Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Virulence ; 9(1): 231-247, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29099326

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Toxinas Bacterianas/metabolismo , Citotoxinas/metabolismo , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Anticuerpos Monoclonales/metabolismo , Toxinas Bacterianas/antagonistas & inhibidores , Citotoxinas/antagonistas & inhibidores , Proteínas Hemolisinas/antagonistas & inhibidores , Proteínas Hemolisinas/metabolismo , Leucocidinas/antagonistas & inhibidores , Leucocidinas/metabolismo , Neutrófilos/inmunología , Neutrófilos/microbiología , Organoides/inmunología , Organoides/microbiología , Organoides/patología , Infecciones Estafilocócicas/prevención & control , Staphylococcus aureus/química
2.
J Infect ; 74(5): 473-483, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28237625

RESUMEN

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.


Asunto(s)
Proteínas Bacterianas/inmunología , Leucocidinas/inmunología , Activación Neutrófila/inmunología , Neutrófilos/inmunología , Antígeno CD11b/análisis , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Humanos , Interleucina-8/análisis , Interleucina-8/inmunología , Interleucina-8/metabolismo , Lipopolisacáridos/inmunología , Modelos Biológicos , Neutrófilos/microbiología , Staphylococcus aureus/inmunología
3.
Virulence ; 8(7): 1203-1215, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28103139

RESUMEN

Klebsiella pneumoniae ST258 is a globally distributed multi-drug resistant pathogen responsible for severe invasive infections. In this study, the different virulence potential of K. pneumoniae ST258 isolates in endotoxin susceptible versus resistant animal models was shown. Furthermore, ST258 clinical isolates were found highly sensitive to the bactericidal effect of naive animal and human serum. These observations imply that LPS, released from the rapidly lysed bacteria, may contribute to the high mortality associated with ST258 bacteremia cases. A humanized version (mAb A1102) of a previously described murine mAb specific for the conserved LPS O-antigen, was tested for endotoxin neutralization. A1102 was able to neutralize TLR-4 activation by ST258-derived LPS in vitro with an efficacy exceeding that of polymyxin B by 3 orders of magnitude. Passive immunization with A1102 afforded a significant level of protection in a galactosamine-sensitized mouse model of endotoxemia, induced by ST258-derived LPS, or upon challenge with live bacteria. Efficacy was retained using an aglycosylated IgG, as well as upon complement depletion, suggesting that Fc-independent endotoxin neutralization may be the main protective mechanism in this model, in spite of the complement-dependent bactericidal and opsonic activities additionally observed for A1102 in vitro. Furthermore, rabbits that are naturally highly susceptible to endotoxin, were also significantly protected by low doses of A1102 when challenged with an ST258 strain. Given this unique mode of action and the high protective efficacy of this mAb, passive immunization, as prophylactic or adjunct therapeutic approach for the treatment of infections caused by ST258 isolates should be considered.


Asunto(s)
Anticuerpos Antibacterianos/inmunología , Anticuerpos Monoclonales/inmunología , Endotoxinas/inmunología , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella pneumoniae/inmunología , Antígenos O/inmunología , Animales , Anticuerpos Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Femenino , Humanos , Inmunización Pasiva , Infecciones por Klebsiella/inmunología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Conejos , Ratas , Ratas Sprague-Dawley
4.
Antimicrob Agents Chemother ; 60(10): 6333-40, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27527081

RESUMEN

Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), especially the USA300 pulsotype, is a frequent cause of skin and soft tissue infections and severe pneumonia. Despite appropriate antibiotic treatment, complications are common and pneumonia is associated with high mortality. S. aureus strains express multiple cytotoxins, including alpha-hemolysin (Hla) and up to five bicomponent leukocidins that specifically target phagocytic cells for lysis. CA-MRSA USA300 strains carry the genes for all six cytotoxins. Species specificity of the leukocidins greatly contributes to the ambiguity regarding their role in S. aureus pathogenesis. We performed a comparative analysis of the leukocidin susceptibility of human, rabbit, and mouse polymorphonuclear leukocytes (PMNs) to assess the translational value of mouse and rabbit S. aureus models. We found that mouse PMNs were largely resistant to LukSF-PV, HlgAB, and HlgCB and susceptible only to LukED, whereas rabbit and human PMNs were highly sensitive to all these cytotoxins. In the rabbit pneumonia model with a USA300 CA-MRSA strain, passive immunization with a previously identified human monoclonal antibody (MAb), Hla-F#5, which cross-neutralizes Hla, LukSF-PV, HlgAB, HlgCB, and LukED, provided full protection, whereas an Hla-specific MAb was only partially protective. In the mouse USA300 CA-MRSA pneumonia model, both types of antibodies demonstrated full protection, suggesting that Hla, but not leukocidin(s), is the principal virulence determinant in mice. As the rabbit recapitulates the high susceptibility to leukocidins characteristic of humans, this species represents a valuable model for assessing novel, cytotoxin-targeting anti-S. aureus therapeutic approaches.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Toxinas Bacterianas/inmunología , Proteínas Hemolisinas/inmunología , Leucocidinas/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Neumonía Necrotizante/prevención & control , Neumonía Estafilocócica/prevención & control , Animales , Anticuerpos Monoclonales/farmacología , Infecciones Comunitarias Adquiridas/microbiología , Modelos Animales de Enfermedad , Femenino , Humanos , Leucocidinas/farmacología , Masculino , Ratones Endogámicos BALB C , Neutrófilos/efectos de los fármacos , Neutrófilos/microbiología , Neumonía Necrotizante/inmunología , Neumonía Necrotizante/microbiología , Neumonía Necrotizante/mortalidad , Neumonía Estafilocócica/inmunología , Conejos
5.
Antimicrob Agents Chemother ; 59(6): 3109-16, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25779571

RESUMEN

The Escherichia coli sequence type 131 (ST131)-O25b:H4 clone has spread worldwide and become responsible for a significant proportion of multidrug-resistant extraintestinal infections. We generated humanized monoclonal antibodies (MAbs) that target the lipopolysaccharide O25b antigen conserved within this lineage. These MAbs bound to the surface of live bacterial cells irrespective of the capsular type expressed. In a serum bactericidal assay in vitro, MAbs induced >95% bacterial killing in the presence of human serum as the complement source. Protective efficacy at low antibody doses was observed in a murine model of bacteremia. The mode of action in vivo was investigated by using aglycosylated derivatives of the protective MAbs. The significant binding to live E. coli cells and the in vitro and in vivo efficacy were corroborated in assays using bacteria grown in human serum to mimic relevant clinical conditions. Given the dry pipeline of novel antibiotics against multidrug-resistant Gram-negative pathogens, passive immunization with bactericidal antibodies offers a therapeutic alternative to control infections caused by E. coli ST131-O25b:H4.


Asunto(s)
Antibacterianos/farmacología , Antibacterianos/uso terapéutico , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Escherichia coli/efectos de los fármacos , Escherichia coli/metabolismo , Antígenos O/metabolismo , Animales , Farmacorresistencia Bacteriana Múltiple , Infecciones por Escherichia coli/tratamiento farmacológico , Infecciones por Escherichia coli/microbiología , Femenino , Ratones
6.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...