Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
MAbs ; 14(1): 2083467, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35730685

RESUMEN

Staphylococcus aureus carries an exceptional repertoire of virulence factors that aid in immune evasion. Previous single-target approaches for S. aureus-specific vaccines and monoclonal antibodies (mAbs) have failed in clinical trials due to the multitude of virulence factors released during infection. Emergence of antibiotic-resistant strains demands a multi-target approach involving neutralization of different, non-overlapping pathogenic factors. Of the several pore-forming toxins that contribute to S. aureus pathogenesis, efforts have largely focused on mAbs that neutralize α-hemolysin (Hla) and target the receptor-binding site. Here, we isolated two anti-Hla and three anti-Panton-Valentine Leukocidin (LukSF-PV) mAbs, and used a combination of hydrogen deuterium exchange mass spectrometry (HDX-MS) and alanine scanning mutagenesis to delineate and validate the toxins' epitope landscape. Our studies identified two novel, neutralizing epitopes targeted by 2B6 and CAN6 on Hla that provided protection from hemolytic activity in vitro and showed synergy in rodent pneumonia model against lethal challenge. Of the anti-LukF mAbs, SA02 and SA131 showed specific neutralization activity to LukSF-PV while SA185 showed cross-neutralization activity to LukSF-PV, γ-hemolysin HlgAB, and leukotoxin ED. We further compared these antigen-specific mAbs to two broadly neutralizing mAbs, H5 (targets Hla, LukSF-PV, HlgAB, HlgCB, and LukED) and SA185 (targeting LukSF-PV, HlgAB, and LukED), and identified molecular level markers for broad-spectrum reactivity among the pore-forming toxins by HDX-MS. To further underscore the need to target the cross-reactive epitopes on leukocidins for the development of broad-spectrum therapies, we annotated Hla sequences isolated from patients in multiple countries for genomic variations within the perspective of our defined epitopes.


Asunto(s)
Infecciones Estafilocócicas , Staphylococcus aureus , Anticuerpos Monoclonales , Proteínas Bacterianas/química , Epítopos , Exotoxinas , Proteínas Hemolisinas , Humanos , Leucocidinas/química , Factores de Virulencia
2.
Biochim Biophys Acta Biomembr ; 1864(9): 183970, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35605647

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas , Staphylococcus aureus Resistente a Meticilina , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/química , Proteínas Hemolisinas/química , Leucocidinas/química , Leucocidinas/metabolismo , Membrana Dobles de Lípidos/metabolismo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Staphylococcus aureus/metabolismo
3.
FEBS J ; 289(12): 3505-3520, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35030303

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas , Proteínas Hemolisinas , Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Hemólisis , Leucocidinas/química , Leucocidinas/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
4.
mBio ; 11(3)2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546616

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Epítopos de Linfocito B/inmunología , Exotoxinas/inmunología , Proteínas Hemolisinas/inmunología , Epítopos Inmunodominantes/inmunología , Animales , Anticuerpos Antibacterianos/sangre , Mapeo Epitopo , Femenino , Humanos , Epítopos Inmunodominantes/genética , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Leucocidinas/química , Leucocidinas/inmunología , Ratones , Ratones Endogámicos BALB C , Biblioteca de Péptidos , Infecciones Estafilocócicas/sangre , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus , Factores de Virulencia
5.
Sci Transl Med ; 11(475)2019 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-30651319

RESUMEN

A key aspect underlying the severity of infections caused by Staphylococcus aureus is the abundance of virulence factors that the pathogen uses to thwart critical components of the human immune response. One such mechanism involves the destruction of host immune cells by cytolytic toxins secreted by S. aureus, including five bicomponent leukocidins: PVL, HlgAB, HlgCB, LukED, and LukAB. Purified leukocidins can lyse immune cells ex vivo, and systemic injections of purified LukED or HlgAB can acutely kill mice. Here, we describe the generation and characterization of centyrins that bind S. aureus leukocidins with high affinity and protect primary human immune cells from toxin-mediated cytolysis. Centyrins are small protein scaffolds derived from the fibronectin type III-binding domain of the human protein tenascin-C. Although centyrins are potent in tissue culture assays, their short serum half-lives limit their efficacies in vivo. By extending the serum half-lives of centyrins through their fusion to an albumin-binding consensus domain, we demonstrate the in vivo efficacy of these biologics in a murine intoxication model and in models of both prophylactic and therapeutic treatment of live S. aureus systemic infections. These biologics that target S. aureus virulence factors have potential for treating and preventing serious staphylococcal infections.


Asunto(s)
Factores Biológicos/farmacología , Leucocidinas/metabolismo , Pruebas de Neutralización , Staphylococcus aureus/metabolismo , Secuencia de Aminoácidos , Animales , Citoprotección/efectos de los fármacos , Citotoxicidad Inmunológica , Hemólisis/efectos de los fármacos , Humanos , Leucocidinas/química , Ratones , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitos/efectos de los fármacos , Infecciones Estafilocócicas/microbiología , Infecciones Estafilocócicas/patología , Staphylococcus aureus/efectos de los fármacos
6.
Toxicon ; 155: 43-48, 2018 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-30312693

RESUMEN

The ß-strand stem release system of staphylococcal ß-barrel pore-forming toxin γ-hemolysin was investigated. Mutations at K15 and R16 in the cap domain of Hlg2 decreased hemolytic activity more markedly than their effect on erythrocyte binding. In addition, D122N mutation of LukF prestem lost the activity with Hlg2 R16A, indicating that electrostatic interactions between residues in the Hlg2 cap and prestem of adjacent LukF in the ring-shaped complex might serve as a switch for stem release.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Proteínas Hemolisinas/química , Proteínas Hemolisinas/genética , Leucocidinas/química , Leucocidinas/genética , Staphylococcus aureus/química , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Eritrocítica/metabolismo , Membrana Eritrocítica/fisiología , Eritrocitos/microbiología , Exotoxinas , Proteínas Hemolisinas/metabolismo , Hemólisis/fisiología , Humanos , Modelos Moleculares , Mutación , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
7.
PLoS One ; 13(9): e0204450, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30261001

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Leucocidinas , Staphylococcus/genética , Staphylococcus/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antibacterianos/inmunología , Anticuerpos Antibacterianos/metabolismo , Proteínas Bacterianas/química , Muerte Celular , Enfermedades de los Perros/inmunología , Perros , Escherichia coli , Exotoxinas/genética , Exotoxinas/metabolismo , Genoma Bacteriano , Leucocidinas/química , Leucocidinas/genética , Leucocidinas/inmunología , Leucocidinas/metabolismo , Leucocitos/metabolismo , Leucocitos/microbiología , Mutación , Filogenia , Proteínas Recombinantes/metabolismo , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/veterinaria , Staphylococcus/inmunología
8.
J Biochem ; 164(2): 93-102, 2018 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-29474554

RESUMEN

Staphylococcus aureus bi-component pore-forming toxins consist of S- and F-components, and form hetero-octameric beta-barrel pores on target blood cell membranes. Among them, γ-haemolysin (Hlg2 and F-component of Luk (LukF)) and LukED (LukE and LukD) possess haemolytic activity, whereas the Panton-Valentine leukocidin (LukS-PV and LukF-PV) does not lyse human erythrocytes. Here, we focussed on four loop structures in the rim domain of S-component, namely loops -1, -2, -3 and -4, and found that replacement of Loop-4 in both Hlg2 and LukE with that of LukS-PV abolished their haemolytic activity. Furthermore, LukS-PV gained haemolytic activity by Loop-4 exchange with Hlg2 or LukE, suggesting that Loop-4 of these S-components determined erythrocyte specificity. LOOP-1 and -2 enhanced the erythrocytes-binding ability of both components. Although Hlg2 and LukE recognize Duffy antigen receptor for chemokines on human erythrocytes, the ability of Loop-4 was not complementary between Hlg2 and LukE. Exchange of Hlg2 with LukE Loop-4 showed weaker activity than intact Hlg2, and LukE mutant with Hlg2 Loop-4 lost its haemolytic activity in combination of LukD. Interestingly, the haemolytic activities of these Loop-4 exchange mutants were affected by F-component, namely LukF enhanced haemolytic activities of these Hlg2 and LukE Loop-4 mutants, and also haemolytic activity of LukS-PV mutant with LukE Loop-4.


Asunto(s)
Eritrocitos/metabolismo , Leucocidinas/metabolismo , Staphylococcus aureus/metabolismo , Humanos , Leucocidinas/química , Leucocidinas/genética , Modelos Moleculares , Mutación , Conformación Proteica
9.
PLoS One ; 12(11): e0187288, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29117225

RESUMEN

A bacterial insertion sequence (IS) is a mobile DNA sequence carrying only the transposase gene (tnp) that acts as a mutator to disrupt genes, alter gene expressions, and cause genomic rearrangements. "Canonical" ISs have historically been characterized by their terminal inverted repeats (IRs), which may form a stem-loop structure, and duplications of a short (non-IR) target sequence at both ends, called target site duplications (TSDs). The IS distributions and virulence potentials of Staphylococcus aureus genomes in familial infection cases are unclear. Here, we determined the complete circular genome sequences of familial strains from a Panton-Valentine leukocidin (PVL)-positive ST50/agr4 S. aureus (GN) infection of a 4-year old boy with skin abscesses. The genomes of the patient strain (GN1) and parent strain (GN3) were rich for "canonical" IS1272 with terminal IRs, both having 13 commonly-existing copies (ce-IS1272). Moreover, GN1 had a newly-inserted IS1272 (ni-IS1272) on the PVL-converting prophage, while GN3 had two copies of ni-IS1272 within the DNA helicase gene and near rot. The GN3 genome also had a small deletion. The targets of ni-IS1272 transposition were IR structures, in contrast with previous "canonical" ISs. There were no TSDs. Based on a database search, the targets for ce-IS1272 were IRs or "non-IRs". IS1272 included a larger structure with tandem duplications of the left (IRL) side sequence; tnp included minor cases of a long fusion form and truncated form. One ce-IS1272 was associated with the segments responsible for immune evasion and drug resistance. Regarding virulence, GN1 expressed cytolytic peptides (phenol-soluble modulin α and δ-hemolysin) and PVL more strongly than some other familial strains. These results suggest that IS1272 transposes through an IR-replacing mechanism, with an irreversible process unlike that of "canonical" transpositions, resulting in genomic variations, and that, among the familial strains, the patient strain has strong virulence potential based on community-associated virulence factors.


Asunto(s)
Elementos Transponibles de ADN/genética , Genómica , Secuencias Invertidas Repetidas/genética , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/aislamiento & purificación , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Secuencia de Bases , Preescolar , Mapeo Cromosómico , Análisis por Conglomerados , ADN Circular/genética , Exotoxinas/química , Exotoxinas/genética , Familia , Femenino , Duplicación de Gen , Regulación Bacteriana de la Expresión Génica , Genes Bacterianos , Genoma Bacteriano , Humanos , Leucocidinas/química , Leucocidinas/genética , Masculino , Mutagénesis Insercional/genética , Reacción en Cadena de la Polimerasa , Profagos/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Infecciones Estafilocócicas/transmisión , Staphylococcus aureus/patogenicidad , Factores de Virulencia/biosíntesis , Factores de Virulencia/genética
10.
Adv Exp Med Biol ; 966: 15-35, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28455832

RESUMEN

Staphylococcus aureus can produce up to five different bi-component cytotoxins: two gamma-hemolysins HlgAB and HlgCB, and leukocidins SF-PV (Panton Valentine leukocidin), ED (LukED) and GH (LukGH, also called LukAB). Their major function in S. aureus pathogenesis is to evade innate immunity by attacking phagocytic cells and to support bacterial growth by lysing red blood cells. The five cytotoxins display different levels of amino acid sequence conservation (30-82%), but all form a remarkably similar beta-barrel type pore structure (greatly resembling the mono-component toxin alpha-hemolysin) that inserts into the target cell membrane leading to necrotic cell death. This review provides an overview of the culmination of decades of research on the structure of these toxins, their unique sequence and structural features that helps to explain the observed functional differences, such as toxin potency towards different cell types and species, receptor specificity and formation of functional non-cognate toxin pairs. The vast knowledge accumulated in this field supports novel approaches and the design of therapeutics targeting these cytotoxins to tame virulence and fight S. aureus infections.


Asunto(s)
Antibacterianos/uso terapéutico , Proteínas Bacterianas/antagonistas & inhibidores , Diseño de Fármacos , Proteínas Hemolisinas/antagonistas & inhibidores , Leucocidinas/antagonistas & inhibidores , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/efectos de los fármacos , Animales , Antibacterianos/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas Hemolisinas/química , Proteínas Hemolisinas/metabolismo , Humanos , Leucocidinas/química , Leucocidinas/metabolismo , Modelos Moleculares , Terapia Molecular Dirigida , Conformación Proteica , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/metabolismo , Staphylococcus aureus/patogenicidad , Relación Estructura-Actividad , Virulencia
11.
Nat Rev Microbiol ; 15(7): 435-447, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28420883

RESUMEN

Staphylococcus aureus is a major bacterial pathogen that causes disease worldwide. The emergence of strains that are resistant to commonly used antibiotics and the failure of vaccine development have resulted in a renewed interest in the pathophysiology of this bacterium. Staphylococcal leukocidins are a family of bi-component pore-forming toxins that are important virulence factors. During the past five years, cellular receptors have been identified for all of the bi-component leukocidins. The identification of the leukocidin receptors explains the cellular tropism and species specificity that is exhibited by these toxins, which has important biological consequences. In this Review, we summarize the recent discoveries that have reignited interest in these toxins and provide an outlook for future research.


Asunto(s)
Citotoxinas , Leucocidinas , Receptores de Superficie Celular , Staphylococcus aureus/patogenicidad , Humanos , Leucocidinas/química , Leucocidinas/fisiología , Receptores de Superficie Celular/metabolismo , Staphylococcus aureus/genética , Tropismo , Factores de Virulencia
12.
MAbs ; 8(7): 1347-1360, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27467113

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/inmunología , Leucocidinas/inmunología , Animales , Proteínas Bacterianas/química , Dimerización , Humanos , Leucocidinas/química
13.
EMBO Rep ; 17(3): 428-40, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26882549

RESUMEN

Staphylococcus aureus (S. aureus) is a human pathogen that relies on the subversion of host phagocytes to support its pathogenic lifestyle. S. aureus strains can produce up to five beta-barrel, bi-component, pore-forming leukocidins that target and kill host phagocytes. Thus, preventing immune cell killing by these toxins is likely to boost host immunity. Here, we describe the identification of glycine-rich motifs within the membrane-penetrating stem domains of the leukocidin subunits that are critical for killing primary human neutrophils. Remarkably, leukocidins lacking these glycine-rich motifs exhibit dominant-negative inhibitory effects toward their wild-type toxin counterparts as well as other leukocidins. Biochemical and cellular assays revealed that these dominant-negative toxins work by forming mixed complexes that are impaired in pore formation. The dominant-negative leukocidins inhibited S. aureus cytotoxicity toward primary human neutrophils, protected mice from lethal challenge by wild-type leukocidin, and reduced bacterial burden in a murine model of bloodstream infection. Thus, we describe the first example of staphylococcal bi-component dominant-negative toxins and their potential as novel therapeutics to combat S. aureus infection.


Asunto(s)
Citotoxinas/genética , Leucocidinas/genética , Mutación , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/patogenicidad , Animales , Citotoxinas/química , Citotoxinas/metabolismo , Citotoxinas/uso terapéutico , Femenino , Glicina/química , Glicina/genética , Humanos , Leucocidinas/química , Leucocidinas/metabolismo , Leucocidinas/uso terapéutico , Ratones , Neutrófilos/microbiología , Fagocitos/microbiología , Dominios Proteicos , Multimerización de Proteína , Staphylococcus aureus/genética , Virulencia/genética
14.
J Biol Chem ; 290(1): 142-56, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25371205

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/química , Antígeno CD11b/química , Proteínas Hemolisinas/química , Leucocidinas/química , Staphylococcus aureus/química , Secuencia de Aminoácidos , Animales , Proteínas Bacterianas/metabolismo , Sitios de Unión , Antígeno CD11b/metabolismo , Cristalografía por Rayos X , Células HL-60 , Proteínas Hemolisinas/metabolismo , Humanos , Leucocidinas/metabolismo , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Unión Proteica , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Staphylococcus aureus/metabolismo , Relación Estructura-Actividad
15.
MAbs ; 7(1): 243-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25523282

RESUMEN

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.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Proteínas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Inmunoglobulina G/inmunología , Leucocidinas/inmunología , Staphylococcus aureus/inmunología , Animales , Anticuerpos Antibacterianos/química , Anticuerpos Monoclonales/química , Especificidad de Anticuerpos , Proteínas Bacterianas/química , Línea Celular , Proteínas Hemolisinas/química , Humanos , Inmunoglobulina G/química , Leucocidinas/química , Conejos , Staphylococcus aureus/química
16.
Nat Commun ; 5: 4897, 2014 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-25263813

RESUMEN

Pathogenic bacteria secrete pore-forming toxins (PFTs) to attack target cells. PFTs are expressed as water-soluble monomeric proteins, which oligomerize into nonlytic prepore intermediates on the target cell membrane before forming membrane-spanning pores. Despite a wealth of biochemical data, the lack of high-resolution prepore structural information has hampered understanding of the ß-barrel formation process. Here, we report crystal structures of staphylococcal γ-haemolysin and leucocidin prepores. The structures reveal a disordered bottom half of the ß-barrel corresponding to the transmembrane region, and a rigid upper extramembrane half. Spectroscopic analysis of fluorescently labelled mutants confirmed that the prepore is distinct from the pore within the transmembrane region. Mutational analysis also indicates a pivotal role for the glycine residue located at the lipid-solvent interface as a 'joint' between the two halves of the ß-barrel. These observations suggest a two-step transmembrane ß-barrel pore formation mechanism in which the upper extramembrane and bottom transmembrane regions are formed independently.


Asunto(s)
Toxinas Bacterianas/química , Staphylococcus aureus/química , Animales , Arginina/química , Membrana Celular/química , Cristalografía por Rayos X , Eritrocitos/efectos de los fármacos , Eritrocitos/microbiología , Glicina/química , Proteínas Hemolisinas/química , Hemólisis , Humanos , Leucocidinas/química , Lípidos/química , Mutación , Fosfolípidos/química , Unión Proteica , Estructura Secundaria de Proteína , Conejos , Solventes/química , Espectrometría de Fluorescencia , Triptófano/química
17.
BMC Infect Dis ; 14: 466, 2014 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-25158781

RESUMEN

BACKGROUND: Prostatic abscesses are an uncommon disease usually caused by enterobacteria. They mostly occur in immunodeficient patients. It is thus extremely rare to have a Staphylococcal prostatic abscess in a young immunocompetent patient. CASE PRESENTATION: A 20-year-old patient was treated with ofloxacin for a suspicion of prostatitis. An ultrasonography was performed because of persisting symptoms and showed acute urinary retention and prostatic abscesses. So the empirical antibiotic therapy was modified with ceftriaxone/amikacin. The disease worsened to severe sepsis and the patient was admitted in ICU. CT-scan and MRI confirmed three abscesses with perirectal infiltration and the bacteriological samples (abscesses and blood cultures) were positive to methicillin-susceptible Staphylococcus aureus producing Panton-Valentine leukocidine. The treatment was changed with fosfomycin/ofloxacin which resulted in a general improvement and the regression of the abscesses. CONCLUSION: Staphyloccocus aureus producing Panton-Valentine leukocidin are most commonly responsible for skin and soft tissue infections. To this day, no other case of prostatic abscess due to this strain but susceptible to methicillin has been described.


Asunto(s)
Absceso/microbiología , Toxinas Bacterianas/química , Exotoxinas/química , Leucocidinas/química , Sepsis/diagnóstico , Sepsis/microbiología , Infecciones Estafilocócicas/microbiología , Absceso/diagnóstico , Humanos , Imagen por Resonancia Magnética , Masculino , Meticilina , Ofloxacino/uso terapéutico , Infecciones de los Tejidos Blandos/microbiología , Infecciones Estafilocócicas/diagnóstico , Tomografía Computarizada por Rayos X , Ultrasonografía , Adulto Joven
18.
Microbiol Mol Biol Rev ; 78(2): 199-230, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24847020

RESUMEN

The ability to produce water-soluble proteins with the capacity to oligomerize and form pores within cellular lipid bilayers is a trait conserved among nearly all forms of life, including humans, single-celled eukaryotes, and numerous bacterial species. In bacteria, some of the most notable pore-forming molecules are protein toxins that interact with mammalian cell membranes to promote lysis, deliver effectors, and modulate cellular homeostasis. Of the bacterial species capable of producing pore-forming toxic molecules, the Gram-positive pathogen Staphylococcus aureus is one of the most notorious. S. aureus can produce seven different pore-forming protein toxins, all of which are believed to play a unique role in promoting the ability of the organism to cause disease in humans and other mammals. The most diverse of these pore-forming toxins, in terms of both functional activity and global representation within S. aureus clinical isolates, are the bicomponent leucocidins. From the first description of their activity on host immune cells over 100 years ago to the detailed investigations of their biochemical function today, the leucocidins remain at the forefront of S. aureus pathogenesis research initiatives. Study of their mode of action is of immediate interest in the realm of therapeutic agent design as well as for studies of bacterial pathogenesis. This review provides an updated perspective on our understanding of the S. aureus leucocidins and their function, specificity, and potential as therapeutic targets.


Asunto(s)
Exotoxinas/metabolismo , Leucocidinas/metabolismo , Staphylococcus aureus/metabolismo , Animales , Exotoxinas/antagonistas & inhibidores , Exotoxinas/química , Exotoxinas/genética , Regulación Bacteriana de la Expresión Génica , Orden Génico , Genoma Bacteriano , Historia del Siglo XIX , Historia del Siglo XX , Interacciones Huésped-Patógeno , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Leucocidinas/antagonistas & inhibidores , Leucocidinas/química , Leucocidinas/genética , Microbiología/historia , Transducción de Señal , Especificidad de la Especie , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/inmunología , Infecciones Estafilocócicas/metabolismo , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Virulencia
19.
PLoS One ; 9(3): e92094, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24643034

RESUMEN

Panton-Valentine leukocidin (PVL), a bicomponent staphylococcal leukotoxin, is involved in the poor prognosis of necrotizing pneumonia. The present study aimed to elucidate the binding mechanism of PVL and in particular its cell-binding domain. The class S component of PVL, LukS-PV, is known to ensure cell targeting and exhibits the highest affinity for the neutrophil membrane (Kd∼10(-10) M) compared to the class F component of PVL, LukF-PV (Kd∼10(-9) M). Alanine scanning mutagenesis was used to identify the residues involved in LukS-PV binding to the neutrophil surface. Nineteen single alanine mutations were performed in the rim domain previously described as implicated in cell membrane interactions. Positions were chosen in order to replace polar or exposed charged residues and according to conservation between leukotoxin class S components. Characterization studies enabled to identify a cluster of residues essential for LukS-PV binding, localized on two loops of the rim domain. The mutations R73A, Y184A, T244A, H245A and Y250A led to dramatically reduced binding affinities for both human leukocytes and undifferentiated U937 cells expressing the C5a receptor. The three-dimensional structure of five of the mutants was determined using X-ray crystallography. Structure analysis identified residues Y184 and Y250 as crucial in providing structural flexibility in the receptor-binding domain of LukS-PV.


Asunto(s)
Toxinas Bacterianas/química , Exotoxinas/química , Leucocidinas/química , Mutación , Neutrófilos/química , Tirosina/química , Alanina/química , Alanina/genética , Secuencia de Aminoácidos , Toxinas Bacterianas/genética , Sitios de Unión , Línea Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Exotoxinas/genética , Expresión Génica , Humanos , Cinética , Leucocidinas/genética , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Alineación de Secuencia , Staphylococcus aureus/química , Tirosina/genética
20.
PLoS One ; 9(2): e88134, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24523877

RESUMEN

Staphylococcus aureus is a main cause of bovine mastitis and a major pathogen affecting human health. The emergence and spread of methicillin-resistant Staphylococcus aureus (MRSA) has become a significant concern for both animal health and public health. This study investigated the incidence of MRSA in milk samples collected from dairy cows with clinical mastitis and characterized the MRSA isolates using antimicrobial susceptibility tests and genetic typing methods. In total, 103 S. aureus isolates were obtained from dairy farms in 4 different provinces in China, including Gansu, Shanghai, Sichuan, and Guizhou. Antimicrobial susceptibility testing of these isolates revealed that the resistance rates to penicillin and sulfamethoxazole were high, while the resistance rates to ciprofloxacin and vancomycin were low. Among the 103 isolates, 49 (47.6%) were found to be mecA-positive, indicating the high incidence of MRSA. However, 37 of the 49 mecA-positive isolates were susceptible to oxacillin as determined by antimicrobial susceptibility assays and were thus classified as oxacillin-susceptible mecA-positive S. aureus (OS-MRSA). These isolates could be misclassified as methicillin susceptible Staphylococcus aureus (MSSA) if genetic detection of mecA was not performed. Molecular characterization of selected mecA-positive isolates showed that they were all negative with Panton-Valentine leukocidin (PVL), but belonged to different spa types and SCCmec types. These results indicate that OS-MRSA is common in bovine mastitis in China and underscore the need for genetic methods (in addition to phenotypic tests) to accurately identify MRSA.


Asunto(s)
Proteínas Bacterianas/metabolismo , Mastitis Bovina/microbiología , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Oxacilina/farmacología , Animales , Bovinos , China/epidemiología , Femenino , Genotipo , Incidencia , Leucocidinas/química , Mastitis Bovina/tratamiento farmacológico , Mastitis Bovina/epidemiología , Pruebas de Sensibilidad Microbiana , Proteínas de Unión a las Penicilinas , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...