Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Protein Sci ; 33(7): e5030, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38864696

ABSTRACT

Bacterial adhesins are cell-surface proteins that anchor to the cell wall of the host. The first stage of infection involves the specific attachment to fibrinogen (Fg), a protein found in human blood. This attachment allows bacteria to colonize tissues causing diseases such as endocarditis. The study of this family of proteins is hence essential to develop new strategies to fight bacterial infections. In the case of the Gram-positive bacterium Staphylococcus aureus, there exists a class of adhesins known as microbial surface components recognizing adhesive matrix molecules (MSCRAMMs). Here, we focus on one of them, the clumping factor A (ClfA), which has been found to bind Fg through the dock-lock-latch mechanism. Interestingly, it has recently been discovered that MSCRAMM proteins employ a catch-bond to withstand forces exceeding 2 nN, making this type of interaction as mechanically strong as a covalent bond. However, it is not known whether this strength is an evolved feature characteristic of the bacterial protein or is typical only of the interaction with its partner. Here, we combine single-molecule force spectroscopy, biophysical binding assays, and molecular simulations to study the intrinsic mechanical strength of ClfA. We find that despite the extremely high forces required to break its interactions with Fg, ClfA is not by itself particularly strong. Integrating the results from both theory and experiments we dissect contributions to the mechanical stability of this protein.


Subject(s)
Coagulase , Fibrinogen , Staphylococcus aureus , Staphylococcus aureus/metabolism , Staphylococcus aureus/chemistry , Coagulase/metabolism , Coagulase/chemistry , Fibrinogen/chemistry , Fibrinogen/metabolism , Protein Binding , Adhesins, Bacterial/metabolism , Adhesins, Bacterial/chemistry , Humans , Protein Stability
2.
J Biol Chem ; 298(1): 101493, 2022 01.
Article in English | MEDLINE | ID: mdl-34915025

ABSTRACT

Fibrin (Fbn) deposits are a hallmark of staphylocoagulase (SC)-positive endocarditis. Binding of the N terminus of Staphylococcus aureus SC to host prothrombin triggers formation of an active SC·prothrombin∗ complex that cleaves host fibrinogen to Fbn. In addition, the C-terminal domain of the prototypical SC contains one pseudorepeat (PR) and seven repeats (R1 → R7) that bind fibrinogen/Fbn fragment D (frag D) by a mechanism that is unclear. Here, we define affinities and stoichiometries of frag D binding to C-terminal SC constructs, using fluorescence equilibrium binding, NMR titration, alanine scanning, and native PAGE. We found that constructs containing the PR and single repeats bound frag D with KD ∼50 to 130 nM and a 1:1 stoichiometry, indicating a conserved binding site bridging the PR and each repeat. NMR titration of PR-R7 with frag D revealed that residues 22 to 49, bridging PR and R7, constituted the minimal peptide (MP) for binding, corroborated by alanine scanning, and binding of labeled MP to frag D. MP alignment with the PR-R and inter-repeat junctions identified critical conserved residues. Full-length PR-(R1 → R7) bound frag D with KD ∼20 nM and a stoichiometry of 1:5, whereas constructs containing the PR and various three repeats competed with PR-(R1 → R7) for frag D binding, with a 1:3 stoichiometry. These findings are consistent with binding at PR-R and R-R junctions with modest inter-repeat sequence variability. CD of PR-R7 and PR-(R1 → R7) suggested a disordered flexible structure, allowing binding of multiple fibrin(ogen) molecules. Taken together, these results provide insights into pathogen localization on host fibrin networks.


Subject(s)
Coagulase , Fibrinogen , Alanine/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Coagulase/chemistry , Coagulase/metabolism , Fibrin/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Protein Binding , Prothrombin/metabolism , Terminal Repeat Sequences
3.
J Biochem ; 169(5): 543-553, 2021 Jul 03.
Article in English | MEDLINE | ID: mdl-33326036

ABSTRACT

In the complement system, the opsonin C3b binds to the bacterial cell surface and mediates the opsonophagocytosis. However, the cell-wall protein SdrE of Staphylococcus aureus inhibits the C3b activity by recruiting the complement regulatory protein factor H (fH). SdrE binds to fH via its N-terminal N2N3 domain, which are also found in six other staphylococcal cell-wall proteins. In this study, we report that not only the N2N3 domain of SdrE but also those of ClfA, FnbpA and FnbpB can bind to fH. When immobilized on a microplate, the N2N3 domains recruited fH and enhanced the factor I (fI)-mediated cleavage of C3b. When mixed with fH and S. aureus cells, the N2N3 domains inhibited the fH binding to S. aureus cells and reduced the fI-mediated C3b cleavage on the bacterial cell surface. The F(ab)'2 fragments of the rabbit N2N3 antibodies also inhibited the fH binding to the S. aureus cell surface. When added to human blood, the N2N3 antibodies or the N2N3 domain proteins significantly increased the bactericidal activity. Based on these results, we conclude that, in S. aureus, not only SdrE but also ClfA, FnbpA and FnbpB can contribute to the inhibition of C3b-mediated opsonophagocytosis.


Subject(s)
Coagulase/chemistry , Staphylococcus aureus/chemistry , Coagulase/genetics , Coagulase/metabolism , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Factor H/metabolism , Humans , Protein Binding , Protein Domains , Staphylococcus aureus/metabolism
4.
J Biol Chem ; 295(17): 5614-5625, 2020 04 24.
Article in English | MEDLINE | ID: mdl-32156702

ABSTRACT

In Staphylococcus aureus-caused endocarditis, the pathogen secretes staphylocoagulase (SC), thereby activating human prothrombin (ProT) and evading immune clearance. A previous structural comparison of the SC(1-325) fragment bound to thrombin and its inactive precursor prethrombin 2 has indicated that SC activates ProT by inserting its N-terminal dipeptide Ile1-Val2 into the ProT Ile16 pocket, forming a salt bridge with ProT's Asp194, thereby stabilizing the active conformation. We hypothesized that these N-terminal SC residues modulate ProT binding and activation. Here, we generated labeled SC(1-246) as a probe for competitively defining the affinities of N-terminal SC(1-246) variants preselected by modeling. Using ProT(R155Q,R271Q,R284Q) (ProTQQQ), a variant refractory to prothrombinase- or thrombin-mediated cleavage, we observed variant affinities between ∼1 and 650 nm and activation potencies ranging from 1.8-fold that of WT SC(1-246) to complete loss of function. Substrate binding to ProTQQQ caused allosteric tightening of the affinity of most SC(1-246) variants, consistent with zymogen activation through occupation of the specificity pocket. Conservative changes at positions 1 and 2 were well-tolerated, with Val1-Val2, Ile1-Ala2, and Leu1-Val2 variants exhibiting ProTQQQ affinity and activation potency comparable with WT SC(1-246). Weaker binding variants typically had reduced activation rates, although at near-saturating ProTQQQ levels, several variants exhibited limiting rates similar to or higher than that of WT SC(1-246). The Ile16 pocket in ProTQQQ appears to favor nonpolar, nonaromatic residues at SC positions 1 and 2. Our results suggest that SC variants other than WT Ile1-Val2-Thr3 might emerge with similar ProT-activating efficiency.


Subject(s)
Bacterial Proteins/metabolism , Coagulase/metabolism , Prothrombin/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/chemistry , Binding Sites , Coagulase/chemistry , Humans , Models, Molecular , Protein Binding , Prothrombin/chemistry , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/chemistry , Substrate Specificity
5.
Proteins ; 88(3): 462-475, 2020 03.
Article in English | MEDLINE | ID: mdl-31589780

ABSTRACT

Protein engineering and synthetic biology stand to benefit immensely from recent advances in silico tools for structural and functional analyses of proteins. In the context of designing novel proteins, current in silico tools inform the user on individual parameters of a query protein, with output scores/metrics unique to each parameter. In reality, proteins feature multiple "parts"/functions and modification of a protein aimed at altering a given part, typically has collateral impact on other protein parts. A system for prediction of the combined effect of design parameters on the overall performance of the final protein does not exist. Function2Form Bridge (F2F-Bridge) attempts to address this by combining the scores of different design parameters pertaining to the protein being analyzed into a single easily interpreted output describing overall performance. The strategy comprises of (a) a mathematical strategy combining data from a myriad of in silico tools into an OP-score (a singular score informing on a user-defined overall performance) and (b) the F2F Plot, a graphical means of informing the wetlab biologist holistically on designed construct suitability in the context of multiple parameters, highlighting scope for improvement. F2F predictive output was compared with wetlab data from a range of synthetic proteins designed, built, and tested for this study. Statistical/machine learning approaches for predicting overall performance, for use alongside the F2F plot, were also examined. Comparisons between wetlab performance and F2F predictions demonstrated close and reliable correlations. This user-friendly strategy represents a pivotal enabler in increasing the accessibility of synthetic protein building and de novo protein design.


Subject(s)
Antibodies/chemistry , Coagulase/chemistry , Machine Learning , Mucin-1/chemistry , Synthetic Biology/methods , Antibodies/metabolism , Coagulase/metabolism , Humans , Models, Statistical , Mucin-1/metabolism , Protein Engineering/methods , Staphylococcus aureus/chemistry , Structure-Activity Relationship
6.
Trends Microbiol ; 27(11): 927-941, 2019 11.
Article in English | MEDLINE | ID: mdl-31375310

ABSTRACT

The microbial surface components recognizing adhesive matrix molecules (MSCRAMMs) are a family of proteins that are defined by the presence of two adjacent IgG-like folded subdomains. These promote binding to ligands by mechanisms that involve major conformational changes exemplified by the binding to fibrinogen by the 'dock-lock-latch' mechanism or to collagen by the 'collagen hug'. Clumping factors A and B are two such MSCRAMMs that have several important roles in the pathogenesis of Staphylococcus aureus infections. MSCRAMM architecture, ligand binding, and roles in infection and colonization are examined with a focus on recent developments with clumping factors.


Subject(s)
Adhesins, Bacterial/physiology , Gram-Positive Cocci/physiology , Adhesins, Bacterial/chemistry , Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Cell Wall/chemistry , Cell Wall/physiology , Coagulase/chemistry , Humans , Ligands , Protein Binding , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Staphylococcus epidermidis/physiology
7.
Front Immunol ; 10: 2967, 2019.
Article in English | MEDLINE | ID: mdl-31921206

ABSTRACT

Staphylococcus aureus causes localized infections or invasive diseases (abscesses or endocarditis). One of its virulence factors is staphylocoagulase (SCG), which binds prothrombin to form a complex with thrombin-like proteolytic activity and leads to uncontrolled fibrin generation at sites of bacterial inoculation. The aim of this study was to characterize the formation, structure, mechanical properties and lysis of SCG-generated clots. Recombinant SCG was expressed in Escherichia coli, purified and the amidolytic activity of its complexes with human prothrombin (SCG-PT) and thrombin (SCG-T) was determined using human thrombin as a reference. Fibrin clots were prepared from purified fibrinogen and human plasma using thrombin, SCG-PT or SCG-T as a coagulase. The kinetics of clot formation and lysis by tissue-type plasminogen activator (tPA) were monitored with turbidimetric assays. Fibrin ultrastructure was examined with scanning electron microscopy and small-angle X-ray scattering (SAXS). Fibrin clot porosity was characterized with fluid permeation assays, whereas the viscoelastic properties and mechanical stability were evaluated with oscillation rheometry. Compared to thrombin, the amidolytic and clotting activity of SCG-PT was 1.6- to 2.5-fold lower on a molar basis. SCG-T had equivalent amidolytic, but reduced clotting activity both on pure fibrinogen (1.6-fold), and in plasma (1.3-fold). The SCG-PT and SCG-T generated fibrin with thicker fibers (10-60% increase in median diameter) than thrombin due to increased number of fibrin protofibrils per fiber cross-section. According to the fluid permeability of the clots SCG-PT and SCG-T promoted the formation of more porous structures. The shear stress resistance in the pure fibrin and plasma clots generated by SCG-PT was significantly lower than in the thrombin clots (243.8 ± 22.0 Pa shear stress was sufficient for disassembly of SCG-PT fibrin vs. 937.3 ± 65.6 Pa in thrombin clots). The tPA-mediated lysis of both pure fibrin and plasma clots produced by SCG-PT or SCG-T was accelerated compared to thrombin, resulting in up to a 2.1-fold increase in tPA potency. Our results indicate that SCG generates a thrombus scaffold with a structure characterized by impaired mechanical stability and increased lytic susceptibility. This proneness to clot disintegration could have implications in the septic embolism from endocardial bacterial vegetation.


Subject(s)
Blood Coagulation , Coagulase/chemistry , Fibrin/chemistry , Plasma/chemistry , Staphylococcus aureus/enzymology , Fibrinogen/chemistry , Humans , Prothrombin/chemistry , Thrombin/chemistry
8.
Microbes Infect ; 21(3-4): 163-169, 2019.
Article in English | MEDLINE | ID: mdl-30447395

ABSTRACT

Coagulase (Coa) secreted by Staphylococcus aureus is associated with the establishment of staphylococcal disease, which activates host prothrombin and generates fibrin shields. The R domain of Coa, consisting of several conserved repeats, is important in immune evasion during S. aureus infection. However, previous research showed that the Coa R domain induced very weak specific antibody responses. In this study, we constructed a new R domain, CoaR6, consisting of 6 repeats that occur most frequently in clinical isolates. By fusing CoaR6 with Hc, the C-terminal fragment of the heavy chain of tetanus neurotoxin, we successfully increased anti-CoaR6 IgG levels in immunized mice which were hardly detected in mice immunized with CoaR6 plus alum. To further improve anti-CoaR6 responses, the combination adjuvants alum plus CpG were formulated with the antigen and exhibited a significantly higher specific antibody response. Moreover, active Th1/Th17 immune responses were observed in Hc-CoaR6 immunized group rather than CoaR6. Active immunization of Hc-CoaR6 with alum plus CpG showed protective effects in a peritonitis model induced by two S. aureus strains with different coagulase types. Our results provided strategies to improve the immunogenicity of R domain and supporting evidences for R domain to be an S. aureus vaccine candidate.


Subject(s)
Bacterial Proteins/immunology , Coagulase/immunology , Staphylococcal Infections/prevention & control , Staphylococcal Vaccines/immunology , Staphylococcus aureus/enzymology , Staphylococcus aureus/immunology , Amino Acid Sequence , Animals , Antibodies, Bacterial/blood , Antigens, Bacterial/immunology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Coagulase/chemistry , Coagulase/genetics , Coagulase/metabolism , Cytokines/immunology , Immunity, Cellular , Metalloendopeptidases/genetics , Mice , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , Staphylococcal Infections/immunology , Staphylococcal Vaccines/administration & dosage , Survival Analysis , Tetanus Toxin/genetics , Vaccination
9.
Biomol NMR Assign ; 11(2): 243-249, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28819722

ABSTRACT

The C-terminal repeat domain of staphylocoagulase that is secreted by the S. aureus is believed to play an important role interacting with fibrinogen and promotes blood clotting. To study this interaction by NMR, full assignment of each amide residue in the HSQC spectrum was required. Despite of the short sequence of the repeat construct, the HSQC spectrum contained a substantial amount of overlapped and exchange broadened resonances, indicating little secondary or tertiary structure. This caused severe problems while using the conventional, amide based NMR method for the backbone assignment. With the growing interest in small apparently disordered proteins, these issues are being faced more frequently. An alternative strategy to improve the backbone assignment capability involved carbon direct detection methods. Circumventing the amide proton detection offers a larger signal dispersion and more uniform signal intensity. For peptides with higher concentrations and in combination with the cold carbon channels of new cryoprobes, higher fields, and sufficiently long relaxation times, the disadvantage of the lower sensitivity of the 13C nucleus can be overcome. Another advantage of this method is the assignment of the proline backbone residues. Complete assignment with the carbon-detected strategy was achieved with a set of only two 3D, one 2D, and a HNCO measurement, which was necessary to translate the information to the HSQC spectrum.


Subject(s)
Amides/chemistry , Carbon/chemistry , Coagulase/chemistry , Nuclear Magnetic Resonance, Biomolecular , Staphylococcus aureus/enzymology
10.
Sci Rep ; 7: 40608, 2017 01 19.
Article in English | MEDLINE | ID: mdl-28102235

ABSTRACT

Staphylococcus aureus is one of major pathogens that can cause a series of diseases in different hosts. In the bovine, it mainly causes subclinical and contagious mastitis, but its mechanisms of infection are not fully understood. Considering the fact that virulence factors play key roles in interactions between the bacterium and host cells, this study aimed to identify if a binding partner of S. aureus clumping factor A (ClfA) exists on the bovine mammary epithelial cells. The ClfA protein was used as a bait to pull down lysates of cultured bovine mammary epithelial cells (MAC-T cells). One pull-down protein was identified through use of mass spectrometry and bioinformatics analyses as bovine AnnexinA2. The Western blot and in vitro binding assay confirmed that the full A domain of ClfA was necessary to bind to AnnexinA2. In addition, the interaction between ClfA and AnnexinA2 was validated biochemically by ELISA with a KD value of 418+/-93 nM. The confocal microscopy demonstrated that ClfA and AnnexinA2 partially co-localized in the plasma membrane and that the majority of them were transported into cytoplasm. Taken together, the results demonstrate that ClfA binds with AnnexinA2 and this interaction could mediate S. aureus invasion into bovine mammary epithelial cells.


Subject(s)
Annexin A2/metabolism , Coagulase/metabolism , Epithelial Cells/metabolism , Mammary Glands, Animal/cytology , Staphylococcus aureus/metabolism , Amino Acid Sequence , Animals , Annexin A2/chemistry , Calcium/pharmacology , Cattle , Cell Line , Coagulase/chemistry , Epithelial Cells/drug effects , Female , Fibrinogen/pharmacology , Protein Domains , Recombinant Proteins/metabolism
11.
EBioMedicine ; 13: 328-338, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27789272

ABSTRACT

The Staphylococcus aureus fibrinogen binding MSCRAMM (Microbial Surface Components Recognizing Adhesive Matrix Molecules), ClfA (clumping factor A) is an important virulence factor in staphylococcal infections and a component of several vaccines currently under clinical evaluation. The mouse monoclonal antibody aurexis (also called 12-9), and the humanized version tefibazumab are therapeutic monoclonal antibodies targeting ClfA that in combination with conventional antibiotics were effective in animal models but showed less impressive efficacy in a limited Phase II clinical trial. We here report the crystal structure and a biochemical characterization of the ClfA/tefibazumab (Fab) complex. The epitope for tefibazumab is located to the "top" of the N3 subdomain of ClfA and partially overlaps with a previously unidentified second binding site for fibrinogen. A high-affinity binding of ClfA to fibrinogen involves both an interaction at the N3 site and the previously identified docking of the C-terminal segment of the fibrinogen γ-chain in the N2N3 trench. Although tefibazumab binds ClfA with high affinity we observe a modest IC50 value for the inhibition of fibrinogen binding to the MSCRAMM. This observation, paired with a common natural occurring variant of ClfA that is not effectively recognized by the mAb, may partly explain the modest effect tefibazumab showed in the initial clinic trail. This information will provide guidance for the design of the next generation of therapeutic anti-staphylococcal mAbs targeting ClfA.


Subject(s)
Antibodies, Monoclonal/chemistry , Coagulase/chemistry , Models, Molecular , Protein Conformation , Amino Acid Substitution , Antibodies, Monoclonal/metabolism , Binding Sites , Coagulase/immunology , Coagulase/metabolism , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/immunology , Epitopes/metabolism , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Protein Binding , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry
13.
J Exp Med ; 213(3): 293-301, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26880578

ABSTRACT

Host immunity against bacteria typically involves antibodies that recognize the microbial surface and promote phagocytic killing. Methicillin-resistant Staphylococcus aureus (MRSA) is a frequent cause of lethal bloodstream infection; however, vaccines and antibody therapeutics targeting staphylococcal surface molecules have thus far failed to achieve clinical efficacy. S. aureus secretes coagulase (Coa), which activates host prothrombin and generates fibrin fibrils that protect the pathogen against phagocytosis by immune cells. Because of negative selection, the coding sequence for the prothrombin-binding D1-D2 domain is highly variable and does not elicit cross-protective immune responses. The R domain, tandem repeats of a 27-residue peptide that bind fibrinogen, is conserved at the C terminus of all Coa molecules, but its functional significance is not known. We show here that the R domain enables bloodstream infections by directing fibrinogen to the staphylococcal surface, generating a protective fibrin shield that inhibits phagocytosis. The fibrin shield can be marked with R-specific antibodies, which trigger phagocytic killing of staphylococci and protect mice against lethal bloodstream infections caused by a broad spectrum of MRSA isolates. These findings emphasize the critical role of coagulase in staphylococcal escape from opsonophagocytic killing and as a protective antigen for S. aureus vaccines.


Subject(s)
Antibodies, Bacterial/immunology , Coagulase/immunology , Phagocytosis , Staphylococcus aureus/immunology , Agglutination , Animals , Antibodies, Monoclonal/immunology , Coagulase/chemistry , Female , Fibrin/metabolism , Humans , Mice, Inbred BALB C , Opsonin Proteins/metabolism , Protein Structure, Tertiary , Repetitive Sequences, Amino Acid , Staphylococcal Infections/blood , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control
14.
mBio ; 7(1): e01885-15, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26733070

ABSTRACT

UNLABELLED: Coagulase (Coa) and Efb, secreted Staphylococcus aureus proteins, are important virulence factors in staphylococcal infections. Coa interacts with fibrinogen (Fg) and induces the formation of fibrin(ogen) clots through activation of prothrombin. Efb attracts Fg to the bacterial surface and forms a shield to protect the bacteria from phagocytic clearance. This communication describes the use of an array of synthetic peptides to identify variants of a linear Fg binding motif present in Coa and Efb which are responsible for the Fg binding activities of these proteins. This motif represents the first Fg binding motif identified for any microbial protein. We initially located the Fg binding sites to Coa's C-terminal disordered segment containing tandem repeats by using recombinant fragments of Coa in enzyme-linked immunosorbent assay-type binding experiments. Sequence analyses revealed that this Coa region contained shorter segments with sequences similar to the Fg binding segments in Efb. An alanine scanning approach allowed us to identify the residues in Coa and Efb that are critical for Fg binding and to define the Fg binding motifs in the two proteins. In these motifs, the residues required for Fg binding are largely conserved, and they therefore constitute variants of a common Fg binding motif which binds to Fg with high affinity. Defining a specific motif also allowed us to identify a functional Fg binding register for the Coa repeats that is different from the repeat unit previously proposed. IMPORTANCE: Staphylococcus aureus infections are a major health problem that affects an estimated 50 million people globally and causes the death of about 20,000 Americans each year. A number of experimental vaccines have been developed during the past years. However, these vaccines have all failed in clinical trials. The ability of S. aureus to form an Fg shield surrounding and protecting bacterial cells from clearance may explain why the vaccines are failing. Furthermore, S. aureus coagulase can induce the formation of a fibrin(ogen) shield in experimental abscess models which surrounds and protects bacteria in the microcolony from clearance. In this study, we identified for the first time a microbial Fg binding motif. Variants of this motif are present in coagulase and Efb. Our results provide a molecular basis for the rational design of inhibitors that could potentially prevent the formation of the obstructing Fg shield.


Subject(s)
Amino Acid Motifs , Bacterial Proteins/metabolism , Coagulase/metabolism , Fibrinogen/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Coagulase/chemistry , Coagulase/genetics , Conserved Sequence , DNA Mutational Analysis , Humans , Protein Binding
15.
Acta Biochim Pol ; 62(3): 517-22, 2015.
Article in English | MEDLINE | ID: mdl-26317125

ABSTRACT

We have discovered that addition of monomeric desAB fibrin to prothrombin leads to appearance of the thrombin-like activity of prothrombin towards S2238 chromogenic substrate. DesA and desABß(15-42)2 fibrin forms did not cause any activation of prothrombin. From this observation we could suggested that amino acid residues of the 15-42 fragment of BßN-domain presented in desAB fibrin, cleaved in desABß(15-42)2 fibrin and protected in desA fibrin, play a crucial role in the non-enzymatic activation of prothrombin. To identify the Bß amino acid residues involved in the fibrin-prothrombin binding we used monoclonal antibodies 1-5G and 2d2a with epitopes in Bß26-42 and Bß12-26 fibrin fragments respectively. The thrombin-like activity in the mixture of prothrombin and desAB fibrin was monitored in the presence of each of these monoclonal antibodies. It was found that anti-Bß12-26 antibody does not exhibit any inhibitory effect on the thombin-like activity of the mixture. In contrast, adding of Bß26-42 antibody into the mixture of desAB fibrin with prothrombin diminished the thrombin-like activity by 70%. Recombinant dimeric peptides Bß(15-44)2 and Bß(15-66)2 that mimic amino acid residues in fibrin were also tested for their ability to activate prothrombin. It was found that both peptides were able to induce non-enzymatic activation of prothrombin. The activation was more evident in the case of Bß(15-44)2 peptide. From the data obtained we can conclude that desAB fibrin binds to prothrombin through the Bß26-42 amino acid residues and the formation of such a complex caused a non-enzymatic activation of prothrombin.


Subject(s)
Fibrin/chemistry , Prothrombin/chemistry , Amino Acids/chemistry , Antibodies, Monoclonal/chemistry , Catalysis , Coagulase/chemistry , Epitopes/chemistry , Fibrin Fibrinogen Degradation Products/chemistry , Fibrinogen/chemistry , Humans , Models, Molecular , Peptide Fragments/chemistry , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary
16.
J Zhejiang Univ Sci B ; 16(1): 70-7, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25559958

ABSTRACT

In many traditional Chinese medicine (TCM) hospitals, most patients are elderly with chronic diseases. Nosocomial bloodstream infections (nBSIs) are an important cause of morbidity and mortality. A retrospective surveillance study was performed to examine the epidemiology and microbiology of nBSIs in a TCM hospital from 2009 to 2011. A total of 482 patients with nBSIs were included in the study period. The incidence rate was 5.7/1000 admissions. Escherichia coli (25.5%) was the most common Gram-negative and coagulase-negative staphylococcus (CoNS) (14.1%) was the most common Gram-positive organism isolated. One-third of the E. coli and Klebsiella pneumoniae isolated from the nBSIs were the third-generation cephalosporin-resistant. Half of the Acinetobacter species isolates were resistant to imipenem. Of all the CoNS isolates, 90.7% were resistant to methicillin. Carbapenems and glycopeptide were the most frequently used for nBSI therapy. Only about one-third of patients (157/482) received appropriate empirical therapy. Septic shock, hemodialysis, Pitt bacteremia score >4, urinary tract infection, and appropriate empirical therapy were most strongly associated with 28-d mortality. The incidence of nBSIs was low in the TCM hospital but the proportion of nBSIs due to antibiotic-resistant organisms was high. A high Pitt bacteremia score was one of the most important risk factors for mortality in nBSIs. Therefore, the implementation of appropriate empirical therapy is crucial to improve the clinical outcome of nBSIs.


Subject(s)
Bacteremia/epidemiology , Bacteremia/microbiology , Cross Infection/epidemiology , Cross Infection/microbiology , Acinetobacter , Adult , Aged , Aged, 80 and over , Cephalosporins/chemistry , China , Coagulase/chemistry , Escherichia coli , Female , Humans , Klebsiella pneumoniae , Male , Medicine, Chinese Traditional , Microbial Sensitivity Tests , Middle Aged , Regression Analysis , Retrospective Studies , Risk Factors , Shock, Septic/metabolism , Staphylococcus , Treatment Outcome
17.
Clin Vaccine Immunol ; 20(8): 1338-40, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23803901

ABSTRACT

The staphylococcal adhesin clumping factor A (ClfA) has a variant amino acid sequence, generating the potential for alterations in epitope structure and immunogenicity of this vaccine candidate. We demonstrated for two recombinant ClfA(40-531) (a slightly truncated version of the fibrinogen-binding domain of ClfA containing amino acids 40 to 531) genetic variants that strain-specific epitopes are immunodominant. This work indicates that immune responses elicited by ClfA may, at least in part, be dependent on the strain of origin of the ClfA.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Coagulase/genetics , Coagulase/immunology , Immunodominant Epitopes/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Animals , Antigens, Bacterial/chemistry , Coagulase/chemistry , Genetic Variation , Mice, Inbred BALB C , Models, Molecular , Protein Conformation
18.
Arq. Inst. Biol ; 80(1): 1-6, jan.-mar.2013.
Article in Portuguese | LILACS | ID: lil-784836

ABSTRACT

A mastite bovina é uma doença importante na exploração leiteira, não apenas pelas perdas econômicas diretas que promove, mas também pelas perdas indiretas e o potencial risco à saúde pública. Dentre as principais causas de infecções intramamárias, destacam-se as bactérias do gênero Staphylococcus spp., sendo que Staphylococcus aureus é o agente etiológico predominante em mastite subclínica. O objetivo desse trabalho foi verificar a frequência de mastite subclínica em oito rebanhos localizados na região Sul do Rio Grande do Sul (Brasil) e a relação da enfermidade com a presença de S. aureus. Adicionalmente, pesquisou-se a presença de S. intermedius e S. hyicus nas amostras de leite obtidas. Para identificação da doença, utilizou-se o California Mastitis Test (CMT). A identificação da espécie de Staphylococcus spp. foi feita em meio de cultura ágar Baird-Parker, com posterior confirmação das colônias suspeitas em coloração de gram, prova de catalase, pesquisa de coagulase livre e pesquisa de termonuclease. A mastite subclínica foi constatada em 53,6% dos animais testados. A presença de Staphylococcus coagulase positiva foi identificada em 12,6% dos animais com mastite subclínica. Nesses mesmos animais, a bactéria identificada como S. aureus foi o agente etiológico presente em 17,6% dos casos. Adicionalmente, pode-se perceber que, dentre o grupo identificado como coagulase positiva, 85,7% corresponderam a S. aureus, enquanto8,5% mostraram características bioquímicas compatíveis com S. intermedius e 5,8% foram consideradas S. hyicus...


Bovine mastitis is an important disease in dairy farming, not only by promoting direct economic losses, but also for indirect losses and the potential risk to public health. The main causes of intramammary infections include the bacteria of the genus Staphylococcus spp., Staphylococcus aureus being the predominant etiologic agent in subclinical mastitis. The aim of this study was to determine the frequency of subclinical mastitis in eight herds from southern Rio Grande do Sul (Brazil) and the relationship of the disease with the presence of S. aureus. In addition, we checked for the presence of S. intermedius and S. hyicus in the milk samples obtained. For identification of the disease, we used the California Mastitis Test (CMT). Identification of Staphylococcus spp. species was made in Barid-Parker agar culture medium, with subsequent confirmation of suspected colonies by way of Gram stain and catalase test along with free-coagulase and thermonuclease research. Subclinical mastitis was identified in 53.6% of animals tested. The presence of coagulase-positive Staphylococcus was identified in 12.6% of animals with subclinical mastitis. In these same animals, bacteria identified as S. aureus were the etiologic agent present in 17.6% of cases. Additionally, it was revealed that among the group identified as coagulase positive, 85.7% corresponded to S. aureus, while 8.5% had biochemical characteristics consistent with S. intermedius and 5.8% were considered S. hyicus...


Subject(s)
Animals , Coagulase/chemistry , Livestock Industry/statistics & numerical data , Mastitis, Bovine/pathology , Public Health , Staphylococcus aureus/pathogenicity
19.
PLoS One ; 7(5): e38407, 2012.
Article in English | MEDLINE | ID: mdl-22675461

ABSTRACT

Similar to other highly successful invasive bacterial pathogens, Staphylococcus aureus recruits the complement regulatory protein factor H (fH) to its surface to inhibit the alternative pathway of complement. Here, we report the identification of the surface-associated protein SdrE as a fH-binding protein using purified fH overlay of S. aureus fractionated cell wall proteins and fH cross-linking to S. aureus followed by mass spectrometry. Studies using recombinant SdrE revealed that rSdrE bound significant fH whether from serum or as a purified form, in both a time- and dose-dependent manner. Furthermore, rSdrE-bound fH exhibited cofactor functionality for factor I (fI)-mediated cleavage of C3b to iC3b which correlated positively with increasing amounts of fH. Expression of SdrE on the surface of the surrogate bacterium Lactococcus lactis enhanced recruitment of fH which resulted in increased iC3b generation. Moreover, surface expression of SdrE led to a reduction in C3-fragment deposition, less C5a generation, and reduced killing by polymorphonuclear cells. Thus, we report the first identification of a S. aureus protein associated with the staphylococcal surface that binds factor H as an immune evasion mechanism.


Subject(s)
Bacterial Proteins/metabolism , Complement Factor H/metabolism , Immune Evasion , Staphylococcus aureus/immunology , Staphylococcus aureus/metabolism , Amino Acid Sequence , Bacterial Proteins/chemistry , Cell Wall/metabolism , Coagulase/chemistry , Coagulase/metabolism , Complement C3b/metabolism , Complement C5a/biosynthesis , Humans , Molecular Sequence Data , Neutrophils/immunology , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
20.
J Biol Chem ; 286(29): 25963-72, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21543319

ABSTRACT

Clumping factor B (ClfB) from Staphylococcus aureus is a bifunctional protein that binds to human cytokeratin 10 (K10) and fibrinogen (Fg). ClfB has been implicated in S. aureus colonization of nasal epithelium and is therefore a key virulence factor. People colonized with S. aureus are at an increased risk for invasive staphylococcal disease. In this study, we have determined the crystal structures of the ligand-binding region of ClfB in an apo-form and in complex with human K10 and Fg α-chain-derived peptides, respectively. We have determined the structures of MSCRAMM binding to two ligands with different sequences in the same site showing the versatile nature of the ligand recognition mode of microbial surface components recognizing adhesive matrix molecules. Both ligands bind ClfB by parallel ß-sheet complementation as observed for the clumping factor A·Î³-chain peptide complex. The ß-sheet complementation is shorter in the ClfB·Fg α-chain peptide complex. The structures show that several residues in ClfB are important for binding to both ligands, whereas others only make contact with one of the ligands. A common motif GSSGXG found in both ligands is part of the ClfB-binding site. This motif is found in many human proteins thus raising the possibility that ClfB recognizes additional ligands.


Subject(s)
Coagulase/chemistry , Coagulase/metabolism , Staphylococcus aureus/enzymology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Amino Acid Motifs , Amino Acid Sequence , Apoenzymes/chemistry , Apoenzymes/genetics , Apoenzymes/metabolism , Binding Sites , Coagulase/genetics , Crystallography, X-Ray , Fibrinogen/chemistry , Fibrinogen/metabolism , Humans , Keratins/chemistry , Keratins/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Structure, Tertiary , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...