Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 123(8): 3363-72, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863624

ABSTRACT

Macrophages play a key role in responding to pathogens and initiate an inflammatory response to combat microbe multiplication. Deactivation of macrophages facilitates resolution of the inflammatory response. Deactivated macrophages are characterized by an immunosuppressive phenotype, but the lack of unique markers that can reliably identify these cells explains the poorly defined biological role of this macrophage subset. We identified lipocalin 2 (LCN2) as both a marker of deactivated macrophages and a macrophage deactivator. We show that LCN2 attenuated the early inflammatory response and impaired bacterial clearance, leading to impaired survival of mice suffering from pneumococcal pneumonia. LCN2 induced IL-10 formation by macrophages, skewing macrophage polarization in a STAT3-dependent manner. Pulmonary LCN2 levels were tremendously elevated during bacterial pneumonia in humans, and high LCN2 levels were indicative of a detrimental outcome from pneumonia with Gram-positive bacteria. Our data emphasize the importance of macrophage deactivation for the outcome of pneumococcal infections and highlight the role of LCN2 and IL-10 as determinants of macrophage performance in the respiratory tract.


Subject(s)
Acute-Phase Proteins/immunology , Lipocalins/immunology , Macrophages, Alveolar/immunology , Oncogene Proteins/immunology , Pneumonia, Pneumococcal/immunology , Proto-Oncogene Proteins/immunology , Acute-Phase Proteins/deficiency , Acute-Phase Proteins/genetics , Adult , Aged , Animals , Female , Humans , Immune Tolerance , Interleukin-10/biosynthesis , Lipocalin-2 , Lipocalins/genetics , Lung/immunology , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oncogene Proteins/deficiency , Oncogene Proteins/genetics , Pneumonia, Pneumococcal/etiology , Transplantation Chimera/immunology
2.
Eur J Immunol ; 42(11): 2983-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22806614

ABSTRACT

Plasma lipoproteins such as LDL (low-density lipoprotein) are important therapeutic targets as they play a crucial role in macrophage biology and metabolic disorders. The impact of lipoprotein profiles on host defense pathways against Gram-positive bacteria is poorly understood. In this report, we discovered that human serum lipoproteins bind to lipoteichoic acid (LTA) from Staphylococcus aureus and thereby alter the immune response to these bacteria. Size-exclusion chromatography and solid-phase-binding analysis of serum revealed the direct interaction of LTA with apolipoproteins (Apo) B100, ApoA1, and ApoA2. Only ApoB100 and the corresponding LDL exerted biological effects as this binding significantly inhibited LTA-induced cytokine releases from human and murine immune cells. Serum from hypercholesterolemic mice or humans significantly diminished cytokine induction in response to S. aureus or its LTA. Sera taken from the patients with familial hypercholesterolemia before and after ApoB100-directed immuno-apheresis confirmed that ApoB100 inhibited LTA-induced inflammation in humans. In addition, mice in which LDL secretion was pharmacologically inhibited, displayed significantly increased serum cytokine levels upon infection with S. aureus in vivo. The present study identifies ApoB100 as an important suppressor of innate immune activation in response to S. aureus and its LTA.


Subject(s)
Apolipoprotein B-100/pharmacology , Lipopolysaccharides/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Teichoic Acids/immunology , Animals , Female , Humans , Hypercholesterolemia/immunology , Immunity, Innate/immunology , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Specific Pathogen-Free Organisms , Staphylococcal Infections/microbiology , Teichoic Acids/antagonists & inhibitors
3.
PLoS Pathog ; 7(5): e1001345, 2011 May.
Article in English | MEDLINE | ID: mdl-21625574

ABSTRACT

Streptococcus pyogenes is a Gram-positive human pathogen that is recognized by yet unknown pattern recognition receptors (PRRs). Engagement of these receptor molecules during infection with S. pyogenes, a largely extracellular bacterium with limited capacity for intracellular survival, causes innate immune cells to produce inflammatory mediators such as TNF, but also type I interferon (IFN). Here we show that signaling elicited by type I IFNs is required for successful defense of mice against lethal subcutaneous cellulitis caused by S. pyogenes. Type I IFN signaling was accompanied with reduced neutrophil recruitment to the site of infection. Mechanistic analysis revealed that macrophages and conventional dendritic cells (cDCs) employ different signaling pathways leading to IFN-beta production. Macrophages required IRF3, STING, TBK1 and partially MyD88, whereas in cDCs the IFN-beta production was fully dependent on IRF5 and MyD88. Furthermore, IFN-beta production by macrophages was dependent on the endosomal delivery of streptococcal DNA, while in cDCs streptococcal RNA was identified as the IFN-beta inducer. Despite a role of MyD88 in both cell types, the known IFN-inducing TLRs were individually not required for generation of the IFN-beta response. These results demonstrate that the innate immune system employs several strategies to efficiently recognize S. pyogenes, a pathogenic bacterium that succeeded in avoiding recognition by the standard arsenal of TLRs.


Subject(s)
DNA, Bacterial/metabolism , Dendritic Cells , Macrophages , RNA, Bacterial/metabolism , Streptococcus pyogenes/immunology , Animals , Cells, Cultured , Cellulitis/microbiology , Cellulitis/mortality , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Gene Silencing , Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Regulatory Factors , Interferon-beta/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Myeloid Differentiation Factor 88 , Neutrophil Infiltration/immunology , Polymerase Chain Reaction , Protein Serine-Threonine Kinases , RNA, Small Interfering , Receptors, Pattern Recognition , Signal Transduction/immunology , Streptococcus pyogenes/genetics
4.
Scand J Clin Lab Invest ; 70(7): 512-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20873968

ABSTRACT

Interactions of bacterial and host products in activating the innate immune system is an important area to address. The role of lipoteichoic acid (LTA) in these interactions is particularly important because it is understudied in comparison to other factors. This study evaluated the effect of cationic peptides (CPs) on LTA-induced proinflammatory cytokine production in human whole blood and on purified leukocytes. Four different CPs of truncated derivatives from the known peptides LL37, BPI, and CP207 were used. Two of the CPs (IG33 and LL33), derivatives from LL37, potentiated S. aureus LTA induced TNFα, IL-6 and IL-1ß production in whole blood. The release of TNFα was increased 30-fold after 16 hours incubation. Intact LL37 also increased LTA-induced TNFα and IL-1ß in a time dependent manner. LTA in combination with either LL33 or IG23 demonstrated a synergistic enhanced TNFα and IL-1ß secretion on isolated leukocytes but not on purified monocytes. When complexed with IG23 and LL33, the electrophoretic mobility of LTA was altered in a non-denaturating gel electrophoresis. LTA was disaggregated and migrated more rapidly, suggesting an amphiphilic effect of CPs on LTA. In conclusion, LTA synergizes with LL37 and its truncated derivatives and this may lead to proinflammatory cytokine production and cause problems in sepsis therapy.


Subject(s)
Cathelicidins/pharmacology , Cytokines/biosynthesis , Cytokines/blood , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Mutant Proteins/pharmacology , Teichoic Acids/pharmacology , Antimicrobial Cationic Peptides , Cell Survival/drug effects , Cytokines/metabolism , Electrophoresis, Polyacrylamide Gel , Hemoglobins/pharmacology , Hemolysis/drug effects , Humans , Leukocytes/drug effects , Leukocytes/metabolism , Monocytes/cytology , Monocytes/drug effects
5.
J Immunol ; 185(6): 3708-17, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20713893

ABSTRACT

Lipoteichoic acid (LTA), a ubiquitous cell wall component of Gram-positive bacteria, represents a potent immunostimulatory molecule. Because LTA of a mutant Staphylococcus aureus strain lacking lipoproteins (Deltalgt-LTA) has been described to be immunobiologically inactive despite a lack of ascertained structural differences to wild-type LTA (wt-LTA), we investigated the functional requirements for the recognition of Deltalgt-LTA by human peripheral blood cells. In this study, we demonstrate that Deltalgt-LTA-induced immune activation critically depends on the immobilization of LTA and the presence of human serum components, which, to a lesser degree, was also observed for wt-LTA. Under experimental conditions allowing LTA-mediated stimulation, we found no differences between the immunostimulatory capacity of Deltalgt-LTA and wt-LTA in human blood cells, arguing for a limited contribution of possible lipoprotein contaminants to wt-LTA-mediated immune activation. In contrast to human blood cells, TLR2-transfected human embryonic kidney 293 cells could be activated only by wt-LTA, whereas activation of these cells by Deltalgt-LTA required the additional expression of TLR6 and CD14, suggesting that activation of human embryonic kidney 293 cells expressing solely TLR2 is probably mediated by residual lipoproteins in wt-LTA. Notably, in human peripheral blood, LTA-specific IgG Abs are essential for Deltalgt-LTA-mediated immune activation and appear to induce the phagocytic uptake of Deltalgt-LTA via engagement of FcgammaRII. In this study, we have elucidated a novel mechanism of LTA-induced cytokine induction in human peripheral blood cells that involves uptake of LTA and subsequent intracellular recognition driven by TLR2, TLR6, and CD14.


Subject(s)
Adjuvants, Immunologic/blood , Lipopolysaccharides/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Teichoic Acids/metabolism , Toll-Like Receptor 2/metabolism , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/physiology , Antigen-Antibody Reactions , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Cell Membrane/microbiology , Cytokines/biosynthesis , Cytokines/blood , Cytokines/metabolism , Humans , Immunoglobulin G/metabolism , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Intracellular Fluid/microbiology , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/physiology , Lipopolysaccharides/blood , Lipopolysaccharides/immunology , Lipoproteins/deficiency , Lipoproteins/genetics , Opsonin Proteins/metabolism , Receptors, IgG/physiology , Staphylococcus aureus/metabolism , Teichoic Acids/blood , Teichoic Acids/immunology , Toll-Like Receptor 2/blood , Toll-Like Receptor 2/physiology , Toll-Like Receptor 6/biosynthesis , Toll-Like Receptor 6/physiology
6.
Exp Dermatol ; 19(8): e296-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19849713

ABSTRACT

BACKGROUND: The cutaneous colonization with Staphylococcus aureus represents a potent trigger factor of atopic dermatitis. Toll-like receptor (TLR)-2 and CD36 have been shown to play a pivotal role in the internalization of staphylococcal components. AIMS: To investigate the impact of TLR-2 ligands on cell surface protein expression in monocytes from wild type (WT) AD patients and TLR-2 R753Q polymorph AD patients. RESULTS: CD36 expression was significantly less downregulated in TLR-2 polymorph AD patients compared to wild type AD patients upon stimulation with peptidoglycan (PGN) and lipoteichoic acid (LTA) and compared to healthy controls upon stimulation with PGN. Expression of CD86 was higher upon N-palmitoyl-S-[2,3-bis(palmitoyl)-(2RS)-propyl]-(R)cysteinyl-alanyl-glycine (Pam3Cys) stimulation in TLR-2 R753Q polymorph AD patients compared to wild type AD patients. Expression of CD80 and CD54 were unaffected. CONCLUSION: The differences in CD36 expression in TLR-2 polymorph AD patients compared to wild type AD patients and healthy controls may be associated with an enhanced susceptibility to skin infections with S. aureus.


Subject(s)
CD36 Antigens/metabolism , Dermatitis, Atopic/metabolism , Monocytes/metabolism , Polymorphism, Single Nucleotide/genetics , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Case-Control Studies , Disease Susceptibility , Humans , Ligands , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Peptidoglycan/pharmacology , Skin/microbiology , Staphylococcal Skin Infections/metabolism , Staphylococcus aureus/isolation & purification , Teichoic Acids/pharmacology
7.
Mol Cancer Res ; 7(10): 1729-35, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19825995

ABSTRACT

Proinflammatory cytokines are centrally involved in tumor progression and survival in non-small cell lung cancer, and both the presence of infiltrating neutrophils and bacterial infection in the lung may indicate a poor prognosis. Against this background, we investigated the effect of the bacterial cell wall component lipopolysaccharide (LPS) on interleukin (IL)-6 and IL-8 synthesis in the non-small cell lung cancer line A549 and in A549-neutrophil cocultures. The LPS induced a dose-dependent and time-dependent release of IL-8 from A549 cells, whereas IL-6 could not be detected. Interestingly, in A549-neutrophil cocultures, IL-8 synthesis was massively amplified and IL-6 was also released, compared with the respective monocultures. The A549 cells were identified as the primary cellular source of these cytokines, as enhanced cytokine mRNA transcription was detected in this cell type, although not in neutrophils in the coculture system. Experiments done in transwells indicated that direct cell-cell contact was a prerequisite for the increased cytokine generation. Inhibition of tumor necrosis factor-alpha bioactivity by neutralizing antibodies and blocking cyclooxygenase-2 activity blunted the enhanced cytokine generation in the coculture system. Amplification of LPS-induced cytokine secretion could be reproduced when the small cell lung cancer cell line H69 was cocultured with neutrophils. When the Gram-positive cell wall component lipoteichoic acid was used instead of LPS, cytokine synthesis was also amplified in A549-neutrophil cocultures, to a similar extent to that observed with LPS. These data indicate that interaction between bacterial pathogens, neutrophils, and tumor cells might amplify the release of proinflammatory cytokines which may promote tumor growth in vivo.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Cytokines/biosynthesis , Inflammation/immunology , Lipopolysaccharides/pharmacology , Lung Neoplasms/immunology , Neutrophils/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/physiopathology , Cell Communication/immunology , Cell Line, Tumor , Cells, Cultured , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Coculture Techniques , Cyclooxygenase 2 Inhibitors/pharmacology , Disease Progression , Dose-Response Relationship, Drug , Humans , Inflammation/genetics , Inflammation/physiopathology , Inflammation Mediators/pharmacology , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , Interleukin-8/drug effects , Lung Neoplasms/genetics , Lung Neoplasms/physiopathology , Neutrophils/drug effects , Pneumonia, Bacterial/complications , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/physiopathology , RNA, Messenger/metabolism , Teichoic Acids/pharmacology , Time Factors , Transcriptional Activation/drug effects
8.
Immunobiology ; 213(3-4): 285-96, 2008.
Article in English | MEDLINE | ID: mdl-18406374

ABSTRACT

Despite similar clinical relevance of Gram-positive and Gram-negative infections, immune activation by Gram-positive bacteria is by far less well understood than immune activation by Gram-negative bacteria. Our group has made available highly purified lipoteichoic acids (LTA) as a key Gram-positive immunostimulatory component. We have characterized the reasons for lower potency of LTA compared to Gram-negative lipopolysaccharide (LPS), identifying lack of IL-12/IFNgamma induction as a general characteristic of TLR2 agonists, and need for presentation of LTA on surfaces for enhanced immunostimulatory potency, as major aspects. Aspects of chemokine induction, where LTA is more potent than LPS, have been addressed. Furthermore, novel complement and plant defence activation, as well as CD36 as a new LTA receptor, were identified. The bacterial costimuli and modulators of LTA inducible responses are being investigated: LTA isolated from so far 16 bacterial species, although different in structure, behave remarkably similar while whole live and killed bacteria differ with regard to the pattern of induced responses. The purification and characterization of the respective components of the bacterial cell wall has begun.


Subject(s)
Cytokines/metabolism , Gram-Positive Bacteria/metabolism , Immunity, Innate , Lipopolysaccharides/metabolism , Monocytes/microbiology , Animals , Bacteria/metabolism , CD36 Antigens/biosynthesis , Cell Wall/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-12/metabolism , Interleukin-8/metabolism , Mice , Models, Biological , Monocytes/metabolism , Teichoic Acids/metabolism
9.
Clin Vaccine Immunol ; 14(12): 1629-33, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17928431

ABSTRACT

Lipoteichoic acid (LTA) from gram-positive bacteria is the counterpart to lipopolysaccharide from gram-negative bacteria. LTA, which activates Toll-like receptor 2 (TLR2), induces a unique cytokine and chemokine pattern. The chemical synthesis of LTA proved its immunostimulatory properties. To determine the minimal active structure of LTA, we reduced synthetic LTA in a number of steps down to the synthetic anchor and employed these molecules to stimulate interleukin-8 (IL-8) release in human whole blood. Ten times more of the synthetic structures with four to six d-alanine-substituted polyglycerophosphate units (50 nM) than of the native LTA preparation was required to induce IL-8 release. A further reduction to three backbone units with two or no d-alanine residues resulted in cytokine induction only from 500 nM. The synthetic anchor was not able to induce IL-8 release even at 5 muM. When the LTA derivatives were used at 500 nM, they induced increasing levels of IL-8 and tumor necrosis factor alpha with increasing elongation of the backbone. Peritoneal macrophages were less responsive than human blood to the synthetic structures. Therefore, TLR2 dependency could be shown only with cells from TLR2-deficient mice for the two largest synthetic structures. This was confirmed by using TLR2-transfected HEK 293 cells. Taken together, these data indicate that although the synthetic anchor (which, unlike the native anchor, contains only myristic acid) cannot induce cytokine release, the addition of three backbone units, even without d-alanine substituents, confers this ability. Lengthening of the chain with d-alanine-substituted backbone units results in increased cytokine-inducing potency and a more sensitive response.


Subject(s)
Cytokines/metabolism , Lipopolysaccharides/chemistry , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Teichoic Acids/chemistry , Teichoic Acids/pharmacology , Animals , Cell Line , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Humans , Kidney/cytology , Lipopolysaccharides/chemical synthesis , Mice , Mice, Knockout , Models, Chemical , Staphylococcus aureus/chemistry , Statistics as Topic , Teichoic Acids/chemical synthesis , Toll-Like Receptor 2/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...