Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Nature ; 505(7483): 412-6, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-24317696

ABSTRACT

Respiratory surfaces are exposed to billions of particulates and pathogens daily. A protective mucus barrier traps and eliminates them through mucociliary clearance (MCC). However, excessive mucus contributes to transient respiratory infections and to the pathogenesis of numerous respiratory diseases. MUC5AC and MUC5B are evolutionarily conserved genes that encode structurally related mucin glycoproteins, the principal macromolecules in airway mucus. Genetic variants are linked to diverse lung diseases, but specific roles for MUC5AC and MUC5B in MCC, and the lasting effects of their inhibition, are unknown. Here we show that mouse Muc5b (but not Muc5ac) is required for MCC, for controlling infections in the airways and middle ear, and for maintaining immune homeostasis in mouse lungs, whereas Muc5ac is dispensable. Muc5b deficiency caused materials to accumulate in upper and lower airways. This defect led to chronic infection by multiple bacterial species, including Staphylococcus aureus, and to inflammation that failed to resolve normally. Apoptotic macrophages accumulated, phagocytosis was impaired, and interleukin-23 (IL-23) production was reduced in Muc5b(-/-) mice. By contrast, in mice that transgenically overexpress Muc5b, macrophage functions improved. Existing dogma defines mucous phenotypes in asthma and chronic obstructive pulmonary disease (COPD) as driven by increased MUC5AC, with MUC5B levels either unaffected or increased in expectorated sputum. However, in many patients, MUC5B production at airway surfaces decreases by as much as 90%. By distinguishing a specific role for Muc5b in MCC, and by determining its impact on bacterial infections and inflammation in mice, our results provide a refined framework for designing targeted therapies to control mucin secretion and restore MCC.


Subject(s)
Lung/immunology , Mucin-5B/metabolism , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Animals , Asthma/immunology , Asthma/metabolism , Bacterial Infections/immunology , Bacterial Infections/microbiology , Cilia/physiology , Ear, Middle/immunology , Ear, Middle/microbiology , Female , Inflammation/pathology , Lung/metabolism , Lung/microbiology , Macrophages/immunology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Mucin 5AC/deficiency , Mucin 5AC/metabolism , Mucin-5B/deficiency , Mucin-5B/genetics , Phagocytosis , Pulmonary Disease, Chronic Obstructive/immunology , Pulmonary Disease, Chronic Obstructive/microbiology , Staphylococcus aureus/immunology , Survival Analysis
2.
J Biol Chem ; 287(13): 9940-9951, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22303018

ABSTRACT

Antibodies that recognize microbial B lymphocyte superantigenic epitopes are produced constitutively with no requirement for adaptive immune maturation. We report cleavage of the Staphylococcus aureus virulence factor extracellular fibrinogen-binding protein (Efb) by catalytic antibodies produced with no exposure to the bacterium and reduction of the catalytic antibody activity following infection. IgG catalytic antibodies that specifically hydrolyzed Efb via a nucleophilic catalytic mechanism were found in the blood of healthy humans and aseptic mice free of S. aureus infection. IgG hydrolyzed peptide bonds on the C-terminal side of basic amino acids, including a bond located within the C3b-binding domain of Efb. Efb digested with the IgG lost its ability to bind C3b and inhibit complement-dependent antibody-mediated red blood cell lysis. In addition to catalysis, the IgG expressed saturable Efb binding activity. IgG from S. aureus-infected mice displayed reduced Efb cleaving activity and increased Efb binding activity compared with uninfected controls, suggesting differing effects of the infection on the antibody subsets responsible for the two activities. IgG from children hospitalized for S. aureus infection also displayed reduced Efb cleavage compared with healthy children. These data suggest a potential defense function for constitutively produced catalytic antibodies to a putative superantigenic site of Efb, but an adaptive catalytic response appears to be proscribed.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Catalytic/immunology , Immunoglobulin G/immunology , Proteolysis , Staphylococcal Infections/immunology , Staphylococcus aureus , Virulence Factors/immunology , Adaptive Immunity/physiology , Adult , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antibodies, Catalytic/blood , Bacterial Proteins , Child , Child, Preschool , Female , Humans , Immunoglobulin G/blood , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Structure, Tertiary , Staphylococcal Infections/blood , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Virulence Factors/blood
3.
J Immunol Methods ; 365(1-2): 142-8, 2011 Feb 28.
Article in English | MEDLINE | ID: mdl-21185300

ABSTRACT

Staphylococcus aureus causes a variety of infections. Knowledge about the physiological role of most S. aureus antigens in colonization and infection is only limited. This can be studied by measuring antigen-specific antibody responses. In this study, we optimized the multiplex microsphere bead-based flow cytometry technique for mouse serum samples. We analysed immunoglobulin G (IgG) levels directed against 26 S. aureus proteins in a single small-volume mouse serum sample. We assessed possible cross reactivity. Furthermore, we analysed serum samples from mice with different types of S. aureus infections caused by different S. aureus strains. The results show that cross reactivity between proteins on microspheres and serum antibodies towards other proteins was limited. We found that lung-infected mice had a higher and broader IgG response than skin-infected mice. Clearly, the site of infection influences the IgG profile. Next, we compared sera from mice with intravenously-induced bacteraemia caused by different S. aureus strains. We showed different IgG responses depending on the causing S. aureus strain. It is concluded that the bead-based multiplex S. aureus antibody assay can be successfully applied to determine the immunogenicity of different S. aureus proteins in relation to the site of infection and the S. aureus strain causing the infection.


Subject(s)
Antibodies, Bacterial/analysis , Immunoassay/methods , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacteremia/immunology , Bacteremia/microbiology , Bacterial Proteins/immunology , Cross Reactions , Female , Flow Cytometry/methods , Immunoglobulin G/analysis , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Microspheres , Pneumonia, Staphylococcal/immunology , Pneumonia, Staphylococcal/microbiology , Reproducibility of Results , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Staphylococcal Vaccines/administration & dosage , Staphylococcus aureus/classification , Staphylococcus aureus/immunology
4.
J Immunol ; 185(12): 7413-25, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21098229

ABSTRACT

Neutrophil extracellular traps (NETs) are webs of DNA covered with antimicrobial molecules that constitute a newly described killing mechanism in innate immune defense. Previous publications reported that NETs take up to 3-4 h to form via an oxidant-dependent event that requires lytic death of neutrophils. In this study, we describe neutrophils responding uniquely to Staphylococcus aureus via a novel process of NET formation that did not require neutrophil lysis or even breach of the plasma membrane. The multilobular nucleus rapidly became rounded and condensed. During this process, we observed the separation of the inner and outer nuclear membranes and budding of vesicles, and the separated membranes and vesicles were filled with nuclear DNA. The vesicles were extruded intact into the extracellular space where they ruptured, and the chromatin was released. This entire process occurred via a unique, very rapid (5-60 min), oxidant-independent mechanism. Mitochondrial DNA constituted very little if any of these NETs. They did have a limited amount of proteolytic activity and were able to kill S. aureus. With time, the nuclear envelope ruptured, and DNA filled the cytoplasm presumably for later lytic NET production, but this was distinct from the vesicular release mechanism. Panton-Valentine leukocidin, autolysin, and a lipase were identified in supernatants with NET-inducing activity, but Panton-Valentine leukocidin was the dominant NET inducer. We describe a new mechanism of NET release that is very rapid and contributes to trapping and killing of S. aureus.


Subject(s)
Bacterial Toxins/immunology , Chromatin/immunology , DNA, Mitochondrial/immunology , Exotoxins/immunology , Immunity, Innate/immunology , Leukocidins/immunology , Neutrophils/immunology , Staphylococcus aureus/immunology , Cytoplasm/immunology , Humans , Oxidation-Reduction
5.
J Biomol Struct Dyn ; 28(1): 1-12, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20476791

ABSTRACT

Panton-Valentine leukocidin (PVL) is a bipartite toxin that plays an important role in the pathogenesis of methicillin-resistant Staphylococcus aureus. Recent clinical data suggest a correlation between PVL and severe cases of S. aureus pneumonia. A clear understanding of the structure and function of PVL is critical to the development of novel, effective treatments. Here, we report an all-atom model of the macromolecular structure of Panton-Valentine leukocidin in its octameric, pre-pore conformation that confirms and extends our understanding of the toxin's mechanism of action.


Subject(s)
Bacterial Toxins/chemistry , Exotoxins/chemistry , Leukocidins/chemistry , Methicillin-Resistant Staphylococcus aureus/chemistry , Models, Molecular , Protein Conformation , Bacterial Toxins/genetics , Crystallography, X-Ray , Exotoxins/genetics , Humans , Hydrogen Bonding , Leukocidins/genetics , Molecular Sequence Data , Mutation
7.
PLoS One ; 4(4): e5042, 2009.
Article in English | MEDLINE | ID: mdl-19347045

ABSTRACT

Staphylococcus aureus necrotizing pneumonia is a severe disease caused by S. aureus strains carrying the Panton Valentine leukocidin (PVL) genes (lukS-PV & lukF-PV) encoded on various bacteriophages (such as phiSLT). Clinical PVL+ strains isolated from necrotizing pneumonia display an increased attachment to matrix molecules (type I and IV collagens and laminin), a phenotype that could play a role in bacterial adhesion to damaged airway epithelium during the early stages of necrotizing pneumonia (J Infect Dis 2004; 190: 1506-15). To investigate the basis of the observed adhesion of S. aureus PVL+ strains, we compared the ability of PVL+ and their isogenic PVL(-) strains to attach to various immobilized matrix molecules. The expression of recombinant fragments of the PVL subunits and the addition of synthetic peptides indicated that the processed LukS-PV signal peptide (LukS-PV SP) was sufficient to significantly enhance the ability of S. aureus to attach to extracellular matrix (ECM) components. Furthermore, we showed that adhesion to ECM components was inhibited by heparin and heparan sulfates (HS) suggesting that in vivo, HS could function as a molecular bridge between the matrix and S. aureus expressing the LukS-PV signal peptide. Site directed mutagenesis, biochemical and structural analyses of the LukS-PV signal peptide indicate that this peptide is present at the S. aureus surface, binds to HS in solid phase assay, and mediates the enhanced S. aureus matrix component adhesion. Our data suggests that after its cleavage by signal peptidase, the signal peptide is released from the membrane and associates to the cell wall through its unique C-terminus sequence, while its highly positively charged N-terminus is exposed on the bacterial surface, allowing its interaction with extracellular matrix-associated HS. This mechanism may provide a molecular bridge that enhances the attachment of the S. aureus PVL+ strains to ECM components exposed at damaged epithelial sites.


Subject(s)
Bacterial Adhesion/physiology , Bacterial Proteins/chemistry , Heparitin Sulfate/metabolism , Leukocidins/chemistry , Protein Sorting Signals/physiology , Staphylococcus aureus/physiology , Bacterial Proteins/genetics , Extracellular Matrix Proteins/metabolism , Leukocidins/genetics , Operon
8.
Clin Vaccine Immunol ; 16(1): 139-41, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19005019

ABSTRACT

We examined the antibody responses of pediatric patients infected with community-associated Staphylococcus aureus isolates. The data show that patients infected with Panton-Valentine leukocidin (PVL)-positive strains developed a dominant immunoglobulin G anti-PVL antibody response that correlates with markers of inflammation.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Toxins/immunology , Community-Acquired Infections/immunology , Exotoxins/immunology , Leukocidins/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Child , Child, Preschool , Humans , Immunoglobulin G/blood , Virulence Factors/immunology
9.
PLoS Pathog ; 4(11): e1000226, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19043557

ABSTRACT

The fibrinogen (Fg) binding MSCRAMM Clumping factor A (ClfA) from Staphylococcus aureus interacts with the C-terminal region of the fibrinogen (Fg) gamma-chain. ClfA is the major virulence factor responsible for the observed clumping of S. aureus in blood plasma and has been implicated as a virulence factor in a mouse model of septic arthritis and in rabbit and rat models of infective endocarditis. We report here a high-resolution crystal structure of the ClfA ligand binding segment in complex with a synthetic peptide mimicking the binding site in Fg. The residues in Fg required for binding to ClfA are identified from this structure and from complementing biochemical studies. Furthermore, the platelet integrin alpha(IIb)beta(3) and ClfA bind to the same segment in the Fg gamma-chain but the two cellular binding proteins recognize different residues in the common targeted Fg segment. Based on these differences, we have identified peptides that selectively antagonize the ClfA-Fg interaction. The ClfA-Fg binding mechanism is a variant of the "Dock, Lock and Latch" mechanism previously described for the Staphylococcus epidermidis SdrG-Fg interaction. The structural insights gained from analyzing the ClfANFg peptide complex and identifications of peptides that selectively recognize ClfA but not alpha(IIb)beta(3) may allow the design of novel anti-staphylococcal agents. Our results also suggest that different MSCRAMMs with similar structural organization may have originated from a common ancestor but have evolved to accommodate specific ligand structures.


Subject(s)
Coagulase/metabolism , Fibrinogen/metabolism , Models, Molecular , Staphylococcal Infections/drug therapy , Binding Sites , Crystallography, X-Ray , Drug Design , Peptides/chemical synthesis , Peptides/pharmacology , Protein Binding/drug effects , Staphylococcus aureus
10.
J Biol Chem ; 283(1): 638-647, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-17991749

ABSTRACT

Staphylococcus epidermidis is an opportunistic pathogen and a major cause of foreign body infections. The S. epidermidis fibrinogen (Fg)-binding adhesin SdrG is necessary and sufficient for the attachment of this pathogen to Fg-coated materials. Based largely on structural analyses of the ligand binding domain of SdrG as an apo-protein and in complex with a Fg-like peptide, we proposed that SdrG follows a "dock, lock, and latch" mechanism to bind to Fg. This binding mechanism involves the docking of the ligand in a pocket formed between two SdrG subdomains followed by the movement of a C-terminal extension of one subdomain to cover the ligand and to insert and complement a beta-sheet in a neighboring subdomain. These proposed events result in a greatly stabilized closed conformation of the MSCRAMM-ligand complex. In this report, we describe a biochemical analysis of the proposed conformational changes that SdrG undergoes upon binding to its ligand. We have introduced disulfide bonds into SdrG to stabilize the open and closed forms of the apo-form of the MSCRAMM. We show that the stabilized closed form does not bind to the ligand and that binding can be restored in the presence of reducing agents such as dithiothreitol. We have also used Förster resonance energy transfer to dynamically show the conformational changes of SdrG upon binding to its ligand. Finally, we have used isothermic calorimetry to determine that hydrophobic interactions between the ligand and the protein are responsible for re-directing the C-terminal extension of the second subdomain required for triggering the beta-strand complementation event.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Ligands , Staphylococcus epidermidis/metabolism , Amino Acid Sequence , Bacterial Proteins/genetics , Carrier Proteins/genetics , Dimerization , Fibrinogen/metabolism , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Sequence Data , Peptides/chemical synthesis , Peptides/chemistry , Peptides/metabolism , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Spectrometry, Fluorescence , Staphylococcus epidermidis/genetics
11.
Science ; 315(5815): 1130-3, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17234914

ABSTRACT

The Staphylococcus aureus Panton-Valentine leukocidin (PVL) is a pore-forming toxin secreted by strains epidemiologically associated with the current outbreak of community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) and with the often-lethal necrotizing pneumonia. To investigate the role of PVL in pulmonary disease, we tested the pathogenicity of clinical isolates, isogenic PVL-negative and PVL-positive S. aureus strains, as well as purified PVL, in a mouse acute pneumonia model. Here we show that PVL is sufficient to cause pneumonia and that the expression of this leukotoxin induces global changes in transcriptional levels of genes encoding secreted and cell wall-anchored staphylococcal proteins, including the lung inflammatory factor staphylococcal protein A (Spa).


Subject(s)
Exotoxins/physiology , Leukocidins/physiology , Lung/pathology , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Staphylococcal Protein A/metabolism , Staphylococcus aureus/pathogenicity , Virulence Factors/physiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Disease Models, Animal , Exotoxins/genetics , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Hemorrhage , Leukocidins/genetics , Lung/microbiology , Methicillin Resistance , Mice , Mice, Inbred BALB C , Necrosis , Oligonucleotide Array Sequence Analysis , Staphylococcal Protein A/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Transcription, Genetic , Virulence , Virulence Factors/genetics
12.
Microbiology (Reading) ; 151(Pt 5): 1453-1464, 2005 May.
Article in English | MEDLINE | ID: mdl-15870455

ABSTRACT

Staphylococcus epidermidis is a ubiquitous human skin commensal that has emerged as a major cause of foreign-body infections. Eleven genes encoding putative cell-wall-anchored proteins were identified by computer analysis of the publicly available S. epidermidis unfinished genomic sequence. Four genes encode previously described proteins (Aap, Bhp, SdrF and SdrG), while the remaining seven have not been characterized. Analysis of primary sequences of the Staphylococcus epidermidis surface (Ses) proteins indicates that they have a structural organization similar to the previously described cell-wall-anchored proteins from S. aureus and other Gram-positive cocci. However, not all of the Ses proteins are direct homologues of the S. aureus proteins. Secondary and tertiary structure predictions suggest that most of the Ses proteins are composed of several contiguous subdomains, and that the majority of these predicted subdomains are folded into beta-rich structures. PCR analysis indicates that certain genes may be found more frequently in disease isolates compared to strains isolated from healthy skin. Patients recovering from S. epidermidis infections had higher antibody titres against some Ses proteins, implying that these proteins are expressed during human infection. Western blot analyses of early-logarithmic and late-stationary in vitro cultures suggest that different regulatory mechanisms control the expression of the Ses proteins.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/metabolism , Membrane Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Gene Expression Regulation, Bacterial , Humans , Immunoglobulin G/blood , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/immunology , Polymerase Chain Reaction/methods , Staphylococcal Infections/immunology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/growth & development , Staphylococcus epidermidis/immunology
13.
Biomol Eng ; 21(3-5): 105-11, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15567104

ABSTRACT

Bacterial adhesion to extracellular matrix proteins plays a major role in infections of host tissue and medical devices. In some species of gram-positive cocci, this adhesion is mediated by specific molecules present on the bacterial cell surface. We have used optical tweezers to dynamically measure the adhesive force between an individual Staphylococcus aureus bacterium and a fibronectin-coated surface. A bacterium was optically trapped and brought in contact with a 10-microm diameter polystyrene microsphere coated with fibronectin. The force required to detach the cell from the microsphere was measured by tracking the displacement signals of the trapped cell on a quadrant photodiode throughout the detachment process for a series of S. aureus strains expressing fibronectin-binding proteins with various degrees of mutation. The single-bond rupture forces ranged between 15 and 26 pN depending on the extent of mutation. No binding was observed in the strain with the highest degree of mutation. These results confirm that multiple regions of the S. aureus fibronectin adhesin participate in the binding process and provide further insight into the role of these regions in the adhesive process.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Adhesion/physiology , Micromanipulation/instrumentation , Micromanipulation/methods , Staphylococcus aureus/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Binding Sites , Extracellular Matrix Proteins/physiology , Extracellular Matrix Proteins/ultrastructure , Mutagenesis, Site-Directed , Optics and Photonics/instrumentation , Protein Binding , Recombinant Proteins/metabolism , Staphylococcus aureus/cytology , Stress, Mechanical , Structure-Activity Relationship
14.
Cell ; 115(2): 217-28, 2003 Oct 17.
Article in English | MEDLINE | ID: mdl-14567919

ABSTRACT

Gram-positive pathogens such as staphylococci contain multiple cell wall-anchored proteins that serve as an interface between the microbe and its environment. Some of these proteins act as adhesins and mediate bacterial attachment to host tissues. SdrG is a cell wall-anchored adhesin from Staphylococcus epidermidis that binds to the Bbeta chain of human fibrinogen (Fg) and is necessary and sufficient for bacterial attachment to Fg-coated biomaterials. Here, we present the crystal structures of the ligand binding region of SdrG as an apoprotein and in complex with a synthetic peptide analogous to its binding site in Fg. Analysis of the crystal structures, along with mutational studies of both the protein and of the peptide, reveals that SdrG binds to its ligand with a dynamic "dock, lock, and latch" mechanism. We propose that this mechanism represents a general mode of ligand binding for structurally related cell wall-anchored proteins of gram-positive bacteria.


Subject(s)
Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Fibrinogen/metabolism , Staphylococcus epidermidis/metabolism , Adhesins, Bacterial/genetics , Alanine/metabolism , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Bacterial Adhesion , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Fibrinogen/chemistry , Fibrinogen/drug effects , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptides/chemistry , Peptides/genetics , Protein Binding , Protein Structure, Tertiary , Serine/metabolism , Thrombin/pharmacology
16.
J Biol Chem ; 277(45): 43017-23, 2002 Nov 08.
Article in English | MEDLINE | ID: mdl-12218064

ABSTRACT

The opportunistic human pathogen Staphylococcus epidermidis is the major cause of nosocomial biomaterial infections. S. epidermidis has the ability to attach to indwelling materials coated with extracellular matrix proteins such as fibrinogen, fibronectin, vitronectin, and collagen. To identify the proteins necessary for S. epidermidis attachment to collagen, we screened an expression library using digoxigenin-labeled collagen as well as two monoclonal antibodies generated against the Staphylococcus aureus collagen-adhesin, Cna, as probes. These monoclonal antibodies recognize collagen binding epitopes on the surface of S. aureus and S. epidermidis cells. Using this approach, we identified GehD, the extracellular lipase originally found in S. epidermidis 9, as a collagen-binding protein. Despite the monoclonal antibody cross-reactivity, the GehD amino acid sequence and predicted structure are radically different from those of Cna. The mature GehD circular dichroism spectra differs from that of Cna but strongly resembles that of a mammalian cell-surface collagen binding receptor, known as the alpha(1) integrin I domain, suggesting that they have similar secondary structures. The GehD protein is translated as a preproenzyme, secreted, and post-translationally processed into mature lipase. GehD does not have the conserved LPXTG C-terminal motif present in cell wall-anchored proteins, but it can be detected in lysostaphin cell wall extracts. A recombinant version of mature GehD binds to collagens type I, II, and IV adsorbed onto microtiter plates in a dose-dependent saturable manner. Recombinant, mature GehD protein and anti-GehD antibodies can inhibit the attachment of S. epidermidis to immobilized collagen. These results provide evidence that GehD may be a bi-functional molecule, acting not only as a lipase but also as a cell surface-associated collagen adhesin.


Subject(s)
Adhesins, Bacterial/metabolism , Carboxylic Ester Hydrolases/metabolism , Collagen/metabolism , Lipase/metabolism , Staphylococcus epidermidis/physiology , Bacterial Adhesion , Base Sequence , Carboxylic Ester Hydrolases/genetics , Circular Dichroism , Cloning, Molecular , DNA Primers , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Enzyme-Linked Immunosorbent Assay , Escherichia coli/enzymology , Escherichia coli/genetics , Gene Library , Lipase/genetics , Protein Binding , Spectrophotometry, Ultraviolet , Staphylococcus epidermidis/enzymology
17.
Lasers Surg Med ; 31(1): 45-52, 2002.
Article in English | MEDLINE | ID: mdl-12124715

ABSTRACT

BACKGROUND AND OBJECTIVES: Biomaterial-mediated infection, a common cause of medical device failure, is initiated by bacterial adhesion to an adsorbed protein layer on the implant surface. This adhesion is thought to be mediated by specific molecules present on the bacterial cell surface. Optical tweezers can be used to measure the adhesive force between a single bacterium and a protein-coated surface. STUDY DESIGN/MATERIALS AND METHODS: Using optical tweezers, a bacterium was trapped and brought in contact with a 10-microm diameter polystyrene microsphere coated with fibronectin. The minimum force required to detach the cell from the bead was determined over a range of fibronectin concentrations and contact times. RESULTS: The detachment forces were integer multiples of an 18-pN base value that was independent of contact time and coating concentration; we propose that the variation in force is related to the number of bonds formed. CONCLUSIONS: These experiments demonstrate that optical tweezers can be used to investigate the adhesion of individual bacteria to surfaces. The results suggest that S. epidermidis has surface proteins capable of binding fibronectin.


Subject(s)
Bacterial Adhesion , Fibronectins/metabolism , Lasers , Staphylococcus epidermidis/physiology , Adhesiveness , Biofilms , Calibration , Extracellular Matrix/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...