Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
Front Immunol ; 9: 602, 2018.
Article in English | MEDLINE | ID: mdl-29686667

ABSTRACT

Group B Streptococcus (GBS) remains an important etiological agent of several infectious diseases including neonatal septicemia, pneumonia, meningitis, and orthopedic device infections. This pathogenicity is due to a variety of virulence factors expressed by Streptococcus agalactiae. Single virulence factors are not sufficient to provoke a streptococcal infection, which is instead promoted by the coordinated activity of several pathogenicity factors. Such determinants, mostly cell wall-associated and secreted proteins, include adhesins that mediate binding of the pathogen to host extracellular matrix/plasma ligands and cell surfaces, proteins that cooperate in the invasion of and survival within host cells and factors that neutralize phagocytosis and/or modulate the immune response. The genome-based approaches and bioinformatics tools and the extensive use of biophysical and biochemical methods and animal model studies have provided a great wealth of information on the molecular structure and function of these virulence factors. In fact, a number of new GBS surface-exposed or secreted proteins have been identified (GBS immunogenic bacterial adhesion protein, leucine-rich repeat of GBS, serine-rich repeat proteins), the three-dimensional structures of known streptococcal proteins (αC protein, C5a peptidase) have been solved and an understanding of the pathogenetic role of "old" and new determinants has been better defined in recent years. Herein, we provide an update of our current understanding of the major surface cell wall-anchored proteins from GBS, with emphasis on their biochemical and structural properties and the pathogenetic roles they may have in the onset and progression of host infection. We also focus on the antigenic profile of these compounds and discuss them as targets for therapeutic intervention.


Subject(s)
Bacterial Proteins/immunology , Cell Wall/immunology , Membrane Proteins/immunology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Vaccines/immunology , Streptococcus agalactiae/immunology , Animals , Biomarkers , Complement System Proteins/immunology , Complement System Proteins/metabolism , Extracellular Matrix , Fimbriae, Bacterial/immunology , Humans , Immunomodulation , Integrins/metabolism , O Antigens/immunology , Streptococcal Infections/metabolism
2.
Microorganisms ; 6(1)2018 Mar 13.
Article in English | MEDLINE | ID: mdl-29533985

ABSTRACT

Staphylococcus aureus is a human pathogen that can cause a wide spectrum of diseases, including sepsis, pneumonia, arthritis, and endocarditis. Ineffective treatment of a number of staphylococcal infections with antibiotics is due to the development and spread of antibiotic-resistant strains following decades of antibiotic usage. This has generated renewed interest within the scientific community in alternative therapeutic agents, such as anti-S. aureus antibodies. Although the role of antibodies in the management of S. aureus diseases is controversial, the success of this pathogen in neutralizing humoral immunity clearly indicates that antibodies offer the host extensive protection. In this review, we report an update on efforts to develop antibody-based agents, particularly monoclonal antibodies, and their therapeutic potential in the passive immunization approach to the treatment and prevention of S. aureus infections.

3.
Methods Mol Biol ; 1627: 309-324, 2017.
Article in English | MEDLINE | ID: mdl-28836210

ABSTRACT

A method is described for the purification of plasma fibronectins based on a combination of gelatin- and arginine-Sepharose chromatography steps. Cellular fibronectin can be purified from an osteosarcoma fibroblast cell line by affinity chromatography using a monoclonal antibody anti-fibronectin as ligand. Furthermore, we also provide a protocol for the purification of fibronectin domains obtained by fractionation of thermolysin-digested plasma fibronectin on ion-exchange/gel filtration chromatography columns. Assessment of the fibronectin purity is performed by SDS-PAGE, while the ligand binding activities of specific fibronectin domains are determined by ELISA.


Subject(s)
Fibronectins/isolation & purification , Peptide Fragments/isolation & purification , Cell Line , Chromatography, Affinity , Chromatography, Gel , Chromatography, Ion Exchange , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Fibronectins/blood , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Peptide Fragments/chemistry
4.
Article in English | MEDLINE | ID: mdl-28529927

ABSTRACT

Neutrophils, complement system and skin collectively represent the main elements of the innate immune system, the first line of defense of the host against many common microorganisms. Bacterial pathogens have evolved strategies to counteract all these defense activities. Specifically, Staphylococcus aureus, a major human pathogen, secretes a variety of immune evasion molecules including proteases, which cleave components of the innate immune system or disrupt the integrity of extracellular matrix and intercellular connections of tissues. Additionally, S. aureus secretes proteins that can activate host zymogens which, in turn, target specific defense components. Secreted proteins can also inhibit the anti-bacterial function of neutrophils or complement system proteases, potentiating S. aureus chances of survival. Here, we review the current understanding of these proteases and modulators of host proteases in the functioning of innate immunity and describe the importance of these mechanisms in the pathology of staphylococcal diseases.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Peptide Hydrolases/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Coagulase/metabolism , Complement System Proteins , Epithelial Cells/immunology , Epithelial Cells/microbiology , Exfoliatins , Humans , Immune Evasion/immunology , Metalloendopeptidases/metabolism , Neutrophils/immunology , RNA-Binding Proteins/metabolism , Serine Endopeptidases/metabolism , Serine Proteases/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Virulence Factors , von Willebrand Factor/metabolism
5.
ACS Nano ; 11(2): 2160-2170, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28151647

ABSTRACT

The collagen-binding protein Cna is a prototype cell surface protein from Staphylococcus aureus which fulfils important physiological functions during pathogenesis. While it is established that Cna binds to collagen (Cn) via the high-affinity collagen hug mechanism, whether this protein is engaged in other ligand-binding mechanisms is poorly understood. Here, we use atomic force microscopy to demonstrate that Cna mediates attachment to two structurally and functionally different host proteins, i.e., the complement system protein C1q and the extracellular matrix protein laminin (Lam), through binding mechanisms that differ from the collagen hug. We show that single Cna-C1q and Cna-Lam bonds are much weaker than the high-affinity Cna-Cn bond and that their formation does not require the B-region of Cna. At the whole cell level, we find that bacterial adhesion to C1q-substrates involves only one (or two) molecular bond(s), while adhesion to Lam is mediated by multiple bonds, thus suggesting that multivalent or cooperative interactions may enhance the strength of adhesion. Both C1q and Lam interactions can be efficiently blocked by monoclonal antibodies directed against the minimal Cn-binding domain of Cna. These results show that Cna is a multifunctional protein capable of binding to multiple host ligands through mechanisms that differ from the classical collagen hug.


Subject(s)
Adhesins, Bacterial/metabolism , Single Molecule Imaging , Single-Cell Analysis , Staphylococcus aureus/chemistry , Adhesins, Bacterial/chemistry , Cell Adhesion , Microscopy, Atomic Force , Staphylococcus aureus/cytology , Staphylococcus aureus/metabolism , Surface Plasmon Resonance
6.
mBio ; 7(5)2016 10 25.
Article in English | MEDLINE | ID: mdl-27795393

ABSTRACT

The bacterial pathogen Staphylococcus aureus expresses a variety of cell surface adhesion proteins that bind to host extracellular matrix proteins. Among these, the collagen (Cn)-binding protein Cna plays important roles in bacterium-host adherence and in immune evasion. While it is well established that the A region of Cna mediates ligand binding, whether the repetitive B region has a dedicated function is not known. Here, we report the direct measurement of the mechanical strength of Cna-Cn bonds on living bacteria, and we quantify the antiadhesion activity of monoclonal antibodies (MAbs) targeting this interaction. We demonstrate that the strength of Cna-Cn bonds in vivo is very strong (~1.2 nN), consistent with the high-affinity "collagen hug" mechanism. The B region is required for strong ligand binding and has been found to function as a spring capable of sustaining high forces. This previously undescribed mechanical response of the B region is of biological significance as it provides a means to project the A region away from the bacterial surface and to maintain bacterial adhesion under conditions of high forces. We further quantified the antiadhesion activity of MAbs raised against the A region of Cna directly on living bacteria without the need for labeling or purification. Some MAbs are more efficient in blocking single-cell adhesion, suggesting that they act as competitive inhibitors that bind Cna residues directly involved in ligand binding. This report highlights the role of protein mechanics in activating the function of staphylococcal adhesion proteins and emphasizes the potential of antibodies to prevent staphylococcal adhesion and biofilm formation. IMPORTANCE: Cna is a collagen (Cn)-binding protein from Staphylococcus aureus that is involved in pathogenesis. Currently, we know little about the functional role of the repetitive B region of the protein. Here, we unravel the mechanical strength of Cna in living bacteria. We show that single Cna-Cn bonds are very strong, reflecting high-affinity binding by the collagen hug mechanism. We discovered that the B region behaves as a nanospring capable of sustaining high forces. This unanticipated mechanical response, not previously described for any staphylococcal adhesin, favors a model in which the B region has a mechanical function that is essential for strong ligand binding. Finally, we assess the antiadhesion activity of monoclonal antibodies against Cna, suggesting that they could be used to inhibit S. aureus adhesion.


Subject(s)
Adhesins, Bacterial/metabolism , Collagen/metabolism , Mechanical Phenomena , Staphylococcus aureus/metabolism , Antibodies, Bacterial/metabolism , Antibodies, Monoclonal/metabolism , Bacterial Adhesion/drug effects , Protein Binding
7.
Sci Rep ; 6: 27996, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27302108

ABSTRACT

During bacterial pathogenesis extensive contacts between the human and the bacterial extracellular proteomes take place. The identification of novel host-pathogen interactions by standard methods using a case-by-case approach is laborious and time consuming. To overcome this limitation, we took advantage of large libraries of human and bacterial recombinant proteins. We applied a large-scale protein microarray-based screening on two important human pathogens using two different approaches: (I) 75 human extracellular proteins were tested on 159 spotted Staphylococcus aureus recombinant proteins and (II) Neisseria meningitidis adhesin (NadA), an important vaccine component against serogroup B meningococcus, was screened against ≈2300 spotted human recombinant proteins. The approach presented here allowed the identification of the interaction between the S. aureus immune evasion protein FLIPr (formyl-peptide receptor like-1 inhibitory protein) and the human complement component C1q, key players of the offense-defense fighting; and of the interaction between meningococcal NadA and human LOX-1 (low-density oxidized lipoprotein receptor), an endothelial receptor. The novel interactions between bacterial and human extracellular proteins here presented might provide a better understanding of the molecular events underlying S. aureus and N. meningitidis pathogenesis.


Subject(s)
Host-Pathogen Interactions , Neisseria meningitidis/physiology , Protein Array Analysis/methods , Staphylococcus aureus/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Animals , Bacterial Proteins/metabolism , Binding Sites , CHO Cells , Complement C1q/metabolism , Cricetulus , Humans , Protein Binding , Recombinant Proteins/metabolism , Scavenger Receptors, Class E/metabolism
8.
J Immunol ; 196(1): 385-94, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26608922

ABSTRACT

The group B Streptococcus (GBS) is a leading cause of neonatal invasive disease. GBS bacteria are surrounded by a thick capsular polysaccharide that is a potent inhibitor of complement deposition via the alternative pathway. Several of its surface molecules can however activate the classical and lectin complement pathways, rendering this species still vulnerable to phagocytic killing. In this study we have identified a novel secreted protein named complement interfering protein (CIP) that downregulates complement activation via the classical and lectin pathways, but not the alternative pathway. The CIP protein showed high affinity toward C4b and inhibited its interaction with C2, presumably preventing the formation of the C4bC2a convertase. Addition of recombinant CIP to GBS cip-negative bacteria resulted in decreased deposition of C3b on their surface and in diminished phagocytic killing in a whole-blood assay. Our data reveal a novel strategy exploited by GBS to counteract innate immunity and could be valuable for the development of anti-infective agents against this important pathogen.


Subject(s)
Bacterial Proteins/immunology , Complement C4b/immunology , Complement Pathway, Classical/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Immune Evasion/immunology , Streptococcus agalactiae/immunology , Adult , Amino Acid Sequence , Complement Activation/immunology , Complement C3b/biosynthesis , Complement C3b/immunology , Complement Pathway, Alternative/immunology , Complement Pathway, Classical/drug effects , Complement Pathway, Mannose-Binding Lectin/drug effects , Humans , Immunity, Innate , Molecular Sequence Data , Phagocytosis/immunology , Protein Binding/immunology , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/metabolism
9.
Infect Immun ; 83(10): 4093-102, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26238710

ABSTRACT

In this study, we investigated the cell wall-anchored fibronectin-binding proteins SpsD and SpsL from the canine commensal and pathogen Staphylococcus pseudintermedius for their role in promoting bacterial invasion of canine progenitor epidermal keratinocytes (CPEK). Invasion was examined by the gentamicin protection assay and fluorescence microscopy. An ΔspsD ΔspsL mutant of strain ED99 had a dramatically reduced capacity to invade CPEK monolayers, while no difference in the invasion level was observed with single mutants. Lactococcus lactis transformed with plasmids expressing SpsD and SpsL promoted invasion, showing that both proteins are important. Soluble fibronectin was required for invasion, and an RGD-containing peptide or antibodies recognizing the integrin α5ß1 markedly reduced invasion, suggesting an important role for the integrin in this process. Src kinase inhibitors effectively blocked internalization, suggesting a functional role for the kinase in invasion. In order to identify the minimal fibronectin-binding region of SpsD and SpsL involved in the internalization process, recombinant fragments of both proteins were produced. The SpsD520-846 and SpsL538-823 regions harboring the major fibronectin-binding sites inhibited S. pseudintermedius internalization. Finally, the effects of staphylococcal invasion on the integrity of different cell lines were examined. Because SpsD and SpsL are critical factors for adhesion and invasion, blocking these processes could provide a strategy for future approaches to treating infections.


Subject(s)
Bacterial Proteins/metabolism , Dog Diseases/microbiology , Epithelial Cells/microbiology , Fibronectins/metabolism , Staphylococcal Infections/veterinary , Staphylococcus/metabolism , Animals , Bacterial Proteins/genetics , Cell Line , Cell Wall/genetics , Cell Wall/metabolism , Dog Diseases/metabolism , Dogs , Protein Binding , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus/genetics , Staphylococcus/pathogenicity , Virulence
10.
J Appl Biomater Funct Mater ; 13(2): e181-5, 2015 Jul 04.
Article in English | MEDLINE | ID: mdl-26045224

ABSTRACT

BACKGROUND: The objective of the present study was to compare solubility and pH of 6 direct pulp capping materials. METHODS: Specimens of each material - i.e., Dycal, Calcicur, Calcimol LC, TheraCal LC, MTA Angelus and ProRoot MTA - were prepared and immersed in water. Solubility was determined after 24 hours and 2 months and analyzed statistically using a 1-way ANOVA and post hoc Tukey test. pH values were measured 3 and 24 hours after manipulation. RESULTS: All direct pulp capping materials showed low solubility; the pH of tested materials ranged from 10 to 12 and showed a nonsignificant increase/reduction after 24 hours. CONCLUSIONS: Within the limitations of this in vitro study, the direct pulp capping materials studied showed different solubility even if no changes were recorded over time. All of the materials showed a very alkaline pH.


Subject(s)
Dental Cements/chemistry , Dental Pulp Capping , Aluminum Compounds/chemistry , Calcium Compounds/chemistry , Calcium Hydroxide/chemistry , Drug Combinations , Hydrogen-Ion Concentration , Materials Testing , Oxides/chemistry , Silicates/chemistry , Solubility
11.
Infect Immun ; 82(6): 2448-59, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24686057

ABSTRACT

Staphylococcus lugdunensis is a coagulase-negative staphylococcus that is a commensal of humans and an opportunistic pathogen. It can cause a spectrum of infections, including those that are associated with the ability to form biofilm, such as occurs with endocarditis or indwelling medical devices. The genome sequences of two strains revealed the presence of orthologues of the ica genes that are responsible for synthesis of poly-N-acetylglucosamine (PNAG) that is commonly associated with biofilm in other staphylococci. However, we discovered that biofilm formed by a panel of S. lugdunensis isolates growing in iron-restricted medium was susceptible to degradation by proteases and not by metaperiodate, suggesting that the biofilm matrix comprised proteins and not PNAG. When the iron concentration was raised to 1 mM biofilm formation by all strains tested was greatly reduced. A mutant of strain N920143 lacking the entire locus that encodes iron-regulated surface determinant (Isd) proteins was defective in biofilm formation under iron-limited conditions. An IsdC-null mutant was defective, whereas IsdK, IsdJ, and IsdB mutants formed biofilm to the same level as the parental strain. Expression of IsdC was required both for the primary attachment to unconditioned polystyrene and for the accumulation phase of biofilm involving cell-cell interactions. Purified recombinant IsdC protein formed dimers in solution and Lactococcus lactis cells expressing only IsdC adhered to immobilized recombinant IsdC but not to IsdJ, IsdK, or IsdB. This is consistent with a specific homophilic interaction between IsdC molecules on neighboring cells contributing to accumulation of S. lugdunensis biofilm in vivo.


Subject(s)
Bacterial Proteins/physiology , Biofilms/growth & development , Carrier Proteins/physiology , Iron/metabolism , Staphylococcal Infections/microbiology , Staphylococcus lugdunensis/physiology , Analysis of Variance , Bacterial Adhesion/physiology , Bacterial Proteins/metabolism , Blotting, Western , Carrier Proteins/metabolism , Culture Media/chemistry , Gene Expression Regulation, Bacterial , Humans , Recombinant Proteins/metabolism
12.
Infect Immun ; 82(7): 2890-901, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24778116

ABSTRACT

Group A streptococcus (GAS) is a human pathogen causing a wide repertoire of mild and severe diseases for which no vaccine is yet available. We recently reported the identification of three protein antigens that in combination conferred wide protection against GAS infection in mice. Here we focused our attention on the characterization of one of these three antigens, Spy0269, a highly conserved, surface-exposed, and immunogenic protein of unknown function. Deletion of the spy0269 gene in a GAS M1 isolate resulted in very long bacterial chains, which is indicative of an impaired capacity of the knockout mutant to properly divide. Confocal microscopy and immunoprecipitation experiments demonstrated that the protein was mainly localized at the cell septum and could interact in vitro with the cell division protein FtsZ, leading us to hypothesize that Spy0269 is a member of the GAS divisome machinery. Predicted structural domains and sequence homologies with known streptococcal adhesins suggested that this antigen could also play a role in mediating GAS interaction with host cells. This hypothesis was confirmed by showing that recombinant Spy0269 could bind to mammalian epithelial cells in vitro and that Lactococcus lactis expressing Spy0269 on its cell surface could adhere to mammalian cells in vitro and to mice nasal mucosa in vivo. On the basis of these data, we believe that Spy0269 is involved both in bacterial cell division and in adhesion to host cells and we propose to rename this multifunctional moonlighting protein as SpyAD (Streptococcus pyogenes Adhesion and Division protein).


Subject(s)
Bacterial Adhesion/physiology , Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/immunology , Streptococcus pyogenes/metabolism , Antigens, Bacterial , Bacterial Proteins/genetics , Cell Line , Cloning, Molecular , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Epithelial Cells/microbiology , Gene Deletion , Humans , Lactococcus lactis/metabolism , Protein Binding , Streptococcus pyogenes/cytology , Streptococcus pyogenes/genetics
13.
PLoS One ; 8(9): e75266, 2013.
Article in English | MEDLINE | ID: mdl-24086487

ABSTRACT

Several species of Gram-positive bacteria can avidly bind soluble and surface-associated fibrinogen (Fng), a property that is considered important in the pathogenesis of human infections. To gain insights into the mechanism by which group B Streptococcus (GBS), a frequent neonatal pathogen, interacts with Fng, we have screened two phage displayed genomic GBS libraries. All of the Fng-binding phage clones contained inserts encoding fragments of FbsA, a protein displaying multiple repeats. Since the functional role of this protein is only partially understood, representative fragments were recombinantly expressed and analyzed for Fng binding affinity and ability to induce immune protection against GBS infection. Maternal immunization with 6pGST, a fragment containing five repeats, significantly protected mouse pups against lethal GBS challenge and these protective effects could be recapitulated by administration of anti-6pGST serum from adult animals. Notably, a monoclonal antibody that was capable of neutralizing Fng binding by 6pGST, but not a non-neutralizing antibody, could significantly protect pups against lethal GBS challenge. These data suggest that FbsA-Fng interaction promotes GBS pathogenesis and that blocking such interaction is a viable strategy to prevent or treat GBS infections.


Subject(s)
Bacterial Proteins/immunology , Carrier Proteins/immunology , Fibrinogen/metabolism , Streptococcus agalactiae/immunology , Animals , Antibodies, Monoclonal/immunology , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Humans , Immunization/methods , Mice , Neutralization Tests , Peptide Library , Protein Binding , Time Factors
14.
PLoS One ; 8(6): e66901, 2013.
Article in English | MEDLINE | ID: mdl-23805283

ABSTRACT

Staphylococcus pseudintermedius, a commensal and pathogen of dogs and occasionally of humans, expresses surface proteins potentially involved in host colonization and pathogenesis. Here, we describe the cloning and characterization of SpsD, a surface protein of S. pseudintermedius reported as interacting with extracellular matrix proteins and corneocytes. A ligand screen and Western immunoblotting revealed that the N-terminal A domain of SpsD bound fibrinogen, fibronectin, elastin and cytokeratin 10. SpsD also interfered with thrombin-induced fibrinogen coagulation and blocked ADP-induced platelet aggregation. The binding site for SpsD was mapped to residues 395-411 in the fibrinogen γ-chain, while binding sites in fibronectin were localized to the N- and C-terminal regions. SpsD also bound to glycine- and serine-rich omega loops within the C-terminal tail region of cytokeratin 10. Ligand binding studies using SpsD variants lacking the C-terminal segment or containing an amino-acid substitution in the putative ligand binding site provided insights into interaction mechanism of SpsD with the different ligands. Together these data demonstrate the multi-ligand binding properties of SpsD and illustrate some interesting differences in the variety of ligands bound by SpsD and related proteins from S. aureus.


Subject(s)
Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Extracellular Matrix Proteins/metabolism , Host-Pathogen Interactions/genetics , Staphylococcus/metabolism , Animals , Bacterial Outer Membrane Proteins/chemistry , Bacterial Outer Membrane Proteins/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Dogs , Elastin/chemistry , Elastin/metabolism , Extracellular Matrix Proteins/chemistry , Fibrinogen/chemistry , Fibrinogen/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Keratin-10/chemistry , Keratin-10/metabolism , Ligands , Mutagenesis, Site-Directed , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification
15.
Int J Artif Organs ; 34(9): 799-810, 2011 Sep.
Article in English | MEDLINE | ID: mdl-22094559

ABSTRACT

Toll-like receptors (TLRs) are the most important class of innate pattern recognition receptors (PRRs) by which host immune and non-immune cells are able to recognize pathogen-associated molecular patterns (PAMPs). Most mammalian species have 10 to 15 types of TLRs. TLRs are believed to function as homo- or hetero-dimers. TLR2, which plays a crucial role in recognizing PAMPs from Staphylococcus aureus, forms heterodimers with TLR1 or TLR6 and each dimer has a different ligand specificity. Staphylococcal lipoproteins, Panton-Valentine toxin and Phenol Soluble Modulins have been identified as potent TLR2 ligands. Conversely, the ligand function attributed to peptidoglycan and LTA remains controversial. TLR2 uses a MyD88-dependent signaling pathway that results in NF-kB translocation into the nucleus and activation of the expression of pro-inflammatory cytokine genes. Recognition rouses both an inflammatory response, culminating in the phagocytosis of bacteria, and an adaptive immune response, with the presentation of resulting bacterial compounds to T cells. Here, recent advances on the recognition of S. aureus by TLRs are presented and discussed, as well as the new therapeutic opportunities deriving from this new knowledge.


Subject(s)
Immunity, Innate , Staphylococcal Infections/prevention & control , Staphylococcus aureus/immunology , Toll-Like Receptors/metabolism , Animals , Bacterial Proteins/metabolism , Humans , Inflammasomes/immunology , Ligands , Signal Transduction , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity
16.
BMC Microbiol ; 10: 160, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20515471

ABSTRACT

BACKGROUND: Fibronectin binding proteins A and B (FnBPA and FnBPB) mediate adhesion of S. aureus to fibrinogen, elastin and fibronectin. We previously identified seven different isotypes of FnBPA based on divergence in the fibrinogen- and elastin-binding A domains. The variation created differences in antigenicity while ligand binding functions were retained. Here, FnBPB variation was examined in both human and bovine isolates and compared to that of FnBPA. RESULTS: Seven different fnbB allelic variants were identified. Some strains that cluster by phylogenetic analysis contain different fnbB variants, whereas more divergent strains contain the same fnbB variant. The phylogeny of fnbB alleles does not match the phylogeny of fnbA alleles. Some FnBPA and FnBPB isotypes that are specified by human S. aureus strains are also found in bovine strains. The seven fnbB allelic variants encode seven distinct isotypes of the FnBPB A domain that are 61 to 85% identical in amino acid sequence. Variant amino acid residues were mapped on a three-dimensional model of the FnBPB A domain and were predicted to be surface-exposed. They are responsible for the antigenic diversity detected with polyclonal antibody and a monoclonal antibody raised against isotype I. Ligand binding by recombinant FnBPB N23 isotypes was compared by ELISA-based solid phase assays and surface plasmon resonance. Each bound to immobilized fibrinogen, elastin and fibronectin dose-dependently and saturably with similar affinities. Binding to fibronectin was surprising because the A domains do not contain any known motifs that mediate binding to fibronectin. This raises the possibility that the A domain of FnBPB contains a novel fibronectin binding motif that binds fibronectin by a novel mechanism. CONCLUSIONS: Seven different isoforms of FnBPB A domain retain ligand-binding functions but are antigenically distinct. The variation in FnBPA and FnBPB occurs in human and bovine S. aureus strains and may act as an immune evasion mechanism. All seven isotypes of FnBPB are capable of binding fibronectin though none contain any known fibronectin-binding motifs. These results have implications for the development of vaccines or immunotherapeutics that target FnBPB.


Subject(s)
Adhesins, Bacterial/metabolism , Genetic Variation , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Adhesins, Bacterial/genetics , Alleles , Amino Acid Sequence , Animals , Antibodies, Bacterial , Antibody Specificity , Antigens, Bacterial , Cloning, Molecular , Gene Expression Regulation, Bacterial , Models, Molecular , Phylogeny , Protein Conformation , Protein Isoforms , Rabbits
17.
Future Microbiol ; 4(10): 1337-52, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19995192

ABSTRACT

Staphylococcus aureus is a versatile and harmful human pathogen in both hospital- and community-acquired infections. S. aureus can initiate host infection by adhering to components of the extracellular matrix. Adherence is mediated by a variety of protein adhesins of the microbial surface component recognizing adhesive matrix molecule (MSCRAMM) family. In this article, we describe these MSCRAMMs in terms of structural organization and ligand-binding capacity and discuss their role as a possible target for immunotherapy.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion , Extracellular Matrix Proteins/metabolism , Staphylococcus aureus/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Humans , Models, Biological , Models, Molecular , Protein Binding , Protein Structure, Quaternary , Protein Structure, Tertiary
18.
Biomaterials ; 30(34): 6621-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19758694

ABSTRACT

Staphylococcus aureus is a major, highly clonal, pathogen causing implant infections. This study aimed at investigating the diverse distribution of bacterial adhesins in most prevalent S. aureus strain types causing orthopaedic implant infections. 200 S. aureus isolates, categorized into ribogroups by automated ribotyping, i.e. rDNA restriction fragment length polymorphism analysis, were screened for the presence of a panel of adhesins genes. Within the collection of isolates, automated ribotyping detected 98 distinct ribogroups. For many ribogroups, characteristic tandem genes arrangements could be identified. In the predominant S. aureus cluster, enlisting 27 isolates, the bbp gene encoding bone sialoprotein-binding protein appeared a typical virulence trait, found in 93% of the isolates. Conversely, the bbp gene was identified in just 10% of the remaining isolates of the collection. In this cluster, co-presence of bbp with the cna gene encoding collagen adhesin was a pattern consistently observed. These findings indicate a crucial role of both these adhesins, able to bind the most abundant bone proteins, in the pathogenesis of orthopaedic implant infections, there where biomaterials interface bone tissues. This study suggests that specific adhesins may synergistically act in the onset of implant infections and that anti-adhesin strategies should be targeted to adhesins conjointly present.


Subject(s)
Adhesins, Bacterial/genetics , Prosthesis-Related Infections/microbiology , Sialoglycoproteins/metabolism , Staphylococcus aureus/genetics , DNA, Ribosomal/genetics , Humans , Polymorphism, Restriction Fragment Length , Staphylococcus aureus/isolation & purification , Staphylococcus aureus/pathogenicity
19.
FASEB J ; 23(9): 3100-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19417080

ABSTRACT

Microbial pathogen entry and survival in the host is mediated by a network of molecular interactions between the two partners, which has been the subject of many research efforts. A complex picture is emerging in which host-pathogen crosstalk involves a high number of proteins, often with redundant functions. In the present study, we investigated the potential of protein microarrays to simultaneously scan interactions between surface proteins from two main human streptococcal pathogens, Streptococcus pyogenes and Streptococcus agalactiae, and three human ligands, fibronectin, fibrinogen, and C4 binding protein, known to play an important role in streptococcal pathogenesis. By using this technology, we confirmed interactions described in the literature and detected a novel set of streptococcal proteins with binding capacities for the human ligands. The observations were validated by Western blot and ELISA techniques. Three of the newly identified proteins were isoforms of a group B streptococcus-secreted component named Fib and displayed differential binding capacities for fibronectin, fibrinogen, and C4BP. The protein regions involved in the interaction with each ligand were identified by constructing fragments of one of the Fib variants. The approach proved valuable for the acquisition of novel insights into the complex network of protein-protein interactions occurring during microbial infection.


Subject(s)
Bacterial Proteins/analysis , Fibrinogen/metabolism , Fibronectins/metabolism , Histocompatibility Antigens/metabolism , Host-Pathogen Interactions , Streptococcus/chemistry , Bacterial Proteins/metabolism , Complement C4b-Binding Protein , Humans , Protein Array Analysis/methods , Protein Binding , Streptococcus agalactiae/chemistry , Streptococcus pyogenes/chemistry
20.
Curr Med Chem ; 15(30): 3185-95, 2008.
Article in English | MEDLINE | ID: mdl-19075662

ABSTRACT

Staphylococcus growth on medical devices represents a common occurrence that can lead to serious illness and death. Biomaterial-associated infection, mostly caused by Staphylococcus epidermidis and Staphylococcus aureus, is fairly complicated by the organism' development of a biofilm, which provides a microenvironment that protects from attack by the host immune system and antibiotics. In this review we present recent insights regarding S. aureus and S. epidermidis structural and functional factors that are effective in biofilm development and describe the regulation of their expression. On the basis of the knowledge gained, we also present the potential and limits of current biochemical and biophysical strategies aimed at preventing biofilm formation or at the treatment of established mature biofilms.


Subject(s)
Biofilms/growth & development , Staphylococcal Infections/prevention & control , Staphylococcal Infections/therapy , Staphylococcus/growth & development , Staphylococcus/chemistry , Staphylococcus/drug effects , Staphylococcus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...