Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Microbiology (Reading) ; 169(11)2023 11.
Article in English | MEDLINE | ID: mdl-37990974

ABSTRACT

Antibiotic persistence is a phenomenon observed when genetically susceptible cells survive long-term exposure to antibiotics. These 'persisters' are an intrinsic component of bacterial populations and stem from phenotypic heterogeneity. Persistence to antibiotics is a concern for public health globally, as it increases treatment duration and can contribute to treatment failure. Furthermore, there is a growing array of evidence that persistence is a 'stepping-stone' for the development of genetic antimicrobial resistance. Urinary tract infections (UTIs) are a major contributor to antibiotic consumption worldwide, and are known to be both persistent (i.e. affecting the host for a prolonged period) and recurring. Currently, in clinical settings, routine laboratory screening of pathogenic isolates does not determine the presence or the frequency of persister cells. Furthermore, the majority of research undertaken on antibiotic persistence has been done on lab-adapted bacterial strains. In the study presented here, we characterized antibiotic persisters in a panel of clinical uropathogenic Escherichia coli isolates collected from hospitals in the UK and Australia. We found that a urine-pH mimicking environment not only induces higher levels of antibiotic persistence to meropenem and colistin than standard laboratory growth conditions, but also results in rapid development of transient colistin resistance, regardless of the genetic resistance profile of the isolate. Furthermore, we provide evidence for the presence of multiple virulence factors involved in stress resistance and biofilm formation in the genomes of these isolates, whose activities have been previously shown to contribute to the formation of persister cells.


Subject(s)
Escherichia coli Infections , Urinary Tract Infections , Uropathogenic Escherichia coli , Humans , Colistin/pharmacology , Meropenem/pharmacology , Meropenem/therapeutic use , Uropathogenic Escherichia coli/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Urinary Tract Infections/drug therapy , Urinary Tract Infections/microbiology , Bacteria/genetics , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology
2.
FASEB J ; 33(10): 10808-10818, 2019 10.
Article in English | MEDLINE | ID: mdl-31262188

ABSTRACT

Colonization of the oropharynx is the initial step in Group A Streptococcus (GAS) pharyngeal infection. We have previously reported that the highly virulent M1T1 GAS clone attaches to oral epithelial cells via M1 protein interaction with blood group antigen carbohydrate structures. Here, we have identified that colonization of human oral epithelial cells by GAS serotypes M3 and M12 is mediated by human blood group antigens [ABO(H)] and Lewis (Le) antigen expression. Removal of linkage-specific fucose, galactose, N-acetylgalactosamine, and sialic acid modulated GAS colonization, dependent on host ABO(H) blood group and Le expression profile. Furthermore, N-linked glycans from human salivary glycoproteins, when released and purified, were potent inhibitors of M1, M3, and M12 GAS colonization ex vivo. These data highlight the important role played by human protein glycosylation patterns in GAS attachment to oral epithelial cell surfaces.-De Oliveira, D. M. P., Everest-Dass, A., Hartley-Tassell, L., Day, C. J., Indraratna, A., Brouwer, S., Cleary, A., Kautto, L., Gorman, J., Packer, N. H., Jennings, M. P., Walker, M. J., Sanderson-Smith, M. L. Human glycan expression patterns influence Group A streptococcal colonization of epithelial cells.


Subject(s)
Host Microbial Interactions/physiology , Polysaccharides/metabolism , Streptococcus pyogenes/pathogenicity , Antigens, Bacterial/physiology , Bacterial Adhesion/immunology , Bacterial Adhesion/physiology , Bacterial Outer Membrane Proteins/physiology , Blood Group Antigens/chemistry , Carrier Proteins/physiology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Glycosylation , Host Microbial Interactions/immunology , Humans , In Vitro Techniques , Polysaccharides/chemistry , Polysaccharides/immunology , Protein Binding , Salivary Proteins and Peptides/chemistry , Salivary Proteins and Peptides/immunology , Salivary Proteins and Peptides/metabolism , Streptococcal Infections/etiology , Streptococcal Infections/microbiology , Streptococcus pyogenes/growth & development , Streptococcus pyogenes/physiology , Virulence/physiology
3.
J Ren Nutr ; 30(5): 462-469, 2020 09.
Article in English | MEDLINE | ID: mdl-32001127

ABSTRACT

OBJECTIVE: Constipation is common in patients with end-stage kidney disease. Nondrug strategies to manage constipation are challenging because of dietary potassium, phosphate, and fluid restrictions. Nuts are a high-fiber food but are excluded from the diet because of the high potassium and phosphate content. The aim of this study was to examine the safety and efficacy of using nuts to improve constipation in adults undertaking hemodialysis (HD). DESIGN AND METHODS: Adult patients undertaking HD were recruited to this nonrandomized, 10-week repeated measures, within-subject, pragmatic clinical trial, conducted in two HD units. The intervention consisted of consumption of 40g of raw almonds daily for four weeks, followed by a two-week washout and four-week control period. The primary safety outcome measures were change in predialysis serum potassium and phosphate levels. The primary efficacy outcome was reduction in constipation, measured using the Bristol Stool Form Scale and Palliative Care Outcome Scale (POS-S) renal symptom score. Secondary outcomes included quality of life, selected uremic toxins, cognition, gut microbiota profile, and symptom burden. RESULTS: Twenty patients completed the trial (median age: 67 [interquartile range: 57.5-77.8] years, 51% male). After controlling for dialysis adequacy, anuria, dietary intake, bicarbonate, and parathyroid hormone, there were no statistically significant changes in serum potassium (P = 0.21) or phosphate (P = 0.16) associated with daily consumption of almonds. However, statistically significant improvements in constipation were seen at weeks 2, 3, 4, and 10. There were statistically significant improvements in quality of life (P = 0.030), overall symptom burden (P = 0.002), vomiting (P = 0.020), itching (P = 0.006), and skin changes (P = 0.002). CONCLUSION: Daily consumption of almonds for four weeks was safe, effective, and well tolerated. Improvements in quality of life and symptom burden warrant further research to elucidate potential mechanisms. The findings support the potential reinclusion of foods such as nuts into the diet of patients who underwent HD.


Subject(s)
Constipation/diet therapy , Constipation/etiology , Diet/methods , Kidney Failure, Chronic/complications , Nuts , Renal Dialysis , Adolescent , Adult , Aged , Aged, 80 and over , Constipation/physiopathology , Female , Humans , Intestines/physiopathology , Kidney Failure, Chronic/therapy , Male , Middle Aged , Young Adult
4.
Clin Infect Dis ; 65(9): 1523-1531, 2017 Oct 16.
Article in English | MEDLINE | ID: mdl-29020160

ABSTRACT

BACKGROUND: Group A Streptococcus (GAS) skin infections are particularly prevalent in developing nations. The GAS M protein, by which strains are differentiated into >220 different emm types, is immunogenic and elicits protective antibodies. A major obstacle for vaccine development has been the traditional understanding that immunity following infection is restricted to a single emm type. However, recent evidence has led to the hypothesis of immune cross-reactivity between emm types. METHODS: We investigated the human serological response to GAS impetigo in Fijian schoolchildren, focusing on 3 major emm clusters (E4, E6, and D4). Pre- and postinfection sera were assayed by enzyme-linked immunosorbent assay with N-terminal M peptides and bactericidal assays using the infecting-type strain, emm cluster-related strains, and nonrelated strains. RESULTS: Twenty of the 53 paired sera demonstrated a ≥4-fold increase in antibody titer against the infecting type. When tested against all cluster-related M peptides, we found that 9 of 17 (53%) paired sera had a ≥4-fold increase in antibody titer to cluster-related strains as well. When grouped by cluster, the mean change to cluster-related emm types in E4 and E6 was >4-fold (5.9-fold and 19.5-fold, respectively) but for D4 was 3.8-fold. The 17 paired sera were tested in bactericidal assays against selected cluster-related and nonrelated strains. While the responses were highly variable, numerous instances of cross-reactive killing were observed. CONCLUSIONS: These data demonstrate that M type-specific and cross-reactive immune responses occur following skin infection. The cross-reactive immune responses frequently align with emm clusters, raising new opportunities to design multivalent vaccines with broad coverage.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Skin Diseases, Bacterial/epidemiology , Skin Diseases, Bacterial/immunology , Streptococcal Infections/epidemiology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Adolescent , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Fiji/epidemiology , Humans , Longitudinal Studies , Students
5.
PLoS Pathog ; 10(8): e1004266, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25165887

ABSTRACT

Infections caused by group A Streptococcus (GAS) are characterized by robust inflammatory responses and can rapidly lead to life-threatening disease manifestations. However, host mechanisms that respond to GAS, which may influence disease pathology, are understudied. Recent works indicate that GAS infection is recognized by multiple extracellular and intracellular receptors and activates cell signalling via discrete pathways. Host leukocyte receptor binding to GAS-derived products mediates release of inflammatory mediators associated with severe GAS disease. GAS induces divergent phagocyte programmed cell death responses and has inflammatory implications. Epithelial cell apoptotic and autophagic components are mobilized by GAS infection, but can be subverted to ensure bacterial survival. Examination of host interactions with GAS and consequences of GAS infection in the context of cellular receptors responsible for GAS recognition, inflammatory mediator responses, and cell death mechanisms, highlights potential avenues for diagnostic and therapeutic intervention. Understanding the molecular and cellular basis of host symptoms during severe GAS disease will assist the development of improved treatment regimens for this formidable pathogen.


Subject(s)
Host-Parasite Interactions/physiology , Inflammation/microbiology , Streptococcal Infections/microbiology , Animals , Cell Death/physiology , Humans , Inflammation/immunology , Inflammation/physiopathology , Streptococcal Infections/immunology , Streptococcal Infections/physiopathology , Streptococcus pyogenes
6.
Clin Microbiol Rev ; 27(2): 264-301, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24696436

ABSTRACT

Streptococcus pyogenes, also known as group A Streptococcus (GAS), causes mild human infections such as pharyngitis and impetigo and serious infections such as necrotizing fasciitis and streptococcal toxic shock syndrome. Furthermore, repeated GAS infections may trigger autoimmune diseases, including acute poststreptococcal glomerulonephritis, acute rheumatic fever, and rheumatic heart disease. Combined, these diseases account for over half a million deaths per year globally. Genomic and molecular analyses have now characterized a large number of GAS virulence determinants, many of which exhibit overlap and redundancy in the processes of adhesion and colonization, innate immune resistance, and the capacity to facilitate tissue barrier degradation and spread within the human host. This improved understanding of the contribution of individual virulence determinants to the disease process has led to the formulation of models of GAS disease progression, which may lead to better treatment and intervention strategies. While GAS remains sensitive to all penicillins and cephalosporins, rising resistance to other antibiotics used in disease treatment is an increasing worldwide concern. Several GAS vaccine formulations that elicit protective immunity in animal models have shown promise in nonhuman primate and early-stage human trials. The development of a safe and efficacious commercial human vaccine for the prophylaxis of GAS disease remains a high priority.


Subject(s)
Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus pyogenes/pathogenicity , Virulence Factors/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Drug Resistance, Bacterial , Host-Pathogen Interactions , Humans , Streptococcal Infections/epidemiology , Streptococcal Infections/mortality , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/immunology , Streptococcus pyogenes/genetics , Virulence , Virulence Factors/genetics
7.
Biochemistry ; 54(25): 3960-8, 2015 Jun 30.
Article in English | MEDLINE | ID: mdl-26029848

ABSTRACT

Plasminogen (Plg) circulates in the host as two predominant glycoforms. Glycoform I Plg (GI-Plg) contains glycosylation sites at Asn289 and Thr346, whereas glycoform II Plg (GII-Plg) is exclusively glycosylated at Thr346. Surface plasmon resonance experiments demonstrated that Plg binding group A streptococcal M protein (PAM) exhibits comparative equal affinity for GI- and GII-Plg in the "closed" conformation (for GII-Plg, KD = 27.4 nM; for GI-Plg, KD = 37.0 nM). When Plg was in the "open" conformation, PAM exhibited an 11-fold increase in affinity for GII-Plg (KD = 2.8 nM) compared with that for GI-Plg (KD = 33.2 nM). The interaction of PAM with Plg is believed to be mediated by lysine binding sites within kringle (KR) 2 of Plg. PAM-GI-Plg interactions were fully inhibited with 100 mM lysine analogue ε-aminocaproic acid (εACA), whereas PAM-GII-Plg interactions were shown to be weakened but not inhibited in the presence of 400 mM εACA. In contrast, binding to the KR1-3 domains of GII-Plg (angiostatin) by PAM was completely inhibited in the presence 5 mM εACA. Along with PAM, emm pattern D GAS isolates express a phenotypically distinct SK variant (type 2b SK) that requires Plg ligands such as PAM to activate Plg. Type 2b SK was able to generate an active site and activate GII-Plg at a rate significantly higher than that of GI-Plg when bound to PAM. Taken together, these data suggest that GAS selectively recruits and activates GII-Plg. Furthermore, we propose that the interaction between PAM and Plg may be partially mediated by a secondary binding site outside of KR2, affected by glycosylation at Asn289.


Subject(s)
Bacterial Proteins/metabolism , Plasminogen/metabolism , Streptococcal Infections/enzymology , Streptococcus pyogenes/metabolism , Aminocaproates/chemistry , Aminocaproates/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Enzyme Activation , Glycosylation , Humans , Kringles , Plasminogen/chemistry , Plasminogen/genetics , Protein Binding , Protein Conformation , Streptococcal Infections/genetics , Streptococcal Infections/microbiology , Streptococcus pyogenes/chemistry , Streptococcus pyogenes/genetics , Streptococcus pyogenes/isolation & purification
8.
PLoS Pathog ; 9(7): e1003469, 2013.
Article in English | MEDLINE | ID: mdl-23853591

ABSTRACT

Recruitment of the serine protease plasmin is central to the pathogenesis of many bacterial species, including Group A streptococcus (GAS), a leading cause of morbidity and mortality globally. A key process in invasive GAS disease is the ability to accumulate plasmin at the cell surface, however the role of host activators of plasminogen in this process is poorly understood. Here, we demonstrate for the first time that the urokinase-type plasminogen activator (uPA) contributes to plasmin recruitment and subsequent invasive disease initiation in vivo. In the absence of a source of host plasminogen activators, streptokinase (Ska) was required to facilitate cell surface plasmin acquisition by GAS. However, in the absence of Ska, host activators were sufficient to promote cell surface plasmin acquisition by GAS strain 5448 during incubation with plasminogen or human plasma. Furthermore, GAS were able mediate a significant increase in the activation of zymogen pro-uPA in human plasma. In order to assess the contribution of uPA to invasive GAS disease, a previously undescribed transgenic mouse model of infection was employed. Both C57/black 6J, and AlbPLG1 mice expressing the human plasminogen transgene, were significantly more susceptible to invasive GAS disease than uPA-/- mice. The observed decrease in virulence in uPA-/-mice was found to correlate directly with a decrease in bacterial dissemination and reduced cell surface plasmin accumulation by GAS. These findings have significant implications for our understanding of GAS pathogenesis, and research aimed at therapeutic targeting of plasminogen activation in invasive bacterial infections.


Subject(s)
Disease Resistance , Plasminogen/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Animals , Crosses, Genetic , Disease Susceptibility , Enzyme Precursors/blood , Enzyme Precursors/metabolism , Fibrinolysin/metabolism , Heterozygote , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plasminogen/genetics , Proteolysis , Streptococcal Infections/blood , Streptococcal Infections/metabolism , Streptococcus pyogenes/pathogenicity , Streptokinase/metabolism , Surface Properties , Urokinase-Type Plasminogen Activator/blood , Urokinase-Type Plasminogen Activator/genetics , Virulence
9.
Biochem J ; 458(1): 23-31, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24266842

ABSTRACT

SK (streptokinase) is a secreted plasminogen activator and virulence factor of GAS (group A Streptococcus). Among GAS isolates, SK gene sequences are polymorphic and are grouped into two sequence clusters (cluster type-1 and cluster type-2) with cluster type-2 being further classified into subclusters (type-2a and type-2b). In the present study, we examined the role of bacterial and host-derived cofactors in SK-mediated plasminogen activation. All SK variants, apart from type-2b, can form an activator complex with Glu-Plg (Glu-plasminogen). Specific ligand-binding-induced conformational changes in Glu-Plg mediated by fibrinogen, PAM (plasminogen-binding group A streptococcal M protein), fibrinogen fragment D or fibrin, were required for type-2b SK to form a functional activator complex with Glu-Plg. In contrast with type-1 and type-2a SK, type-2b SK activator complexes were inhibited by α2-antiplasmin unless bound to fibrin or to the GAS cell-surface via PAM in combination with fibrinogen. Taken together, these data suggest that type-2b SK plasminogen activation may be restricted to specific microenvironments within the host such as fibrin deposits or the bacterial cell surface through the action of α2-antiplasmin. We conclude that phenotypic SK variation functionally underpins a pathogenic mechanism whereby SK variants differentially focus plasminogen activation, leading to specific niche adaption within the host.


Subject(s)
Plasminogen/metabolism , Streptococcus pyogenes/enzymology , Streptokinase/metabolism , Catalytic Domain
10.
Curr Top Microbiol Immunol ; 368: 29-48, 2013.
Article in English | MEDLINE | ID: mdl-23179674

ABSTRACT

Diseases caused by Streptococcus pyogenes (Group A streptococcus, GAS) range from superficial infections such as pharyngitis and impetigo to potentially fatal rheumatic heart disease and invasive disease. Studies spanning emm-typing surveillance to population genomics are providing new insights into the epidemiology, pathogenesis, and biology of this organism. Such studies have demonstrated the differences that exist in the epidemiology of streptococcal disease between developing and developed nations. In developing nations, where streptococcal disease is endemic, the diversity of GAS emm-types circulating is much greater than that found in developed nations. An association between emm-type and disease, as observed in developed countries is also lacking. Intriguingly, comparative genetic studies suggest that emm-type is not always a good predictor of the evolutionary relatedness of geographically distant isolates. A view of GAS as a highly dynamic organism, in possession of a core set of virulence genes that contribute to host niche specialization and common pathogenic processes, augmented by accessory genes that change the relative virulence of specific lineages is emerging. Our inability to definitively identify genetic factors that contribute to specific disease outcome underscores the complex nature of streptococcal diseases.


Subject(s)
Streptococcal Infections/epidemiology , Streptococcus pyogenes/genetics , Humans , Molecular Epidemiology , Multilocus Sequence Typing , Streptococcus pyogenes/pathogenicity , Virulence Factors/genetics
11.
FASEB J ; 27(7): 2633-43, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23531597

ABSTRACT

In Western countries, invasive infections caused by M1T1 serotype group A Streptococcus (GAS) are epidemiologically linked to mutations in the control of virulence regulatory 2-component operon (covRS). In indigenous communities and developing countries, severe GAS disease is associated with genetically diverse non-M1T1 GAS serotypes. Hypervirulent M1T1 covRS mutant strains arise through selection by human polymorphonuclear cells for increased expression of GAS virulence factors such as the DNase Sda1, which promotes neutrophil resistance. The GAS bacteremia isolate NS88.2 (emm 98.1) is a covS mutant that exhibits a hypervirulent phenotype and neutrophil resistance yet lacks the phage-encoded Sda1. Here, we have employed a comprehensive systems biology (genomic, transcriptomic, and proteomic) approach to identify NS88.2 virulence determinants that enhance neutrophil resistance in the non-M1T1 GAS genetic background. Using this approach, we have identified streptococcal collagen-like protein A and general stress protein 24 proteins as NS88.2 determinants that contribute to survival in whole blood and neutrophil resistance in non-M1T1 GAS. This study has revealed new factors that contribute to GAS pathogenicity that may play important roles in resisting innate immune defenses and the development of human invasive infections.


Subject(s)
Bacterial Proteins/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Bacterial Adhesion/genetics , Bacterial Adhesion/immunology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line , Electrophoresis, Gel, Two-Dimensional , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genome, Bacterial/genetics , Genomics/methods , Host-Pathogen Interactions/immunology , Humans , Mice , Microbial Viability/genetics , Microbial Viability/immunology , Mutation , Neutrophil Activation/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Oligonucleotide Array Sequence Analysis , Proteome/genetics , Proteome/metabolism , Proteomics/methods , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Streptococcal Infections/blood , Streptococcal Infections/microbiology , Streptococcus pyogenes/genetics , Streptococcus pyogenes/pathogenicity , Virulence/genetics , Virulence/immunology
12.
Nat Med ; 13(8): 981-5, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17632528

ABSTRACT

Most invasive bacterial infections are caused by species that more commonly colonize the human host with minimal symptoms. Although phenotypic or genetic correlates underlying a bacterium's shift to enhanced virulence have been studied, the in vivo selection pressures governing such shifts are poorly understood. The globally disseminated M1T1 clone of group A Streptococcus (GAS) is linked with the rare but life-threatening syndromes of necrotizing fasciitis and toxic shock syndrome. Mutations in the GAS control of virulence regulatory sensor kinase (covRS) operon are associated with severe invasive disease, abolishing expression of a broad-spectrum cysteine protease (SpeB) and allowing the recruitment and activation of host plasminogen on the bacterial surface. Here we describe how bacteriophage-encoded GAS DNase (Sda1), which facilitates the pathogen's escape from neutrophil extracellular traps, serves as a selective force for covRS mutation. The results provide a paradigm whereby natural selection exerted by the innate immune system generates hypervirulent bacterial variants with increased risk of systemic dissemination.


Subject(s)
Deoxyribonuclease I/metabolism , Streptococcal Infections/microbiology , Streptococcus pyogenes/enzymology , Streptococcus pyogenes/pathogenicity , Animals , Cell Survival , Deoxyribonuclease I/genetics , Humans , Immunity, Innate , Mice , Neutrophils/cytology , Neutrophils/microbiology , Phenotype , Selection, Genetic , Streptococcal Infections/pathology , Streptococcus pyogenes/genetics , Virulence
13.
Talanta ; 276: 126221, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38776768

ABSTRACT

Streptococcus pyogenes (Group A Streptococcus; GAS) is a Gram-positive bacterium responsible for substantial human mortality and morbidity. Conventional diagnosis of GAS pharyngitis relies on throat swab culture, a low-throughput, slow, and relatively invasive 'gold standard'. While molecular approaches are becoming increasingly utilized, the potential of saliva as a diagnostic fluid for GAS infection remains largely unexplored. Here, we present a novel, high-throughput, sensitive, and robust speB qPCR assay that reliably detects GAS in saliva using innovative 3base™ technology (Genetic Signatures Limited, Sydney, Australia). The assay has been validated on baseline, acute, and convalescent saliva samples generated from the Controlled Human Infection for Vaccination Against Streptococcus (CHIVAS-M75) trial, in which healthy adult participants were challenged with emm75 GAS. In these well-defined samples, our high-throughput assay outperforms throat culture and conventional qPCR in saliva respectively, affirming the utility of the 3base™ platform, demonstrating the feasibility of saliva as a diagnostic biofluid, and paving the way for the development of novel non-invasive approaches for the detection of GAS and other oropharyngeal pathogens.


Subject(s)
Pharyngitis , Saliva , Streptococcus pyogenes , Humans , Streptococcus pyogenes/isolation & purification , Saliva/microbiology , Pharyngitis/microbiology , Pharyngitis/diagnosis , Streptococcal Infections/diagnosis , Streptococcal Infections/microbiology , Adult , Real-Time Polymerase Chain Reaction/methods
14.
Virulence ; 14(1): 2264090, 2023 12.
Article in English | MEDLINE | ID: mdl-37830540

ABSTRACT

ABBREVIATIONS: CovRS, control of virulence regulatory system; GAS, Group A Streptococcus; PMN, polymorphonuclear leukocyte.


Subject(s)
Neutrophils , Streptococcal Infections , Humans , Streptococcus pyogenes/genetics , Caspases/genetics , Bacterial Proteins/genetics , Virulence
15.
J Biomed Biotechnol ; 2012: 272148, 2012.
Article in English | MEDLINE | ID: mdl-23118502

ABSTRACT

Multiple species of bacteria are able to sequester the host zymogen plasminogen to the cell surface. Once localised to the bacterial surface, plasminogen can act as a cofactor in adhesion, or, following activation to plasmin, provide a source of potent proteolytic activity. Numerous bacterial plasminogen receptors have been identified, and the mechanisms by which they interact with plasminogen are diverse. Here we provide an overview of bacterial plasminogen receptors and discuss the diverse role bacterial plasminogen acquisition plays in the relationship between bacteria and the host.


Subject(s)
Bacteria/metabolism , Receptors, Urokinase Plasminogen Activator/metabolism , Bacteria/cytology , Host-Pathogen Interactions , Humans , Models, Biological , Plasminogen/chemistry , Plasminogen/metabolism , Protein Binding
16.
Biochim Biophys Acta ; 1804(6): 1342-9, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20152941

ABSTRACT

In vertebrates, fibrinolysis is primarily carried out by the serine protease plasmin (Pm), which is derived from activation of the zymogen precursor, plasminogen (Pg). One of the most distinctive features of Pg/Pm is the presence of five homologous kringle (K) domains. These structural elements possess conserved Lys-binding sites (LBS) that facilitate interactions with substrates, activators, inhibitors and receptors. In human Pg (hPg), K2 displays weak Lys affinity, however the LBS of this domain has been implicated in an atypical interaction with the N-terminal region of a bacterial surface protein known as PAM (Pg-binding group A streptococcal M-like protein). A direct correlation has been established between invasiveness of group A streptococci and their ability to bind Pg. It has been previously demonstrated that a 30-residue internal peptide (VEK-30) from the N-terminal region of PAM competitively inhibits binding of the full-length parent protein to Pg. We have attempted to determine the effects of this ligand-protein interaction on the regulation of Pg zymogen activation and conformation. Our results show minimal effects on the sedimentation velocity coefficients (S degrees (20,w)) of Pg when associated to VEK-30 and a direct relationship between the concentration of VEK-30 or PAM and the activation rate of Pg. These results are in contrast with the major conformational changes elicited by small-molecule activators of Pg, and point towards a novel mechanism of Pg activation that may underlie group A streptococcal (GAS) virulence.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Outer Membrane Proteins/chemistry , Bacterial Proteins/chemistry , Carrier Proteins/chemistry , Peptides/chemistry , Plasminogen/chemistry , Streptococcus pyogenes/chemistry , Virulence Factors/chemistry , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Enzyme Activation , Fibrinolysin/metabolism , Fibrinolysis/physiology , Humans , Peptides/metabolism , Plasminogen/metabolism , Protein Structure, Tertiary , Streptococcus pyogenes/metabolism , Virulence Factors/metabolism
17.
Int J Med Microbiol ; 301(2): 157-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20951639

ABSTRACT

Increasing awareness of the relevance of Streptococcus dysgalactiae ssp. equisimilis as a human pathogen motivates the analysis of its pathomechanisms. One of the mechanisms that increases infectivity and dissemination of several streptococcal species is the recruitment and subsequent activation of host plasminogen on the streptococcal surface. This study identified GCS3 as a novel plasminogen-binding M protein of S. dysgalactiae ssp. equisimilis and revealed a difference in the mode of binding as compared to the plasminogen-binding protein PAM of S. pyogenes. In contrast to PAM, GCS3 did not bind to the kringle 1-3 region of plasminogen. Despite this difference, GCS3 exerts the same function of recruiting plasminogen to the streptococcal surface, which can be activated by streptokinase and host plasminogen activators to serve as a spreading factor. Moreover, we demonstrate a role of GCS3 in plasminogen-dependent streptococcal adherence to human pharyngeal cells (cell line Detroit 562) that indicates an additional function of the protein as an adhesin in the oral cavity.


Subject(s)
Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Carrier Proteins/metabolism , Plasminogen/metabolism , Streptococcus/pathogenicity , Virulence Factors/metabolism , Adhesins, Bacterial/genetics , Bacterial Adhesion , Bacterial Proteins/genetics , Carrier Proteins/genetics , Cell Line , Epithelial Cells/microbiology , Humans , Streptococcus/genetics , Virulence Factors/genetics
18.
Sci Rep ; 11(1): 8200, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859234

ABSTRACT

Group A Streptococcus (GAS) causes 700 million infections and accounts for half a million deaths per year. Biofilm formation has been implicated in both pharyngeal and dermal GAS infections. In vitro, plate-based assays have shown that several GAS M-types form biofilms, and multiple GAS virulence factors have been linked to biofilm formation. Although the contributions of these plate-based studies have been valuable, most have failed to mimic the host environment, with many studies utilising abiotic surfaces. GAS is a human specific pathogen, and colonisation and subsequent biofilm formation is likely facilitated by distinct interactions with host tissue surfaces. As such, a host cell-GAS model has been optimised to support and grow GAS biofilms of a variety of GAS M-types. Improvements and adjustments to the crystal violet biofilm biomass assay have also been tailored to reproducibly detect delicate GAS biofilms. We propose 72 h as an optimal growth period for yielding detectable biofilm biomass. GAS biofilms formed are robust and durable, and can be reproducibly assessed via staining/washing intensive assays such as crystal violet with the aid of methanol fixation prior to staining. Lastly, SEM imaging of GAS biofilms formed by this model revealed GAS cocci chains arranged into three-dimensional aggregated structures with EPS matrix material. Taken together, we outline an efficacious GAS biofilm pharyngeal cell model that can support long-term GAS biofilm formation, with biofilms formed closely resembling those seen in vivo.


Subject(s)
Biofilms/growth & development , Pharynx/microbiology , Streptococcus pyogenes/physiology , Calibration , Cell Culture Techniques/standards , Cells, Cultured , Humans , Microbiological Techniques/standards , Models, Biological , Pharynx/cytology , Streptococcal Infections/microbiology , Streptococcus pyogenes/growth & development , Streptococcus pyogenes/pathogenicity , Virulence Factors/metabolism
19.
Antibiotics (Basel) ; 9(11)2020 Nov 04.
Article in English | MEDLINE | ID: mdl-33158121

ABSTRACT

Group A Streptococcus (GAS) causes 700 million infections and accounts for half a million deaths per year. Antibiotic treatment failure rates of 20-40% have been observed. The role host cell glycans play in GAS biofilm formation in the context of GAS pharyngitis and subsequent antibiotic treatment failure has not been previously investigated. GAS serotype M12 GAS biofilms were assessed for biofilm formation on Detroit 562 pharyngeal cell monolayers following enzymatic removal of all N-linked glycans from pharyngeal cells with PNGase F. Removal of N-linked glycans resulted in an increase in biofilm biomass compared to untreated controls. Further investigation into the removal of terminal mannose and sialic acid residues with α1-6 mannosidase and the broad specificity sialidase (Sialidase A) also found that biofilm biomass increased significantly when compared to untreated controls. Increases in biofilm biomass were associated with increased production of extracellular polymeric substances (EPS). Furthermore, it was found that M12 GAS biofilms grown on untreated pharyngeal monolayers exhibited a 2500-fold increase in penicillin tolerance compared to planktonic GAS. Pre-treatment of monolayers with exoglycosidases resulted in a further doubling of penicillin tolerance in resultant biofilms. Lastly, an additional eight GAS emm-types were assessed for biofilm formation in response to terminal mannose and sialic acid residue removal. As seen for M12, biofilm biomass on monolayers increased following removal of terminal mannose and sialic acid residues. Collectively, these data demonstrate that pharyngeal cell surface glycan structures directly impact GAS biofilm formation in a strain and glycan specific fashion.

20.
Front Cell Infect Microbiol ; 10: 596023, 2020.
Article in English | MEDLINE | ID: mdl-33585270

ABSTRACT

Invasive infections due to group A Streptococcus (GAS) advance rapidly causing tissue degradation and unregulated inflammation. Neutrophils are the primary immune cells that respond to GAS. The neutrophil response to GAS was characterised in response to two M1T1 isolates; 5448 and animal passaged variant 5448AP. Co-incubation of neutrophils with 5448AP resulted in proliferation of GAS and lowered the production of reactive oxygen species when compared with 5448. Infection with both strains invoked neutrophil death, however apoptosis was reduced in response to 5448AP. Both strains induced neutrophil caspase-1 and caspase-4 expression in vitro, with inflammatory caspase activation detected in vitro and in vivo. GAS infections involving strains such as 5448AP that promote an inflammatory neutrophil phenotype may contribute to increased inflammation yet ineffective bacterial eradication, contributing to the severity of invasive GAS infections.


Subject(s)
Streptococcal Infections , Streptococcus pyogenes , Animals , Caspases/genetics , Neutrophils , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL