Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Immunol ; 9: 2405, 2018.
Article in English | MEDLINE | ID: mdl-30405609

ABSTRACT

Streptococcus pneumoniae is endowed with a variety of surface-exposed proteins representing putative vaccine candidates. Lipoproteins are covalently anchored to the cell membrane and highly conserved among pneumococcal serotypes. Here, we evaluated these lipoproteins for their immunogenicity and protective potential against pneumococcal colonisation. A multiplex-based immunoproteomics approach revealed the immunogenicity of selected lipoproteins. High antibody titres were measured in sera from mice immunised with the lipoproteins MetQ, PnrA, PsaA, and DacB. An analysis of convalescent patient sera confirmed the immunogenicity of these lipoproteins. Examining the surface localisation and accessibility of the lipoproteins using flow cytometry indicated that PnrA and DacB were highly abundant on the surface of the bacteria. Mice were immunised intranasally with PnrA, DacB, and MetQ using cholera toxin subunit B (CTB) as an adjuvant, followed by an intranasal challenge with S. pneumoniae D39. PnrA protected the mice from pneumococcal colonisation. For the immunisation with DacB and MetQ, a trend in reducing the bacterial load could be observed, although this effect was not statistically significant. The reduction in bacterial colonisation was correlated with the increased production of antigen-specific IL-17A in the nasal cavity. Immunisation induced high systemic IgG levels with a predominance for the IgG1 isotype, except for DacB, where IgG levels were substantially lower compared to MetQ and PnrA. Our results indicate that lipoproteins are interesting targets for future vaccine strategies as they are highly conserved, abundant, and immunogenic.


Subject(s)
Pneumococcal Infections/immunology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibody Specificity/immunology , Female , Flow Cytometry , Humans , Immunogenicity, Vaccine , Immunoglobulin G/blood , Immunoglobulin G/immunology , Lipoproteins/immunology , Mutation , Pneumococcal Infections/microbiology , Pneumococcal Vaccines/administration & dosage , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/genetics , Vaccination
2.
Antioxid Redox Signal ; 28(6): 410-430, 2018 02 20.
Article in English | MEDLINE | ID: mdl-27967218

ABSTRACT

AIMS: Bacillithiol (BSH) is the major low-molecular-weight thiol of the human pathogen Staphylococcus aureus. In this study, we used OxICAT and Voronoi redox treemaps to quantify hypochlorite-sensitive protein thiols in S. aureus USA300 and analyzed the role of BSH in protein S-bacillithiolation. RESULTS: The OxICAT analyses enabled the quantification of 228 Cys residues in the redox proteome of S. aureus USA300. Hypochlorite stress resulted in >10% increased oxidation of 58 Cys residues (25.4%) in the thiol redox proteome. Among the highly oxidized sodium hypochlorite (NaOCl)-sensitive proteins are five S-bacillithiolated proteins (Gap, AldA, GuaB, RpmJ, and PpaC). The glyceraldehyde-3-phosphate (G3P) dehydrogenase Gap represents the most abundant S-bacillithiolated protein contributing 4% to the total Cys proteome. The active site Cys151 of Gap was very sensitive to overoxidation and irreversible inactivation by hydrogen peroxide (H2O2) or NaOCl in vitro. Treatment with H2O2 or NaOCl in the presence of BSH resulted in reversible Gap inactivation due to S-bacillithiolation, which could be regenerated by the bacilliredoxin Brx (SAUSA300_1321) in vitro. Molecular docking was used to model the S-bacillithiolated Gap active site, suggesting that formation of the BSH mixed disulfide does not require major structural changes. Conclusion and Innovation: Using OxICAT analyses, we identified 58 novel NaOCl-sensitive proteins in the pathogen S. aureus that could play protective roles against the host immune defense and include the glycolytic Gap as major target for S-bacillithiolation. S-bacillithiolation of Gap did not require structural changes, but efficiently functions in redox regulation and protection of the active site against irreversible overoxidation in S. aureus. Antioxid. Redox Signal. 28, 410-430.


Subject(s)
Bacterial Proteins/metabolism , Cysteine/analogs & derivatives , Glucosamine/analogs & derivatives , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Cysteine/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , Glucosamine/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/chemistry , Humans , Hydrogen Peroxide/metabolism , Hypochlorous Acid/toxicity , Protein Conformation/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Stress, Physiological/drug effects , Stress, Physiological/genetics
3.
Sci Rep ; 7(1): 5020, 2017 07 10.
Article in English | MEDLINE | ID: mdl-28694441

ABSTRACT

Mycothiol (MSH) is the major low molecular weight (LMW) thiol in Actinomycetes and functions in post-translational thiol-modification by protein S-mycothiolation as emerging thiol-protection and redox-regulatory mechanism. Here, we have used shotgun-proteomics to identify 26 S-mycothiolated proteins in the pathogen Corynebacterium diphtheriae DSM43989 under hypochlorite stress that are involved in energy metabolism, amino acid and nucleotide biosynthesis, antioxidant functions and translation. The glyceraldehyde-3-phosphate dehydrogenase (GapDH) represents the most abundant S-mycothiolated protein that was modified at its active site Cys153 in vivo. Exposure of purified GapDH to H2O2 and NaOCl resulted in irreversible inactivation due to overoxidation of the active site in vitro. Treatment of GapDH with H2O2 or NaOCl in the presence of MSH resulted in S-mycothiolation and reversible GapDH inactivation in vitro which was faster compared to the overoxidation pathway. Reactivation of S-mycothiolated GapDH could be catalyzed by both, the Trx and the Mrx1 pathways in vitro, but demycothiolation by Mrx1 was faster compared to Trx. In summary, we show here that S-mycothiolation can function in redox-regulation and protection of the GapDH active site against overoxidation in C. diphtheriae which can be reversed by both, the Mrx1 and Trx pathways.


Subject(s)
Corynebacterium diphtheriae/enzymology , Cysteine/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Glycopeptides/chemistry , Inositol/chemistry , Proteomics/methods , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Hydrogen Peroxide/pharmacology , Oxidation-Reduction , Oxidative Stress , Protein Processing, Post-Translational , Sodium Hypochlorite/pharmacology
4.
Sci Rep ; 6: 38094, 2016 12 05.
Article in English | MEDLINE | ID: mdl-27917891

ABSTRACT

The human pathogen Streptococcus pneumoniae is decorated with a special class of surface-proteins known as choline-binding proteins (CBPs) attached to phosphorylcholine (PCho) moieties from cell-wall teichoic acids. By a combination of X-ray crystallography, NMR, molecular dynamics techniques and in vivo virulence and phagocytosis studies, we provide structural information of choline-binding protein L (CbpL) and demonstrate its impact on pneumococcal pathogenesis and immune evasion. CbpL is a very elongated three-module protein composed of (i) an Excalibur Ca2+-binding domain -reported in this work for the very first time-, (ii) an unprecedented anchorage module showing alternate disposition of canonical and non-canonical choline-binding sites that allows vine-like binding of fully-PCho-substituted teichoic acids (with two choline moieties per unit), and (iii) a Ltp_Lipoprotein domain. Our structural and infection assays indicate an important role of the whole multimodular protein allowing both to locate CbpL at specific places on the cell wall and to interact with host components in order to facilitate pneumococcal lung infection and transmigration from nasopharynx to the lungs and blood. CbpL implication in both resistance against killing by phagocytes and pneumococcal pathogenesis further postulate this surface-protein as relevant among the pathogenic arsenal of the pneumococcus.


Subject(s)
Carrier Proteins/metabolism , Choline/metabolism , Pneumococcal Infections/metabolism , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Teichoic Acids/metabolism , Animals , Binding Sites/physiology , Calcium/metabolism , Cell Wall/metabolism , Cell Wall/microbiology , Crystallography, X-Ray/methods , Female , Immune Evasion/physiology , Mice , Models, Molecular , Nasopharynx/metabolism , Nasopharynx/microbiology , Phagocytes/metabolism , Phagocytes/microbiology , Phosphorylcholine/metabolism , Pneumococcal Infections/microbiology , Respiratory Tract Infections/metabolism , Respiratory Tract Infections/microbiology , Virulence/physiology
5.
J Infect Dis ; 214(6): 953-61, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27368348

ABSTRACT

BACKGROUND: Pneumococcal proteins involved in the resistance against oxidative stress are present in all strains and therefore are potential antigens that could be suitable for new therapies and/or vaccines. Their role in the pathogenesis of pneumococcal meningitis has not been addressed. METHODS: We investigated the individual contributions of extracellular thioredoxin lipoproteins (Etrx1 and Etrx2) and the intracellular and extracellular methionine sulfoxide reductases (SpMsrAB1 and SpMsrAB2, respectively) in the progression and outcome of pneumococcal meningitis, using Kaplan-Meier survival curves, bacteriological and histological studies, and measurements of proinflammatory mediators. RESULTS: The absence of Etrx1, Etrx2, or SpMsrAB1 moderately attenuated virulence as compared to the wild-type strain but did not significantly affect bacterial growth in the brain and bloodstream. Loss of function of SpMsrAB2 alone, both Etrx proteins, or both SpMsrAB proteins resulted in a less severe course of infection, with low numbers of animals dying of infection, a lower risk of associated meningeal inflammation, and reduced bacterial densities in the cerebellum, blood, and spleen. CONCLUSIONS: Our data support the importance of the extracellular redox repair system in virulence and its potential as a target for the design of new antimicrobials and vaccine formulations against Streptococcus pneumoniae.


Subject(s)
Meningitis, Pneumococcal/pathology , Methionine Sulfoxide Reductases/metabolism , Streptococcus pneumoniae/pathogenicity , Thioredoxins/metabolism , Virulence Factors/metabolism , Animals , Blood/microbiology , Brain/microbiology , Disease Models, Animal , Female , Gene Deletion , Meningitis, Pneumococcal/immunology , Methionine Sulfoxide Reductases/genetics , Mice, Inbred C57BL , Oxidation-Reduction , Oxidative Stress , Spleen , Streptococcus pneumoniae/genetics , Survival Analysis , Thioredoxins/genetics , Virulence , Virulence Factors/genetics
6.
Int J Med Microbiol ; 304(8): 1233-46, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25455218

ABSTRACT

Colonization of Streptococcus pneumoniae (pneumococci) is a prerequisite for bacterial dissemination and their capability to enter the bloodstream. Pneumococci have evolved various successful strategies to colonize the mucosal epithelial barrier of humans. A pivotal mechanism of host cell invasion implicated with invasive diseases is promoted by the interaction of pneumococcal PspC with the polymeric Ig-receptor (pIgR). However, the mechanism(s) of pneumococcal endocytosis and the intracellular route of pneumococci upon uptake by the PspC-pIgR-interaction are not known. Here, we demonstrate by using a combination of pharmacological inhibitors and genetics interference approaches the involvement of active dynamin-dependent caveolae and clathrin-coated vesicles for pneumococcal uptake via the PspC-pIgR mechanism. Depleting cholesterol from host cell membranes and disruption of lipid microdomains impaired pneumococcal internalization. Moreover, chemical inhibition of clathrin or functional inactivation of dynamin, caveolae or clathrin by RNA interference significantly affected pneumococcal internalization suggesting that clathrin-mediated endocytosis (CME) and caveolae are involved in the bacterial uptake process. Confocal fluorescence microscopy of pIgR-expressing epithelial cells infected with pneumococci or heterologous Lactococcus lactis expressing PspC demonstrated bacterial co-localization with fluorescent-tagged clathrin and early as well as recycling or late endosomal markers such as Lamp1, Rab5, Rab4, and Rab7, respectively. In conclusion these data suggest that PspC-promoted uptake is mediated by both CME and caveolae. After endocytosis pneumococci are routed via the endocytic pathway into early endosomes and are then sorted into recycling or late endosomes, which can result in pneumococcal killing in phagolysosomes or transcytosis via recycling endosomes.


Subject(s)
Bacterial Adhesion , Caveolins/metabolism , Clathrin/metabolism , Endocytosis , Epithelial Cells/physiology , Receptors, Polymeric Immunoglobulin/metabolism , Streptococcus pneumoniae/physiology , Animals , Bacterial Proteins/metabolism , Cell Line , Dogs , Epithelial Cells/microbiology , Humans , Protein Binding
7.
Mol Microbiol ; 93(6): 1183-206, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25060741

ABSTRACT

Bacterial cell wall hydrolases are essential for peptidoglycan turnover and crucial to preserve cell shape. The d,d-carboxypeptidase DacA and l,d-carboxypeptidase DacB of Streptococcus pneumoniae function in a sequential manner. Here, we determined the structure of the surface-exposed lipoprotein DacB. The crystal structure of DacB, radically different to that of DacA, contains a mononuclear Zn(2+) catalytic centre located in the middle of a large and fully exposed groove. Two different conformations were found presenting a different arrangement of the active site topology. The critical residues for catalysis and substrate specificity were identified. Loss-of-function of DacA and DacB altered the cell shape and this was consistent with a modified peptidoglycan peptide composition in dac mutants. Contrary, an lgt mutant lacking lipoprotein diacylglyceryl transferase activity required for proper lipoprotein maturation retained l,d-carboxypeptidase activity and showed an intact murein sacculus. In addition we demonstrated pathophysiological effects of disabled DacA or DacB activities. Real-time bioimaging of intranasal infected mice indicated a substantial attenuation of ΔdacB and ΔdacAΔdacB pneumococci, while ΔdacA had no significant effect. In addition, uptake of these mutants by professional phagocytes was enhanced, while the adherence to lung epithelial cells was decreased. Thus, structural and functional studies suggest DacA and DacB as optimal drug targets.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Carboxypeptidases/chemistry , Carboxypeptidases/genetics , Pneumococcal Infections/veterinary , Streptococcus pneumoniae/enzymology , Animals , Bacterial Proteins/metabolism , Carboxypeptidases/metabolism , Catalytic Domain , Cell Wall/physiology , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Enzymologic , Mice , Models, Molecular , Phenotype , Pneumococcal Infections/metabolism , Protein Structure, Secondary , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity
8.
J Vis Exp ; (84): e51174, 2014 Feb 23.
Article in English | MEDLINE | ID: mdl-24637643

ABSTRACT

Pneumonia is one of the major health care problems in developing and industrialized countries and is associated with considerable morbidity and mortality. Despite advances in knowledge of this illness, the availability of intensive care units (ICU), and the use of potent antimicrobial agents and effective vaccines, the mortality rates remain high(1). Streptococcus pneumoniae is the leading pathogen of community-acquired pneumonia (CAP) and one of the most common causes of bacteremia in humans. This pathogen is equipped with an armamentarium of surface-exposed adhesins and virulence factors contributing to pneumonia and invasive pneumococcal disease (IPD). The assessment of the in vivo role of bacterial fitness or virulence factors is of utmost importance to unravel S. pneumoniae pathogenicity mechanisms. Murine models of pneumonia, bacteremia, and meningitis are being used to determine the impact of pneumococcal factors at different stages of the infection. Here we describe a protocol to monitor in real-time pneumococcal dissemination in mice after intranasal or intraperitoneal infections with bioluminescent bacteria. The results show the multiplication and dissemination of pneumococci in the lower respiratory tract and blood, which can be visualized and evaluated using an imaging system and the accompanying analysis software.


Subject(s)
Luminescent Measurements/methods , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/pathogenicity , Virulence Factors/analysis , Animals , Bacteremia/blood , Bacteremia/microbiology , Disease Models, Animal , Female , Lung/microbiology , Mice , Pneumonia, Pneumococcal/blood , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/metabolism , Virulence Factors/metabolism
9.
J Proteome Res ; 13(2): 650-67, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24387739

ABSTRACT

Surface proteins are important for the fitness and virulence of the Gram-positive pathogen Streptococcus pneumoniae. They are crucial for interaction of the pathogen with its human host during infection. Therefore, the analysis of the pneumococcal surface proteome is an important task that requires powerful tools. In this study, two different methods, an optimized biotinylation approach and shaving with trypsin beads, were applied to study the pneumococcal surface proteome and to identify surface-exposed protein domains, respectively. The identification of nearly 95% of the predicted lipoproteins and 75% of the predicted sortase substrates reflects the high coverage of the two classical surface protein classes accomplished in this study. Furthermore, the biotinylation approach was applied to study the impact of an impaired lipoprotein maturation pathway on the cell envelope proteome and exoproteome. Loss of the lipoprotein diacylglyceryl transferase Lgt leads to striking changes in the lipoprotein distribution. Many lipoproteins disappear from the surface proteome and accumulate in the exoproteome. Further insights into lipoprotein processing in pneumococci are provided by immunoblot analyses of bacterial lysates and corresponding supernatant fractions. Taken together, the first comprehensive overview of the pneumococcal surface and exoproteome is presented, and a model for lipoprotein processing in S. pneumoniae is proposed.


Subject(s)
Bacterial Proteins/biosynthesis , Lipoproteins/biosynthesis , Proteome , Streptococcus pneumoniae/metabolism , Bacterial Proteins/metabolism , Base Sequence , Biotin/metabolism , DNA Primers , Electrophoresis, Polyacrylamide Gel , Lipoproteins/metabolism , Polymerase Chain Reaction , Subcellular Fractions/metabolism , Trypsin/metabolism
10.
EMBO Mol Med ; 5(12): 1852-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24136784

ABSTRACT

The respiratory pathogen Streptococcus pneumoniae has evolved efficient mechanisms to resist oxidative stress conditions and to displace other bacteria in the nasopharynx. Here we characterize at physiological, functional and structural levels two novel surface-exposed thioredoxin-family lipoproteins, Etrx1 and Etrx2. The impact of both Etrx proteins and their redox partner methionine sulfoxide reductase SpMsrAB2 on pneumococcal pathogenesis was assessed in mouse virulence studies and phagocytosis assays. The results demonstrate that loss of function of either both Etrx proteins or SpMsrAB2 dramatically attenuated pneumococcal virulence in the acute mouse pneumonia model and that Etrx proteins compensate each other. The deficiency of Etrx proteins or SpMsrAB2 further enhanced bacterial uptake by macrophages, and accelerated pneumococcal killing by H2 O2 or free methionine sulfoxides (MetSO). Moreover, the absence of both Etrx redox pathways provokes an accumulation of oxidized SpMsrAB2 in vivo. Taken together our results reveal insights into the role of two extracellular electron pathways required for reduction of SpMsrAB2 and surface-exposed MetSO. Identification of this system and its target proteins paves the way for the design of novel antimicrobials.


Subject(s)
Bacterial Proteins/metabolism , Streptococcus pneumoniae/metabolism , Animals , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Catalytic Domain , Crystallography, X-Ray , Disease Models, Animal , Female , Hydrogen Peroxide/pharmacology , Macrophages/immunology , Macrophages/physiology , Methionine/analogs & derivatives , Methionine/pharmacology , Mice , Molecular Sequence Data , Oxidative Stress/drug effects , Phagocytosis , Pneumonia/immunology , Pneumonia/microbiology , Pneumonia/mortality , Protein Structure, Tertiary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Streptococcus pneumoniae/pathogenicity , Survival Analysis , Virulence
11.
J Biol Chem ; 288(22): 15614-27, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23603906

ABSTRACT

Adherence of Streptococcus pneumoniae is directly mediated by interactions of adhesins with eukaryotic cellular receptors or indirectly by exploiting matrix and serum proteins as molecular bridges. Pneumococci engage vitronectin, the human adhesive glycoprotein and complement inhibitor, to facilitate attachment to epithelial cells of the mucosal cavity, thereby modulating host cell signaling. In this study, we identified PspC as a vitronectin-binding protein interacting with the C-terminal heparin-binding domain of vitronectin. PspC is a multifunctional surface-exposed choline-binding protein displaying various adhesive properties. Vitronectin binding required the R domains in the mature PspC protein, which are also essential for the interaction with the ectodomain of the polymeric immunoglobulin receptor and secretory IgA. Consequently, secretory IgA competitively inhibited binding of vitronectin to purified PspC and to PspC-expressing pneumococci. In contrast, Factor H, which binds to the N-terminal part of mature PspC molecules, did not interfere with the PspC-vitronectin interaction. Using a series of vitronectin peptides, the C-terminal heparin-binding domain was shown to be essential for the interaction of soluble vitronectin with PspC. Binding experiments with immobilized vitronectin suggested a region N-terminal to the identified heparin-binding domain as an additional binding region for PspC, suggesting that soluble, immobilized, as well as cellularly bound vitronectin possesses different conformations. Finally, vitronectin bound to PspC was functionally active and inhibited the deposition of the terminal complement complex. In conclusion, this study identifies and characterizes (on the molecular level) the interaction between the pneumococcal adhesin PspC and the human glycoprotein vitronectin.


Subject(s)
Bacterial Proteins/metabolism , Streptococcus pneumoniae/metabolism , Vitronectin/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Protein Binding , Protein Structure, Tertiary , Streptococcus pneumoniae/chemistry , Streptococcus pneumoniae/genetics , Vitronectin/chemistry , Vitronectin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...