36 and CD204/SR-A as accessory LPS receptors that can contribute to pro-inflammatory and microbicidal activation of macrophages. We have found that CD36 differently regulates activation of mouse macrophages by S-LPS versus R-LPS. The ability of CD36 to substitute for CD14 in loading R-LPS, but not S-LPS onto TLR4/MD-2 allows CD14-independent macrophage responses to R-LPS. Conversely, S-LPS, but not R-LPS effectively stimulates CD14 binding to CD36, which favors S-LPS transfer from CD14 onto TLR4/MD-2 under conditions of low CD14 occupancy with S-LPS in serum-free medium. In contrast, in the presence of serum, CD36 reduces S-LPS binding to TLR4/MD-2 and the subsequent MyD88-dependent signaling, by mediating internalization of S-LPS/CD14 complexes. Additionally, CD36 positively regulates activation of TRIF-dependent signaling by both S-LPS and R-LPS, by promoting TLR4/MD-2 endocytosis. In contrast, we have found that SR-A does not function as a S-LPS receptor. Thus, by co-operating with CD14 in both R- and S-LPS loading onto TLR4/MD-2, CD36 can enhance the sensitivity of tissue-resident macrophages in detecting infections by Gram-negative bacteria. However, in later phases, following influx of serum to the infection site, the CD36-mediated negative regulation of MyD88-dependent branch of S-LPS-induced TLR4 signaling might constitute a mechanism to prevent an excessive inflammatory response, while preserving the adjuvant effect of S-LPS for adaptive immunity.


Subject(s)
CD36 Antigens/metabolism , Cytokines/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Animals , CD36 Antigens/genetics , Cell Membrane/metabolism , Lipopolysaccharide Receptors/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism
6.
Immunol Cell Biol ; 94(2): 164-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26215791

ABSTRACT

Janeway's pattern recognition theory holds that the immune system detects infection through a limited number of the so-called pattern recognition receptors (PRRs). These receptors bind specific chemical compounds expressed by entire groups of related pathogens, but not by host cells (pathogen-associated molecular patterns (PAMPs). In contrast, Matzinger's danger hypothesis postulates that products released from stressed or damaged cells have a more important role in the activation of immune system than the recognition of nonself. These products, named by analogy to PAMPs as danger-associated molecular patterns (DAMPs), are proposed to act through the same receptors (PRRs) as PAMPs and, consequently, to stimulate largely similar responses. Herein, I review direct and indirect evidence that contradict the widely accepted danger theory, and suggest that it may be false.


Subject(s)
Immune System , Immunity , Models, Immunological , Receptors, Pattern Recognition/immunology , Animals , Host-Pathogen Interactions , Humans
8.
PLoS One ; 10(4): e0123293, 2015.
Article in English | MEDLINE | ID: mdl-25849867

ABSTRACT

The initiation of adaptive immune responses to protein antigens has to be preceded by their uptake by antigen presenting cells and intracellular proteolytic processing. Paradoxically, endocytic receptors involved in antigen uptake do not bind the majority of proteins, which may be the main reason why purified proteins stimulate at most weak immune responses. A shared feature of different types of adjuvants, capable of boosting immunogenicity of protein vaccines, is their ability to induce acute inflammation, characterized by early influx of activated neutrophils. Neutrophils are also rapidly recruited to sites of tissue injury or infection. These cells are the source of potent oxidants, including hypochlorous acid (HOCl), causing oxidation of proteins present in inflammatory foci. We demonstrate that oxidation of proteins by endogenous, neutrophils-derived HOCl increases their immunogenicity. Upon oxidation, different, randomly chosen simple proteins (yeast alcohol dehydrogenase, human and bovine serum albumin) and glycoproteins (human apo-transferrin, ovalbumin) gain the ability to bind with high affinity to several endocytic receptors on antigen presenting cells, which seems to be the major mechanism of their increased immunogenicity. The mannose receptor (CD206), scavenger receptors A (CD204) and CD36 were responsible for the uptake and presentation of HOCl-modified proteins by murine dendritic cells and macrophages. Other scavenger receptors, SREC-I and LOX-1, as well as RAGE were also able to bind HOCl-modified proteins, but they did not contribute significantly to these ligands uptake by dendritic cells because they were either not expressed or exhibited preference for more heavily oxidised proteins. Our results indicate that oxidation by neutrophils-derived HOCl may be a physiological mechanism of conferring immunogenicity on proteins which in their native forms do not bind to endocytic receptors. This mechanism might enable the immune system to detect infections caused by pathogens not recognized by pattern recognition receptors.


Subject(s)
Alcohol Dehydrogenase/immunology , Glycoproteins/immunology , Hypochlorous Acid/metabolism , Neutrophils/chemistry , Serum Albumin/immunology , Animals , Antigen Presentation , CD36 Antigens/metabolism , CHO Cells , Cricetulus , Dendritic Cells/immunology , Humans , Lectins, C-Type/metabolism , Macrophages/immunology , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mice , Oxidation-Reduction , Receptors, Cell Surface/metabolism , Scavenger Receptors, Class A/metabolism
9.
Elife ; 32014 Oct 15.
Article in English | MEDLINE | ID: mdl-25317947

ABSTRACT

To identify new approaches to enhance innate immunity to bacterial pneumonia, we investigated the natural experiment of gender differences in resistance to infections. Female and estrogen-treated male mice show greater resistance to pneumococcal pneumonia, seen as greater bacterial clearance, diminished lung inflammation, and better survival. In vitro, lung macrophages from female mice and humans show better killing of ingested bacteria. Inhibitors and genetically altered mice identify a critical role for estrogen-mediated activation of lung macrophage nitric oxide synthase-3 (NOS3). Epidemiologic data show decreased hospitalization for pneumonia in women receiving estrogen or statins (known to activate NOS3). Pharmacologic targeting of NOS3 with statins or another small-molecule compound (AVE3085) enhanced macrophage bacterial killing, improved bacterial clearance, and increased host survival in both primary and secondary (post-influenza) pneumonia. The data identify a novel mechanism for host defense via NOS3 and suggest a potential therapeutic strategy to reduce secondary bacterial pneumonia after influenza.


Subject(s)
Immunity, Innate , Macrophages, Alveolar/enzymology , Molecular Targeted Therapy , Nitric Oxide Synthase Type III/metabolism , Pneumonia, Pneumococcal/enzymology , Pneumonia, Pneumococcal/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Benzodioxoles/pharmacology , Benzodioxoles/therapeutic use , Case-Control Studies , Estrogens/pharmacology , Female , Hospitalization , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Immunity, Innate/drug effects , Indans/pharmacology , Indans/therapeutic use , Macrophage Activation/drug effects , Macrophages, Alveolar/drug effects , Male , Mice, Inbred C57BL , Microbial Viability/drug effects , Middle Aged , Odds Ratio , Phagocytosis/drug effects , Pneumonia, Pneumococcal/pathology , Pneumonia, Pneumococcal/therapy , Young Adult
10.
Innate Immun ; 20(8): 826-47, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24257313

ABSTRACT

SR-A/CD204 and CD36 are major receptors responsible for oxidized lipoproteins uptake by macrophages in atherosclerotic plaques. Both receptors also share the role as receptors for different pathogens, but studies on their signaling have been hampered by the lack of selective ligands. We report that, upon specific ligation by Ab, SR-A does not induce cytokine production, but mediates inhibition of LPS-stimulated production of IL-6 and IL-12/23p40, enhancement of IL-10 release, and has no effect on TNF-α and RANTES production in murine macrophages. In contrast, anti-CD36 Ab alone stimulated production of all these cytokines, with IL-10 production being exceptionally high. Effects of anti-CD36 Ab, except of IL-10 production, were mediated by CD14 and TLR2, whereas those of SR-A ligation by heterotrimeric Gi/o proteins and by phosphatidylinositol 3-kinases. Surprisingly, we found that LPS uptake by macrophages was mediated in part by CD36 cooperating with CD14, whereas SR-A was not involved in this process. Finely, during in vitro Ag presentation to naïve CD4(+) lymphocytes, pre-incubation of macrophages with anti-CD36 Ab enhanced IFN-γ production in the co-culture, but exerted the opposite effect under conditions enabling IL-10 accumulation. In contrast, anti-SR-A Ab was ineffective alone, but reversed the Th1-polarizing effect of LPS.


Subject(s)
CD36 Antigens/physiology , Immunity, Cellular/physiology , Macrophages/immunology , Macrophages/physiology , Scavenger Receptors, Class A/physiology , Androstadienes/pharmacology , Animals , CD36 Antigens/genetics , Cells, Cultured , Cytokines/biosynthesis , Female , Lipopolysaccharide Receptors/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Pertussis Toxin/pharmacology , Scavenger Receptors, Class A/genetics , Toll-Like Receptor 2/drug effects , Wortmannin
11.
Postepy Hig Med Dosw (Online) ; 66: 104-19, 2012 Feb 29.
Article in Polish | MEDLINE | ID: mdl-22470185

ABSTRACT

Recognition of pathogens by innate immune cells is mediated by pattern recognition receptors (PRR), which include scavenger receptors (SR). The class A SR, SR-A/CD204 and MARCO, are characterized by the presence of collagenous and SR cysteine-rich domains in their extracellular portions. Both receptors are expressed mainly on macrophages and dendritic cells. Thanks to their ability to bind to a wide range of polyanionic ligands, the class A SR may participate in numerous functions of these cells, such as endocytosis, and adhesion to extracellular matrix and to other cells. Among SR-A ligands are oxidized lipoproteins and ß-amyloid fibrils, which link SR-A to the pathogenesis of arteriosclerosis and Alzheimer's disease. Despite the demonstration of class A SR involvement in so many processes, the lack of selective ligands precluded reaching definite conclusions concerning their signaling abilities. Using specific receptor ligation with antibodies, we showed that SR-A and MARCO trigger intracellular signaling, modulating pro-inflammatory and microbicidal activities of macrophages. Surprisingly, despite similarities in structure and ligand binding repertoires, SR-A and MARCO exert opposite effects on interleukin-12 (IL-12) production in macrophages. SR-A ligation also stimulated H2O2 and IL-10 production, but had no effect on the release of several other cytokines. These limited effects of specific SR-A ligation contrast with generalized enhancement of immune responses observed in SR-A-deficient mice. Recent studies have revealed that many of these effects of SR-A deficiency may be caused by compensatory changes in the expression of other receptors and/or disinhibition of signal transduction from receptors belonging to the Toll/IL-1R family, rather than by the loss of the receptor function of SR-A.


Subject(s)
Cell Communication/immunology , Dendritic Cells/immunology , Macrophages/immunology , Receptors, Immunologic/immunology , Scavenger Receptors, Class A/immunology , Signal Transduction/physiology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Arteriosclerosis/metabolism , Cytoplasm/metabolism , Humans , Interleukin-10/biosynthesis , Interleukin-12/biosynthesis , Ligands , Lipoproteins/metabolism , Macrophages/metabolism , Receptors, Immunologic/metabolism , Scavenger Receptors, Class A/chemistry
12.
Postepy Hig Med Dosw (Online) ; 66: 120-31, 2012 Feb 29.
Article in Polish | MEDLINE | ID: mdl-22470186

ABSTRACT

Recognition of pathogens by innate immune cells is mediated by pattern recognition receptors (PRR), which include the class A scavenger receptors (SR), SR-A/CD204 and MARCO. It seems that in addition to activating innate immune responses, phagocytosis and inflammation, this initial, PRR-mediated recognition also determines polarization of adaptive immune responses (Th1, Th2, Th17 or Treg). It has been demonstrated that class A SR are major PRR mediating opsonin-independent phagocytosis. SR-A- or MARCO-deficient mice exhibit impaired ability to control bacterial infections, resulting in increased mortality. Our results suggest that in addition to impaired bacterial destruction by macrophages, dysregulation of immune responses may contribute to defective antibacterial defense in class A SR-deficient mice. Using specific receptor ligation with antibodies, we showed that SR-A and MARCO regulate in an opposite manner production of IL-12 in macrophages, the cytokine playing a crucial role in Th1/Th2 polarization of adaptive immune responses. Together with the observation that expression of MARCO is increased by different Th1-polarizing factors and decreased by Th2-polarizing factors, these results suggest that changes in relative expression levels of SR-A and MARCO may be a mechanism of sustained polarization of adaptive immune responses.


Subject(s)
Bacterial Infections/immunology , Immunologic Memory/immunology , Macrophages/immunology , Phagocytosis/immunology , Receptors, Immunologic/immunology , Scavenger Receptors, Class A/immunology , Animals , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Humans , Interleukin-12/biosynthesis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Opsonin Proteins/immunology , Receptors, Immunologic/metabolism , Scavenger Receptors, Class A/metabolism , Th1 Cells/immunology
13.
Inflamm Res ; 61(2): 113-26, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22116297

ABSTRACT

OBJECTIVES: Numerous receptors have been implicated in recognition of pathogenic fungi by macrophages, including the ß-glucan receptor dectin-1. The role of scavenger receptors (SRs) in anti-fungal immunity is not well characterized. METHODS: We studied uptake of unopsonized Saccharomycetes cerevisiae (zymosan) and live Candida albicans yeasts as well as zymosan-stimulated H(2)O(2) production in J774 macrophage-like cells and peritoneal exudate macrophages (PEMs). The role of different receptors was assessed with the use of competitive ligands, transfected cells and receptor-deficient macrophages. RESULTS: The uptake of zymosan by untreated J774 cells was mediated approximately half by SRs and half by a ß-glucan receptor which was distinct from dectin-1 and not linked to stimulation of H(2)O(2) production. Ligands of ß-glucan receptors and of SRs also inhibited uptake of C. albicans by macrophages (J774 cells and PEMs). In macrophages pretreated with a CpG motif-containing oligodeoxynucleotide (CpG-ODN) the relative contribution of SRs to yeast uptake increased and that of ß-glucan receptors decreased. Whereas the class A SR MARCO participated in the uptake of both zymosan and C. albicans by CpG-ODN-pretreated, but not untreated macrophages, the related receptor SR-A/CD204 was involved in the uptake of zymosan, but not of C. albicans. The reduction of zymosan-stimulated H(2)O(2) production observed in DS-pretreated J774 cells and in class A SRs-deficient PEMs suggest that class A SRs mediate part of this process. CONCLUSIONS: Our results revealed that SRs belong to a redundant system of receptors for yeasts. Binding of yeasts to different receptors in resting versus CpG-ODN-pre-exposed macrophages may differentially affect polarization of adaptive immune responses.


Subject(s)
Candida albicans/immunology , Macrophages, Peritoneal/immunology , Receptors, Immunologic/immunology , Saccharomyces cerevisiae/immunology , Scavenger Receptors, Class A/immunology , Adaptive Immunity , Animals , CHO Cells , Cell Line , Cricetinae , Cricetulus , Female , Hydrogen Peroxide/metabolism , Macrophages, Peritoneal/microbiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides/pharmacology , Phagocytosis/drug effects , Phagocytosis/immunology , Zymosan/immunology , Zymosan/metabolism
14.
Anal Biochem ; 413(2): 185-91, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21354097

ABSTRACT

Lipopolysaccharide (LPS) is recognized by Toll-like receptor 4 (TLR4) of macrophages triggering production of pro-inflammatory mediators. One of the factors determining the magnitude of responses to LPS, which may even lead to life-threatening septic shock, is the cell surface abundance of TLR4. However, quantitation of the surface TLR4 is difficult due to the low level of receptor expression. To develop a method of TLR4 assessment, we labeled the receptor on the cell surface with a rabbit antibody followed by either anti-rabbit immunoglobulin G-fluorescein isothiocyanate (IgG-FITC) for flow cytometry or with anti-rabbit IgG-peroxidase for a cellular enzyme-linked immunosorbent assay (ELISA). Alternatively, the anti-TLR4 antibody was detected by anti-rabbit IgG labeled with (125)I. Flow cytometry did not allow detection of TLR4 on the surface of J774 cells or human macrophages. In contrast, application of cellular ELISA or the radiolabeling technique combined with effective blockage of nonspecific binding of antibodies provided TLR4-specific signals. The level of TLR4 on the surface of J774 cells did not change on treatment with 1-100ng/ml LPS; however, it was reduced by approximately 30-40% after 2 h of treatment with 1 µg/ml LPS. These data indicate that down-regulation of surface TLR4 can serve as a means of negative regulation of cell responses toward high doses of LPS.


Subject(s)
Cell Membrane/metabolism , Toll-Like Receptor 4/metabolism , Animals , Cell Line , Culture Media, Conditioned , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression , Humans , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Mice , Radioligand Assay , Toll-Like Receptor 4/chemistry , Tumor Necrosis Factor-alpha/metabolism
15.
Microb Pathog ; 50(6): 350-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21419839

ABSTRACT

Lipoarabinomannan capped with terminal oligomannosides (ManLAM) is a component of mycobacteria cell wall enabling Mycobacterium tuberculosis to infect macrophages. We found that short treatment (3.5h) of macrophage-like J774 cells and thioglycollate-elicited peritoneal murine macrophages with ManLAM and its deacylated form enhanced LPS-stimulated release of tumor necrosis factor-α (TNF-α). In contrast, prolong incubation of J774 cells with ManLAM (16h) led to inhibition of LPS-stimulated TNF-α production. LPS-triggered secretion of nitric oxide (NO) was suppressed by ManLAM and its deacylated form. Effects of ManLAM and its deacylated derivative were mimicked by dextran sulfate, a general ligand of scavenger receptors. The enhancement of LPS-induced TNF-α production by dextran sulfate was partially reversed by an antibody neutralizing scavenger receptor SR-PSOX/CXCL16 while the stimulatory activity of deacylated ManLAM was reversed by an antibody neutralizing class B scavenger receptor CD36. Our data suggest that CD36 mediates the activity of ManLAM and its deacylated form leading to TNF-α release in LPS-stimulated J774 cells and peritoneal murine macrophages, while NO production is modulated by unknown scavenger receptors.


Subject(s)
Lipopolysaccharides/pharmacology , Mycobacterium tuberculosis/drug effects , Nitric Oxide/metabolism , Receptors, Scavenger/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Antigens, Bacterial/biosynthesis , CD36 Antigens/metabolism , Chemokine CXCL16 , Chemokine CXCL6/metabolism , Dextran Sulfate/pharmacology , Drug Synergism , Lipopolysaccharide Receptors/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mannose-Binding Lectin/metabolism , Mice , Nitric Oxide/antagonists & inhibitors , Nitric Oxide/biosynthesis , Toll-Like Receptor 2/metabolism
16.
J Immunol ; 185(11): 6960-73, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21041721

ABSTRACT

LPS is a constituent of cell walls of Gram-negative bacteria that, acting through the CD14/TLR4 receptor complex, causes strong proinflammatory activation of macrophages. In murine peritoneal macrophages and J774 cells, LPS at 1-2 ng/ml induced maximal TNF-α and MIP-2 release, and higher LPS concentrations were less effective, which suggested a negative control of LPS action. While studying the mechanism of this negative regulation, we found that in J774 cells, LPS activated both acid sphingomyelinase and neutral sphingomyelinase and moderately elevated ceramide, ceramide 1-phosphate, and sphingosine levels. Lowering of the acid sphingomyelinase and neutral sphingomyelinase activities using inhibitors or gene silencing upregulated TNF-α and MIP-2 production in J774 cells and macrophages. Accordingly, treatment of those cells with exogenous C8-ceramide diminished TNF-α and MIP-2 production after LPS stimulation. Exposure of J774 cells to bacterial sphingomyelinase or interference with ceramide hydrolysis using inhibitors of ceramidases also lowered the LPS-induced TNF-α production. The latter result indicates that ceramide rather than sphingosine suppresses TNF-α and MIP-2 production. Of these two cytokines, only TNF-α was negatively regulated by ceramide 1-phosphate as was indicated by upregulated TNF-α production after silencing of ceramide kinase gene expression. None of the above treatments diminished NO or RANTES production induced by LPS. Together the data indicate that ceramide negatively regulates production of TNF-α and MIP-2 in response to LPS with the former being sensitive to ceramide 1-phosphate as well. We hypothesize that the ceramide-mediated anti-inflammatory pathway may play a role in preventing endotoxic shock and in limiting inflammation.


Subject(s)
Ceramides/physiology , Down-Regulation/immunology , Lipopolysaccharides/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Animals , Cell Line , Ceramides/metabolism , Chemokine CXCL2/antagonists & inhibitors , Chemokine CXCL2/biosynthesis , Down-Regulation/genetics , Female , Gene Silencing/immunology , Inflammation Mediators/antagonists & inhibitors , Inflammation Mediators/pharmacology , Inflammation Mediators/physiology , Lipopolysaccharides/antagonists & inhibitors , Lysophospholipids/physiology , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred BALB C , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/biosynthesis , Phosphotransferases (Alcohol Group Acceptor)/genetics , Sphingosine/analogs & derivatives , Sphingosine/physiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors
17.
Inflamm Res ; 59(4): 277-89, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19784549

ABSTRACT

OBJECTIVE: Denatured proteins are deposited in damaged tissues, around implanted biomaterials, during natural aging as well as in a heterogeneous group of amyloid diseases, such as Alzheimer's disease. There is evidence that tissue damage observed in amyloidosis is mediated mainly by factors released from activated macrophages, such as superoxide and nitric oxide (NO), as opposed to direct interaction between amyloid fibrils and nonimmune cells. METHODS: Mouse resident peritoneal macrophages were stimulated in serum-free medium with different preparations of nonamyloidogenic proteins: alcohol dehydrogenase (AD), bovine serum albumin (BSA) or fibrinogen (FG). Intra- and extracellular superoxide production was measured by, respectively, nitro blue tetrazolium (NBT) reduction and lucigenin-enhanced chemiluminescence. Levels of nitrite (reflecting NO release) were measured in culture supernatants. RESULTS: Aggregates of denatured, nonamyloidogenic proteins, but not their native or denatured but not aggregated counterparts, stimulated superoxide and/or NO production in macrophages. The NO production was mediated by beta(1) and beta(2) integrins, with a possible contribution of receptor for advanced glycation end products (RAGE). It was catalyzed by inducible NO synthase (iNOS), enhanced synergistically by interferon-gamma (IFN-gamma), and inhibited by covalently modified proteins-components of advanced glycation end products. Although intracellular superoxide production was stimulated significantly by denatured BSA and AD, but not by FG, both denatured BSA and FG strongly enhanced zymosan-stimulated extracellular release of reactive oxygen species. CONCLUSION: Our results point at similarities in macrophage responses to denatured nonamyloidogenic proteins and to amyloid fibrils. Thus, the tissue injury observed in amyloidosis may result from overstimulation of mechanisms that, under physiological conditions, enable macrophages to recognize and remove denatured proteins.


Subject(s)
Macrophages/metabolism , Nitric Oxide/biosynthesis , Proteins/pharmacology , Superoxides/metabolism , Alcohol Dehydrogenase/chemistry , Alcohol Dehydrogenase/pharmacology , Animals , Cell Lineage/drug effects , Cell Survival/drug effects , Cells, Cultured , Female , Fibrinogen/chemistry , Fibrinogen/pharmacology , Hot Temperature , Integrins/physiology , Interleukin-6/metabolism , Luminescence , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Protein Denaturation , Proteins/chemistry , Serum Albumin/chemistry , Serum Albumin/pharmacology
18.
Bioessays ; 30(10): 943-54, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18800365

ABSTRACT

Mycobacterium tuberculosis is the causative agent of pulmonary tuberculosis which has infected one third of the mankind and causes 2-3 million deaths worldwide each year. The persistence of the infection ensues from the ability of M. tuberculosis to subvert host immune responses in favor of survival and growth of mycobacteria in macrophages. The mechanisms by which M. tuberculosis manipulates the host immune system have only recently come to light. These activities are attributed to lipoarabinomannans (LAM) and their precursors lipomannans (LM), two predominant glycolipids of M. tuberculosis cell wall. LM are able to skew anti-mycobacterial immune responses into un-protective ones, while LAM evoke immunosupression upon binding to macrophage and dendritic cell receptors specialized in binding to "self" host components. A newly emerging idea implicates plasma membrane rafts in LM and LAM signaling. Depending on acylation patterns, the glycolipids may either directly incorporate into the raft membrane via mannosylphosphatidylinositol anchors or interact with raft-associated proteins to affect the assembly of receptor signaling complexes.


Subject(s)
Host-Pathogen Interactions/immunology , Macrophages/immunology , Membrane Microdomains/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis, Pulmonary/immunology , Acylation , Cell Wall/microbiology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Humans , Lipopolysaccharides/immunology , Macrophages/metabolism , Macrophages/microbiology , Membrane Microdomains/metabolism , Mycobacterium tuberculosis/metabolism , Phosphatidylinositols/immunology , Th1 Cells/immunology , Th1 Cells/microbiology , Th2 Cells/immunology , Th2 Cells/microbiology
19.
J Leukoc Biol ; 80(4): 870-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16882874

ABSTRACT

The macrophage Class A scavenger receptor MARCO (macrophage receptor with a collagenous structure) functions as a pattern-recognition receptor for bacterial components, but its role in responses to CpG oligonucleotide sequences (CpG-ODN) in microbial DNA has not been characterized. Phosphorothioate (PS)-linked CpG-ODN stimulated IL-12 and NO production in wild-type but not in MARCO-deficient, thioglycollate-elicited peritoneal macrophages. MARCO and the related class A receptor SR-A belong to a redundant system of receptors for PS ODNs. The ability of MARCO to bind CpG-ODNs and conversely, to costimulate IL-12 and NO production upon specific ligation with immobilized mAb is consistent with MARCO being a signaling receptor for CpG-ODNs, costimulating TLR9-mediated NO and IL-12 production in macrophages. In contrast to MARCO, SR-A is likely to mediate negative regulation of macrophage responses to CpG-ODNs. In particular, increased affinity toward SR-A may contribute to decreased potency of oligo G-modified CpG-ODNs in stimulating IL-12 production. The results suggest that differential involvement of activating and inhibitory membrane receptors, such as SR-A and MARCO, may underlie profound differences observed in biological activities of different ODN sequences.


Subject(s)
Macrophages/immunology , Oligodeoxyribonucleotides/pharmacology , Receptors, Immunologic/physiology , Scavenger Receptors, Class A/physiology , Animals , CHO Cells , Cricetinae , Cricetulus , Interleukin-12/biosynthesis , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide/biosynthesis , Receptors, Immunologic/deficiency , Scavenger Receptors, Class A/deficiency , Structure-Activity Relationship
20.
J Immunol ; 175(12): 8032-41, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16339540

ABSTRACT

The macrophage class A scavenger receptors, macrophage receptor with a collagenous structure (MARCO) and type I/II class A scavenger receptor (SR-AI/II), share structural features and roles in host defense, but little is known about their regulation and signaling properties. Ligation of MARCO on mouse thioglycollate-elicited peritoneal macrophages (PEMs) with immobilized mAb costimulated IL-12 production, in contrast to previously reported inhibition by SR-AI/II. PEMs from MARCO-deficient mice exhibited 2.7 times lower IL-12 production in responses to stimulation with LPS and IFN-gamma and lack of significant IL-12 production on stimulation with LPS alone. Conversely, SR-AI/II-deficient PEMs produced 2.4 and 1.8 times more IL-12 than wild-type PEMs in response to LPS or LPS and IFN-gamma, respectively. Corresponding differences in regulation of SR-A and MARCO expression were also observed. Th1 adjuvants (LPS, a CpG motif-containing oligodeoxynucleotide (CpG-ODN), IL-12, and GM-CSF) increased, whereas Th2-polarizing factors (IL-4, M-CSF, and non-CpG ODN) decreased expression of MARCO on J774 macrophage-like cells. Expression of SR-A was regulated in the opposite manner to MARCO or not affected. Whereas MARCO was involved in opsonin-independent phagocytosis in CpG-ODN-pretreated but not in IL-4-pretreated J774 cells, anti-SR-A Abs inhibited particle uptake in untreated and IL-4-pretreated but not in CpG-ODN-pretreated cells. SR-A and MARCO are regulated differently and mediate distinct negative and positive effects on IL-12 production in macrophages. These differences may contribute to sustained Th1 or Th2 polarization of ongoing immune responses.


Subject(s)
Gene Expression Regulation/immunology , Receptors, Immunologic/physiology , Scavenger Receptors, Class A/physiology , Animals , Immunity , Interleukin-12/biosynthesis , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Phagocytosis , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics , Scavenger Receptors, Class A/deficiency , Scavenger Receptors, Class A/genetics , Th1 Cells/metabolism , Th2 Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...