Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Curr Microbiol ; 69(6): 894-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25108378

ABSTRACT

Group B protective surface protein (BPS) is expressed on the cell surface of some group B streptococcal (GBS) (Streptococcus agalactiae) strains and adds to the identification by capsular polysaccharide (CPS), and c or R proteins. We investigated the prevalence of BPS among GBS clinical isolates (303 invasive, 4122 colonizing) collected over 11 years in four American cities. Hot HCl cell extracts were tested by immunoprecipitation in agarose with rabbit antisera to BPS; the alpha (α) and beta (ß) components of c protein; R1, R3, and R4 species of R protein; and CPS serotypes Ia-VIII. BPS was found in 155 isolates (seven invasive, 148 colonizing). Of these, 87 were Ia, 37 II, 20 V; none were III. BPS was expressed usually with another protein: a species of R by 87 or a component of c by 39. The predominant CPS/protein profiles with BPS were Ia/R1,BPS and II/c(α + ß),BPS. Thus, along with CPS serotype and other surface proteins, BPS can be a valuable marker for precise strain characterization of unique GBS clinical isolates with complex surface protein profiles.


Subject(s)
Antigens, Bacterial/analysis , Antigens, Surface/analysis , Streptococcal Infections/microbiology , Streptococcus agalactiae/chemistry , Streptococcus agalactiae/isolation & purification , Americas , Antigens, Bacterial/classification , Antigens, Surface/classification , Carrier State/microbiology , Cities , Humans , Immunoprecipitation , Meningitis, Bacterial/microbiology , Sepsis/microbiology , Streptococcus agalactiae/classification
2.
Thromb Haemost ; 111(3): 401-16, 2014 Mar 03.
Article in English | MEDLINE | ID: mdl-24196407

ABSTRACT

Streptococcus pneumoniae is not only a commensal of the nasopharyngeal epithelium, but may also cause life-threatening diseases. Immune-electron microscopy studies revealed that the bacterial glycolytic enzyme, phosphoglycerate kinase (PGK), is localised on the pneumococcal surface of both capsulated and non-capsulated strains and colocalises with plasminogen. Since pneumococci may concentrate host plasminogen (PLG) together with its activators on the bacterial cell surface to facilitate the formation of plasmin, the involvement of PGK in this process was studied. Specific binding of human or murine PLG to strain-independent PGK was documented, and surface plasmon resonance analyses indicated a high affinity interaction with the kringle domains 1-4 of PLG. Crystal structure determination of pneumococcal PGK together with peptide array analysis revealed localisation of PLG-binding site in the N-terminal region and provided structural motifs for the interaction with PLG. Based on structural analysis data, a potential interaction of PGK with tissue plasminogen activator (tPA) was proposed and experimentally confirmed by binding studies, plasmin activity assays and thrombus degradation analyses.


Subject(s)
Bacterial Proteins/metabolism , Phosphoglycerate Kinase/metabolism , Plasminogen/metabolism , Streptococcus pneumoniae/physiology , Tissue Plasminogen Activator/metabolism , Animals , Bacterial Proteins/genetics , Cloning, Molecular , Crystallography, X-Ray , Fibrinolysin/metabolism , Humans , Laryngeal Mucosa/microbiology , Mice , Nasal Mucosa/microbiology , Phosphoglycerate Kinase/genetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs/genetics , Protein Transport , Surface Plasmon Resonance
3.
Vet Microbiol ; 162(1): 270-7, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-22939986

ABSTRACT

Although Streptococcus (S.) canis is known to cause severe infections in dogs and cats and harbors a clear zoonotic potential, knowledge about physiology and pathogenesis is mostly elusive. The arginine deiminase system (ADS) has been described in certain streptococcal species and its role in the establishment of infection has been suggested. In this study we focused on the identification and characterization of the ADS in S. canis. Using genome sequencing and subsequent in silico analysis we identified the ADS of S. canis as a gene cluster composed of seven genes. RT-PCR analysis revealed that the ADS of S. canis is transcribed in four transcriptional units, comprising three monocistronical mRNAs and one operon structure. As a secondary metabolic pathway, the ADS of S. canis is strictly regulated by carbon catabolite repression (CCR) and arginine as demonstrated on transcriptional, translational, and enzymatical level, respectively. Furthermore, growth kinetics with a chemically defined medium clearly showed that arginine, the substrate of the ADS, is essential for the biological fitness of S. canis. Using Immuno-electron microscopy analysis, we observed a surface-exposed localization of the ADS enzymes arginine deiminase (ArcA), ornithine carbamoyltransferase (ArcB), and carbamate kinase (ArcC), respectively, which might suggest the contribution of the ADS to the development of streptococcal infections.


Subject(s)
Hydrolases/genetics , Hydrolases/metabolism , Streptococcus/enzymology , Arginine/metabolism , Base Sequence , Hydrolases/chemistry , Multigene Family , Operon , Ornithine Carbamoyltransferase/metabolism , Phosphotransferases (Carboxyl Group Acceptor)/metabolism , Streptococcal Infections/enzymology , Streptococcal Infections/microbiology , Streptococcus/genetics , Streptococcus/metabolism
4.
J Mol Med (Berl) ; 90(10): 1197-207, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22527883

ABSTRACT

Streptococcus pyogenes (group A Streptococcus (GAS)) causes ∼700 million human infections each year, resulting in over 500,000 deaths. The development of a commercial GAS vaccine is hampered by the occurrence of many unique GAS serotypes, antigenic variation within the same serotype, differences in serotype geographical distribution, and the production of antibodies cross-reactive with human tissue that may lead to autoimmune disease. Several independent studies have documented a number of GAS cell wall-associated or secreted metabolic enzymes that contain neither N-terminal leader sequences nor C-terminal cell wall anchors. Here, we applied a proteomic analysis of serotype M1T1 GAS cell wall extracts for the purpose of vaccine development. This approach catalogued several anchorless proteins and identified two protective vaccine candidates, arginine deiminase and trigger factor. These surface-exposed enzymes are expressed across multiple GAS serotypes exhibiting ≥99% amino acid sequence identity. Vaccine safety concerns are alleviated by the observation that these vaccine candidates lack human homologs, while sera from human populations suffering repeated GAS infections and high levels of autoimmune complications do not recognize these enzymes. Our study demonstrates anchorless cell surface antigens as promising vaccine candidates for the prevention of GAS disease.


Subject(s)
Bacterial Proteins/metabolism , Cell Wall/metabolism , Hydrolases/metabolism , Peptidylprolyl Isomerase/metabolism , Streptococcal Infections/prevention & control , Streptococcal Vaccines/metabolism , Streptococcus pyogenes/immunology , Adolescent , Animals , Bacterial Proteins/immunology , Cell Wall/immunology , Child , Female , Humans , Hydrolases/immunology , Immune Sera/immunology , Immunity, Active , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microscopy, Immunoelectron , Peptidylprolyl Isomerase/immunology , Proteome/immunology , Proteome/metabolism , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Streptococcal Infections/immunology , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/immunology , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/ultrastructure , Vaccination , Young Adult
5.
FASEB J ; 22(8): 2715-22, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18467595

ABSTRACT

The human protease plasmin plays a crucial role in the capacity of the group A streptococcus (GAS; Streptococcus pyogenes) to initiate invasive disease. The GAS strain NS88.2 was isolated from a case of bacteremia from the Northern Territory of Australia, a region with high rates of GAS invasive disease. Mutagenesis of the NS88.2 plasminogen binding M protein Prp was undertaken to examine the contribution of plasminogen binding and cell surface plasmin acquisition to virulence. The isogenic mutant NS88.2prp was engineered whereby four amino acid residues critical for plasminogen binding were converted to alanine codons in the GAS genome sequence. The mutated residues were reverse complemented to the wild-type sequence to construct GAS strain NS88.2prpRC. In comparison to NS88.2 and NS88.2prpRC, the NS88.2prp mutant exhibited significantly reduced ability to bind human plasminogen and accumulate cell surface plasmin activity during growth in human plasma. Utilizing a humanized plasminogen mouse model of invasive infection, we demonstrate that the capacity to bind plasminogen and accumulate surface plasmin activity plays an essential role in GAS virulence.


Subject(s)
Antigens, Bacterial/physiology , Bacterial Outer Membrane Proteins/physiology , Carrier Proteins/physiology , Plasminogen/metabolism , Streptococcus pyogenes/physiology , Streptococcus pyogenes/pathogenicity , Animals , Antigens, Bacterial/genetics , Bacterial Outer Membrane Proteins/genetics , Base Sequence , Carrier Proteins/genetics , DNA Primers/genetics , DNA, Bacterial/genetics , Disease Models, Animal , Fibrinogen/metabolism , Genes, Bacterial , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Phagocytosis , Plasminogen/genetics , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Streptococcal Infections/etiology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/isolation & purification , Virulence/genetics , Virulence/physiology
6.
J Biol Chem ; 283(10): 6359-66, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18180300

ABSTRACT

Serum opacity factor (SOF) is a unique multifunctional virulence determinant expressed at the surface of Streptococcus pyogenes and has been shown to elicit protective immunity against GAS infection in a murine challenge model. SOF consists of two distinct domains with different binding capacities: an N-terminal domain that binds apolipoprotein AI and a C-terminal repeat domain that binds fibronectin and fibrinogen. The capacity of SOF to opacify serum by disrupting the structure of high density lipoproteins may preclude its use as a vaccine antigen in humans. This study generated mutant forms of recombinant SOF with reduced (100-fold) or abrogated opacity factor (OF) activity, for use as vaccine antigens. However, alterations introduced into the N-terminal SOF peptide (SOFDeltaFn) by mutagenesis to abrogate OF activity, abolish the capacity of SOF to protect against lethal systemic S. pyogenes challenge in a murine model. Mutant forms of purified SOFDeltaFn peptide were also used to assess the contribution of OF activity to the pathogenic processes of cell adhesion and cell invasion. Using latex beads coated with full-length SOF, SOFDeltaFn peptide, or a peptide encompassing the C-terminal repeats (FnBD), we demonstrate that adhesion to HEp-2 cells is mediated by both SOFDeltaFn and FnBD. The HEp-2 cell binding displayed by the N-terminal SOFDeltaFn peptide is independent of OF activity. We demonstrate that while the N terminus of SOF does not directly mediate intracellular uptake by epithelial cells, this domain enhances epithelial cell uptake mediated by full-length SOF, in comparison to the FnBD alone.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Epithelial Cells/immunology , Peptide Hydrolases/immunology , Streptococcal Infections/immunology , Streptococcal Vaccines/immunology , Streptococcus pyogenes/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/pharmacology , Apolipoprotein A-I/genetics , Apolipoprotein A-I/immunology , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/genetics , Cell Line , Disease Models, Animal , Epithelial Cells/microbiology , Fibrinogen/genetics , Fibrinogen/immunology , Fibronectins/genetics , Fibronectins/immunology , Immunization , Mice , Mice, Inbred BALB C , Mutation , Peptide Hydrolases/genetics , Peptides/genetics , Peptides/immunology , Peptides/pharmacology , Protein Binding/genetics , Protein Binding/immunology , Protein Structure, Tertiary/genetics , Streptococcal Infections/genetics , Streptococcal Infections/prevention & control , Streptococcal Vaccines/genetics , Streptococcal Vaccines/pharmacology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity
7.
J Clin Microbiol ; 45(8): 2392-7, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17553974

ABSTRACT

Viridans streptococci (VS) are responsible for several systemic diseases, such as endocarditis, abscesses, and septicemia. Unfortunately, species identification by conventional methods seems to be more difficult than species identification of other groups of bacteria. The aim of the present study was to evaluate the use of cell matrix-assisted laser desorption ionization-time of flight mass spectrometry (MALDI-TOF-MS) for the rapid identification of 10 different species of VS. A total of 99 VS clinical isolates, 10 reference strains, and 20 strains from our in-house culture collection were analyzed by MALDI-TOF-MS. To evaluate the mass-spectrometric discrimination results, all strains were identified in parallel by phenotypic and genotypic methods. MALDI-TOF-MS identified 71 isolates as the mitis group, 23 as the anginosus group, and 5 as Streptococcus salivarius. Comparison of the species identification results obtained by the MALDI-TOF-MS analyses and with the phenotypic/genotypic identification systems showed 100% consistency at the species level. Thus, MALDI-TOF-MS seems to be a rapid and reliable method for the identification of species of VS from clinical samples.


Subject(s)
Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcal Infections/microbiology , Viridans Streptococci/chemistry , Viridans Streptococci/classification , Bacterial Typing Techniques , Humans , Polymerase Chain Reaction , Sensitivity and Specificity , Sequence Analysis, DNA
8.
Clin Infect Dis ; 43(7): 884-91, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16941370

ABSTRACT

BACKGROUND: The factors behind the reemergence of severe, invasive group A streptococcal (GAS) diseases are unclear, but it could be caused by altered genetic endowment in these organisms. However, data from previous studies assessing the association between single genetic factors and invasive disease are often conflicting, suggesting that other, as-yet unidentified factors are necessary for the development of this class of disease. METHODS: In this study, we used a targeted GAS virulence microarray containing 226 GAS genes to determine the virulence gene repertoires of 68 GAS isolates (42 associated with invasive disease and 28 associated with noninvasive disease) collected in a defined geographic location during a contiguous time period. We then employed 3 advanced machine learning methods (genetic algorithm neural network, support vector machines, and classification trees) to identify genes with an increased association with invasive disease. RESULTS: Virulence gene profiles of individual GAS isolates varied extensively among these geographically and temporally related strains. Using genetic algorithm neural network analysis, we identified 3 genes with a marginal overrepresentation in invasive disease isolates. Significantly, 2 of these genes, ssa and mf4, encoded superantigens but were only present in a restricted set of GAS M-types. The third gene, spa, was found in variable distributions in all M-types in the study. CONCLUSIONS: Our comprehensive analysis of GAS virulence profiles provides strong evidence for the incongruent relationships among any of the 226 genes represented on the array and the overall propensity of GAS to cause invasive disease, underscoring the pathogenic complexity of these diseases, as well as the importance of multiple bacteria and/or host factors.


Subject(s)
Streptococcal Infections/metabolism , Streptococcus/pathogenicity , Virulence Factors/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Streptococcal Infections/physiopathology , Streptococcus/genetics , Streptococcus/isolation & purification , Virulence , Virulence Factors/genetics
9.
FASEB J ; 20(10): 1745-7, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16790522

ABSTRACT

The globally disseminated Streptococcus pyogenes M1T1 clone causes a number of highly invasive human diseases. The transition from local to systemic infection occurs by an unknown mechanism; however invasive M1T1 clinical isolates are known to express significantly less cysteine protease SpeB than M1T1 isolates from local infections. Here, we show that in comparison to the M1T1 strain 5448, the isogenic mutant delta speB accumulated 75-fold more human plasmin activity on the bacterial surface following incubation in human plasma. Human plasminogen was an absolute requirement for M1T1 strain 5448 virulence following subcutaneous (s.c.) infection of humanized plasminogen transgenic mice. S. pyogenes M1T1 isolates from the blood of infected humanized plasminogen transgenic mice expressed reduced levels of SpeB in comparison with the parental 5448 used as inoculum. We propose that the human plasminogen system plays a critical role in group A streptococcal M1T1 systemic disease initiation. SpeB is required for S. pyogenes M1T1 survival at the site of local infection, however, SpeB also disrupts the interaction of S. pyogenes M1T1 with the human plasminogen activation system. Loss of SpeB activity in a subpopulation of S. pyogenes M1T1 at the site of infection results in accumulation of surface plasmin activity thus triggering systemic spread.


Subject(s)
Plasminogen/physiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/genetics , Exotoxins/genetics , Fibrinolysin/metabolism , Gene Expression Regulation, Bacterial , Humans , Mice , Mice, Transgenic , Mutation , Streptococcal Infections/etiology , Streptococcus pyogenes/chemistry , Virulence
10.
Infect Immun ; 72(12): 7342-5, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15557665

ABSTRACT

Fibronectin binding protein F1 (Sfb1) of Streptococcus pyogenes (group A streptococcus [GAS]) is a well-characterized adhesin that has been shown to induce protection in mice against a lethal intranasal GAS challenge after intranasal immunization with cholera toxin B subunit (CTB) as adjuvant. With a murine skin infection model, we have shown that Sfb1/CTB vaccination neither elicits opsonizing antibodies nor prevents systemic bacterial growth and dissemination to internal organs after a subcutaneous GAS challenge. These results indicate that an Sfb1-based vaccine should be complemented with additional protective antigens in order to be used in areas such as the tropical north of Australia, where the skin is the primary route of entry for invasive streptococcal diseases.


Subject(s)
Adhesins, Bacterial/immunology , Skin Diseases, Bacterial/prevention & control , Streptococcal Infections/prevention & control , Streptococcus pyogenes/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/immunology , Blood Bactericidal Activity , Disease Models, Animal , Mice , Mice, Inbred BALB C , Vaccination
11.
Indian J Med Res ; 119 Suppl: 115-20, 2004 May.
Article in English | MEDLINE | ID: mdl-15232175

ABSTRACT

BACKGROUND & OBJECTIVES: The fibronectin binding protein Sfb1 of Streptococcus pyogenes is a well characterised antigen which induces protection against lethal challenge with group A streptococcus (GAS) when adjuvanted with cholera toxin B-subunit (CTB). As an alternative to CTB adjuvanted intranasal immunisations we investigated the immune responses generated in mice using Sfb1 incorporated in to the skin and mucosal adjuvant SAMA4. METHODS: Mice (BALB/c) were vaccinated intradermally with 100 microl of either SAMA4 (adjuvant only group) or SAMA4/Sfb1 and were boosted 7 days later. Mice vaccinated with CTB based vaccines were immunised by intranasal inoculation with a mixture containing 30 microg Sfb1 and 10 microg CTB on days 1, 3, 5 and 15. At 14 days after the last booster immunisation the immune response was characterised and mice were challenged with 10(8) CFU of S. pyogenes. RESULTS: Mice vaccinated with SAMA4/Sfb1 elicited a Sfb1-specific IgG response in the sera that was significantly higher than that seen in control mice and mice immunised with the adjuvant only (P<0.05). No significant differences were seen for specific IgA antibodies in the sera in all groups examined. Compared with non-immunised and adjuvant only immunised controls, mice immunised with the Sfb1/SAMA4 vaccine exhibited a significant increase (P<0.05) in the number of Sfb1 reactive spleen cells in lymphoproliferation assays which were three fold higher than those seen for mice vaccinated with the Sfb1/CTB vaccine. Mice vaccinated with CTB/Sfb1 had the highest level of protection (80%) as where mice vaccinated with SAMA4 and SAMA4/Sfb1 displayed no protection (20% and 40%). INTERPRETATION & CONCLUSION: These data suggest that the SAMA4 adjuvant used in this study fails to elicit protective immunity in BALB/c mice when used to adjuvant the known protective antigen Sfb1.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Vaccines/immunology , ISCOMs , Streptococcus pyogenes/immunology , Animals , Antibody Formation , Enzyme-Linked Immunosorbent Assay , Liposomes , Lymphocytes/immunology , Mice , Mice, Inbred BALB C
12.
Indian J Med Res ; 119 Suppl: vii-viii, 2004 May.
Article in English | MEDLINE | ID: mdl-15232209
13.
Infect Immun ; 70(9): 5202-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12183571

ABSTRACT

Caenorhabditis elegans is currently introduced as a new, facile, and cheap model organism to study the pathogenesis of gram-negative bacteria such as Pseudomonas aeruginosa and Salmonella enterica serovar Typhimurium. The mechanisms of killing involve either diffusible exotoxins or infection-like processes. Recently, it was shown that also some gram-positive bacteria kill C. elegans, although the precise mechanisms of killing remained open. We examined C. elegans as a pathogenesis model for the gram-positive bacterium Streptococcus pyogenes, a major human pathogen capable of causing a wide spectrum of diseases. We demonstrate that S. pyogenes kills C. elegans, both on solid and in liquid medium. Unlike P. aeruginosa and S. enterica serovar Typhimurium, the killing by S. pyogenes is solely mediated by hydrogen peroxide. Killing required live streptococci; the killing capacity depends on the amount of hydrogen peroxide produced, and killing can be inhibited by catalase. Major exotoxins of S. pyogenes are not involved in the killing process as confirmed by using specific toxin inhibitors and knockout mutants. Moreover, no accumulation of S. pyogenes in C. elegans is observed, which excludes the involvement of infection-like processes. Preliminary results show that S. pneumoniae can also kill C. elegans by hydrogen peroxide production. Hydrogen peroxide-mediated killing might represent a common mechanism by which gram-positive, catalase-negative pathogens kill C. elegans.


Subject(s)
Bacterial Proteins , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/toxicity , Membrane Proteins , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Toxins/toxicity , Exotoxins/toxicity , Models, Animal , Virulence/physiology
14.
J Infect Dis ; 184(7): 846-52, 2001 Oct 01.
Article in English | MEDLINE | ID: mdl-11550125

ABSTRACT

The influence of genetic background on the ability to control infection with group A streptococci was investigated in different inbred strains of mice. Whereas BALB/c, C57BL/10, and DBA/2 mice were the most resistant strains, with lower bacteria loads and higher survival times, C3H/HeN and CBA/J mice exhibited substantially higher bacterial growth and 100% mortality. Differences in susceptibility were not dependent on the inoculum size. Resistance was influenced by sex, with males being much more susceptible than females. B cell- and T cell-deficient mice from the resistant background were as resistant to infection as were immunocompetent mice, which suggests that the effector mechanisms are independent of adaptive immunity. These results demonstrate for the first time the influence of genetic background and sex on susceptibility to infection with Streptococcus pyogenes in mice. The use of this mouse model of group A streptococcal infection will allow for a better definition of parameters involved in the outcome of the disease.


Subject(s)
Genetic Predisposition to Disease , Streptococcal Infections/genetics , Streptococcus pyogenes , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred DBA , Mice, SCID , Sex Factors , Species Specificity , Streptococcal Infections/immunology , Streptococcal Infections/microbiology
15.
Infect Immun ; 69(8): 4851-7, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11447160

ABSTRACT

Immunoglobulin binding proteins are one of several pathogenicity factors which have been associated with invasive disease caused by group A streptococci. The surface-bound M and M-like proteins of Streptococcus pyogenes are the most characterized of these immunoglobulin binding proteins, and in most cases they bind only a single antibody class. Here we report the identification of a novel non-M-type secreted protein, designated SibA (for secreted immunoglobulin binding protein from group A streptococcus), which binds all immunoglobulin G (IgG) subclasses, the Fc and Fab fragments, and also IgA and IgM. SibA has no significant sequence homology to any M-related proteins, is not found in the vir regulon, and contains none of the characteristic M-protein regions, such as the A or C repeats. Like M proteins, however, SibA does have relatively high levels of alanine, lysine, glutamic acid, leucine, and glycine. SibA and M proteins also share an alpha-helical N-terminal secondary structure which has been previously implicated in immunoglobulin binding in M proteins. Evidence presented here indicates that this is also the case for SibA. SibA also has regions of local similarity with other coiled-coil proteins such as Listeria monocytogenes P45 autolysin, human myosin heavy chain, macrogolgin, and Schistoma mansoni paramyosin, some of which are of potential significance since cross-reactive antibodies between myosin proteins and M proteins have been implicated in the development of the autoimmune sequelae of streptococcal disease.


Subject(s)
Bacterial Proteins/genetics , Carrier Proteins/genetics , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Amino Acid Sequence , Bacterial Proteins/immunology , Base Sequence , Carrier Proteins/immunology , DNA, Bacterial , Humans , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Immunoglobulin M/immunology , Immunoglobulins/metabolism , Molecular Sequence Data , Sequence Analysis, DNA , Streptococcal Infections/epidemiology , Streptococcus pyogenes/immunology
16.
Mol Microbiol ; 40(6): 1273-87, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11442827

ABSTRACT

Binding of human plasminogen to Streptococcus pneumoniae and its subsequent activation promotes penetration of bacteria through reconstituted basement membranes. In this study, we have characterized a novel pneumococcal surface protein with a molecular mass of 47 kDa, designated Eno, which specifically binds human plasmin(ogen), exhibits alpha-enolase activity and is necessary for viability. Using enzyme assays, we have confirmed the alpha-enolase activity of both pneumococcal surface-displayed Eno and purified recombinant Eno protein. Immunoelectron microscopy indicated the presence of Eno in the cytoplasm as well as on the surface of encapsulated and unencapsulated pneumococci. Plasminogen-binding activity was demonstrated with whole pneumococcal cells and purified Eno protein. Binding of activated plasminogen was also shown for Eno; however, the affinity for plasmin is significantly reduced compared with plasminogen. Results from competitive inhibition assays indicate that binding is mediated through the lysine binding sites in plasmin(ogen). Carboxypeptidase B treatment and amino acid substitutions of the C-terminal lysyl residues of Eno indicated that the C-terminal lysine is pivotal for plasmin(ogen)-binding activity. Eno is ubiquitously distributed among pneumococcal serotypes, and binding experiments suggested the reassociation of secreted Eno to the bacterial cell surface. The reassociation was also confirmed by immunoelectron microscopy. The results suggest a mechanism of plasminogen activation for human pathogens that might contribute to their virulence potential in invasive infectious processes.


Subject(s)
Cell Membrane/metabolism , Fibrinolysin/metabolism , Phosphopyruvate Hydratase/metabolism , Plasminogen/metabolism , Streptococcus pneumoniae/enzymology , Amino Acid Sequence , Bacterial Outer Membrane Proteins/metabolism , Carboxypeptidase B , Carboxypeptidases/chemistry , Carboxypeptidases/metabolism , Cell Survival , Cross Reactions , Cytoplasm/metabolism , Gene Expression Regulation, Bacterial , Humans , Lysine/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/immunology , Sequence Analysis, Protein , Serotyping , Streptococcus pneumoniae/classification , Streptococcus pneumoniae/metabolism , Streptococcus pyogenes/enzymology
17.
Mol Microbiol ; 40(1): 99-114, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11298279

ABSTRACT

Group A streptococci (GAS) specifically attach to and internalize into human epithelial host cells. In some GAS isolates, fibronectin-binding proteins were identified as being responsible for these virulence traits. In the present study, the previously identified global negative regulator Nra was shown to control the binding of soluble fibronectin probably via regulation of protein F2 and/or SfbII expression in the serotype M49 strain 591. According to results from a conventional invasion assay based on the recovery of viable intracellular bacteria, the increased fibronectin binding did not affect bacterial adherence to HEp-2 epithelial cells, but was associated with a reduction in the internalization rates. However, when examined by confocal and electron microscopy techniques, the nra-mutant bacteria were shown to exhibit higher adherence and internalization rates than the corresponding wild type. The mutant bacteria escaped from the phagocytic vacuoles much faster, promoting consistent morphological changes which resulted in severe host cell damage. The apoptotic and lytic processes observed in nra-mutant infected host cells were correlated with an increased expression of the genes encoding superantigen SpeA, the cysteine protease SpeB, and streptolysin S in the nra-mutant bacteria. Adherence and internalization rates of a nra/speB-double mutant at wild-type levels indicated that the altered speB expression in the nra mutant contributed to the observed changes in both processes. The Nra-dependent effects on bacterial virulence were confined to infections carried out with stationary growth phase bacteria. In conclusion, the obtained results demonstrated that the global GAS regulator Nra modulates virulence genes, which are involved in host cell damage. Thus, by helping to achieve a critical balance of virulence factor expression that avoids the injury of target cells, Nra may facilitate GAS persistence in a safe intracellular niche.


Subject(s)
Bacterial Adhesion , Bacterial Proteins/physiology , Streptococcus pyogenes/physiology , Transcription Factors , Cell Line , Epithelial Cells/microbiology , Fluorescent Antibody Technique , Humans , Microscopy, Confocal , Microscopy, Electron , Streptococcus pyogenes/pathogenicity , Streptococcus pyogenes/ultrastructure , Virulence/genetics
18.
J Bacteriol ; 183(4): 1175-83, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11157929

ABSTRACT

Group B streptococcus (GBS) is the leading cause of bacterial sepsis and meningitis in neonates. N-terminal sequencing of major proteins in the culture supernatant of a clinical isolate of GBS identified a protein of about 50 kDa which could be detected in all of 27 clinical isolates tested. The corresponding gene, designated pcsB, was isolated from a GBS cosmid library and subsequently sequenced. The deduced PcsB polypeptide consists of 447 amino acid residues (M(r), 46,754), carries a potential N-terminal signal peptide sequence of 25 amino acids, and shows significant similarity to open reading frames of unknown function from different organisms and to the murein hydrolase P45 from Listeria monocytogenes. Northern blot analysis revealed a monocistronic transcriptional organization for pcsB in GBS. Insertional inactivation of pcsB in the genome of GBS resulted in mutant strain Sep1 exhibiting a drastically reduced growth rate compared to the parental GBS strain and showing an increased susceptibility to osmotic pressure and to various antibiotics. Electron microscopic analysis of GBS mutant Sep1 revealed growth in clumps, cell separation in several planes, and multiple division septa within single cells. These data suggest a pivotal role of PcsB for cell division and antibiotic tolerance of GBS.


Subject(s)
Bacterial Proteins/genetics , Cell Cycle Proteins , Cell Wall , Streptococcus agalactiae/genetics , Streptococcus agalactiae/ultrastructure , Anti-Bacterial Agents/pharmacology , Cell Division/genetics , Genes , Genes, Bacterial , Gram-Positive Bacteria/genetics , Microbial Sensitivity Tests , Molecular Sequence Data , Mutagenesis , N-Acetylmuramoyl-L-alanine Amidase/analysis , Streptococcus agalactiae/drug effects , Transcription, Genetic
19.
J Biol Chem ; 276(12): 9537-42, 2001 Mar 23.
Article in English | MEDLINE | ID: mdl-11124969

ABSTRACT

Clostridium botulinum C3 is the prototype of the family of the C3-like transferases that ADP-ribosylate exclusively RhoA, -B and -C. The ADP-ribose at Asn-41 results in functional inactivation of Rho reflected by disaggregation of the actin cytoskeleton. We report on a new C3-like transferase produced by a pathogenic Staphylococcus aureus strain. The transferase designated C3(Stau) was cloned from the genomic DNA. At the amino acid level, C3(Stau) revealed an identity of 35% to C3 from C. botulinum and Clostridium limosum exoenzyme, respectively, and of 78% to EDIN from S. aureus. In addition to RhoA, which is the target of the other C3-like transferases, C3(Stau) modified RhoE and Rnd3. RhoE was ADP-ribosylated at Asn-44, which is equivalent to Asn-41 of RhoA. RhoE and Rnd3 are members of the Rho subfamily, which are deficient in intrinsic GTPase activity and possess a RhoA antagonistic cell function. The protein substrate specificity found with recombinant Rho proteins was corroborated by expression of RhoE in Xenopus laevis oocytes showing that RhoE was also modified in vivo by C3(Stau) but not by C3 from C. botulinum. The poor cell accessibility of C3(Stau) was overcome by generation of a chimeric toxin recruiting the cell entry machinery of C. botulinum C2 toxin. The chimeric C3(Stau) caused the same morphological and cytoskeletal changes as the chimeric C. botulinum C3. C3(Stau) is a new member of the family of the C3-like transferases but is also the prototype of a subfamily of RhoE/Rnd modifying transferases.


Subject(s)
ADP Ribose Transferases/metabolism , Botulinum Toxins , GTPase-Activating Proteins/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Staphylococcus aureus/enzymology , Amino Acid Sequence , Base Sequence , DNA, Bacterial , Molecular Sequence Data , Sequence Homology, Amino Acid
20.
Infect Immun ; 69(1): 622-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11119566

ABSTRACT

Fibronectin-binding protein I (SfbI) represents a major adhesin of Streptococcus pyogenes. Mice were intranasally immunized with recombinant proteins spanning different portions of SfbI to identify the minimal fragment able to elicit a protective response against a lethal challenge with S. pyogenes. The strongest cellular responses and the highest levels of antigen-specific secretory immunoglobulin A (IgA) were detected in mice immunized with the fibronectin-binding region of SfbI. In contrast, animals vaccinated with a polypeptide spanning the aromatic and proline-rich regions showed the highest titers and fastest IgG response in serum. Vaccination with either SfbI without a membrane anchor and signal peptide or a polypeptide encompassing its fibronectin-binding regions resulted in efficient protection against heterologous challenge (60% and 80%, respectively), whereas the use of a polypeptide lacking this region conferred marginal protection (10%) with respect to the control group (0%). These results demonstrate that the fibronectin-binding region of SfbI is a promising candidate antigen for developing anti-S. pyogenes vaccines.


Subject(s)
Adhesins, Bacterial , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Carrier Proteins/immunology , Streptococcus pyogenes/immunology , Administration, Intranasal , Animals , Antibodies, Bacterial/blood , Binding Sites , Fishes , Immunization , Immunoglobulin A, Secretory/biosynthesis , Immunoglobulin G/biosynthesis , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...