Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters










Publication year range
1.
FEMS Microbiol Lett ; 369(1)2022 02 26.
Article in English | MEDLINE | ID: mdl-35191469

ABSTRACT

Staphylococcus pseudintermedius is one of the major pathogens causing canine skin infection. In canine atopic dermatitis (AD), heterogeneous strains of S. pseudintermedius reside on the affected skin site. Because an increase in specific IgE to this bacterium has been reported, S. pseudintermedius is likely to exacerbate the severity of canine AD. In this study, the IgE reactivities to various S. pseudintermedius strains and the IgE-reactive molecules of S. pseudintermedius were investigated. First, examining the IgE reactivities to eight strains of S. pseudintermedius using 141 sera of AD dogs, strain variation of S. pseudintermedius showed 10-63% of the IgE reactivities. This is different from the expected result based on the concept of Staphylococcus aureus clonality in AD patients. Moreover, according to the western blot analysis, there were more than four proteins reactive to IgE. Subsequently, the analysis of the common IgE-reactive protein at ∼15 kDa confirmed that the DM13-domain-containing protein was reactive in AD dogs, which is not coincident with any S. aureus IgE-reactive molecules. Considering these, S. pseudintermedius is likely to exacerbate AD severity in dogs, slightly different from the case of S. aureus in human AD.


Subject(s)
Dermatitis, Atopic , Animals , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/veterinary , Dogs , Humans , Immunoglobulin E/metabolism , Staphylococcus/genetics , Staphylococcus aureus/genetics
2.
Infect Immun ; 87(12)2019 12.
Article in English | MEDLINE | ID: mdl-31548327

ABSTRACT

Upon microbial infection, host immune cells recognize bacterial cell envelope components through cognate receptors. Although bacterial cell envelope components function as innate immune molecules, the role of the physical state of the bacterial cell envelope (i.e., particulate versus soluble) in host immune activation has not been clearly defined. Here, using two different forms of the staphylococcal cell envelope of Staphylococcus aureus RN4220 and USA300 LAC strains, we provide biochemical and immunological evidence that the particulate state is required for the effective activation of host innate immune responses. In a murine model of peritoneal infection, the particulate form of the staphylococcal cell envelope (PCE) induced the production of chemokine (C-X-C motif) ligand 1 (CXCL1) and CC chemokine ligand 2 (CCL2), the chemotactic cytokines for neutrophils and monocytes, respectively, resulting in a strong influx of the phagocytes into the peritoneal cavity. In contrast, compared with PCE, the soluble form of cell envelope (SCE), which was derived from PCE by treatment with cell wall-hydrolyzing enzymes, showed minimal activity. PCE also induced the secretion of calprotectin (myeloid-related protein 8/14 [MRP8/14] complex), a phagocyte-derived antimicrobial protein, into the peritoneal cavity at a much higher level than did SCE. The injected PCE particles were phagocytosed by the infiltrated neutrophils and monocytes and then delivered to mediastinal draining lymph nodes. More importantly, intraperitoneally (i.p.) injected PCE efficiently protected mice from S. aureus infection, which was abolished by the depletion of either monocytes/macrophages or neutrophils. This study demonstrated that the physical state of bacterial cells is a critical factor for efficient host immune activation and the protection of hosts from staphylococcal infections.


Subject(s)
Cell Wall/immunology , Monocytes/immunology , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Chemokine CCL2/metabolism , Chemokine CXCL1/metabolism , Female , Immunity, Innate/immunology , Leukocyte L1 Antigen Complex/metabolism , Macrophages/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Staphylococcal Infections/microbiology
3.
J Gen Virol ; 98(8): 2171-2180, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28730979

ABSTRACT

The group of phages belonging to the family Podoviridae, genus P68virus, including Staphylococcus viruses S13' and S24-1, are important because of their benefits in phage therapy against Staphylococcus aureus infections. The O-glycosidic linkage patterns of wall teichoic acids (WTAs) in S. aureus cell walls seem to be important for adsorption of this phage group. In this study, the adsorption of Staphylococcus viruses S13' and S24-1 to S. aureus was examined using strains with modified WTA glycosidic linkage patterns. We found that the ß-O-N-acetylglucosamine of WTAs was essential for S13' adsorption, while N-acetylglucosamine, regardless of the α- and ß-O-glycosidic linkages of the WTAs, was essential for S24-1 adsorption. Next, examining the binding activities of their receptor-binding proteins (RBPs) to cell walls with different WTA glycosidic patterns, the ß-O-N-acetylglucosamine of the WTAs was essential for S13' RBP binding, while N-acetylglucosamine, regardless of the α- and ß-O-glycosidic linkages of the WTAs, was essential for S24-1 RBP binding. Therefore, the results of the RBP binding assays were consistent with those of the phage adsorption assays. Bioinformatic analysis suggested that the RBPs of Staphylococcus viruses S13' and S24-1 were structurally similar to the RBPs of phage phi11 of thefamily Siphoviridae. Phylogenetic analysis of the RBPs indicated that two phylogenetic subclusters in the family Podoviridae were related to the glycosidic linkage patterns required for phage adsorption, possibly mediated by RBPs. We hope that this study will encourage the future development of therapeutic phages.


Subject(s)
Receptors, Virus/metabolism , Staphylococcus Phages/physiology , Staphylococcus aureus/virology , Teichoic Acids/metabolism , Virus Attachment , Podoviridae/physiology , Receptors, Virus/chemistry , Teichoic Acids/chemistry
4.
Immunobiology ; 221(10): 1091-101, 2016 10.
Article in English | MEDLINE | ID: mdl-27424796

ABSTRACT

Staphylococcus aureus is a Gram-positive bacterial pathogen that is decorated by glycopolymers, including wall teichoic acid (WTA), peptidoglycan, lipoteichoic acid, and capsular polysaccharides. These bacterial surface glycopolymers are recognized by serum antibodies and a variety of pattern recognition molecules, including mannose-binding lectin (MBL). Recently, we demonstrated that human serum MBL senses staphylococcal WTA. Whereas MBL in infants who have not yet fully developed adaptive immunity binds to S. aureus WTA and activates complement serum, MBL in adults who have fully developed adaptive immunity cannot bind to WTA because of an inhibitory effect of serum anti-WTA IgG. Furthermore, we showed that human anti-WTA IgGs purified from pooled adult serum IgGs triggered activation of classical complement-dependent opsonophagocytosis against S. aureus. Because the epitopes of WTA that are recognized by anti-WTA IgG and MBL have not been determined, we constructed several S. aureus mutants with altered WTA glycosylation. Our intensive biochemical studies provide evidence that the ß-GlcNAc residues of WTA are required for the induction of anti-WTA IgG-mediated opsonophagocytosis and that both ß- and α-GlcNAc residues are required for MBL-mediated complement activation. The molecular interactions of other S. aureus cell wall components and host recognition proteins are also discussed. In summary, in this review, we discuss the biological importance of S. aureus cell surface glycopolymers in complement activation and host defense responses.


Subject(s)
Cell Wall/immunology , Complement Activation/immunology , Complement System Proteins/immunology , Polysaccharides, Bacterial/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Teichoic Acids/immunology , Adaptive Immunity , Age Factors , Animals , Antibodies, Bacterial/immunology , Epitopes/immunology , Host-Pathogen Interactions/immunology , Humans , Immunization , Immunoglobulin G/immunology , Mannose-Binding Lectin/blood , Mannose-Binding Lectin/immunology , Mannose-Binding Lectin/metabolism , Opsonin Proteins/immunology , Phagocytosis/immunology , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/metabolism , Protein Binding , Serum Amyloid P-Component , Staphylococcal Infections/microbiology , Teichoic Acids/chemistry , Teichoic Acids/metabolism
5.
Infect Immun ; 83(11): 4247-55, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26283333

ABSTRACT

The cell envelopes of many Gram-positive bacteria contain wall teichoic acids (WTAs). Staphylococcus aureus WTAs are composed of ribitol phosphate (RboP) or glycerol phosphate (GroP) backbones substituted with D-alanine and N-acetyl-D-glucosamine (GlcNAc) or N-acetyl-D-galactosamine (GalNAc). Two WTA glycosyltransferases, TarM and TarS, are responsible for modifying the RboP WTA with α-GlcNAc and ß-GlcNAc, respectively. We recently reported that purified human serum anti-WTA IgG specifically recognizes ß-GlcNAc of the staphylococcal RboP WTA and then facilitates complement C3 deposition and opsonophagocytosis of S. aureus laboratory strains. This prompted us to examine whether anti-WTA IgG can induce C3 deposition on a diverse set of clinical S. aureus isolates. To this end, we compared anti-WTA IgG-mediated C3 deposition and opsonophagocytosis abilities using 13 different staphylococcal strains. Of note, the majority of S. aureus strains tested was recognized by anti-WTA IgG, resulting in C3 deposition and opsonophagocytosis. A minority of strains was not recognized by anti-WTA IgG, which correlated with either extensive capsule production or an alteration in the WTA glycosylation pattern. Our results demonstrate that the presence of WTAs with TarS-mediated glycosylation with ß-GlcNAc in clinically isolated S. aureus strains is an important factor for induction of anti-WTA IgG-mediated C3 deposition and opsonophagocytosis.


Subject(s)
Cell Wall/immunology , Complement C3/immunology , Immunoglobulin G/immunology , Phagocytosis , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Teichoic Acids/immunology , Bacterial Proteins/metabolism , Complement Activation , Glycosyltransferases/metabolism , Humans , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics
6.
BMB Rep ; 48(1): 36-41, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24856825

ABSTRACT

Recently, we demonstrated that human serum amyloid P component (SAP) specifically recognizes exposed bacterial peptidoglycan (PGN) of wall teichoic acid (WTA)-deficient Staphylococcus aureus ΔtagO mutant cells and then induces complement-independent phagocytosis. In our preliminary experiments, we found the existence of human serum immunoglobulins that recognize S. aureus PGN (anti-PGNIgGs), which may be involved in complement-dependent opsonophagocytosis against infected S. aureus cells. We assumed that purified serum anti-PGN-IgGs and S. aureus ΔtagO mutant cells are good tools to study the molecular mechanism of anti-PGN-IgG-mediated phagocytosis. Therefore, we tried to identify the intracellular molecule(s) that is involved in the anti-PGN-IgG-mediated phagocytosis using purified human serum anti-PGN-IgGs and different S. aureus mutant cells. Here, we show that anti-PGN-IgG-mediated phagocytosis in phorbol myristate acetate-treated U937 cells is mediated by Ca2(+) release from intracellular Ca2(+) stores and anti-PGN-IgG dependent Ca2(+) mobilization is controlled via a phospholipase Cγ-2-mediated pathway.


Subject(s)
Antibodies/immunology , Calcium/metabolism , Immunoglobulin G/blood , Peptidoglycan/immunology , Phagocytosis/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Antibodies/blood , Humans , Immunoglobulin G/immunology , Phospholipase C gamma/metabolism , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , U937 Cells
7.
J Biol Chem ; 288(43): 30956-68, 2013 Oct 25.
Article in English | MEDLINE | ID: mdl-24045948

ABSTRACT

Serum antibodies and mannose-binding lectin (MBL) are important host defense factors for host adaptive and innate immunity, respectively. Antibodies and MBL also initiate the classical and lectin complement pathways, respectively, leading to opsonophagocytosis. We have shown previously that Staphylococcus aureus wall teichoic acid (WTA), a cell wall glycopolymer consisting of ribitol phosphate substituted with α- or ß-O-N-acetyl-d-glucosamine (GlcNAc) and d-alanine, is recognized by MBL and serum anti-WTA IgG. However, the exact antigenic determinants to which anti-WTA antibodies or MBL bind have not been determined. To answer this question, several S. aureus mutants, such as α-GlcNAc glycosyltransferase-deficient S. aureus ΔtarM, ß-GlcNAc glycosyltransferase-deficient ΔtarS, and ΔtarMS double mutant cells, were prepared from a laboratory and a community-associated methicillin-resistant S. aureus strain. Here, we describe the unexpected finding that ß-GlcNAc WTA-deficient ΔtarS mutant cells (which have intact α-GlcNAc) escape from anti-WTA antibody-mediated opsonophagocytosis, whereas α-GlcNAc WTA-deficient ΔtarM mutant cells (which have intact ß-GlcNAc) are efficiently engulfed by human leukocytes via anti-WTA IgG. Likewise, MBL binding in S. aureus cells was lost in the ΔtarMS double mutant but not in either single mutant. When we determined the serum concentrations of the anti-α- or anti-ß-GlcNAc-specific WTA IgGs, anti-ß-GlcNAc WTA-IgG was dominant in pooled human IgG fractions and in the intact sera of healthy adults and infants. These data demonstrate the importance of the WTA sugar conformation for human innate and adaptive immunity against S. aureus infection.


Subject(s)
Antibodies, Bacterial/immunology , Cell Wall/immunology , Epitopes/immunology , Immunoglobulin G/immunology , Leukocytes/immunology , Mannose-Binding Lectin/immunology , Phagocytosis/immunology , Staphylococcus aureus/chemistry , Teichoic Acids/immunology , Adaptive Immunity/physiology , Adult , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Cell Wall/chemistry , Epitopes/chemistry , Female , Humans , Immunity, Innate/physiology , Infant , Infant, Newborn , Leukocytes/microbiology , Male , Mannose-Binding Lectin/blood , Mutation , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/immunology , Staphylococcus aureus/enzymology , Staphylococcus aureus/immunology , Teichoic Acids/chemistry
8.
J Immunol ; 191(6): 3319-27, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23966633

ABSTRACT

The human pathogen Staphylococcus aureus is responsible for many community-acquired and hospital-associated infections and is associated with high mortality. Concern over the emergence of multidrug-resistant strains has renewed interest in the elucidation of host mechanisms that defend against S. aureus infection. We recently demonstrated that human serum mannose-binding lectin binds to S. aureus wall teichoic acid (WTA), a cell wall glycopolymer--a discovery that prompted further screening to identify additional serum proteins that recognize S. aureus cell wall components. In this report, we incubated human serum with 10 different S. aureus mutants and determined that serum amyloid P component (SAP) bound specifically to a WTA-deficient S. aureus ΔtagO mutant, but not to tagO-complemented, WTA-expressing cells. Biochemical characterization revealed that SAP recognizes bacterial peptidoglycan as a ligand and that WTA inhibits this interaction. Although SAP binding to peptidoglycan was not observed to induce complement activation, SAP-bound ΔtagO cells were phagocytosed by human polymorphonuclear leukocytes in an FcγR-dependent manner. These results indicate that SAP functions as a host defense factor, similar to other peptidoglycan recognition proteins and nucleotide-binding oligomerization domain-like receptors.


Subject(s)
Carrier Proteins/immunology , Phagocytosis/immunology , Serum Amyloid P-Component/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Blotting, Western , Flow Cytometry , Humans
9.
PLoS One ; 8(8): e69739, 2013.
Article in English | MEDLINE | ID: mdl-23936347

ABSTRACT

The objectives of this study were to investigate the immune response to intradermal immunization with wall teichoic acid (WTA) and the effect of MBL deficiency in a murine model of infection with methicillin-resistant Staphylococcus aureus (MRSA). WTA is a bacterial cell wall component that is implicated in invasive infection. We tested susceptibility to MRSA infection in wild type (WT) and MBL deficient mice using two strains of MRSA: MW2, a community-associated MRSA (CA-MRSA); and COL, a healthcare-associated MRSA (HA-MRSA). We also performed in vitro assays to investigate the effects of anti-WTA IgG containing murine serum on complement activation and bacterial growth in whole blood. We found that MBL knockout (KO) mice are relatively resistant to a specific MRSA strain, MW2 CA-MRSA, compared to WT mice, while both strains of mice had similar susceptibility to a different strain, COL HA-MRSA. Intradermal immunization with WTA elicited and augmented an anti-WTA IgG response in both WT and MBL KO mice. WTA immunization significantly reduced susceptibility to both MW2 CA-MRSA and COL HA-MRSA, independent of the presence of MBL. The protective mechanisms of anti-WTA IgG are mediated at least in part by complement activation and clearance of bacteria from blood. The significance of these findings is that 1) Intradermal immunization with WTA induces production of anti-WTA IgG; and 2) This anti-WTA IgG response protects from infection with both MW2 CA-MRSA and COL HA-MRSA even in the absence of MBL, the deficiency of which is common in humans.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Cell Wall/immunology , Immunoglobulin G/immunology , Mannose-Binding Lectins/physiology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Infections/prevention & control , Teichoic Acids/pharmacology , Animals , Antibodies, Anti-Idiotypic/metabolism , Cell Wall/drug effects , Complement Activation , Female , Immunization , Immunoglobulin G/metabolism , Injections, Intradermal , Mannose-Binding Lectin/deficiency , Metabolism, Inborn Errors , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Teichoic Acids/immunology
10.
Proc Natl Acad Sci U S A ; 110(26): E2381-9, 2013 Jun 25.
Article in English | MEDLINE | ID: mdl-23757494

ABSTRACT

Many bacteria accumulate granules of polyhydroxyalkanoate (PHA) within their cells, which confer resistance to nutritional depletion and other environmental stresses. Here, we report an unexpected involvement of the bacterial endocellular storage polymer, PHA, in an insect-bacterium symbiotic association. The bean bug Riptortus pedestris harbors a beneficial and specific gut symbiont of the ß-proteobacterial genus Burkholderia, which is orally acquired by host nymphs from the environment every generation and easily cultivable and genetically manipulatable. Biochemical and cytological comparisons between symbiotic and cultured Burkholderia detected more PHA granules consisting of poly-3-hydroxybutyrate and associated phasin (PhaP) protein in the symbiotic Burkholderia. Among major PHA synthesis genes, phaB and phaC were disrupted by homologous recombination together with the phaP gene, whereby ΔphaB, ΔphaC, and ΔphaP mutants were generated. Both in culture and in symbiosis, accumulation of PHA granules was strongly suppressed in ΔphaB and ΔphaC, but only moderately in ΔphaP. In symbiosis, the host insects infected with ΔphaB and ΔphaC exhibited significantly lower symbiont densities and smaller body sizes. These deficient phenotypes associated with ΔphaB and ΔphaC were restored by complementation of the mutants with plasmids encoding a functional phaB/phaC gene. Retention analysis of the plasmids revealed positive selection acting on the functional phaB/phaC in symbiosis. These results indicate that the PHA synthesis genes of the Burkholderia symbiont are required for normal symbiotic association with the Riptortus host. In vitro culturing analyses confirmed vulnerability of the PHA gene mutants to environmental stresses, suggesting that PHA may play a role in resisting stress under symbiotic conditions.


Subject(s)
Burkholderia/genetics , Burkholderia/metabolism , Genes, Bacterial , Heteroptera/microbiology , Polyhydroxyalkanoates/biosynthesis , Polyhydroxyalkanoates/genetics , Symbiosis/genetics , Acyltransferases/genetics , Acyltransferases/metabolism , Amino Acid Sequence , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Digestive System/microbiology , Genetic Complementation Test , Molecular Sequence Data , Mutation , Phenotype , Stress, Physiological/genetics
11.
J Bone Miner Res ; 28(11): 2381-91, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23633269

ABSTRACT

Bacterial infection can cause inflammatory bone diseases accompanied by the bone destruction resulting from excess generation of osteoclasts. Although lipoproteins are one of the major immunostimulating components of bacteria, little is known about their effects on bone metabolism. In this study, we investigated the role of lipoproteins in bacteria-induced bone destruction using Staphylococcus aureus wild type, its lipoprotein-deficient mutant, and synthetic lipopeptides Pam2CSK4 and Pam3CSK4 known to mimic bacterial lipoproteins. Formaldehyde-inactivated S. aureus or the synthetic lipopeptides induced severe bone loss in the femurs of mice after intraperitoneal administration and in a calvarial bone implantation model, whereas the lipoprotein-deficient S. aureus did not show such effects. Mechanism studies further identified three action mechanisms for the lipopeptide-induced osteoclast differentiation and bone resorption via (i) enhancement of osteoclast differentiation through Toll-like receptor 2 and MyD88-dependent signaling pathways; (ii) induction of pro-inflammatory cytokines, TNF-α and IL-6; and (iii) upregulation of RANKL expression with downregulation of osteoprotegerin expression in osteoblasts. Taken together, these results suggest that lipoprotein might be an important bacterial component responsible for bone destruction during bacterial infections through augmentation of osteoclast differentiation and activation.


Subject(s)
Bone Resorption/microbiology , Bone Resorption/pathology , Cell Differentiation/drug effects , Lipoproteins/pharmacology , Osteoclasts/pathology , Staphylococcus aureus/chemistry , Animals , Bone Resorption/diagnostic imaging , Bone Resorption/genetics , DNA/metabolism , Down-Regulation/drug effects , Enzyme Activation/drug effects , Interleukin-6/metabolism , Lipopeptides/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Mutation/genetics , Myeloid Differentiation Factor 88/metabolism , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoprotegerin/metabolism , Protein Binding/drug effects , RANK Ligand/metabolism , Radiography , Skull/diagnostic imaging , Skull/drug effects , Skull/pathology , Toll-Like Receptor 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
12.
J Immunol ; 189(12): 5903-11, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23152562

ABSTRACT

The innate immune system has developed to acquire a wide variety of pattern-recognition receptors (PRRs) to identify potential pathogens, whereas pathogens have also developed to escape host innate immune responses. ITIM-bearing receptors are attractive targets for pathogens to attenuate immune responses against them; however, the in vivo role of the inhibitory PRRs in host-bacteria interactions remains unknown. We demonstrate in this article that Staphylococcus aureus, a major Gram-positive bacteria, exploits inhibitory PRR paired Ig-like receptor (PIR)-B on macrophages to suppress ERK1/2 and inflammasome activation, and subsequent IL-6 and IL-1ß secretion. Consequently, Pirb(-/-) mice infected with S. aureus showed enhanced inflammation and more effective bacterial clearance, resulting in resistance to the sepsis. Screening of S. aureus mutants identified lipoteichoic acid (LTA) as an essential bacterial cell wall component required for binding to PIR-B and modulating inflammatory responses. In vivo, however, an LTA-deficient S. aureus mutant was highly virulent and poorly recognized by macrophages in both wild-type and Pirb(-/-) mice, demonstrating that LTA recognition by PRRs other than PIR-B mediates effective bacterial elimination. These results provide direct evidence that bacteria exploit the inhibitory receptor for virulence, and host immune system counterbalances the infection.


Subject(s)
Receptors, Immunologic/physiology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Virulence/immunology , Animals , Down-Regulation/immunology , Female , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Inflammasomes/metabolism , MAP Kinase Signaling System/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Receptors, Immunologic/deficiency , Receptors, Immunologic/genetics
13.
FEBS J ; 279(23): 4247-68, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23094979

ABSTRACT

Bacterial lipoproteins are characterized by the presence of a conserved N-terminal lipid-modified cysteine residue that allows the hydrophilic protein to anchor onto bacterial cell membranes. These proteins play important roles in a wide variety of bacterial physiological processes, including virulence, and induce innate immune reactions by functioning as ligands of the mammalian Toll-like receptor 2. We review recent advances in our understanding of bacterial lipoprotein structure, biosynthesis and structure-function relationships between bacterial lipoproteins and Toll-like receptor 2. Notably, 40 years after the first report of the triacyl structure of Braun's lipoprotein in Escherichia coli, recent intensive MS-based analyses have led to the discovery of three new lipidated structures of lipoproteins in monoderm bacteria: the lyso, N-acetyl and peptidyl forms. Moreover, the bacterial lipoprotein structure is considered to be constant in each bacterium; however, lipoprotein structures in Staphylococcus aureus vary between the diacyl and triacyl forms depending on the environmental conditions. Thus, the lipidation state of bacterial lipoproteins, particularly in monoderm bacteria, is more complex than previously assumed.


Subject(s)
Bacteria/metabolism , Lipoproteins/metabolism , Bacteria/genetics , Gram-Positive Bacteria/genetics , Gram-Positive Bacteria/metabolism , Lipoproteins/genetics , Protein Processing, Post-Translational , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism
14.
J Immunol ; 189(10): 4951-9, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23071283

ABSTRACT

Wall teichoic acid (WTA) of Staphylococcus aureus is a major cell envelope-associated glycopolymer that is a key molecule in promoting colonization during S. aureus infection. The complement system plays a key role in the opsonization and clearance of pathogens. We recently reported that S. aureus WTA functions as a ligand of human serum mannose-binding lectin (MBL), a recognition molecule of the lectin complement pathway. Intriguingly, serum MBL in adults does not bind to WTA because of an inhibitory effect of serum anti-WTA-IgG. In this study, serum anti-WTA-IgG was purified to homogeneity using a purified S. aureus WTA-coupled affinity column to examine the biological function of human anti-WTA-IgG. The purified anti-WTA-IgG contained the IgG2 subclass as a major component and specifically induced C4 and C3 deposition on the S. aureus surface in the anti-WTA-IgG-depleted serum, but not in C1q-deficient serum. Furthermore, the anti-WTA-IgG-dependent C3 deposition induced phagocytosis of S. aureus cells by human polymorphonuclear leukocytes. These results demonstrate that serum anti-WTA-IgG is a real trigger for the induction of classical complement-dependent opsonophagocytosis against S. aureus. Our results also support the fact that a lack of the lectin complement pathway in MBL-deficient adults is compensated by Ag-specific, Ab-mediated adaptive immunity.


Subject(s)
Antibodies, Bacterial/immunology , Cell Wall/immunology , Immunoglobulin G/immunology , Neutrophils/immunology , Phagocytosis/immunology , Staphylococcus aureus/immunology , Teichoic Acids/immunology , Adult , Antigen-Antibody Complex/immunology , Complement C3/immunology , Complement C4/immunology , Complement Pathway, Classical/immunology , Humans , Neutrophils/cytology
15.
J Bacteriol ; 194(13): 3299-306, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22467779

ABSTRACT

Bacterial lipoproteins are believed to exist in only one specific lipid-modified structure, such as the diacyl form or the triacyl form, in each bacterium. In the case of Staphylococcus aureus, recent extensive matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry analysis revealed that S. aureus lipoproteins exist in the α-aminoacylated triacyl form. Here, we discovered conditions that induce the accumulation of diacyl lipoproteins that lack α-aminoacylation in S. aureus. The accumulation of diacyl lipoproteins required a combination of conditions, including acidic pH and a post-logarithmic-growth phase. High temperatures and high salt concentrations additively accelerated the accumulation of the diacyl lipoprotein form. Following a post-logarithmic-growth phase where S. aureus MW2 cells were grown at pH 6, SitC lipoprotein was found almost exclusively in its diacyl structure rather than in its triacyl structure. This is the first report showing that the environment mediates lipid-modified structural alterations of bacterial lipoproteins.


Subject(s)
Gene Expression Regulation, Bacterial , Lipoproteins/metabolism , Methicillin-Resistant Staphylococcus aureus/growth & development , Staphylococcus aureus/growth & development , Acylation , Anti-Bacterial Agents/pharmacology , Culture Media/chemistry , Hot Temperature , Humans , Hydrogen-Ion Concentration , Lipoproteins/chemistry , Lipoproteins/genetics , Methicillin/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/metabolism , Methicillin-Resistant Staphylococcus aureus/physiology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Staphylococcus aureus/physiology
16.
J Biol Chem ; 287(16): 13170-81, 2012 Apr 13.
Article in English | MEDLINE | ID: mdl-22303020

ABSTRACT

Bacterial lipoproteins/lipopeptides inducing host innate immune responses are sensed by mammalian Toll-like receptor 2 (TLR2). These bacterial lipoproteins are structurally divided into two groups, diacylated or triacylated lipoproteins, by the absence or presence of an amide-linked fatty acid. The presence of diacylated lipoproteins has been predicted in low-GC content gram-positive bacteria and mycoplasmas based on the absence of one modification enzyme in their genomes; however, we recently determined triacylated structures in low-GC gram-positive Staphylococcus aureus, raising questions about the actual lipoprotein structure in other low-GC content gram-positive bacteria. Here, through intensive MS analyses, we identified a novel and unique bacterial lipoprotein structure containing an N-acyl-S-monoacyl-glyceryl-cysteine (named the lyso structure) from low-GC gram-positive Enterococcus faecalis, Bacillus cereus, Streptococcus sanguinis, and Lactobacillus bulgaricus. Two of the purified native lyso-form lipoproteins induced proinflammatory cytokine production from mice macrophages in a TLR2-dependent and TLR1-independent manner but with a different dependence on TLR6. Additionally, two other new lipoprotein structures were identified. One is the "N-acetyl" lipoprotein structure containing N-acetyl-S-diacyl-glyceryl-cysteine, which was found in five gram-positive bacteria, including Bacillus subtilis. The N-acetyl lipoproteins induced the proinflammatory cytokines through the TLR2/6 heterodimer. The other was identified in a mycoplasma strain and is an unusual diacyl lipoprotein structure containing two amino acids before the lipid-modified cysteine residue. Taken together, our results suggest the existence of novel TLR2-stimulating lyso and N-acetyl forms of lipoproteins that are conserved in low-GC content gram-positive bacteria and provide clear evidence for the presence of yet to be identified key enzymes involved in the bacterial lipoprotein biosynthesis.


Subject(s)
Gram-Positive Bacteria/immunology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/microbiology , Toll-Like Receptor 2/immunology , Toll-Like Receptor 2/metabolism , Animals , Bacillus cereus/immunology , Bacillus cereus/metabolism , Bacillus subtilis/immunology , Bacillus subtilis/metabolism , Enterococcus faecalis/immunology , Enterococcus faecalis/metabolism , Geobacillus/immunology , Geobacillus/metabolism , Gram-Positive Bacteria/metabolism , Lactobacillus/immunology , Lactobacillus/metabolism , Mice , Mice, Inbred C57BL , Pneumonia, Mycoplasma/immunology , Pneumonia, Mycoplasma/metabolism , Streptococcus sanguis/immunology , Streptococcus sanguis/metabolism , Toll-Like Receptor 1/genetics , Toll-Like Receptor 1/immunology , Toll-Like Receptor 1/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 6/genetics , Toll-Like Receptor 6/immunology , Toll-Like Receptor 6/metabolism
17.
Dev Comp Immunol ; 36(3): 540-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22001126

ABSTRACT

The biochemical characterization of novel antimicrobial peptides (AMPs) and the determination of ligand molecules that induce AMP production are essential for understanding the host innate immune response in insects. Here, we purified a new 14-kDa AMP, named tenecin 4, from the larval hemolymph of the beetle Tenebrio molitor. Tenecin 4 contains 14% glycine residues and has moderate similarities both to the C-terminal region of Drosophila attacin and to silk-moth gloverin proteins. Purified tenecin 4 showed bactericidal activity against Gram-negative Escherichia coli but not against Gram-positive Bacillus subtilis or the fungus Candida albicans. Tenecin 4 production was induced by Toll cascade-activating ligands, such as ß-1,3-glucan, lysine-type peptidoglycan and active Spätzle, and by the probable Imd pathway-activating ligand monomeric meso-diaminopimelic acid-type peptidoglycan. Taken together, these data show that tenecin 4 is a defense protein against Gram-negative pathogens and is induced by multiple ligands in Tenebrio larvae.


Subject(s)
Antimicrobial Cationic Peptides/isolation & purification , Antimicrobial Cationic Peptides/pharmacology , Insect Proteins/isolation & purification , Insect Proteins/pharmacology , Tenebrio/immunology , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/immunology , Bacillus subtilis/drug effects , Candida albicans/drug effects , Cloning, Molecular , Escherichia coli/drug effects , Fat Body/immunology , Fat Body/metabolism , Hemocytes/immunology , Hemocytes/metabolism , Hemolymph/chemistry , Hemolymph/immunology , Immunity, Innate , Insect Proteins/chemistry , Insect Proteins/immunology , Larva/chemistry , Larva/immunology , Molecular Sequence Data , Tenebrio/chemistry , Tenebrio/growth & development , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism
18.
J Biol Chem ; 286(40): 35087-95, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21862574

ABSTRACT

Serpins are protease inhibitors that play essential roles in the down-regulation of extracellular proteolytic cascades. The core serpin domain is highly conserved, and typical serpins are encoded with a molecular size of 35-50 kDa. Here, we describe a novel 93-kDa protein that contains two complete, tandemly arrayed serpin domains. This twin serpin, SPN93, was isolated from the larval hemolymph of the large beetle Tenebrio molitor. The N-terminal serpin domain of SPN93 forms a covalent complex with the Spätzle-processing enzyme, a terminal serine protease of the Toll signaling cascade, whereas the C-terminal serpin domain of SPN93 forms complexes with a modular serine protease and the Spätzle-processing enzyme-activating enzyme, which are two different enzymes of the cascade. Consequently, SPN93 inhibited ß-1,3-glucan-mediated Toll proteolytic cascade activation in an in vitro system. Site-specific proteolysis of SPN93 at the N-terminal serpin domain was observed after activation of the Toll proteolytic cascade in vivo, and down-regulation of SPN93 by RNAi sensitized ß-1,3-glucan-mediated larval death. Therefore, SPN93 is the first serpin that contains twin tandemly arrayed and functionally active serpin domains that have a regulatory role in the larval Toll proteolytic signaling cascade.


Subject(s)
Serine Proteinase Inhibitors/chemistry , Serpins/chemistry , Serpins/metabolism , Toll-Like Receptors/metabolism , Animals , Chromatography/methods , Cloning, Molecular , Coleoptera , Humans , Melanins/chemistry , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA Interference , Signal Transduction
19.
Dev Comp Immunol ; 35(8): 835-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21453720

ABSTRACT

We recently reported that D-alanylation of Staphylococcus aureus wall teichoic acid (WTA) mitigates an induction of the Toll-mediated humoral response in Drosophila by interfering with peptidoglycan (PG) recognition by PG recognition protein-SA (PGRP-SA). Here, we investigated the mode of this interference by using an in vitro cell free system from larvae of the coleoptran insect Tenebrio molitor. WTA modification on PG had a potent inhibitory effect on PGRP-SA-mediated Toll proteolytic cascade activation, and the D-alanylation of WTA enhanced its inhibitory effect. Purified D-alanylated WTA released from PG lost its inhibitory action on both Toll cascade activation and PGRP-SA binding to insoluble PG. The inhibition of PGRP-SA binding to PG by D-alanylated WTA took place not only on polymeric PG but also on WTA-attached disaccharide units of monomeric PG. These results suggest that D-alanylation-mediated evasion requires the covalent bonding of D-alanylated WTA to PG, but not net-like cross-linking structure of PG.


Subject(s)
Cell Wall/metabolism , Immune Evasion , Immunity, Innate , Larva/immunology , Peptidoglycan/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/metabolism , Teichoic Acids/metabolism , Tenebrio/immunology , Alanine/chemistry , Amidohydrolases/chemistry , Amidohydrolases/metabolism , Animals , Carrier Proteins/chemistry , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cell Wall/chemistry , Cell Wall/immunology , Cell-Free System , Enzyme Assays , Insect Proteins/chemistry , Insect Proteins/immunology , Insect Proteins/metabolism , Peptidoglycan/chemistry , Peptidoglycan/immunology , Protein Binding , Signal Transduction/immunology , Staphylococcus aureus/immunology , Teichoic Acids/chemistry , Teichoic Acids/immunology , Toll-Like Receptors/chemistry , Toll-Like Receptors/metabolism
20.
Biochem Biophys Res Commun ; 406(3): 449-53, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21333631

ABSTRACT

Cytokine responses to microbes are triggered by pattern recognition receptors, such as Toll-like receptors (TLRs), which sense pathogen-associated molecular patterns. Cell wall-associated triacylated lipoproteins in Staphylococcus aureus are known to be native TLR2 ligands that mediate host inflammatory responses against S. aureus. However, the mechanism by which these lipidated lipoproteins, which are buried under the thick S. aureus cell wall, work to stimulate TLR2 remains unclear. Heat-killed wild type S. aureus cells activated human monocytic THP-1 cells to produce proinflammatory cytokines, including interleukin (IL)-8, whereas the lipoprotein lipidation-deficient lgt mutant induced less than an eighth of the amount of IL-8 induced by the wild type. IL-8 induction in response to heat-killed S. aureus cells in THP-1 cells was not inhibited by a blocking antibody against cell surface TLR2, suggesting that intracellular TLR2 might be involved in the induction of IL-8 by S. aureus lipoprotein. The relationship between phagocytosis and IL-8 production in THP-1 cells was analyzed on a single-cell level by flow cytometry using fluorescein-labeled S. aureus cells and phycoerythrin-labeled anti-IL-8 antibody. Production of intracellular IL-8 was correlated with phagocytosis of S. aureus cells in THP-1 cells and in human peripheral blood mononuclear cells. Opsonization of S. aureus cells enhanced both the phagocytosis of S. aureus cells and the production of intracellular IL-8 in THP-1 cells. These results suggest that lipidated lipoproteins on S. aureus cells stimulate human monocytes after phagocytosis.


Subject(s)
Interleukin-8/immunology , Lipoproteins/immunology , Monocytes/immunology , Phagocytosis , Staphylococcus aureus/immunology , Toll-Like Receptor 2/immunology , Antibodies, Blocking/immunology , Cell Line , Humans , Ligands , Lipoproteins/genetics , Staphylococcus aureus/genetics , Toll-Like Receptor 2/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...