Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 307
Filter
1.
J Biol Chem ; 299(12): 105321, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37802313

ABSTRACT

Staphylococcus aureus (S. aureus) is a serious global pathogen that causes a diverse range of invasive diseases. S. aureus utilizes a family of pore-forming toxins, known as bi-component leukocidins, to evade the host immune response and promote infection. Among these is LukAB (leukocidin A/leukocidin B), a toxin that assembles into an octameric ß-barrel pore in the target cell membrane, resulting in host cell death. The established cellular receptor for LukAB is CD11b of the Mac-1 complex. Here, we show that hydrogen voltage-gated channel 1 is also required for the cytotoxicity of all major LukAB variants. We demonstrate that while each receptor is sufficient to recruit LukAB to the plasma membrane, both receptors are required for maximal lytic activity. Why LukAB requires two receptors, and how each of these receptors contributes to pore-formation remains unknown. To begin to resolve this, we performed an alanine scanning mutagenesis screen to identify mutations that allow LukAB to maintain cytotoxicity without CD11b. We discovered 30 mutations primarily localized in the stem domains of LukA and LukB that enable LukAB to exhibit full cytotoxicity in the absence of CD11b. Using crosslinking, electron microscopy, and hydroxyl radical protein footprinting, we show these mutations increase the solvent accessibility of the stem domain, priming LukAB for oligomerization. Together, our data support a model in which CD11b binding unlatches the membrane penetrating stem domains of LukAB, and this change in flexibility promotes toxin oligomerization.


Subject(s)
Bacterial Proteins , Leukocidins , Staphylococcus aureus , Toxins, Biological , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Membrane/metabolism , Leukocidins/genetics , Leukocidins/metabolism , Leukocidins/toxicity , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Toxins, Biological/metabolism , Mutation , Protein Binding/genetics , Protein Domains , Cell Line , CHO Cells , Cricetulus , Animals
2.
Microbiol Spectr ; 11(4): e0107323, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37347186

ABSTRACT

Staphylococcus aureus gamma-hemolysin CB (HlgCB) is a core-genome-encoded pore-forming toxin that targets the C5a receptor, similar to the phage-encoded Panton-Valentine leucocidin (PVL). Absolute quantification by mass spectrometry of HlgCB in 39 community-acquired pneumonia (CAP) isolates showed considerable variations in the HlgC and HlgB yields between isolates. Moreover, although HlgC and HlgB are encoded on a single operon, their levels were dissociated in 10% of the clinical strains studied. To decipher the molecular basis for the variation in hlgCB expression and protein production among strains, different regulation levels were analyzed in representative clinical isolates and reference strains. Both the HlgCB level and the HlgC/HlgB ratio were found to depend on hlgC promoter activity and mRNA processing and translation. Strikingly, only one single nucleotide polymorphism (SNP) in the 5' untranslated region (UTR) of hlgCB mRNA strongly impaired hlgC translation in the USA300 strain, leading to a strong decrease in the level of HlgC but not in HlgB. Finally, we found that high levels of HlgCB synthesis led to mortality in a rabbit model of pneumonia, correlated with the implication of the role of HlgCB in severe S. aureus CAP. Taken together, this work illustrates the complexity of virulence factor expression in clinical strains and demonstrates a butterfly effect where subtle genomic variations have a major impact on phenotype and virulence. IMPORTANCE S. aureus virulence in pneumonia results in its ability to produce several virulence factors, including the leucocidin PVL. Here, we demonstrate that HlgCB, another leucocidin, which targets the same receptors as PVL, highly contributes to S. aureus virulence in pvl-negative strains. In addition, considerable variations in HlgCB quantities are observed among clinical isolates from patients with CAP. Biomolecular analyses have revealed that a few SNPs in the promoter sequences and only one SNP in the 5' UTR of hlgCB mRNA induce the differential expression of hlgCB, drastically impacting hlgC mRNA translation. This work illustrates the subtlety of regulatory mechanisms in bacteria, especially the sometimes major effects on phenotypes of single nucleotide variation in noncoding regions.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Animals , Rabbits , Staphylococcus aureus/metabolism , Leukocidins/genetics , Leukocidins/metabolism , Leukocidins/pharmacology , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Virulence/genetics , Exotoxins/genetics , Exotoxins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
3.
Cell Host Microbe ; 31(5): 751-765.e11, 2023 05 10.
Article in English | MEDLINE | ID: mdl-37098341

ABSTRACT

Treating and preventing infections by antimicrobial-resistant bacterial pathogens is a worldwide problem. Pathogens such as Staphylococcus aureus produce an array of virulence determinants, making it difficult to identify single targets for the development of vaccines or monoclonal therapies. We described a human-derived anti-S. aureus monoclonal antibody (mAb)-centyrin fusion protein ("mAbtyrin") that simultaneously targets multiple bacterial adhesins, resists proteolysis by bacterial protease GluV8, avoids Fc engagement by S. aureus IgG-binding proteins SpA and Sbi, and neutralizes pore-forming leukocidins via fusion with anti-toxin centyrins, while maintaining Fc- and complement-mediated functions. Compared with the parental mAb, mAbtyrin protected human phagocytes and boosted phagocyte-mediated killing. The mAbtyrin also reduced pathology, reduced bacterial burden, and protected from different types of infections in preclinical animal models. Finally, mAbtyrin synergized with vancomycin, enhancing pathogen clearance in an animal model of bacteremia. Altogether, these data establish the potential of multivalent mAbs for treating and preventing S. aureus diseases.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Humans , Staphylococcus aureus , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control , Staphylococcal Infections/microbiology , Antibodies, Monoclonal/therapeutic use , Phagocytes/metabolism , Leukocidins/metabolism , Leukocidins/therapeutic use
4.
Int J Mol Sci ; 24(8)2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37108277

ABSTRACT

The gamma-hemolysin protein is one of the most common pore-forming toxins expressed by the pathogenic bacterium Staphylococcus aureus. The toxin is used by the pathogen to escape the immune system of the host organism, by assembling into octameric transmembrane pores on the surface of the target immune cell and leading to its death by leakage or apoptosis. Despite the high potential risks associated with Staphylococcus aureus infections and the urgent need for new treatments, several aspects of the pore-formation process from gamma-hemolysin are still unclear. These include the identification of the interactions between the individual monomers that lead to the formation of a dimer on the cell membrane, which represents the unit for further oligomerization. Here, we employed a combination of all-atom explicit solvent molecular dynamics simulations and protein-protein docking to determine the stabilizing contacts that guide the formation of a functional dimer. The simulations and the molecular modeling reveal the importance of the flexibility of specific protein domains, in particular the N-terminus, to drive the formation of the correct dimerization interface through functional contacts between the monomers. The results obtained are compared with the experimental data available in the literature.


Subject(s)
Bacterial Toxins , Hemolysin Proteins , Hemolysin Proteins/metabolism , Bacterial Toxins/metabolism , Leukocidins/metabolism , Bacterial Proteins/metabolism , Cell Membrane/metabolism
5.
Microbiol Spectr ; 11(1): e0257422, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36688711

ABSTRACT

Staphylococcus aureus is a clinically important bacterial pathogen that has become resistant to treatment with most routinely used antibiotics. Alternative strategies, such as vaccination and phage therapy, are therefore actively being investigated to prevent or combat staphylococcal infections. Vaccination requires that vaccine targets are expressed at sufficient quantities during infection so that they can be targeted by the host's immune system. While our knowledge of in vitro expression levels of putative vaccine candidates is comprehensive, crucial in vivo expression data are scarce and promising vaccine candidates during in vitro assessment often prove ineffective in preventing S. aureus infection. Here, we show how a newly developed high-throughput quantitative reverse transcription-PCR (qRT-PCR) assay monitoring the expression of 84 staphylococcal genes encoding mostly virulence factors can inform the selection and design of effective vaccine candidates against staphylococcal infections. We show that this assay can accurately quantify mRNA expression levels of these genes in several host organs relying only on very limited amounts of bacterial mRNA in each sample. We selected two highly expressed genes, lukE and lukD, encoding pore-forming leukotoxins, to inform the design of detoxified recombinant proteins and showed that immunization with recombinant genetically detoxified LukED antigens conferred protection against staphylococcal skin infection in mice. Consequently, knowledge of in vivo-expressed virulence determinants can be successfully deployed to identify and select promising candidates for optimized design of effective vaccine antigens against S. aureus. Notably, this approach should be broadly applicable to numerous other pathogens. IMPORTANCE Vaccination is an attractive strategy for preventing bacterial infections in an age of increased antimicrobial resistance. However, vaccine development frequently suffers significant setbacks when candidate antigens that show promising results in in vitro experimentation fail to protect from disease. An alluring strategy is to focus resources on developing bacterial virulence factors that are expressed during disease establishment or maintenance and are critical for bacterial in-host survival as vaccine targets. While expression profiles of many virulence factors have been characterized in detail in vitro, our knowledge of their in vivo expression profiles is still scarce. Here, using a high-throughput qRT-PCR approach, we identified two highly expressed leukotoxins in a murine infection model and showed that genetically detoxified derivatives of these elicited a protective immune response in a murine skin infection model. Therefore, in vivo gene expression can inform the selection of promising candidates for the design of effective vaccine antigens.


Subject(s)
Staphylococcal Infections , Vaccines , Animals , Mice , Staphylococcus aureus/metabolism , Bacterial Proteins/metabolism , Leukocidins/genetics , Leukocidins/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism , Vaccines/metabolism , Staphylococcal Infections/microbiology , Gene Expression Profiling
6.
Clin Exp Dermatol ; 47(12): 2150-2158, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36040400

ABSTRACT

Panton-Valentine leucocidin (PVL) is a virulence factor produced by certain strains of Staphylococcus aureus (SA). Through its cytolytic action on the cell membranes of human polymorphonuclear neutrophils, PVL causes a range of pathologies collectively known as PVL-SA disease. The hallmark clinical signs of PVL-SA are recurrent boils and necrotizing skin and soft tissue infections (SSTIs) in otherwise healthy patients; however, it can lead to more severe and invasive presentations, including necrotizing haemorrhagic pneumonia, necrotizing fasciitis and purpura fulminans. Young adults with minimal previous exposure to healthcare settings tend to be at highest risk for acquiring PVL-SA disease, with close physical contact playing a central role in disease transmission. The prevalence of PVL-SA varies globally; however, this is often underestimated owing to a lack of routine PVL testing. In the UK, PVL-positive SA isolates have been rising over the past decade alongside an increasing prevalence of multidrug resistance in larger cities. This review article aims to raise awareness of the PVL toxin, to aid clinicians with diagnostic pointers and to provide guidance with treatment, with an emphasis on the need for further population-based studies.


Subject(s)
Soft Tissue Infections , Staphylococcal Infections , Humans , Exotoxins/metabolism , Leukocidins/metabolism , Prevalence , Soft Tissue Infections/epidemiology , Staphylococcal Infections/epidemiology , Staphylococcus aureus
7.
Biochim Biophys Acta Biomembr ; 1864(9): 183970, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35605647

ABSTRACT

Methicillin-resistant Staphylococcus aureus is among those pathogens currently posing the highest threat to public health. Its host immune evasion strategy is mediated by pore-forming toxins (PFTs), among which the bi-component γ-hemolysin is one of the most common. The complexity of the porogenesis mechanism by γ-hemolysin poses difficulties in the development of antivirulence therapies targeting PFTs from S. aureus, and sparse and apparently contrasting experimental data have been produced. Here, through a large set of molecular dynamics simulations at different levels of resolution, we investigate the first step of pore formation, and in particular the effect of membrane composition on the ability of γ-hemolysin components, LukF and Hlg2, to steadily adhere to the lipid bilayer in the absence of proteinaceous receptors. Our simulations are in agreement with experimental data of γ-hemolysin pore formation on model membranes, which are here explained on the basis of the bilayer properties. Our computational investigation suggests a possible rationale to explain experimental data on phospholipid binding to the LukF component, and to hypothesise a mechanism by which, on purely lipidic bilayers, the stable anchoring of LukF to the cell surface facilitates Hlg2 binding, through the exposure of its N-terminal region. We expect that further insights on the mechanism of transition between soluble and membrane bound-forms and on the role played by the lipid molecules will contribute to the design of antivirulence agents with enhanced efficacy against methicillin-resistant S. aureus infections.


Subject(s)
Bacterial Toxins , Methicillin-Resistant Staphylococcus aureus , Bacterial Proteins/metabolism , Bacterial Toxins/chemistry , Hemolysin Proteins/chemistry , Leukocidins/chemistry , Leukocidins/metabolism , Lipid Bilayers/metabolism , Methicillin-Resistant Staphylococcus aureus/metabolism , Staphylococcus aureus/metabolism
8.
J Thromb Haemost ; 20(6): 1464-1475, 2022 06.
Article in English | MEDLINE | ID: mdl-35303391

ABSTRACT

BACKGROUND: Toxins are key virulence determinants of pathogens and can impair the function of host immune cells, including platelets. Insights into pathogen toxin interference with platelets will be pivotal to improve treatment of patients with bacterial bloodstream infections. MATERIALS AND METHODS: In this study, we deciphered the effects of Staphylococcus aureus toxins α-hemolysin, LukAB, LukDE, and LukSF on human platelets and compared the effects with the pore forming toxin pneumolysin of Streptococcus pneumoniae. Activation of platelets and loss of platelet function were investigated by flow cytometry, aggregometry, platelet viability, fluorescence microscopy, and intracellular calcium release. Thrombus formation was assessed in whole blood. RESULTS: α-hemolysin (Hla) is known to be a pore-forming toxin. Hla-induced calcium influx initially activates platelets as indicated by CD62P and αIIbß3 integrin activation, but also induces finally alterations in the phenotype of platelets. In contrast to Hla and pneumolysin, S. aureus bicomponent pore-forming leukocidins LukAB, LukED, and LukSF do not bind to platelets and had no significant effect on platelet activation and viability. The presence of small amounts of Hla (0.2 µg/ml) in whole blood abrogates thrombus formation indicating that in systemic infections with S. aureus the stability of formed thrombi is impaired. Damage of platelets by Hla was not neutralized by intravenous immune globulins. CONCLUSION: Our findings might be of clinical relevance for S. aureus induced endocarditis. Stabilizing the aortic-valve thrombi by inhibiting Hla-induced impairment of platelets might reduce the risk for septic (micro-)embolization.


Subject(s)
Staphylococcal Infections , Thrombosis , Bacterial Proteins/metabolism , Bacterial Proteins/pharmacology , Calcium , Hemolysin Proteins/metabolism , Hemolysin Proteins/pharmacology , Humans , Leukocidins/metabolism , Staphylococcus aureus
9.
Elife ; 112022 03 21.
Article in English | MEDLINE | ID: mdl-35311641

ABSTRACT

Staphylococcus aureus (SA) leukocidin ED (LukED) belongs to a family of bicomponent pore forming toxins that play important roles in SA immune evasion and nutrient acquisition. LukED targets specific G protein-coupled chemokine receptors to lyse human erythrocytes (red blood cells) and leukocytes (white blood cells). The first recognition step of receptors is critical for specific cell targeting and lysis. The structural and molecular bases for this mechanism are not well understood but could constitute essential information to guide antibiotic development. Here, we characterized the interaction of LukE with chemokine receptors ACKR1, CCR2, and CCR5 using a combination of structural, pharmacological, and computational approaches. First, crystal structures of LukE in complex with a small molecule mimicking sulfotyrosine side chain (p-cresyl sulfate) and with peptides containing sulfotyrosines issued from receptor sequences revealed the location of receptor sulfotyrosine binding sites in the toxins. Then, by combining previous and novel experimental data with protein docking, classical and accelerated weight histogram (AWH) molecular dynamics we propose models of the ACKR1-LukE and CCR5-LukE complexes. This work provides novel insights into chemokine receptor recognition by leukotoxins and suggests that the conserved sulfotyrosine binding pocket could be a target of choice for future drug development.


Subject(s)
Staphylococcal Infections , Staphylococcus aureus , Humans , Immune Evasion , Leukocidins/metabolism , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Staphylococcus aureus/genetics
10.
J Immunol ; 208(5): 1170-1179, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35140134

ABSTRACT

Mucosa-associated invariant T (MAIT) cells recognize bacterial riboflavin metabolite Ags presented by MHC class Ib-related protein (MR1) and play important roles in immune control of microbes that synthesize riboflavin. This includes the pathobiont Staphylococcus aureus, which can also express a range of virulence factors, including the secreted toxin leukocidin ED (LukED). In this study, we found that human MAIT cells are hypersensitive to LukED-mediated lysis and lost on exposure to the toxin, leaving a T cell population devoid of MAIT cells. The cytolytic effect of LukED on MAIT cells was rapid and occurred at toxin concentrations lower than those required for toxicity against conventional T cells. Furthermore, this coincided with high MAIT cell expression of CCR5, and loss of these cells was efficiently inhibited by the CCR5 inhibitor maraviroc. Interestingly, exposure and preactivation of MAIT cells with IL-12 and IL-18, or activation via TCR triggering, partially protected from LukED toxicity. Furthermore, analysis of NK cells indicated that LukED targeted the mature cytotoxic CD57+ NK cell subset in a CCR5-independent manner. Overall, these results indicate that LukED efficiently eliminates immune cells that can respond rapidly to S. aureus in an innate fashion without the need for clonal expansion, and that MAIT cells are exceptionally vulnerable to this toxin. Thus, the findings support a model where LukED secretion may allow S. aureus to avoid recognition by the rapid cell-mediated responses mediated by MAIT cells and NK cells.


Subject(s)
Immune Evasion/immunology , Killer Cells, Natural/immunology , Leukocidins/metabolism , Mucosal-Associated Invariant T Cells/pathology , Receptors, CCR5/metabolism , Staphylococcus aureus/pathogenicity , CCR5 Receptor Antagonists/pharmacology , Cell Line , Humans , Interleukin-12 Subunit p35/metabolism , Interleukin-18/metabolism , Lymphocyte Activation/immunology , Maraviroc/pharmacology , Mucosal-Associated Invariant T Cells/immunology , Staphylococcal Infections/pathology , Staphylococcus aureus/immunology , THP-1 Cells , Virulence Factors/metabolism
11.
FEBS J ; 289(12): 3505-3520, 2022 06.
Article in English | MEDLINE | ID: mdl-35030303

ABSTRACT

Staphylococcus aureus expresses several hemolytic pore-forming toxins (PFTs), which are all commonly composed of three domains: cap, rim and stem. PFTs are expressed as soluble monomers and assemble to form a transmembrane ß-barrel pore in the erythrocyte cell membrane. The stem domain undergoes dramatic conformational changes to form a pore. Staphylococcal PFTs are classified into two groups: one-component α-hemolysin (α-HL) and two-component γ-hemolysin (γ-HL). The α-HL forms a homo-heptamer, whereas γ-HL is an octamer composed of F-component (LukF) and S-component (Hlg2). Because PFTs are used as materials for nanopore-based sensors, knowledge of the functional properties of PFTs is used to develop new, engineered PFTs. However, it remains challenging to design PFTs with a ß-barrel pore because their formation as transmembrane protein assemblies requires large conformational changes. In the present study, aiming to investigate the design principles of the ß-barrel formed as a consequence of the conformational change, chimeric mutants composed of the cap/rim domains of α-HL and the stem of LukF or Hlg2 were prepared. Biochemical characterization and electron microscopy showed that one of them assembles as a heptameric one-component PFT, whereas another participates as both a heptameric one- and heptameric/octameric two-component PFT. All chimeric mutants intrinsically assemble into SDS-resistant oligomers. Based on these observations, the role of the stem domain of these PFTs is discussed. These findings provide clues for the engineering of staphylococcal PFT ß-barrels for use in further promising applications.


Subject(s)
Bacterial Toxins , Hemolysin Proteins , Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Hemolysis , Leukocidins/chemistry , Leukocidins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism
12.
Nat Commun ; 12(1): 6193, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34702812

ABSTRACT

Staphylococcus aureus bi-component pore-forming leukocidins are secreted toxins that directly target and lyse immune cells. Intriguingly, one of the leukocidins, Leukocidin AB (LukAB), is found associated with the bacterial cell envelope in addition to secreted into the extracellular milieu. Here, we report that retention of LukAB on the bacterial cells provides S. aureus with a pre-synthesized active toxin that kills immune cells. On the bacteria, LukAB is distributed as discrete foci in two distinct compartments: membrane-proximal and surface-exposed. Through genetic screens, we show that a membrane lipid, lysyl-phosphatidylglycerol (LPG), and lipoteichoic acid (LTA) contribute to LukAB deposition and release. Furthermore, by studying non-covalently surface-bound proteins we discovered that the sorting of additional exoproteins, such as IsaB, Hel, ScaH, and Geh, are also controlled by LPG and LTA. Collectively, our study reveals a multistep secretion system that controls exoprotein storage and protein translocation across the S. aureus cell wall.


Subject(s)
Cell Membrane/metabolism , Cell Wall/metabolism , Staphylococcus aureus/metabolism , Virulence Factors/metabolism , Animals , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Cytotoxins/metabolism , Cytotoxins/toxicity , Humans , Leukocidins/metabolism , Leukocidins/toxicity , Lipopolysaccharides/genetics , Lipopolysaccharides/metabolism , Lysine/genetics , Lysine/metabolism , Mice , Phagocytes/drug effects , Phosphatidylglycerols/genetics , Phosphatidylglycerols/metabolism , Protein Transport , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Teichoic Acids/genetics , Teichoic Acids/metabolism , Virulence Factors/toxicity
13.
Microbiologyopen ; 10(5): e1235, 2021 10.
Article in English | MEDLINE | ID: mdl-34713607

ABSTRACT

OBJECTIVE: To characterize the Staphylococcus aureus strains colonizing healthy Spanish children. METHODS: Between March and July 2018, 1876 Spanish children younger than 14 years attending primary healthcare centers were recruited from rural and urban areas. Staphylococcus aureus colonization of the anterior nostrils was analyzed. MecA and mecC genes, antibiotic susceptibility, and genotyping according to the spa were determined in all strains, and the following toxins were examined: Panton-Valentine leucocidin (pvl), toxic shock syndrome toxin (tst), and exfoliative toxins (eta, etb, etd). Multilocus sequence typing (MLST) and staphylococcal cassette chromosome (SCCmec) typing were performed on methicillin-resistant Staphylococcus aureus (MRSA) strains, as well as pulsed-field gel electrophoresis (PFGE). RESULTS: 619 strains were isolated in 1876 children (33%), and 92% of them were sent for characterization to the Spanish National Centre of Microbiology (n = 572). Twenty (3.5%) of these strains were mecA-positive. Several spa types were detected among MRSA, being t002 the most frequently observed (30%), associating with SCCmec IVc. Among MSSA, 33% were positive for tst, while only 0.73% were positive for pvl. The 20 MRSA strains were negative for pvl, and 6 (30%) harbored the tst gene. CONCLUSIONS: methicillin-resistant Staphylococcus aureus nasal colonization in Spanish children is rare, with t002 being the most observed spa type, associated with SCCmec IVc. None of the MRSA strains produced pvl, but up to 30% of S. aureus strains were positive for tst.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/classification , Methicillin-Resistant Staphylococcus aureus/genetics , Nose/microbiology , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Child , Child, Preschool , DNA, Bacterial , Drug Resistance, Bacterial , Exfoliatins/metabolism , Exotoxins/metabolism , Female , Genotyping Techniques/methods , Humans , Leukocidins/metabolism , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Microbial Sensitivity Tests , Multilocus Sequence Typing/methods , Penicillin-Binding Proteins/metabolism , Spain , Staphylococcal Protein A/metabolism
14.
BMC Infect Dis ; 21(1): 372, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33882854

ABSTRACT

BACKGROUND: Some Staphylococcus aureus strains produce Panton-Valentine leukocidin (PVL), a bi-component pore-forming toxin, which causes leukocyte lysis and tissue necrosis. Currently, there is very limited information on the molecular epidemiology of PVL-encoding S. aureus strains in Iran. This study aimed to determine the molecular epidemiology and genetic background of PVL-positive S. aureus clinical strains isolated from Iranian patients. METHODS: A total of 28 PVL-positive S. aureus strains were detected from 600 S. aureus isolates between February 2015 and March 2018 from different hospitals in Tehran, Iran. Antimicrobial susceptibility testing was performed according to the Clinical and Laboratory Standards Institute (CLSI) guidelines. Molecular genotyping was performed using SCCmec and accessory gene regulator (agr) typing, PVL haplotyping, multilocus sequence typing (MLST), and pulsed-field gel electrophoresis (PFGE). RESULTS: The highest antibiotic resistance rate was found to be against erythromycin (57.1%), followed by ciprofloxacin (42.8%) and clindamycin (35.7%). Moreover, 19 (67.9%) out of 28 S. aureus isolates were identified as MRSA, including CA-MRSA (14/19, 73.7%) and HA-MRSA (5/19, 26.3%). SCCmec type IVa was detected as the predominant type (10/19, 52.6%), followed by type III (5/19, 26.3%) and type V (4/19, 21.1%). The agr type I was identified as the most common type (14/28, 50%), and H and R haplotype groups were observed at frequencies of 67.9 and 32.1%, respectively. Among H variants, the predominant variant was H2 (78/9%). The isolates encompassed 21 different sequence types (STs), including 16 new STs (ST5147 to ST5162). Based on eBURST analysis, the isolates were clustered into five CCs, including CC30, CC22, CC1, CC8, and CC5 (ST5160), and nine singletons. PFGE typing showed that 24 isolates were clustered into A (4 pulsotypes), B (9 pulsotypes), and C (11 pulsotypes) clusters. CONCLUSIONS: A high prevalence of PVL-positive CA-MRSA strains was detected in Iran. The majority of PVL-positive isolates were of H (mostly H2) variant, while R variant was harbored by 100% of PVL-positive MRSA strains. Also, CC8, CC22, and CC30 were identified as the dominant clones among PVL-encoding S. aureus strains. This study promotes a better understanding of the molecular epidemiology and evolution of PVL-positive S. aureus strains in Iran.


Subject(s)
Bacterial Toxins/genetics , Exotoxins/genetics , Haplotypes , Leukocidins/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Polymorphism, Single Nucleotide , Staphylococcal Infections/epidemiology , Anti-Bacterial Agents/therapeutic use , Bacterial Toxins/metabolism , Drug Resistance, Multiple, Bacterial/genetics , Electrophoresis, Gel, Pulsed-Field , Exotoxins/metabolism , Genomics , Humans , Iran/epidemiology , Leukocidins/metabolism , Male , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Multilocus Sequence Typing , Phylogeny , Prevalence , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
15.
Nat Microbiol ; 6(6): 731-745, 2021 06.
Article in English | MEDLINE | ID: mdl-33875847

ABSTRACT

Staphylococcus aureus has evolved into diverse lineages, known as clonal complexes (CCs), which exhibit differences in the coding sequences of core virulence factors. Whether these alterations affect functionality is poorly understood. Here, we studied the highly polymorphic pore-forming toxin LukAB. We discovered that the LukAB toxin variants produced by S. aureus CC30 and CC45 kill human phagocytes regardless of whether CD11b, the previously established LukAB receptor, is present, and instead target the human hydrogen voltage-gated channel 1 (HVCN1). Biochemical studies identified the domain within human HVCN1 that drives LukAB species specificity, enabling the generation of humanized HVCN1 mice with enhanced susceptibility to CC30 LukAB and to bloodstream infection caused by CC30 S. aureus strains. Together, this work advances our understanding of an important S. aureus toxin and underscores the importance of considering genetic variation in characterizing virulence factors and understanding the tug of war between pathogens and the host.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Ion Channels/metabolism , Leukocidins/genetics , Leukocidins/metabolism , Staphylococcal Infections/metabolism , Staphylococcus aureus/metabolism , Animals , CD11b Antigen/genetics , CD11b Antigen/metabolism , Genetic Variation , Humans , Ion Channels/genetics , Mice, Inbred C57BL , Phagocytes/metabolism , Phagocytes/microbiology , Staphylococcal Infections/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics
16.
Int J Mol Sci ; 23(1)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-35008761

ABSTRACT

Staphylococcus aureus is a vital bovine mastitis pathogen causing huge economic losses to the dairy industry worldwide. In our previous studies, leukotoxin ED (LukED) was detected in most S. aureus strains isolated from bovine mastitis. Here, four single-chain fragment variables (scFvs) (ZL8 and ZL42 targeting LukE, ZL22 and ZL23 targeting LukD) were obtained using purified LukE and LukD proteins as the antigens after five rounds of bio-panning. The complementarity-determining region 3 (CDR3) of the VH domain of these scFvs exhibited significant diversities. In vitro, the scFvs significantly decreased LukED-induced cell killing by inhibiting the binding of LukED to chemokine receptors (CCR5 and CXCR2) and reduced the death rates of bovine neutrophils and MAC-T cells caused by LukED and S. aureus (p < 0.05). In an S. aureus-induced mouse mastitis model, histopathology and MPO results revealed that scFvs ameliorated the histopathological damages and reduced the infiltration of inflammatory cells (p < 0.05). The ELISA and qPCR assays showed that scFvs reduced the transcription and expression levels of Tumor Necrosis Factor-alpha (TNF-α), interleukin-1ß (IL-1ß), IL-6, IL-8 and IL-18 (p < 0.05). The overall results demonstrated the protective anti-inflammatory effect of scFvs in vitro and in vivo, enlightening the potential role of scFvs in the prevention and treatment of S. aureus-induced mastitis.


Subject(s)
Inflammation/pathology , Leukocidins/metabolism , Mastitis/microbiology , Single-Chain Antibodies/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Animals , Cattle , Cell Death , Cytokines/metabolism , Female , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Leukocidins/isolation & purification , Mammary Glands, Animal/pathology , Mice , Mice, Inbred BALB C , Neutrophils/metabolism , Peptide Library , Peroxidase/metabolism , Receptors, Chemokine/metabolism , Single-Chain Antibodies/isolation & purification , Staphylococcal Infections/pathology , Staphylococcus aureus/growth & development
17.
FASEB J ; 35(2): e21167, 2021 02.
Article in English | MEDLINE | ID: mdl-33241563

ABSTRACT

Panton-Valentine Leukocidin (PVL) is a bicomponent leukotoxin produced by 3%-10% of clinical Staphylococcus aureus (SA) strains involved in the severity of hospital and community-acquired infections. Although PVL was long known as a pore-forming toxin, recent studies have challenged the formation of a pore at the plasma membrane, while its endocytosis and the exact mode of action remain to be defined. In vitro immunolabeling of human neutrophils shows that Neutrophil Extracellular Traps (NETosis) is triggered by the action of purified PVL, but not by Gamma hemolysin CB (HlgCB), a structurally similar SA leukotoxin. PVL causes the ejection of chromatin fibers (NETs) decorated with antibacterial peptides independently of the NADPH oxidase oxidative burst. Leukotoxin partially colocalizes with mitochondria and enhances the production of reactive oxygen species from these organelles, while showing an increased autophagy, which results unnecessary for NETs ejection. PVL NETosis is elicited through Ca2+ -activated SK channels and Myeloperoxidase activity but is abolished by Allopurinol pretreatment of neutrophils. Moreover, massive citrullination of the histone H3 is performed by peptidyl arginine deiminases. Inhibition of this latter enzymes fails to abolish NET extrusion. Unexpectedly, PVL NETosis does not seem to involve Src kinases, which is the main kinase family activated downstream the binding of PVL F subunit to CD45 receptor, while the specific kinase pathway differs from the NADPH oxidase-dependent NETosis. PVL alone causes a different and specific form of NETosis that may rather represent a bacterial strategy conceived to disarm and disrupt the immune response, eventually allowing SA to spread.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Exotoxins/metabolism , Extracellular Traps/immunology , Leukocidins/metabolism , Mitochondria/metabolism , Neutrophils/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/metabolism , Adult , Cells, Cultured , Female , Healthy Volunteers , Hemolysin Proteins/metabolism , Humans , Male , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Respiratory Burst , Staphylococcal Infections/microbiology
18.
Microb Drug Resist ; 27(6): 823-834, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33232626

ABSTRACT

Bronchiectasis, which is an abnormal and irreversible dilation of one or several bronchial segments, causes significant morbidity and impaired quality of life to patients, mainly as the result of recurrent and chronic respiratory infections. Staphylococcus aureus is a microorganism known for its high infectious potential related to the production of molecules with great pathogenic power, such as enzymes, toxins, adhesins, and biofilm, which determine the degree of severity of systemic symptoms and can induce exacerbated immune response. This review highlighted the clinical significance of S. aureus colonization/infection in bronchiectasis patients, since little is known about it, despite its increasing frequency of isolation and potential serious morbidity.


Subject(s)
Bronchiectasis/complications , Staphylococcal Infections/etiology , Staphylococcus aureus/physiology , Adhesins, Escherichia coli/genetics , Adhesins, Escherichia coli/metabolism , Bacterial Toxins/metabolism , Biofilms/growth & development , Bronchiectasis/mortality , Exotoxins/metabolism , Humans , Leukocidins/metabolism , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/physiology , Microbiota/physiology , Prognosis , Staphylococcus aureus/genetics , Superantigens/immunology
19.
J Dairy Sci ; 104(1): 969-976, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33162097

ABSTRACT

We identified 199 Staphylococcus aureus isolates from quarter milk samples of 1,289 dairy cattle between 2014 and 2018. About 66% of the isolates were resistant to at least 1 antimicrobial agent; the highest rate of resistance was to penicillin, followed by resistance to ampicillin, erythromycin, and sulfadimethoxine. We obtained 30 methicillin-resistant S. aureus (MRSA) strains from 6 farms in 3 provinces. The MRSA strains exhibited a significantly higher resistance rate to most of the tested antimicrobials than the oxacillin-susceptible strains. The MRSA strains represented 5 genotypes: ST72-t324-SCCmec IV (n = 14), ST30-t1752-SCCmec IV (n = 8), ST188-t189-SCCmec NT (n = 6), ST188-t2284-SCCmec NT (n = 1), and NT-NT-SCCmec IV (n = 1). One of the ST188 MRSA strains represented a novel staphylococcal protein A (spa) type (t2284). In addition, 7 of the 8 ST30 MRSA strains were Panton-Valentine leukocidin (PVL)-positive and carried various staphylococcal enterotoxin encoding genes. This is the first report of PVL-positive ST30 MRSA-t1752-SCCmec IV from bovine mastitis in Korea. All of ST72-t324-SCCmec IV MRSA strains carried staphylococcal enterotoxin and leukotoxin encoding genes. They were also sensitive to most of the tested non-ß-lactam antimicrobials. In contrast, ST188-t189 MRSA strains were resistant to multiple antimicrobials and predominantly carried the leukotoxin encoding gene. Taken together, these findings may indicate that dairy cows could be a major source for spreading MRSA strains, and contaminated milk could be a vehicle for transmission. Suitable hygienic measures should be established in dairy farms and processing plants to limit the likelihood of introducing MRSA into the food chain.


Subject(s)
Bacterial Toxins/metabolism , Exotoxins/metabolism , Leukocidins/metabolism , Mastitis, Bovine/microbiology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Milk/microbiology , Staphylococcal Infections/veterinary , Animals , Cattle , Enterotoxins/genetics , Exotoxins/genetics , Female , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/metabolism , Republic of Korea , Staphylococcal Infections/microbiology
20.
Eur J Clin Microbiol Infect Dis ; 40(1): 77-83, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32767177

ABSTRACT

Panton-Valentine leukocidin (PVL) appears to be a virulence factor which, among others, can exacerbate the pathogenicity of Staphylococcus aureus infections, especially inducing severe necrotic, deep-seated skin infections, abscesses, and recurrences. These peculiarities have some overlaps with hidradenitis suppurativa (HS). Our main aim was to assess if S. aureus producing PVL could have some role in influencing clinical features and/or course of HS, specifically in the suppuration and recurrence of lesions. This pilot, mono-centric, observational study included all adult subjects affected with HS consecutively referring to our HS clinic over a 3-month period. Clinically evident suppuration and at least 2 weeks wash out from any antibiotic were the main inclusion criteria. Purulent material from HS skin lesions was collected with swabs in order to isolate micro-organisms, with specific regard to S. aureus. Detection of PVL was performed by real-time quantitative PCR (RT-qPCR). We also analyzed purulent material from suppurative skin lesions other than HS, as a control. Thirty HS patients were included; 29 purulent lesions (96.7%) harbored at least one bacterial species. Five (16.7%) swab samples were positive for S. aureus, none of which was positive for PVL genes. Among the 30 purulent disorders included as controls, 8 (26.3%) were positive for S. aureus; of these, 4 strains (50%) expressed LPV. The study results seem to exclude the pathogenetic involvement of S. aureus producing PVL in HS; as a result, PVL does not seem to represent a potential target in the future development of HS treatments.


Subject(s)
Bacterial Toxins/metabolism , Exotoxins/metabolism , Hidradenitis Suppurativa/microbiology , Leukocidins/metabolism , Staphylococcus aureus/metabolism , Adult , Female , Humans , Male , Pilot Projects , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...