Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
BMC Microbiol ; 22(1): 219, 2022 09 17.
Article in English | MEDLINE | ID: mdl-36115948

ABSTRACT

BACKGROUND: The prevalence of Staphylococcus aureus isolates carrying the Panton-Valentine leukocidin (PVL) gene is higher in Africa (≈50%) compared to Europe (< 5%). The study aimed to measure anti-PVL-antibodies in Africans and Germans in a multi-center study and to test whether detected antibodies can neutralize the cytotoxic effect of PVL on polymorphonuclear leukocytes (PMNs). METHODS: Sera from asymptomatic Africans (n = 22, Nigeria, Gabon) and Caucasians (n = 22, Germany) were used to quantify antibody titers against PVL and α-hemolysin (in arbitrary units [AU]) by ELISA. PMNs from one African and German donor were exposed to 5 nM recombinant PVL to measure the neutralizing effect of serial dilutions of pooled sera from African and Caucasian participants, or donor sera at 0.625 and 2.5% (v/v). RESULTS: Anti-PVL-antibodies were significantly higher in Africans than in Germans (1.9 vs. 0.7 AU, p < 0.0001). The pooled sera from the study participants neutralized the cytotoxic effect of PVL on African and German PMNs in a dose dependent manner. Also, neutralization of PVL on PMNs from the African and German donors had a stronger effect with African sera (half-maximal inhibitory concentration (IC50) = 0.27 and 0.47%, respectively) compared to Caucasian sera (IC50 = 3.51 and 3.59% respectively). CONCLUSION: Africans have higher levels of neutralizing anti-PVL-antibodies. It remains unclear if or at what level these antibodies protect against PVL-related diseases.


Subject(s)
Antibodies, Neutralizing/blood , Leukocidins , Neutrophils , Staphylococcal Infections , Staphylococcus aureus , Antibodies, Neutralizing/immunology , Bacterial Toxins/blood , Bacterial Toxins/immunology , Exotoxins/blood , Exotoxins/immunology , Germany/epidemiology , Hemolysin Proteins , Humans , Leukocidins/blood , Leukocidins/immunology , Neutrophils/immunology , Nigeria/epidemiology , Staphylococcal Infections/blood , Staphylococcal Infections/epidemiology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity
2.
J Infect Dis ; 225(1): 177-185, 2022 01 05.
Article in English | MEDLINE | ID: mdl-34145461

ABSTRACT

BACKGROUND: Staphylococcus aureus infections are common throughout the lifespan, with recurrent infections occurring in nearly half of infected children. There is no licensed vaccine, underscoring the need to better understand how S. aureus evades protective immunity. Despite much study, the relative contributions of antibodies and T cells to protection against S. aureus infections in humans are not fully understood. METHODS: We prospectively quantified S. aureus-specific antibody levels by ELISA and T-cell responses by ELISpot in S. aureus-infected and healthy children. RESULTS: S. aureus-specific antibody levels and T-cell responses increased with age in healthy children, suggesting a coordinated development of anti-staphylococcal immunity. Antibody levels against leukotoxin E (LukE) and Panton-Valentine leukocidin (LukS-PV), but not α-hemolysin (Hla), were higher in younger infected children, compared with healthy children; these differences disappeared in older children. We observed a striking impairment of global and S. aureus-specific T-cell function in children with invasive and noninvasive infection, suggesting that S. aureus-specific immune responses are dysregulated during childhood infection regardless of the infection phenotype. CONCLUSIONS: These findings identify a potential mechanism by which S. aureus infection actively evades adaptive immune responses, thereby preventing the development of protective immunity and maintaining susceptibility to recurrent infection.


Subject(s)
Antibodies, Bacterial/blood , Exotoxins/immunology , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections/epidemiology , Staphylococcal Infections/immunology , Staphylococcus aureus , Adolescent , Bacterial Toxins , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Hemolysin Proteins/immunology , Humans , Infant , Male , Prospective Studies , Seroepidemiologic Studies , T-Lymphocytes , Young Adult
3.
Infect Immun ; 89(10): e0034221, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34227839

ABSTRACT

In the Gram-positive pathogen Staphylococcus aureus, pore-forming toxins (PFTs), such as leukocidins and hemolysins, play prominent roles in staphylococcal pathogenesis by killing host immune cells and red blood cells (RBCs). However, it remains unknown which combination of toxin antigens would induce the broadest protective immune response against those toxins. In this study, by targeting six major staphylococcal PFTs (i.e., gamma-hemolysin AB [HlgAB], gamma-hemolysin CB [HlgCB], leukocidin AB [LukAB], leukocidin ED [LukED], Panton-Valentine leukocidin [LukSF-PV], and alpha-hemolysin [Hla]), we generated 10 recombinant toxins or toxin subunits, 3 toxoids, and their rabbit antibodies. Using the cytolytic assay for RBCs and polymorphonuclear cells (PMNs), we determined the best combination of toxin antibodies conferring the broadest protection against those staphylococcal PFTs. Although anti-HlgA IgG (HlgA-IgG) showed low cross-reactivity to other toxin components, it was essential to protect rabbit and human RBCs and human PMNs. For the protection of rabbit RBCs, HlaH35L toxoid-IgG was also required, whereas for human PMNs, LukS-IgG and LukAE323AB-IgG were essential too. When the toxin/toxoid antigens HlgA, LukS-PV, HlaH35L, and LukAE323AB were used to immunize rabbits, they increased rabbit survival; however, they did not block staphylococcal abscess formation in kidneys. Based on these results, we proposed that the combination of HlgA, LukS, HlaH35L, and LukAE323AB is the optimal vaccine component to protect human RBCs and PMNs from staphylococcal PFTs. We also concluded that a successful S. aureus vaccine requires not only those toxin antigens but also other antigens that can induce immune responses blocking staphylococcal colonization.


Subject(s)
Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Vaccines, Combined/immunology , Animals , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Cross Reactions/immunology , Erythrocytes/immunology , Erythrocytes/microbiology , Exotoxins/immunology , Hemolysin Proteins/immunology , Humans , Immunization/methods , Leukocidins/immunology , Neutrophils/immunology , Neutrophils/microbiology , Rabbits , Staphylococcal Infections/microbiology , Toxoids/immunology
4.
Vet Immunol Immunopathol ; 230: 110149, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33197719

ABSTRACT

Staphylococcus aureus is the leading cause of clinical mastitis and is associated with persistent subclinical infections in ewes, significantly compromising the quality and quantity of milk productions. To date, vaccines intended for use in sheep have been mainly focused on biofilm production traits, but many S. aureus pathogenic isolates do not produce biofilm, including those circulating in Sardinia, one of the leading sheep milk producers in Europe. The aim of this work was to identify suitable immunodominant, alternative candidates to biofilm components for vaccine and diagnostic development. An immunoproteomics study was carried out by testing sera from naturally infected sheep with a prevalent S. aureus lineage against cellular and secreted antigens, followed by tandem mass spectrometry identification of the most prominent immunogens. Four cellular and three secreted S. aureus antigens elicited a strong humoral host immune response. The four cellular antigens were the housekeeping proteins pyruvate kinase, elongation Factor Tu, dihydrolipoyl dehydrogenase, and alpha-keto acid dehydrogenase. The three secreted antigens were the bifunctional autolysin (Atl) and the two components of the Panton-Valentine leukocidin, lukF-PV/lukM, demonstrating the carriage of prophage phiPV83 in a sheep isolate and the strong response of the sheep host against them. In consideration of the key role played by these secreted proteins in S. aureus replication and immune evasion, these antigens may represent suitable candidates for developing vaccines eliciting a more successful immunological protection in areas where non-biofilm forming Staphylococcus spp. are the most widespread intramammary pathogens.


Subject(s)
Antigens, Bacterial/blood , Antigens, Bacterial/immunology , Mastitis, Bovine/microbiology , Sheep Diseases/microbiology , Staphylococcal Infections/veterinary , Staphylococcus aureus/chemistry , Staphylococcus aureus/immunology , Animals , Antigens, Bacterial/administration & dosage , Bacterial Toxins/immunology , Cattle , Cattle Diseases/immunology , Cattle Diseases/microbiology , Cattle Diseases/prevention & control , Exotoxins/immunology , Female , Immunity, Humoral , Leukocidins/immunology , Mastitis, Bovine/prevention & control , Proteomics/methods , Sheep , Sheep Diseases/immunology , Sheep Diseases/prevention & control , Staphylococcal Infections/immunology , Staphylococcal Vaccines , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Tandem Mass Spectrometry
5.
Infect Immun ; 88(10)2020 09 18.
Article in English | MEDLINE | ID: mdl-32719153

ABSTRACT

Bacterial biofilms are linked with chronic infections and have properties distinct from those of planktonic, single-celled bacteria. The virulence mechanisms associated with Staphylococcus aureus biofilms are becoming better understood. Human neutrophils are critical for the innate immune response to S. aureus infection. Here, we describe two virulence strategies that converge to promote the ability of S. aureus biofilms to evade killing by neutrophils. Specifically, we show that while neutrophils exposed to S. aureus biofilms produce extracellular traps (NETs) and phagocytose bacteria, both mechanisms are inefficient in clearance of the biofilm biomass. This is attributed to the leukocidin LukAB, which promotes S. aureus survival during phagocytosis. We also show that the persistence of biofilm bacteria trapped in NETs is facilitated by S. aureus nuclease (Nuc)-mediated degradation of NET DNA. This study describes key aspects of the interaction between primary human neutrophils and S. aureus biofilms and provides insight into how S. aureus evades the neutrophil response to cause persistent infections.


Subject(s)
Bacterial Proteins/immunology , Biofilms , Immune Evasion , Leukocidins/immunology , Micrococcal Nuclease/immunology , Neutrophils/immunology , Staphylococcus aureus/pathogenicity , Bacterial Proteins/genetics , Biofilms/growth & development , Extracellular Traps/immunology , Extracellular Traps/metabolism , Extracellular Traps/microbiology , Humans , Leukocidins/genetics , Microbial Viability , Micrococcal Nuclease/genetics , Neutrophils/microbiology , Neutrophils/pathology , Phagocytosis , Staphylococcus aureus/immunology , Virulence
6.
mBio ; 11(3)2020 06 16.
Article in English | MEDLINE | ID: mdl-32546616

ABSTRACT

Staphylococcus aureus infection is a major public health threat in part due to the spread of antibiotic resistance and repeated failures to develop a protective vaccine. Infection is associated with production of virulence factors that include exotoxins that attack host barriers and cellular defenses, such as the leukocidin (Luk) family of bicomponent pore-forming toxins. To investigate the structural basis of antibody-mediated functional inactivation of Luk toxins, we generated a panel of murine monoclonal antibodies (MAbs) that neutralize host cell killing by the γ-hemolysin HlgCB. By biopanning these MAbs against a phage-display library of random Luk peptide fragments, we identified a small subregion within the rim domain of HlgC as the epitope for all the MAbs. Within the native holotoxin, this subregion folds into a conserved ß-hairpin structure, with exposed key residues, His252 and Tyr253, required for antibody binding. On the basis of the phage-display results and molecular modeling, a 15-amino-acid synthetic peptide representing the minimal epitope on HlgC (HlgC241-255) was designed, and preincubation with this peptide blocked antibody-mediated HIgCB neutralization. Immunization of mice with HlgC241-255 or the homologous LukS246-260 subregion peptide elicited serum antibodies that specifically recognized the native holotoxin subunits. Furthermore, serum IgG from patients who were convalescent for invasive S. aureus infection showed neutralization of HlgCB toxin activity ex vivo, which recognized the immunodominant HlgC241-255 peptide and was dependent on His252 and Tyr253 residues. We have thus validated an efficient, rapid, and scalable experimental workflow for identification of immunodominant and immunogenic leukotoxin-neutralizing B-cell epitopes that can be exploited for new S. aureus-protective vaccines and immunotherapies.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Epitopes, B-Lymphocyte/immunology , Exotoxins/immunology , Hemolysin Proteins/immunology , Immunodominant Epitopes/immunology , Animals , Antibodies, Bacterial/blood , Epitope Mapping , Female , Humans , Immunodominant Epitopes/genetics , Immunoglobulin G/blood , Immunoglobulin G/immunology , Leukocidins/chemistry , Leukocidins/immunology , Mice , Mice, Inbred BALB C , Peptide Library , Staphylococcal Infections/blood , Staphylococcal Infections/immunology , Staphylococcus aureus , Virulence Factors
7.
J Infect Dis ; 221(2): 267-275, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31504652

ABSTRACT

Staphylococcus aureus is a common pathogen causing infections in humans with various degrees of severity, with pneumonia being one of the most severe infections. In as much as staphylococcal pneumonia is a disease driven in large part by α-hemolysin (Hla) and Panton-Valentine leukocidin (PVL), we evaluated whether active immunization with attenuated forms of Hla (HlaH35L/H48L) alone, PVL components (LukS-PVT28F/K97A/S209A and LukF-PVK102A) alone, or combination of all 3 toxoids could prevent lethal challenge in a rabbit model of necrotizing pneumonia caused by the USA300 community-associated methicillin-resistant S. aureus (MRSA). Rabbits vaccinated with Hla toxoid alone or PVL components alone were only partially protected against lethal pneumonia, whereas those vaccinated with all 3 toxoids had 100% protection against lethality. Vaccine-mediated protection correlated with induction of polyclonal antibody response that neutralized not only α-hemolysin and PVL, but also other related toxins, produced by USA300 and other epidemic MRSA clones.


Subject(s)
Bacterial Toxins/immunology , Exotoxins/immunology , Hemolysin Proteins/immunology , Leukocidins/immunology , Pneumonia, Necrotizing/prevention & control , Pneumonia, Staphylococcal/prevention & control , Animals , Bacterial Toxins/administration & dosage , Disease Models, Animal , Exotoxins/administration & dosage , Hemolysin Proteins/administration & dosage , Humans , Leukocidins/administration & dosage , Methicillin-Resistant Staphylococcus aureus , Pneumonia, Necrotizing/immunology , Pneumonia, Staphylococcal/immunology , Rabbits , Vaccination
8.
Proc Natl Acad Sci U S A ; 117(1): 317-327, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31852826

ABSTRACT

Host-pathogen interactions are central to understanding microbial pathogenesis. The staphylococcal pore-forming cytotoxins hijack important immune molecules but little is known about the underlying molecular mechanisms of cytotoxin-receptor interaction and host specificity. Here we report the structures of a staphylococcal pore-forming cytotoxin, leukocidin GH (LukGH), in complex with its receptor (the α-I domain of complement receptor 3, CD11b-I), both for the human and murine homologs. We observe 2 binding interfaces, on the LukG and the LukH protomers, and show that human CD11b-I induces LukGH oligomerization in solution. LukGH binds murine CD11b-I weakly and is inactive toward murine neutrophils. Using a LukGH variant engineered to bind mouse CD11b-I, we demonstrate that cytolytic activity does not only require binding but also receptor-dependent oligomerization. Our studies provide an unprecedented insight into bicomponent leukocidin-host receptor interaction, enabling the development of antitoxin approaches and improved animal models to explore these approaches.


Subject(s)
Bacterial Proteins/metabolism , CD11b Antigen/metabolism , Leukocidins/metabolism , Macrophage-1 Antigen/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Animals , Bacterial Proteins/immunology , Bacterial Proteins/ultrastructure , CD11b Antigen/immunology , CD11b Antigen/ultrastructure , Cell Line , Cell Membrane/metabolism , Crystallography, X-Ray , Humans , Leukocidins/immunology , Macrophage-1 Antigen/immunology , Macrophage-1 Antigen/ultrastructure , Mice , Models, Molecular , Neutrophils/cytology , Neutrophils/immunology , Neutrophils/metabolism , Protein Domains/immunology , Protein Multimerization/immunology , Rabbits , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure , Species Specificity , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity
9.
Toxins (Basel) ; 11(6)2019 06 14.
Article in English | MEDLINE | ID: mdl-31207937

ABSTRACT

Staphylococcus aureus (SA) infections cause high mortality and morbidity in humans. Being central to its pathogenesis, S. aureus thwarts the host defense by secreting a myriad of virulence factors, including bicomponent, pore-forming leukotoxins. While all vaccine development efforts that aimed at achieving opsonophagocytic killing have failed, targeting virulence by toxoid vaccines represents a novel approach to preventing mortality and morbidity that are caused by SA. The recently discovered leukotoxin LukAB kills human phagocytes and monocytes and it is present in all known S. aureus clinical isolates. While using a structure-guided approach, we generated a library of mutations that targeted functional domains within the LukAB heterodimer to identify attenuated toxoids as potential vaccine candidates. The mutants were evaluated based on expression, solubility, yield, biophysical properties, cytotoxicity, and immunogenicity, and several fully attenuated LukAB toxoids that were capable of eliciting high neutralizing antibody titers were identified. Rabbit polyclonal antibodies against the lead toxoid candidate provided potent neutralization of LukAB. While the neutralization of LukAB alone was not sufficient to fully suppress leukotoxicity in supernatants of S. aureus USA300 isolates, a combination of antibodies against LukAB, α-toxin, and Panton-Valentine leukocidin completely neutralized the cytotoxicity of these strains. These data strongly support the inclusion of LukAB toxoids in a multivalent toxoid vaccine for the prevention of S. aureus disease.


Subject(s)
Bacterial Proteins/immunology , Bacterial Vaccines , Leukocidins/immunology , Staphylococcal Infections/prevention & control , Toxoids/immunology , Animals , Bacterial Proteins/genetics , Cell Survival , Escherichia coli/genetics , Female , HL-60 Cells , Humans , Leukocidins/genetics , Mice, Inbred ICR , Monocytes , THP-1 Cells , Toxoids/genetics
10.
Article in English | MEDLINE | ID: mdl-31138566

ABSTRACT

Surgical site infections (SSIs) are commonly caused by Staphylococcus aureus We report that a combination of three monoclonal antibodies (MEDI6389) that neutralize S. aureus alpha-toxin, clumping factor A, and four leukocidins (LukSF, LukED, HlgAB, and HlgCB) plus vancomycin had enhanced efficacy compared with control antibody plus vancomycin in two mouse models of S. aureus SSI. Therefore, monoclonal antibody-based neutralization of multiple S. aureus virulence factors may provide an adjunctive perioperative approach to combat S. aureus SSIs.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Staphylococcal Infections/drug therapy , Surgical Wound Infection/drug therapy , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Bacterial Proteins/immunology , Broadly Neutralizing Antibodies/pharmacology , Coagulase/immunology , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice, Inbred C57BL , Mice, Inbred Strains , Staphylococcal Infections/microbiology , Surgical Wound Infection/microbiology , Vancomycin/pharmacology
11.
Article in English | MEDLINE | ID: mdl-31138568

ABSTRACT

ASN100 is a novel antibody combination of two fully human IgG1(κ) monoclonal antibodies (MAbs), ASN-1 and ASN-2, which neutralize six Staphylococcus aureus cytotoxins, alpha-hemolysin (Hla) and five bicomponent leukocidins. We assessed the safety, tolerability, and serum and lung pharmacokinetics of ASN100 in a randomized, double-blind, placebo-controlled single-dose-escalation first-in-human study. Fifty-two healthy volunteers were enrolled and randomized to receive either ASN-1, ASN-2, a combination of both MAbs (ASN100), or a corresponding placebo. Thirty-two subjects in the double-blind dose escalation portion of the study received ASN-1 or ASN-2 at a 200-, 600-, 1,800-, or 4,000-mg dose, or placebo. Eight subjects received both MAbs simultaneously in a 1:1 ratio (ASN100) at 3,600 or 8,000 mg, or they received placebos. Twelve additional subjects received open-label ASN100 at 3,600 or 8,000 mg to assess the pharmacokinetics of ASN-1 and ASN-2 in epithelial lining fluid (ELF) by bronchoalveolar lavage fluid sampling. Subjects were monitored for 98 days (double-blind cohorts) or 30 days (open-label cohorts) for safety assessment. No dose-limiting toxicities were observed, and all adverse events were mild and transient, with only two adverse events considered possibly related to the investigational product. ASN100 exhibited linear serum pharmacokinetics with a half-life of approximately 3 weeks and showed detectable penetration into the ELF. No treatment-emergent anti-drug antibody responses were detected. The toxin neutralizing potency of ASN100 in human serum was confirmed up to 58 days postdosing. The favorable safety profile, ELF penetration, and maintained functional activity in serum supported the further clinical development of ASN100.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacokinetics , Bacterial Toxins/antagonists & inhibitors , Cytotoxins/immunology , Adult , Anti-Bacterial Agents/pharmacokinetics , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Bronchoalveolar Lavage Fluid , Cytotoxins/antagonists & inhibitors , Cytotoxins/metabolism , Double-Blind Method , Female , Healthy Volunteers , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/immunology , Humans , Leukocidins/antagonists & inhibitors , Leukocidins/immunology , Male , Placebos , Staphylococcal Infections , Staphylococcus aureus/immunology
12.
Clin Lab ; 65(1)2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30775902

ABSTRACT

BACKGROUND: We aimed to investigate the frequency of fibronectin binding protein (FBP), which is part of the first step of adhesion, and Panton-Valentine leukocidin (PVL) toxin, which contributes to the destruction of host leukocytes and tissue necrosis, in clinical S. aureus strains. METHODS: One hundred S. aureus strains were included in the study and distributed as follows; 33 from skinwound swabs and catheter tips (SWCT), 33 from body fluid and secretion specimens (BSFS) such as tracheal aspirate, sputum, and pleural effusion fluid, 18 from tissue biopsy specimens (TBS), 10 specimens from blood, and related specimens (BRS) such as bone marrow, and cerebral spinal fluid, and six specimens from mucosal membrane of pharynx, nose, and vagina (MMS). Methicillin resistance was tested by disk diffusion method. mecA (methicillin resistance coded gene), pvl and fnbA genes were investigated by using a PCR method. RESULTS: Thirty-seven strains (37.0%) were identified as methicillin resistant S. aureus (MRSA) and 63 (63.0%) as methicillin susceptible S. aureus (MSSA) strains. fnbA was more frequent in S. aureus isolates of MMSs (100.0%); followed by BRSs (80.0%), SWCTs (78.8%), TBS (72.3%), and BSFs (66.7%), whereas pvl gene was more frequent in isolates of BRS (60.0%), followed by TBSs (50.0%), SWCTs (33.4%), BSFs (30.3%), and MMSs (16.7%). fnbA existed in 85.7% of MSSA and 56.8% of MRSA in contrast to pvl, which was more frequent in MRSA (70.3%) than those of MSSA strains (17.4%). These differences were statistically significant (p < 0.05). CONCLUSIONS: Our different clinical specimens contained a high rate of fnbA (75.0%) and low-moderate frequency of pvl (37.0%). fnbA was most frequent in S. aureus of MMSs, followed by BRSs, and SWCTs, whereas pvl was ex-isted in high proportion in S. aureus of BRSs, followed by TBSs, and SWCTs. Presence of PVL in a high proportion in MRSA strains of superfical specimens such SWCT (24.4%) and deeper serious specimens such as BRS (16.3%) compared to MSSA strains from the same specimens, 3.2% and 0%, respectively, have shown that MRSA infections still threatens patients' lives and control of their spread is urgently needed.


Subject(s)
Adhesins, Bacterial/immunology , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Exotoxins/immunology , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Infections/immunology , Virulence Factors/immunology , Adhesins, Bacterial/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Drug Resistance, Microbial/drug effects , Drug Resistance, Microbial/genetics , Drug Resistance, Microbial/immunology , Exotoxins/genetics , Exotoxins/metabolism , Humans , Leukocidins/genetics , Leukocidins/metabolism , Methicillin/pharmacology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/immunology , Penicillin-Binding Proteins/metabolism , Staphylococcal Infections/microbiology , Virulence Factors/genetics , Virulence Factors/metabolism
13.
Anaerobe ; 56: 78-87, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30771460

ABSTRACT

Clostridium chauvoei is the etiologic agent of blackleg in cattle, inducing fever, severe myonecrosis, oedemic lesions and ultimately death of infected animals. The pathogen often results in such rapid death that antibiotic therapy is futile and thus vaccination is the only efficient strategy in order to control the disease. The ß-barrel pore forming leucocidin Clostridium chauvoei toxin A (CctA) is one of the best characterised toxins of C. chauvoei and has been shown to be an important virulence factor. It has been reported to induce protective immunity and is conserved across C. chauvoei strains collected from diverse geographical locations for more than 50 years. The aim of this study was to identify the location of the CctA toxin during liquid culture fermentation and to use CctA to develop an in vitro assay to replace the current guinea pig challenge assay for vaccine potency in standard batch release procedures. We report that CctA is fully secreted in C. chauvoei culture and show that it is found abundantly in the supernatant of liquid cultures. Sera from cattle vaccinated with a commercial blackleg vaccine revealed strong haemolysin-neutralizing activity against recombinant CctA which reached titres of 1000 times 28 days post-vaccination. Similarly, guinea pig sera from an official potency control test reached titres of 600 times 14 days post-vaccination. In contrast, ELISA was not able to specifically measure anti-CctA antibodies in cattle serum due to strong cross-reactions with antibodies against other proteins present pre-vaccination. We conclude that haemolysin-neutralizing antibodies are a valuable measurement for protective immunity against blackleg and have the potential to be a suitable replacement of the guinea pig challenge potency test, which would forego the unnecessary challenge of laboratory animals.


Subject(s)
Antibodies, Bacterial/blood , Antibodies, Neutralizing/blood , Bacterial Toxins/immunology , Bacterial Vaccines/immunology , Cattle Diseases/prevention & control , Clostridium Infections/veterinary , Clostridium chauvoei/immunology , Animals , Bacterial Toxins/metabolism , Bacterial Vaccines/administration & dosage , Cattle , Clostridium Infections/prevention & control , Clostridium chauvoei/metabolism , Culture Media/chemistry , Enzyme-Linked Immunosorbent Assay , Guinea Pigs , Leukocidins/immunology , Leukocidins/metabolism , Neutralization Tests , Virulence Factors/immunology
14.
J Pediatric Infect Dis Soc ; 8(2): 128-135, 2019 May 11.
Article in English | MEDLINE | ID: mdl-29538707

ABSTRACT

BACKGROUND: Staphylococcus aureus is among the most commonly identified causes of invasive bacterial infection in children; however, reliable results from cultures of sterile-site samples often cannot be obtained, which necessitates prescription of a broad empiric antimicrobial agent(s). Children with invasive S aureus infection rapidly generate high antibody titers to the cytotoxin LukAB; therefore, the aim of this study was to assess the diagnostic utility of an anti-LukAB antibody assay for children with musculoskeletal infection (MSKI). METHODS: We conducted a 2-year prospective study of all eligible children admitted to Vanderbilt Children's Hospital with an MSKI. Acute and convalescent sera were obtained, and antibodies that target LukAB were measured by an enzyme-linked immunosorbent assay. RESULTS: Forty-two children were enrolled. The median concentrations of LukAB antibodies for children with S aureus infection were 130.3 U/mL in the acute phase and 455 U/mL in the convalescent phase (P < .001). The median concentrations of LukAB antibodies in children with a non-S aureus MSKI were 8.6 U/mL in the acute phase and 9.7 U/mL in the convalescent phase. The assay discriminated between S aureus and non-S aureus infection with areas under the receiver operating characteristic curve of 0.81 (95% confidence interval, 0.67-0.95; P < .001) and 0.95 (95% confidence interval, 0.86-1; P < .001) for samples tested in the acute and follow-up periods, respectively. With no false-negative results, the assay accurately ruled out S aureus in samples obtained during the convalescent phase. CONCLUSION: Culture-independent diagnostics have the potential to improve care by narrowing antimicrobial therapy on the basis of the likelihood of S aureus infection. The results of this proof-of-concept study suggest that a LukAB serologic assay might be useful in the diagnosis of invasive bacterial infections, and larger-scale validation studies are warranted.


Subject(s)
Antibodies, Bacterial/blood , Bacterial Proteins/immunology , Leukocidins/immunology , Staphylococcal Infections/diagnosis , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Child , Child, Preschool , Enzyme-Linked Immunosorbent Assay/methods , Female , Histocompatibility Antigens Class II , Hospitals, Pediatric , Humans , Immunoglobulin G/blood , Infant , Male , Musculoskeletal Diseases/complications , Musculoskeletal Diseases/microbiology , Prospective Studies , Staphylococcal Infections/microbiology , United States
15.
BMC Microbiol ; 18(1): 181, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30419818

ABSTRACT

BACKGROUND: Staphylococcus aureus is a leading cause of Gram-positive bacterial infections worldwide; however, the treatment of S. aureus infection has become increasingly difficult due to the prevalence of methicillin-resistant S. aureus strains, highlighting the urgent need for the development of novel strategies. The complexity of S. aureus pathogenesis relies on virulence factors. Recent studies have demonstrated that leukocidins expressed by the majority of clinical isolates play important roles in the pathogenesis of S. aureus. RESULTS: In this study, we developed three human monoclonal antibodies against all F-components of leukocidins HlgABC, LukSF, and LukED with high affinity. These antibodies were found to be capable of blocking leukocidin-mediated cell lysis in vitro. Furthermore, the antibodies dramatically reduced disease progression and mortality after S. aureus infection in vivo. CONCLUSIONS: Our findings revealed that neutralizing bicomponent leukocidins may be a promising strategy to combat infections caused by S. aureus.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Hemolysin Proteins/immunology , Leukocidins/immunology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/mortality , Staphylococcus aureus/physiology , Animals , Antibodies, Monoclonal/immunology , Disease Progression , Erythrocytes/drug effects , Female , Hemolysin Proteins/toxicity , Hemolysis/drug effects , Humans , Leukocidins/toxicity , Mice, Inbred BALB C , Neutralization Tests , Rabbits , Staphylococcal Infections/immunology , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics
16.
PLoS One ; 13(9): e0204450, 2018.
Article in English | MEDLINE | ID: mdl-30261001

ABSTRACT

Bacterial infections from Staphylococcus pseudintermedius are the most common cause of skin infections (pyoderma) affecting dogs. Two component pore-forming leukocidins are a family of potent toxins secreted by staphylococci and consist of S (slow) and F (fast) components. They impair the innate immune system, the first line of defense against these pathogens. Seven different leukocidins have been characterized in Staphylococcus aureus, some of which are host and cell specific. Through genome sequencing and analysis of the S. pseudintermedius secretome using liquid chromatography mass spectrometry we identified two proteins, named "LukS-I" and "LukF-I", encoded on a degenerate prophage contained in the genome of S. pseudintermedius isolates. Phylogenetic analysis of LukS-I components in comparison to the rest of the leukocidin family showed that LukS-I was most closely related to S. intermedius LukS-I, S. aureus LukE and LukP, whereas LukF-I was most similar to S. intermedius LukF-I S. aureus gamma hemolysin subunit B. The killing effect of recombinant S. pseudintermedius LukS-I and LukF-I on canine polymorphonuclear leukocytes was determined using a flow cytometry cell permeability assay. The cytotoxic effect occurred only when the two recombinant proteins were combined. Engineered mutant versions of the two-component pore-forming leukocidins, produced through amino acids substitutions at selected points, were not cytotoxic. Anti-Luk-I produced in dogs against attenuated proteins reduced the cytotoxic effect of native canine leukotoxin which highlights the importance of Luk-I as a promising component in a vaccine against canine S. pseudintermedius infections.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Leukocidins , Staphylococcus/genetics , Staphylococcus/metabolism , Amino Acid Sequence , Animals , Antibodies, Bacterial/immunology , Antibodies, Bacterial/metabolism , Bacterial Proteins/chemistry , Cell Death , Dog Diseases/immunology , Dogs , Escherichia coli , Exotoxins/genetics , Exotoxins/metabolism , Genome, Bacterial , Leukocidins/chemistry , Leukocidins/genetics , Leukocidins/immunology , Leukocidins/metabolism , Leukocytes/metabolism , Leukocytes/microbiology , Mutation , Phylogeny , Recombinant Proteins/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/veterinary , Staphylococcus/immunology
17.
Proc Natl Acad Sci U S A ; 115(28): 7416-7421, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29941565

ABSTRACT

Bacterial biofilms efficiently evade immune defenses, greatly complicating the prognosis of chronic infections. How methicillin-resistant Staphylococcus aureus (MRSA) biofilms evade host immune defenses is largely unknown. This study describes some of the major mechanisms required for S. aureus biofilms to evade the innate immune response and provides evidence of key virulence factors required for survival and persistence of bacteria during chronic infections. Neutrophils are the most abundant white blood cells in circulation, playing crucial roles in the control and elimination of bacterial pathogens. Specifically, here we show that, unlike single-celled populations, S. aureus biofilms rapidly skew neutrophils toward neutrophil extracellular trap (NET) formation through the combined activity of leukocidins Panton-Valentine leukocidin and γ-hemolysin AB. By eliciting this response, S. aureus was able to persist, as the antimicrobial activity of released NETs was ineffective at clearing biofilm bacteria. Indeed, these studies suggest that NETs could inadvertently potentiate biofilm infections. Last, chronic infection in a porcine burn wound model clearly demonstrated that leukocidins are required for "NETosis" and facilitate bacterial survival in vivo.


Subject(s)
Bacterial Proteins/immunology , Biofilms , Extracellular Traps/immunology , Immune Evasion , Leukocidins/immunology , Neutrophils/immunology , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/physiology , Wound Infection/immunology , Animals , Extracellular Traps/microbiology , Humans , Staphylococcal Skin Infections/pathology , Swine , Wound Infection/microbiology , Wound Infection/pathology
18.
J Infect Chemother ; 24(5): 383-388, 2018 May.
Article in English | MEDLINE | ID: mdl-29409694

ABSTRACT

Community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) infects healthy individuals, although the precise cause remains unclear. CA-MRSA produces Panton-Valentine leukocidin (PVL), which often causes severe invasive infection; however, antitoxin drugs against PVL are limited. Intravenous immunoglobulin (IVIg) possesses antitoxin activity, but unfortunately, the optimal dose is unknown. Here, we measured the PVL neutralizing antibody titer in the plasma of Japanese individuals and sera of American donors. Next, we compared the cytotoxic effects of PVL on neutrophils in phosphate buffered saline (PBS) or whole blood to determine the effect of the neutralizing antibody. Finally, we evaluated the effective concentration of IVIg required to neutralize PVL in PBS and whole blood. We observed that the titer of PVL neutralizing antibody in healthy individuals polarized as high and low/none group. Additionally, the PVL neutralizing antibody titer considerably affected the concentration at which IVIg elicited its effect. This suggests that PVL-producing CA-MRSA might be involved in determining the severity of infection in healthy individuals without neutralizing antibody against PVL. The neutralizing effect of IVIg was observed in both PBS and whole blood. However, the optimal concentration of IVIg required for neutralizing PVL varied between PBS and whole blood. In addition, since the PVL-neutralizing activity of IVIg also largely depends on blood composition, such as neutralizing antibody concentration, the optimal dosage of IVIg as an antitoxin drug should be decided in a timely manner after considering the patient's medical background.


Subject(s)
Antibodies, Neutralizing/blood , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/blood , Community-Acquired Infections/drug therapy , Exotoxins/antagonists & inhibitors , Exotoxins/blood , Immunoglobulins, Intravenous/administration & dosage , Leukocidins/antagonists & inhibitors , Leukocidins/blood , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Infections/drug therapy , Antibodies, Neutralizing/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Buffers , Community-Acquired Infections/immunology , Exotoxins/immunology , Humans , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Neutrophils/drug effects , Neutrophils/immunology , Staphylococcal Infections/immunology
19.
Article in English | MEDLINE | ID: mdl-28874371

ABSTRACT

The pathogenesis of Staphylococcus aureus is mediated by an array of important virulence factors, including the two-component leukocidin family of toxins. LukAB (also known as LukGH), the most recently discovered leukocidin, is potently lethal to phagocytes, produced during invasive human disease, and present in all known clinical isolates of S. aureus Intravenous immunoglobulin (IVIg) is often used clinically in severe S. aureus infections. The primary aim of this study was to assess the binding and neutralization potential of IVIg against LukAB. A secondary aim was to examine the lot-to-lot variability of IVIg in the binding and neutralization of LukAB. We studied 24 distinct lots of IVIg and compared them to serum from children with invasive S. aureus infection (in the acute and convalescent phases) and from healthy, uninfected controls. We found that all lots of IVIg contained functional antibodies targeting LukAB. After adjusting for total antibody content per sample, we found that the amount of anti-LukAB antibody in IVIg was similar to that seen with healthy controls and less than that seen with patients with invasive S. aureus infection. IVIg samples had lower neutralization capacity than samples from healthy controls and children with invasive infection. IVIg had remarkably little lot-to-lot variation in LukAB binding but had significantly more variation in toxin neutralization. These results represent the first report of functional antibodies against the important S. aureus leukocidin LukAB in IVIg. Given the frequent clinical use of IVIg for severe S. aureus infections, improving our understanding of functional antibody properties exhibited by this therapeutic is essential.


Subject(s)
Antibodies, Neutralizing/immunology , Bacterial Proteins/immunology , Immunoglobulins, Intravenous/immunology , Leukocidins/immunology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Antibodies, Neutralizing/blood , Antibody Affinity/immunology , Child , Child, Preschool , Humans , Staphylococcal Infections/immunology , Staphylococcal Infections/therapy , Staphylococcus aureus/immunology , Virulence Factors/immunology
20.
J Allergy Clin Immunol ; 140(4): 1054-1067.e10, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28216434

ABSTRACT

BACKGROUND: The Nod-like receptor NACHT, LRR, and PYD domain-containing protein 3 (NLRP3) and Bruton tyrosine kinase (BTK) are protagonists in innate and adaptive immunity, respectively. NLRP3 senses exogenous and endogenous insults, leading to inflammasome activation, which occurs spontaneously in patients with Muckle-Wells syndrome; BTK mutations cause the genetic immunodeficiency X-linked agammaglobulinemia (XLA). However, to date, few proteins that regulate NLRP3 inflammasome activity in human primary immune cells have been identified, and clinically promising pharmacologic targeting strategies remain elusive. OBJECTIVE: We sought to identify novel regulators of the NLRP3 inflammasome in human cells with a view to exploring interference with inflammasome activity at the level of such regulators. METHODS: After proteome-wide phosphoproteomics, the identified novel regulator BTK was studied in human and murine cells by using pharmacologic and genetic BTK ablation. RESULTS: Here we show that BTK is a critical regulator of NLRP3 inflammasome activation: pharmacologic (using the US Food and Drug Administration-approved inhibitor ibrutinib) and genetic (in patients with XLA and Btk knockout mice) BTK ablation in primary immune cells led to reduced IL-1ß processing and secretion in response to nigericin and the Staphylococcus aureus toxin leukocidin AB (LukAB). BTK affected apoptosis-associated speck-like protein containing a CARD (ASC) speck formation and caspase-1 cleavage and interacted with NLRP3 and ASC. S aureus infection control in vivo and IL-1ß release from cells of patients with Muckle-Wells syndrome were impaired by ibrutinib. Notably, IL-1ß processing and release from immune cells isolated from patients with cancer receiving ibrutinib therapy were reduced. CONCLUSION: Our data suggest that XLA might result in part from genetic inflammasome deficiency and that NLRP3 inflammasome-linked inflammation could potentially be targeted pharmacologically through BTK.


Subject(s)
Agammaglobulinemia/genetics , Cryopyrin-Associated Periodic Syndromes/genetics , Genetic Diseases, X-Linked/genetics , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein-Tyrosine Kinases/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Adaptive Immunity , Adaptor Proteins, Signal Transducing , Agammaglobulinaemia Tyrosine Kinase , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/immunology , Cells, Cultured , Humans , Immunity, Innate , Leukocidins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , NLR Proteins , Nigericin/immunology , Protein-Tyrosine Kinases/genetics , Proteomics , Pyrin Domain/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Lamin B Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...