Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Med Virol ; 95(11): e29245, 2023 11.
Article in English | MEDLINE | ID: mdl-38009693

ABSTRACT

Arthropod-borne flaviviruses include a number of medically relevant human pathogens such as the mosquito-borne dengue (DEN), Zika, and yellow fever (YF) viruses as well as tick-borne encephalitis virus (TBEV). All flaviviruses are antigenically related and anamnestic responses due to prior immunity can modulate antibody specificities in subsequent infections or vaccinations. In our study, we analyzed the induction of broadly flavivirus cross-reactive antibodies in tick-borne encephalitis (TBE) and DEN patients without or with prior flavivirus exposure through TBE and/or YF vaccination, and determined the contribution of these antibodies to TBE and dengue virus (DENV) neutralization. In addition, we investigated the formation of cross-reactive antibodies in TBE-vaccination breakthroughs (VBTs). A TBEV infection without prior YF or TBE vaccination induced predominantly type-specific antibodies. In contrast, high levels of broadly cross-reactive antibodies were found in samples from TBE patients prevaccinated against YF as well as in DEN patients prevaccinated against TBE and/or YF. While these cross-reactive antibodies did not neutralize TBEV, they were effective in neutralizing DENV. This discrepancy points to structural differences between the two viruses and indicates that broadly cross-reactive epitopes are less accessible in TBEV than in DENV. In TBE VBT infections, type-specific antibodies dominated the antibody response, thus revealing no difference from that of unvaccinated TBE patients. Our results emphasize significant differences in the structural properties of different flaviviruses that have an impact on the induction of broadly cross-reactive antibodies and their functional activities in virus neutralization.


Subject(s)
Dengue , Encephalitis Viruses, Tick-Borne , Encephalitis, Tick-Borne , Flavivirus Infections , Zika Virus Infection , Zika Virus , Animals , Humans , Encephalitis, Tick-Borne/prevention & control , Antibody Formation , Antibodies, Viral , Flavivirus Infections/prevention & control , Vaccination , Dengue/prevention & control
2.
Viruses ; 13(4)2021 03 31.
Article in English | MEDLINE | ID: mdl-33807442

ABSTRACT

Flaviviruses circulate worldwide and cause a number of medically relevant human diseases, such as dengue, Zika, yellow fever, and tick-borne encephalitis (TBE). Serology plays an important role in the diagnosis of flavivirus infections, but can be impeded by antigenic cross-reactivities among flaviviruses. Therefore, serological diagnosis of a recent infection can be insufficiently specific, especially in areas where flaviviruses co-circulate and/or vaccination coverage against certain flaviviruses is high. In this study, we developed a new IgM assay format, which is well suited for the specific diagnosis of TBE, Zika and dengue virus infections. In the case of TBE and Zika, the IgM response proved to be highly specific for the infecting virus. In contrast, primary dengue virus infections induced substantial amounts of cross-reactive IgM antibodies, which is most likely explained by structural peculiarities of dengue virus particles. Despite the presence of cross-reactive IgM, the standardized nature and the quantitative read-out of the assay even allowed the serotype-specific diagnosis of recent dengue virus infections in most instances.


Subject(s)
Antibodies, Viral/blood , Antigens, Viral/immunology , Cross Reactions/immunology , Flavivirus Infections/diagnosis , Flavivirus/immunology , Immunoglobulin M/blood , Serologic Tests/methods , Antigens, Viral/classification , Cohort Studies , Dengue/blood , Dengue/diagnosis , Dengue/immunology , Dengue Virus/immunology , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/diagnosis , Encephalitis, Tick-Borne/immunology , Flavivirus/classification , Flavivirus Infections/blood , Flavivirus Infections/virology , Humans , Serogroup , Serologic Tests/standards , Zika Virus/immunology , Zika Virus Infection/blood , Zika Virus Infection/diagnosis , Zika Virus Infection/immunology
3.
Front Immunol ; 11: 16, 2020.
Article in English | MEDLINE | ID: mdl-32038660

ABSTRACT

West Nile (WN) virus infection of humans is frequently asymptomatic, but can also lead to WN fever or neuroinvasive disease. CD4 T cells and B cells are critical in the defense against WN virus, and neutralizing antibodies, which are directed against the viral glycoprotein E, are an accepted correlate of protection. For the efficient production of these antibodies, B cells interact directly with CD4 helper T cells that recognize peptides from E or the two other structural proteins (capsid-C and membrane-prM/M) of the virus. However, the specific protein sites yielding such helper epitopes remain unknown. Here, we explored the CD4 T cell response in humans after WN virus infection using a comprehensive library of overlapping peptides covering all three structural proteins. By measuring T cell responses in 29 individuals with either WN virus disease or asymptomatic infection, we showed that CD4 T cells focus on peptides in specific structural elements of C and at the exposed surface of the pre- and postfusion forms of the E protein. Our data indicate that these immunodominant epitopes are recognized in the context of multiple different HLA molecules. Furthermore, we observed that immunodominant antigen regions are structurally conserved and similarly targeted in other mosquito-borne flaviviruses, including dengue, yellow fever, and Zika viruses. Together, these findings indicate a strong impact of virion protein structure on epitope selection and antigenicity, which is an important issue to consider in future vaccine design.


Subject(s)
Asymptomatic Infections , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , West Nile Fever/immunology , West Nile virus/immunology , Adult , Aged , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Capsid Proteins/immunology , Cohort Studies , Dengue Virus/chemistry , Dengue Virus/immunology , Epitopes, T-Lymphocyte/chemistry , Female , HLA-D Antigens/genetics , Humans , Immunodominant Epitopes/immunology , Male , Middle Aged , Peptide Library , RNA, Viral/blood , Viral Envelope Proteins/immunology , West Nile Fever/virology , West Nile virus/chemistry , Yellow fever virus/chemistry , Yellow fever virus/immunology , Zika Virus/chemistry , Zika Virus/immunology
4.
PLoS Negl Trop Dis ; 14(2): e0008034, 2020 02.
Article in English | MEDLINE | ID: mdl-32017766

ABSTRACT

BACKGROUND: Zika virus has recently spread to South- and Central America, causing congenital birth defects and neurological complications. Many people at risk are flavivirus pre-immune due to prior infections with other flaviviruses (e.g. dengue virus) or flavivirus vaccinations. Since pre-existing cross-reactive immunity can potentially modulate antibody responses to Zika virus infection and may affect the outcome of disease, we analyzed fine-specificity as well as virus-neutralizing and infection-enhancing activities of antibodies induced by a primary Zika virus infection in flavivirus-naïve as well as yellow fever- and/or tick-borne encephalitis-vaccinated individuals. METHODOLOGY: Antibodies in sera from convalescent Zika patients with and without vaccine-induced immunity were assessed by ELISA with respect to Zika virus-specificity and flavivirus cross-reactivity. Functional analyses included virus neutralization and infection-enhancement. The contribution of IgM and cross-reactive antibodies to these properties was determined by depletion experiments. PRINCIPAL FINDINGS: Pre-existing flavivirus immunity had a strong influence on the antibody response in primary Zika virus infections, resulting in higher titers of broadly flavivirus cross-reactive antibodies and slightly lower levels of Zika virus-specific IgM. Antibody-dependent enhancement (ADE) of Zika virus was mediated by sub-neutralizing concentrations of specific IgG but not by cross-reactive antibodies. This effect was potently counteracted by the presence of neutralizing IgM. Broadly cross-reactive antibodies were able to both neutralize and enhance infection of dengue virus but not Zika virus, indicating a different exposure of conserved sequence elements in the two viruses. CONCLUSIONS: Our data point to an important role of flavivirus-specific IgM during the transient early stages of infection, by contributing substantially to neutralization and by counteracting ADE. In addition, our results highlight structural differences between strains of Zika and dengue viruses that are used for analyzing infection-enhancement by cross-reactive antibodies. These findings underscore the possible impact of specific antibody patterns on flavivirus disease and vaccination efficacy.


Subject(s)
Antibodies, Viral/blood , Viral Vaccines/immunology , Zika Virus Infection/prevention & control , Zika Virus/immunology , Antibody Affinity , Antigens, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Neutralization Tests , Polyethylene Glycols , Viral Envelope Proteins/immunology , Zika Virus/genetics
5.
NPJ Vaccines ; 4: 38, 2019.
Article in English | MEDLINE | ID: mdl-31508246

ABSTRACT

Flaviviruses have an increasing global impact as arthropod-transmitted human pathogens, exemplified by Zika, dengue, yellow fever (YF), West Nile, Japanese encephalitis, and tick-borne encephalitis (TBE) viruses. Since all flaviviruses are antigenically related, they are prone to phenomena of immunological memory ('original antigenic sin'), which can modulate immune responses in the course of sequential infections and/or vaccinations. In our study, we analyzed the influence of pre-existing YF vaccine-derived immunity on the antibody response to TBE vaccination. By comparing samples from YF pre-vaccinated and flavivirus-naive individuals, we show that YF immunity not only caused a significant impairment of the neutralizing antibody response to TBE vaccination but also a reduction of the specific TBE virus neutralizing activities (NT/ELISA-titer ratios). Our results point to a possible negative effect of pre-existing cross-reactive immunity on the outcome of flavivirus vaccination that may also pertain to other combinations of sequential flavivirus infections and/or vaccinations.

7.
Front Immunol ; 9: 1196, 2018.
Article in English | MEDLINE | ID: mdl-29899743

ABSTRACT

Zika virus (ZIKV) has recently caused explosive outbreaks in Pacific islands, South- and Central America. Like with other flaviviruses, protective immunity is strongly dependent on potently neutralizing antibodies (Abs) directed against the viral envelope protein E. Such Ab formation is promoted by CD4 T cells through direct interaction with B cells that present epitopes derived from E or other structural proteins of the virus. Here, we examined the extent and epitope dominance of CD4 T cell responses to capsid (C) and envelope proteins in Zika patients. All patients developed ZIKV-specific CD4 T cell responses, with substantial contributions of C and E. In both proteins, immunodominant epitopes clustered at sites that are structurally conserved among flaviviruses but have highly variable sequences, suggesting a strong impact of protein structural features on immunodominant CD4 T cell responses. Our data are particularly relevant for designing flavivirus vaccines and their evaluation in T cell assays and provide insights into the importance of viral protein structure for epitope selection and antigenicity.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Capsid Proteins/genetics , Epitopes, T-Lymphocyte/genetics , Immunodominant Epitopes/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/immunology , Zika Virus Infection/immunology , Zika Virus/physiology , Adult , Aged , Antibodies, Neutralizing/metabolism , Antibodies, Viral/metabolism , Capsid Proteins/immunology , Cells, Cultured , Conserved Sequence/genetics , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunodominant Epitopes/immunology , Male , Middle Aged , Structure-Activity Relationship , Viral Envelope Proteins/immunology , Young Adult
8.
Virulence ; 9(1): 231-247, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29099326

ABSTRACT

Pathogenesis of Staphylococcus aureus is increasingly recognized to be driven by powerful toxins. Staphylococcus aureus employs up to six pore-forming toxins to subvert the human host defense and to promote bacterial invasion: alpha-hemolysin that disrupts epithelial and endothelial barriers and five leukocidins that lyse phagocytes involved in bacterial clearance. Previously, we described two human monoclonal antibodies (mAbs), ASN-1 that neutralizes alpha-hemolysin and four leukocidins (LukSF-PV, LukED, HlgAB, HlgCB), and ASN-2 that inactivates the 5th leukocidin, LukGH. In this study we tested the individual and combined effects of ASN-1 and ASN-2 in multiple in vitro models employing relevant human target cells. We found that diverse S. aureus isolates with different genetic backgrounds (based on MLST- and spa-typing) and antibiotic sensitivity (both MRSA and MSSA) displayed greatly different cytotoxin expression patterns influenced by the type of growth medium used. Both mAbs were required to fully prevent the lysis of human neutrophils exposed to the mixture of recombinant cytotoxins or native toxins present in the culture supernatants of S. aureus isolates. Flow cytometry confirmed the protective effects of ASN-1 + ASN-2 (known as ASN100) on granulocytes, monocytes, NK-cells and T-lymphocytes. ASN-1 alone preserved the integrity of a 3D-primary culture of human tracheal/bronchial mucociliary epithelial tissue infected with S. aureus. We conclude that simultaneous inhibition of alpha-hemolysin and five leukocidins by ASN100 blocks cytolytic activity of S. aureus towards human target cells in vitro.


Subject(s)
Antibodies, Monoclonal/immunology , Bacterial Toxins/metabolism , Cytotoxins/metabolism , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Antibodies, Monoclonal/metabolism , Bacterial Toxins/antagonists & inhibitors , Cytotoxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism , Leukocidins/antagonists & inhibitors , Leukocidins/metabolism , Neutrophils/immunology , Neutrophils/microbiology , Organoids/immunology , Organoids/microbiology , Organoids/pathology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/chemistry
9.
Article in English | MEDLINE | ID: mdl-28874372

ABSTRACT

The multidrug-resistant H30 subclone of extraintestinal pathogenic Escherichia coli sequence type 131 (ST131-H30) has spread worldwide. This clone expresses a conserved lipopolysaccharide (LPS) O antigen, O25b. Previously, we described monoclonal antibodies (MAbs) specific to the O25b antigen and characterized them as diagnostic and therapeutic tools. In this study, evidence is provided that besides the previously shown complement-mediated bactericidal effect, an O25b-specific humanized MAb, A1124, also enhances opsonophagocytic uptake by the murine macrophage cell line RAW 264.7. Both phagocyte-dependent killing and phagocyte-independent killing, triggered by A1124, were confirmed in human whole blood. Furthermore, A1124 was shown to neutralize endotoxin activity of purified LPS of clinical isolates. This activity was demonstrated in vitro using both RAW 264.7 cells and a human Toll-like receptor 4 (TLR4) reporter cell line, as well as in a murine model of endotoxemia using purified LPS for challenge. Significant protective efficacy of A1124 at low doses (<1 mg/kg of body weight) was shown in murine and rat models of bacteremia. The contribution of the bactericidal and anti-inflammatory effects was dissected in the mouse bacteremia model through depletion of complement with cobra venom factor (CVF). Protective efficacy was lost in complement-depleted mice, suggesting the essential role of complement-mediated activities for protection in this model. These data suggest that A1124 exhibits different mechanisms of action, namely, direct complement-mediated and opsonophagocytic killing as well as endotoxin neutralization in various challenge models. Which of these activities are the most relevant in a clinical setting will need to be addressed by future translational studies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antibodies, Monoclonal/pharmacology , Drug Resistance, Multiple, Bacterial/drug effects , Escherichia coli Infections/drug therapy , Extraintestinal Pathogenic Escherichia coli/drug effects , Animals , Blood/microbiology , Cell Line , Complement System Proteins/metabolism , Disease Models, Animal , Endotoxemia/drug therapy , Endotoxemia/microbiology , Endotoxins/metabolism , Extraintestinal Pathogenic Escherichia coli/isolation & purification , Female , Humans , Macrophages/microbiology , Mice, Inbred BALB C , O Antigens/immunology , Rats, Sprague-Dawley
10.
J Infect ; 74(5): 473-483, 2017 05.
Article in English | MEDLINE | ID: mdl-28237625

ABSTRACT

OBJECTIVES: Staphylococcus aureus produces up to five bi-component leukocidins - LukSF-PV, gamma-hemolysins AB and CB, LukGH (LukAB) and LukED - to evade innate immunity by lysing phagocytic cells. Species specificity of these leukocidins limits the relevance of animal models, therefore we assessed their individual contribution using human neutrophils. METHODS: Human polymorphonuclear leukocytes (PMNs) were activated with stimuli relevant during bacterial infections and sensitivity to recombinant leukocidins was measured in cell-viability assays. Leukocidin receptor expression was quantified by flow cytometry. RESULTS: We observed greatly variable sensitivities of different PMN preparations towards LukGH. Activation of PMNs by lipopolysaccharide (LPS) or S. aureus culture supernatant (CS) lacking all leukocidins resulted in higher surface expression of CD11b, the LukGH receptor, and greatly enhanced the sensitivity towards LukGH, eliminating the variability observed with unstimulated cells. In contrast, CS induced a decrease in sensitivity of PMNs to the other four leukocidins and reduced surface staining for their cognate receptors (CXCR1, CXCR2, C5aR, C5L2). Delta-toxin and peptidoglycan mimicked the effect of CS. Moreover, IL-8, an important cytokine in neutrophil activation, also selectively increased LukGH sensitivity. Deletion of lukGH, but not other leukocidin genes, prevented PMN killing upon infection with USA300 CA-MRSA. CONCLUSION: Inflammatory signals enhance the susceptibility of human PMNs to lysis by LukGH rendering this toxin dominant among the S. aureus leukocidins in vitro.


Subject(s)
Bacterial Proteins/immunology , Leukocidins/immunology , Neutrophil Activation/immunology , Neutrophils/immunology , CD11b Antigen/analysis , CD11b Antigen/immunology , CD11b Antigen/metabolism , Humans , Interleukin-8/analysis , Interleukin-8/immunology , Interleukin-8/metabolism , Lipopolysaccharides/immunology , Models, Biological , Neutrophils/microbiology , Staphylococcus aureus/immunology
11.
MAbs ; 8(7): 1347-1360, 2016 10.
Article in English | MEDLINE | ID: mdl-27467113

ABSTRACT

LukGH (LukAB) is a potent leukocidin of Staphylococcus aureus that lyses human phagocytic cells and is thought to contribute to immune evasion. Unlike the other bi-component leukocidins of S. aureus, LukGH forms a heterodimer before binding to its receptor, CD11b expressed on professional phagocytic cells, and displays significant sequence variation. We employed a high diversity human IgG1 library presented on yeast cells to discover monoclonal antibodies (mAbs) neutralizing the cytolytic activity of LukGH. Recombinant LukG and LukH monomers or a LukGH dimer were used as capture antigens in the library selections. We found that mAbs identified with LukG or LukH as bait had no or very low toxin neutralization potency. In contrast, LukGH dimer-selected antibodies proved to be highly potent, and several mAbs were able to neutralize even the most divergent LukGH variants. Based on biolayer interferometry and mesoscale discovery, the high affinity antibody binding site on the LukGH complex was absent on the individual monomers, suggesting that it was generated upon formation of the LukG-LukH dimer. X-ray crystallography analysis of the complex between the LukGH dimer and the antigen-binding fragment of a very potent mAb (PDB code 5K59) indicated that the epitope is located in the predicted cell binding region (rim domain) of LukGH. The corresponding IgG inhibited the binding of LukGH dimer to target cells. Our data suggest that knowledge of the native conformation of target molecules is essential to generate high affinity and functional mAbs.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Bacterial Proteins/immunology , Leukocidins/immunology , Animals , Bacterial Proteins/chemistry , Dimerization , Humans , Leukocidins/chemistry
12.
J Biol Chem ; 290(1): 142-56, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25371205

ABSTRACT

The bi-component leukocidins of Staphylococcus aureus are important virulence factors that lyse human phagocytic cells and contribute to immune evasion. The γ-hemolysins (HlgAB and HlgCB) and Panton-Valentine leukocidin (PVL or LukSF) were shown to assemble from soluble subunits into membrane-bound oligomers on the surface of target cells, creating barrel-like pore structures that lead to cell lysis. LukGH is the most distantly related member of this toxin family, sharing only 30-40% amino acid sequence identity with the others. We observed that, unlike other leukocidin subunits, recombinant LukH and LukG had low solubility and were unable to bind to target cells, unless both components were present. Using biolayer interferometry and intrinsic tryptophan fluorescence we detected binding of LukH to LukG in solution with an affinity in the low nanomolar range and dynamic light scattering measurements confirmed formation of a heterodimer. We elucidated the structure of LukGH by x-ray crystallography at 2.8-Šresolution. This revealed an octameric structure that strongly resembles that reported for HlgAB, but with important structural differences. Structure guided mutagenesis studies demonstrated that three salt bridges, not found in other bi-component leukocidins, are essential for dimer formation in solution and receptor binding. We detected weak binding of LukH, but not LukG, to the cellular receptor CD11b by biolayer interferometry, suggesting that in common with other members of this toxin family, the S-component has the primary contact role with the receptor. These new insights provide the basis for novel strategies to counteract this powerful toxin and Staphylococcus aureus pathogenesis.


Subject(s)
Bacterial Proteins/chemistry , CD11b Antigen/chemistry , Hemolysin Proteins/chemistry , Leukocidins/chemistry , Staphylococcus aureus/chemistry , Amino Acid Sequence , Animals , Bacterial Proteins/metabolism , Binding Sites , CD11b Antigen/metabolism , Crystallography, X-Ray , HL-60 Cells , Hemolysin Proteins/metabolism , Humans , Leukocidins/metabolism , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Protein Binding , Protein Multimerization , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Staphylococcus aureus/metabolism , Structure-Activity Relationship
13.
MAbs ; 7(1): 243-54, 2015.
Article in English | MEDLINE | ID: mdl-25523282

ABSTRACT

Staphylococcus aureus is a major human pathogen associated with high mortality. The emergence of antibiotic resistance and the inability of antibiotics to counteract bacterial cytotoxins involved in the pathogenesis of S. aureus call for novel therapeutic approaches, such as passive immunization with monoclonal antibodies (mAbs). The complexity of staphylococcal pathogenesis and past failures with single mAb products represent considerable barriers for antibody-based therapeutics. Over the past few years, efforts have focused on neutralizing α-hemolysin. Recent findings suggest that the concerted actions of several cytotoxins, including the bi-component leukocidins play important roles in staphylococcal pathogenesis. Therefore, we aimed to isolate mAbs that bind to multiple cytolysins by employing high diversity human IgG1 libraries presented on the surface of yeast cells. Here we describe cross-reactive antibodies with picomolar affinity for α-hemolysin and 4 different bi-component leukocidins that share only ∼26% overall amino acid sequence identity. The molecular basis of cross-reactivity is the recognition of a conformational epitope shared by α-hemolysin and F-components of gamma-hemolysin (HlgAB and HlgCB), LukED and LukSF (Panton-Valentine Leukocidin). The amino acids predicted to form the epitope are conserved and known to be important for cytotoxic activity. We found that a single cross-reactive antibody prevented lysis of human phagocytes, epithelial and red blood cells induced by α-hemolysin and leukocidins in vitro, and therefore had superior effectiveness compared to α-hemolysin specific antibodies to protect from the combined cytolytic effect of secreted S. aureus toxins. Such mAb afforded high levels of protection in murine models of pneumonia and sepsis.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Monoclonal/immunology , Bacterial Proteins/immunology , Hemolysin Proteins/immunology , Immunoglobulin G/immunology , Leukocidins/immunology , Staphylococcus aureus/immunology , Animals , Antibodies, Bacterial/chemistry , Antibodies, Monoclonal/chemistry , Antibody Specificity , Bacterial Proteins/chemistry , Cell Line , Hemolysin Proteins/chemistry , Humans , Immunoglobulin G/chemistry , Leukocidins/chemistry , Rabbits , Staphylococcus aureus/chemistry
14.
Am J Respir Crit Care Med ; 190(10): 1139-48, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25303310

ABSTRACT

RATIONALE: Colonization of lower airways by Staphylococcus aureus is a risk factor for the development of ventilator-associated tracheobronchitis (VAT) and ventilator-associated pneumonia (VAP). However, little is known about the virulence factors of methicillin-sensitive and -resistant S. aureus (MSSA and MRSA) that may influence host colonization and progression to VAT and VAP. OBJECTIVES: We evaluated MRSA and MSSA endotracheal aspirates (ETA) for genotype and α-hemolysin activity in relation to the development of VAT and VAP. METHODS: Serial S. aureus ETA isolates from ventilated patients were analyzed for methicillin resistance, molecular type by Multi-Locus Sequence Typing and spa-typing, and α-hemolysin activity by semiquantitative analysis of hemolysis on sheep blood agar and quantitative measurement of cytolysis of human lung epithelial cells. The virulence of selected strains was assessed in mice by intranasal challenge. MEASUREMENTS AND MAIN RESULTS: We detected S. aureus from ETA samples in a quarter of the 231 ventilated patients analyzed; one-third of them developed VAP. VAP patients (n = 15) were mainly infected by MSSA strains (87%), whereas colonized individuals (n = 18) not progressing to disease mainly carried MRSA strains (68%). MSSA isolates from colonized or VAT patients exhibited significantly lower α-hemolysin activity than those from VAP cases; however, no such relationship was found with MRSA strains. α-Hemolysin activity of S. aureus isolates was predictive for virulence in mouse pneumonia model. CONCLUSIONS: MSSA strains with strong blood agar hemolysis and high α-hemolysin activity are markers for VAP, but not VAT, and might be considered in differential diagnosis and initiation of therapy.


Subject(s)
Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Pneumonia, Ventilator-Associated/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/pathogenicity , Aged , Aged, 80 and over , Animals , Bronchitis/diagnosis , Bronchitis/metabolism , Bronchitis/microbiology , Cohort Studies , Disease Models, Animal , Female , Humans , Male , Methicillin-Resistant Staphylococcus aureus/metabolism , Mice , Middle Aged , Pneumonia, Ventilator-Associated/diagnosis , Pneumonia, Ventilator-Associated/metabolism , Risk Factors , Staphylococcal Infections/diagnosis , Staphylococcal Infections/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/metabolism , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL