Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
PLoS Pathog ; 13(9): e1006461, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28880920

ABSTRACT

Bacterial superantigens (SAgs) cause Vß-dependent T-cell proliferation leading to immune dysregulation associated with the pathogenesis of life-threatening infections such as toxic shock syndrome, and necrotizing pneumonia. Previously, we demonstrated that staphylococcal enterotoxin-like toxin X (SElX) from Staphylococcus aureus is a classical superantigen that exhibits T-cell activation in a Vß-specific manner, and contributes to the pathogenesis of necrotizing pneumonia. Here, we discovered that SElX can also bind to neutrophils from human and other mammalian species and disrupt IgG-mediated phagocytosis. Site-directed mutagenesis of the conserved sialic acid-binding motif of SElX abolished neutrophil binding and phagocytic killing, and revealed multiple glycosylated neutrophil receptors for SElX binding. Furthermore, the neutrophil binding-deficient mutant of SElX retained its capacity for T-cell activation demonstrating that SElX exhibits mechanistically independent activities on distinct cell populations associated with acquired and innate immunity, respectively. Finally, we demonstrated that the neutrophil-binding activity rather than superantigenicity is responsible for the SElX-dependent virulence observed in a necrotizing pneumonia rabbit model of infection. Taken together, we report the first example of a SAg, that can manipulate both the innate and adaptive arms of the human immune system during S. aureus pathogenesis.


Subject(s)
Enterotoxins/metabolism , Exfoliatins/pharmacology , Neutrophils/drug effects , Staphylococcal Infections , Superantigens/pharmacology , Animals , Exfoliatins/metabolism , Humans , Lymphocyte Activation/immunology , Neutrophils/immunology , Rabbits , Receptors, Antigen, T-Cell, alpha-beta/immunology , Staphylococcus aureus/chemistry , Staphylococcus aureus/metabolism , Superantigens/immunology
2.
PLoS Pathog ; 12(6): e1005696, 2016 06.
Article in English | MEDLINE | ID: mdl-27280885

ABSTRACT

Mycobacteria produce a capsule layer, which consists of glycan-like polysaccharides and a number of specific proteins. In this study, we show that, in slow-growing mycobacteria, the type VII secretion system ESX-5 plays a major role in the integrity and stability of the capsule. We have identified PPE10 as the ESX-5 substrate responsible for this effect. Mutants in esx-5 and ppe10 both have impaired capsule integrity as well as reduced surface hydrophobicity. Electron microscopy, immunoblot and flow cytometry analyses demonstrated reduced amounts of surface localized proteins and glycolipids, and morphological differences in the capsular layer. Since capsular proteins secreted by the ESX-1 system are important virulence factors, we tested the effect of the mutations that cause capsular defects on virulence mechanisms. Both esx-5 and ppe10 mutants of Mycobacterium marinum were shown to be impaired in ESX-1-dependent hemolysis. In agreement with this, the ppe10 and esx5 mutants showed reduced recruitment of ubiquitin in early macrophage infection and intermediate attenuation in zebrafish embryos. These results provide a pivotal role for the ESX-5 secretion system and its substrate PPE10, in the capsular integrity of pathogenic mycobacteria. These findings open up new roads for research on the mycobacterial capsule and its role in virulence and immune modulation.


Subject(s)
Bacterial Capsules/metabolism , Mycobacterium Infections, Nontuberculous/metabolism , Mycobacterium marinum/pathogenicity , Type VII Secretion Systems/metabolism , Virulence/physiology , Animals , Cell Line , Chromatography, Thin Layer , Disease Models, Animal , Flow Cytometry , Humans , Immunoblotting , Microscopy, Electron , Mycobacterium marinum/metabolism , Virulence Factors/metabolism , Zebrafish
3.
Front Immunol ; 5: 467, 2014.
Article in English | MEDLINE | ID: mdl-25309547

ABSTRACT

Initial elimination of invading Staphylococcus aureus from the body is mediated by professional phagocytes. The neutrophil is the major phagocyte of the innate immunity and plays a key role in the host defense against staphylococcal infections. Opsonization of the bacteria with immunoglobulins and complement factors enables efficient recognition by the neutrophil that subsequently leads to intracellular compartmentalization and killing. Here, we provide a review of the key processes evolved in neutrophil-mediated phagocytosis of S. aureus and briefly describe killing. As S. aureus is not helpless against the professional phagocytes, we will also highlight its immune evasion arsenal related to phagocytosis.

4.
Cell Microbiol ; 16(11): 1646-65, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24840181

ABSTRACT

In order to cause colonization and invasive disease, pathogenic bacteria secrete proteins that modulate host immune defences. Identification and characterization of these proteins leads to a better understanding of the pathological processes underlying infectious and inflammatory diseases and is essential in the development of new strategies for their prevention and treatment. Current techniques to functionally characterize these proteins are laborious and inefficient. Here we describe a high-throughput functional selection strategy using phage display in order to identify immune evasion proteins. Using this technique we identified two previously uncharacterized proteins secreted by Staphylococcus aureus, SElX and SSL6 that bind to neutrophil surface receptors. SElX binds PSGL-1 on neutrophils and thereby inhibits the interaction between PSGL-1 and P-selectin, a crucial step in the recruitment of neutrophils to the site of infection. SSL6 is the first bacterial protein identified that binds CD47, a widely expressed cell surface protein recently described as an interesting target in anti-cancer therapy. Our findings provide new insights into the pathogenesis of S. aureus infections and support phage display as an efficient method to identify bacterial secretome proteins interacting with humoral or cellular immune components.


Subject(s)
Bacterial Proteins/metabolism , Cell Surface Display Techniques/methods , Host-Pathogen Interactions , Membrane Proteins/metabolism , Neutrophils/microbiology , Staphylococcus aureus/physiology , CD47 Antigen , Membrane Glycoproteins
5.
Traffic ; 14(10): 1029-41, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23837619

ABSTRACT

Macroautophagy (hereafter referred to as autophagy) is an evolutionarily conserved intracellular catabolic transport route that generally allows the lysosomal degradation of cytoplasmic components, including bulk cytosol, protein aggregates, damaged or superfluous organelles and invading microbes. Target structures are sequestered by double-membrane vesicles called autophagosomes, which are formed through the concerted action of the autophagy (ATG)-related proteins. Until recently it was assumed that ATG proteins were exclusively involved in autophagy. A growing number of studies, however, have attributed functions to some of them that are distinct from their classical role in autophagosome biogenesis. Autophagy-independent roles of the ATG proteins include the maintenance of cellular homeostasis and resistance to pathogens. For example, they assist and enhance the turnover of dead cells and microbes upon their phagocytic engulfment, and inhibit murine norovirus replication. Moreover, bone resorption by osteoclasts, innate immune regulation triggered by cytoplasmic DNA and the ER-associated degradation regulation all have in common the requirement of a subset of ATG proteins. Microorganisms such as coronaviruses, Chlamydia trachomatis or Brucella abortus have even evolved ways to manipulate autophagy-independent functions of ATG proteins in order to ensure the completion of their intracellular life cycle. Taken together these novel mechanisms add to the repertoire of functions and extend the number of cellular processes involving the ATG proteins.


Subject(s)
Autophagy/physiology , Proteins/metabolism , Animals , Homeostasis/physiology , Humans , Phagosomes/metabolism , Phagosomes/physiology
6.
Mol Microbiol ; 86(2): 472-84, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22925462

ABSTRACT

Pathogenic mycobacteria require type VII secretion (T7S) systems to transport virulence factors across their complex cell envelope. These bacteria have up to five of these systems, termed ESX-1 to ESX-5. Here, we show that ESX-5 of Mycobacterium tuberculosis mediates the secretion of EsxN, PPE and PE_PGRS proteins, indicating that ESX-5 is a major secretion pathway in this important pathogen. Using the ESX-5 system of Mycobacterium marinum and Mycobacterium bovis BCG as a model, we have purified and analysed the T7S membrane complex under native conditions. blue native-PAGE and immunoprecipitation experiments showed that the ESX-5 membrane complex of both species has a size of ~ 1500 kDa and is composed of four conserved membrane proteins, i.e. EccB(5) , EccC(5) , EccD(5) and EccE(5) . Subsequent limited proteolysis suggests that EccC(5) and EccE(5) mostly reside on the periphery of the complex. We also observed that EccC(5) and EccD(5) expression is essential for the formation of a stable membrane complex. These are the first data on a T7S membrane complex and, given the high conservation of its components, our data can likely be generalized to most mycobacterial T7S systems.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Secretion Systems , Cell Membrane/metabolism , Mycobacterium marinum/metabolism , Mycobacterium tuberculosis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Cell Membrane/chemistry , Cell Membrane/genetics , Mycobacterium marinum/chemistry , Mycobacterium marinum/genetics , Mycobacterium tuberculosis/chemistry , Mycobacterium tuberculosis/genetics , Protein Structure, Tertiary , Protein Transport
7.
J Immunol ; 187(9): 4744-53, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21957139

ABSTRACT

During infection of humans and animals, pathogenic mycobacteria manipulate the host cell causing severe diseases such as tuberculosis and leprosy. To understand the basis of mycobacterial pathogenicity, it is crucial to identify the molecular virulence mechanisms. In this study, we address the contribution of ESX-1 and ESX-5--two homologous type VII secretion systems of mycobacteria that secrete distinct sets of immune modulators--during the macrophage infection cycle. Using wild-type, ESX-1- and ESX-5-deficient mycobacterial strains, we demonstrate that these secretion systems differentially affect subcellular localization and macrophage cell responses. We show that in contrast to ESX-1, the effector proteins secreted by ESX-5 are not required for the translocation of Mycobacterium tuberculosis or Mycobacterium marinum to the cytosol of host cells. However, the M. marinum ESX-5 mutant does not induce inflammasome activation and IL-1ß activation. The ESX-5 system also induces a caspase-independent cell death after translocation has taken place. Importantly, by means of inhibitory agents and small interfering RNA experiments, we reveal that cathepsin B is involved in both the induction of cell death and inflammasome activation upon infection with wild-type mycobacteria. These results reveal distinct roles for two different type VII secretion systems during infection and shed light on how virulent mycobacteria manipulate the host cell in various ways to replicate and spread.


Subject(s)
Homeodomain Proteins/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Membrane Transport Proteins/metabolism , Mycobacterium marinum/immunology , Mycobacterium tuberculosis/immunology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Death/immunology , Cell Line , Cell Line, Tumor , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Interleukin-1beta/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/microbiology , Mice , Mycobacterium marinum/pathogenicity , Mycobacterium tuberculosis/pathogenicity
8.
Cell Microbiol ; 12(10): 1506-16, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20545943

ABSTRACT

The human pathogen Staphylococcus aureus has a plethora of virulence factors that promote its colonization and survival in the host. Among such immune modulators are staphylococcal superantigen-like (SSL) proteins, comprising a family of 14 small, secreted molecules that seem to interfere with the host innate immune system. SSL7 has been described to bind immunoglobulin A (IgA) and complement C5, thereby inhibiting IgA-FcαRI binding and serum killing of Escherichia coli. As C5a generation, in contrast to C5b-9-mediated lysis, is crucial for immune defence against staphylococci, we investigated the impact of SSL7 on staphylococcal-induced C5a-mediated effects. Here, we show that SSL7 inhibits C5a generation induced by staphylococcal opsonization, slightly enhanced by its IgA-binding capacity. Moreover, we demonstrate a strong protective activity of SSL7 against staphylococcal clearance in human whole blood. SSL7 strongly inhibited the C5a-induced phagocytosis of S. aureus and oxidative burst in an in vitro whole-blood inflammation model. Furthermore, we found that SSL7 affects all three pathways of complement activation and inhibits the cleavage of C5 by interference of its binding to C5 convertases. Finally, SSL7 effects were also demonstrated in vivo. In a murine model of immune complex peritonitis, SSL7 abrogated the C5a-driven influx of neutrophils in mouse peritoneum.


Subject(s)
Complement C5a/immunology , Exotoxins/immunology , Immune Evasion , Staphylococcus aureus/pathogenicity , Animals , Blood/immunology , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Peritonitis/immunology , Peritonitis/microbiology , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology
9.
Cell Oncol ; 32(1-2): 1-10, 2010.
Article in English | MEDLINE | ID: mdl-20208131

ABSTRACT

Bacterial proteins provide promising tools for novel anticancer therapies. Staphylococcal superantigen-like 5 (SSL5) was recently described to bind P-selectin glycoprotein ligand-1 (PSGL-1) on leukocytes and to inhibit neutrophil rolling on a P-selectin surface. As leukocytes and tumor cells share many characteristics in migration and dissemination, we explored the potential of SSL5 as an antagonist of malignant cell behavior. Previously, it was demonstrated that rolling of human HL-60 leukemia cells on activated endothelial cells was mediated by P-selectin. In this study, we show that SSL5 targets HL-60 cells. Binding of SSL5 was rapid and without observed toxicity. Competition of SSL5 with the binding of three anti-PSGL-1 antibodies and P-selectin to HL-60 cells identified PSGL-1 as the ligand on HL-60 cells. Presence of sialyl Lewis x epitopes on PSGL-1 was crucial for its interaction with SSL5. Importantly, SSL5 not only inhibited the interaction of HL-60 cells with activated endothelial cells but also with platelets, which both play an important role in growth and metastasis of cancers. These data support the concept that SSL5 could be a lead in the search for novel strategies against hematological malignancies.


Subject(s)
Bacterial Proteins/metabolism , Blood Platelets/physiology , Down-Regulation , Endothelial Cells/physiology , Leukemia/physiopathology , Staphylococcus aureus/metabolism , Superantigens/metabolism , Bacterial Proteins/genetics , Cell Adhesion , Cells, Cultured , HL-60 Cells , Humans , Leukemia/immunology , Leukemia/metabolism , Neutrophils/physiology , Protein Binding , Staphylococcus aureus/genetics , Superantigens/genetics
10.
FEMS Microbiol Rev ; 34(3): 395-414, 2010 May.
Article in English | MEDLINE | ID: mdl-20059549

ABSTRACT

Microorganisms have developed several mechanisms to modulate the host immune system to increase their survival and propagation in the host. Their presence in the host is not only revealed by self-produced peptides but also through host-derived chemokines and active complement fragments. These so-called chemoattractants are recognized by G protein-coupled receptors (GPCRs) expressed on leukocyte cell membranes. Activation of GPCRs triggers leukocyte activation and guides their recruitment to the site of infection. Therefore, GPCRs play a central role in leukocyte trafficking leading to microbial clearance. It is therefore not surprising that microorganisms are able to sabotage this arm of the immune response. Different microorganisms have evolved a variety of tactics to modulate GPCR activation. Here, we review the mechanisms and proteins used by major human pathogens and less virulent microorganisms that affect GPCR signaling. While viruses generally produce receptor and chemoattractant mimics, parasites and bacteria such as Staphylococcus aureus, Streptococcus pyogenes, Porphyromonas gingivalis, and Bordetella pertussis secrete proteins that affect receptor signaling, directly antagonize receptors, cleave stimuli, and even prevent stimulus generation. As the large arsenal of GPCR modulators aids prolonged microbial persistence in the host, their study provides us a better understanding of microbial pathogenesis.


Subject(s)
Cell Movement , Gram-Negative Bacteria/immunology , Gram-Positive Bacteria/immunology , Immune Evasion , Phagocytes/physiology , Virulence Factors/metabolism , Animals , Gram-Negative Bacteria/metabolism , Gram-Negative Bacteria/pathogenicity , Gram-Positive Bacteria/metabolism , Gram-Positive Bacteria/pathogenicity , Humans , Phagocytes/immunology , Virulence
11.
J Immunol ; 183(10): 6569-78, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19846866

ABSTRACT

The members of the formyl peptide receptor (FPR) family are involved in the sensing of chemoattractant substances, including bacteria-derived N-formylated peptides and host-derived peptides and proteins. We have recently described two chemoattractant receptor inhibitors from Staphylococcus aureus. Chemotaxis inhibitory protein of S. aureus (CHIPS) blocks the formyl peptide receptor (FPR) and the receptor for complement C5a (C5aR), while FPR-like 1 (FPRL1) inhibitory protein (FLIPr) blocks the FPRL1. Here, we describe another staphylococcal chemoattractant-inhibiting protein with 73% overall homology to FLIPr and identical first 25 aa, which we termed FLIPr-like. This protein inhibits neutrophil calcium mobilization and chemotaxis induced by the FPRL1-ligand MMK-1 and FPR-ligand fMLP. While its FPRL1-inhibitory activity lies in the comparable nanomolar range of FLIPr, its antagonism of the FPR is approximately 100-fold more potent than that of FLIPr and comparable to that of CHIPS. The second N-terminal phenylalanine was required for its inhibition of the FPR, but it was dispensable for the FPRL1. Furthermore, the deletion of the first seven amino acids reduced its antagonism of the FPRL1, and the exchange of the first six amino acids with that of CHIPS-conferred receptor specificity. Finally, studies with cells transfected with several chemoattractant receptors confirmed that FLIPr-like specifically binds to the FPR and FPRL1. In conclusion, the newly described excreted protein from S. aureus, FLIPr-like, is a potent inhibitor of the FPR- and FPRL1-mediated neutrophil responses and may be used to selectively modulate these chemoattractant receptors.


Subject(s)
Bacterial Proteins/pharmacology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Chemotaxis/drug effects , Humans , Molecular Sequence Data , Neutrophils/drug effects , Neutrophils/immunology , Peptides/immunology , Peptides/metabolism , Receptor, Anaphylatoxin C5a/immunology , Receptor, Anaphylatoxin C5a/metabolism , Receptors, Formyl Peptide/immunology , Receptors, Formyl Peptide/metabolism , Staphylococcus aureus/immunology
12.
Curr Pharm Des ; 15(29): 3396-416, 2009.
Article in English | MEDLINE | ID: mdl-19860687

ABSTRACT

Metastases are the cause of 90% of human cancer deaths. The current treatment of cancer with chemo,- and/or radiotherapy is based on cell death by DNA damage neglecting the fact that cancer cell invasion into surrounding tissues and metastasizing are fundamental features of neoplasms and the major reason for treatment failure. Metastasis is the result of several sequential steps and represents a highly organized, non-random, and organ-selective process. A number of in vitro and in vivo models show that tumor cells use chemokine-mediated mechanisms during this metastasizing process, comparable to those observed in the regulation of leukocyte trafficking. Furthermore, chemokines modulate tumor behavior such as the regulation of tumor-associated angiogenesis, activation of host tumor-specific immunological responses, and direct stimulation of tumor cell proliferation in an autocrine fashion. These findings may lead to new drugs that target chemokines or their receptors and will likely be of great additional value for treatment of cancer patients.


Subject(s)
Chemokines/physiology , Neoplasms/metabolism , Receptors, Chemokine/physiology , Animals , Chemokines/genetics , Female , Humans , Neoplasms/genetics , Pregnancy , Receptors, Chemokine/genetics , Receptors, G-Protein-Coupled/physiology
13.
Neoplasia ; 11(4): 333-44, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19308288

ABSTRACT

PURPOSE: Tumor cell migration and metastasis share many similarities with leukocyte trafficking, which is critically regulated by chemokines and their receptors. CXCR4 is the most widely expressed chemokine receptor in many different types of cancer and has been linked to tumor dissemination and poor prognosis. Several CXCR4 antagonists have been synthesized. A totally novel approach to discover chemokine receptor antagonists is the use of bacteria. Bacteria produce chemokine receptor inhibitors to prevent neutrophil extravasation and migration toward the infection site to escape clearance by innate immune cells. The aim of the current study was to find and identify the mechanism of a bacterial protein that specifically targets CXCR4, a chemokine receptor shared by neutrophils and cancer cells. EXPERIMENTAL DESIGN: Several staphylococcal proteins were screened for their capacity to prevent binding of a function-blocking antibody against CXCR4. RESULTS: Staphylococcal superantigen-like 10 was found to bind CXCR4 expressed on human T acute lymphoblastic leukemia, lymphoma, and cervical carcinoma cell lines. It potently inhibited CXCL12-induced calcium mobilization and cell migration. CONCLUSIONS: Staphylococcal superantigen-like 10 is a potential lead in the development of new anticancer compounds preventing metastasis by targeting CXCR4.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Chemokine CXCL12/metabolism , Neoplasms/immunology , Staphylococcus/immunology , Superantigens/pharmacology , Blotting, Western , Cell Line, Tumor , Humans , Neoplasms/metabolism , Polymerase Chain Reaction , Receptors, CXCR4/antagonists & inhibitors , Transfection
14.
Blood ; 113(2): 328-37, 2009 Jan 08.
Article in English | MEDLINE | ID: mdl-18941109

ABSTRACT

Staphylococcus aureus secretes several virulence factors modulating immune responses. Staphylococcal superantigen-like (SSL) proteins are a family of 14 exotoxins with homology to superantigens, but with generally unknown function. Recently, we showed that SSL5 binds to P-selectin glycoprotein ligand 1 dependently of sialyl Lewis X and inhibits P-selectin-dependent neutrophil rolling. Here, we show that SSL5 potently and specifically inhibits leukocyte activation by anaphylatoxins and all classes of chemokines. SSL5 inhibited calcium mobilization, actin polymerization, and chemotaxis induced by chemokines and anaphylatoxins but not by other chemoattractants. Antibody competition experiments showed that SSL5 targets several chemokine and anaphylatoxin receptors. In addition, transfection studies showed that SSL5 binds glycosylated N-termini of all G protein-coupled receptors (GPCRs) but only inhibits stimuli of protein nature that require the receptor N-terminus for activation. Furthermore, SSL5 increased binding of chemokines to cells independent of chemokine receptors through their common glycosaminoglycan-binding site. Importance of glycans was shown for both GPCR and chemokine binding. Thus, SSL5 is an important immunomodulatory protein of S aureus that targets several crucial, initial stages of leukocyte extravasation. It is therefore a potential new antiinflammatory compound for diseases associated with chemoattractants and their receptors and disorders characterized by excessive recruitment of leukocytes.


Subject(s)
Anaphylatoxins/immunology , Bacterial Proteins/immunology , Exotoxins/immunology , Immunologic Factors/immunology , Leukocyte Rolling/immunology , Membrane Glycoproteins/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , Virulence Factors/immunology , Anaphylatoxins/metabolism , Bacterial Proteins/metabolism , Calcium Signaling/immunology , Cell Line , Chemokines/immunology , Chemokines/metabolism , Exotoxins/metabolism , Humans , Immunologic Factors/metabolism , Lewis X Antigen/analogs & derivatives , Membrane Glycoproteins/metabolism , Neutrophils/metabolism , Protein Binding/immunology , Receptors, Chemokine/immunology , Receptors, Chemokine/metabolism , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Trisaccharides/immunology , Trisaccharides/metabolism , Virulence Factors/metabolism
15.
Blood ; 109(7): 2936-43, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17132726

ABSTRACT

Staphylococcus aureus secretes several virulence factors interfering with host-cell functions. Staphylococcal superantigen-like (SSL) proteins are a family of 11 exotoxins with structural homology to superantigens but with generally unknown functions. Recently, we described that chemotaxis inhibitory protein of Staphylococcus aureus (CHIPS(31-121)), a potent inhibitor of C5a-induced responses, is structurally homologous to the C-terminal domain of SSL5. Here, we identify P-selectin glycoprotein ligand-1 (PSGL-1), involved in the initial rolling of neutrophils along the endothelium, as a target for SSL5. SSL5 specifically bound to Chinese hamster ovary cells stably expressing PSGL-1 (CHO-PSGL-1), which was dependent of sulfation and sialylation. Furthermore, SSL5 bound to PSGL-1/Ig fusion protein immobilized on a biosensor chip. SSL5 affected binding of soluble P-selectin/Fc chimera, the principle ligand of PSGL-1, to CHO-PSGL-1 cells and inhibited adhesion of neutrophils to immobilized P-selectin under static conditions. Under flow conditions SSL5 strongly decreased neutrophil rolling on immobilized P-selectin/Fc and activated human endothelial cells. In conclusion, SSL5 interferes with the interaction between PSGL-1 and P-selectin, suggesting that S aureus uses SSL5 to prevent neutrophil extravasation toward the site of infection. This makes SSL5 a potential lead for the development of new anti-inflammatory compounds for disorders characterized by excessive recruitment of leukocytes.


Subject(s)
Leukocyte Rolling/drug effects , Membrane Glycoproteins/metabolism , Neutrophils/drug effects , Neutrophils/physiology , P-Selectin/metabolism , Staphylococcus aureus/pathogenicity , Superantigens/metabolism , Superantigens/toxicity , Animals , Bacterial Toxins/metabolism , Bacterial Toxins/toxicity , Base Sequence , Binding, Competitive , CHO Cells , Cells, Cultured , Cloning, Molecular , Cricetinae , Cricetulus , DNA, Bacterial/genetics , Exotoxins/metabolism , Exotoxins/toxicity , Humans , In Vitro Techniques , Leukocyte Rolling/physiology , Protein Binding , Recombinant Proteins/metabolism , Recombinant Proteins/toxicity , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Superantigens/genetics
16.
J Immunol ; 177(11): 8017-26, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17114475

ABSTRACT

Bacteria have developed mechanisms to escape the first line of host defense, which is constituted by the recruitment of phagocytes to the sites of bacterial invasion. We previously described the chemotaxis inhibitory protein of Staphylococcus aureus, a protein that blocks the activation of neutrophils via the formyl peptide receptor (FPR) and C5aR. We now describe a new protein from S. aureus that impaired the neutrophil responses to FPR-like1 (FPRL1) agonists. FPRL1 inhibitory protein (FLIPr) inhibited the calcium mobilization in neutrophils stimulated with MMK-1, WKYMVM, prion-protein fragment PrP(106-126), and amyloid beta(1-42). Stimulation with low concentrations of fMLP was partly inhibited. Directed migration was also completely prevented toward MMK-1 and partly toward fMLP. Fluorescence-labeled FLIPr efficiently bound to neutrophils, monocytes, B cells, and NK cells. HEK293 cells transfected with human C5aR, FPR, FPRL1, and FPRL2 clearly showed that FLIPr directly bound to FPRL1 and, at higher concentrations, also to FPR but not to C5aR and FPRL2. FLIPr can reveal unknown inflammatory ligands crucial during S. aureus infections. As a novel described FPRL1 antagonist, it might lead to the development of therapeutic agents in FPRL1-mediated inflammatory components of diseases such as systemic amyloidosis, Alzheimer's, and prion disease.


Subject(s)
Anti-Inflammatory Agents/metabolism , Bacterial Proteins/metabolism , Chemotaxis, Leukocyte/immunology , Receptors, Formyl Peptide/antagonists & inhibitors , Receptors, Lipoxin/antagonists & inhibitors , Bacterial Proteins/genetics , Calcium/metabolism , Cells, Cultured , Cloning, Molecular , Humans , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Polymerase Chain Reaction , Receptors, Formyl Peptide/metabolism , Staphylococcus aureus
SELECTION OF CITATIONS
SEARCH DETAIL
...