Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 68
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32479075

RESUMEN

Optimization of small-molecule probes or drugs is a synthetically lengthy, challenging, and resource-intensive process. Lack of automation and reliance on skilled medicinal chemists is cumbersome in both academic and industrial settings. Here, we demonstrate a high-throughput hit-to-lead process based on the biocompatible sulfur(VI) fluoride exchange (SuFEx) click chemistry. A high-throughput screening hit benzyl (cyanomethyl)carbamate (Ki = 8 µM) against a bacterial cysteine protease SpeB was modified with a SuFExable iminosulfur oxydifluoride [RN═S(O)F2] motif, rapidly diversified into 460 analogs in overnight reactions, and the products were directly screened to yield drug-like inhibitors with 480-fold higher potency (Ki = 18 nM). We showed that the improved molecule is active in a bacteria-host coculture. Since this SuFEx linkage reaction succeeds on picomole scale for direct screening, we anticipate our methodology can accelerate the development of robust biological probes and drug candidates.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/farmacología , Exotoxinas/antagonistas & inhibidores , Compuestos de Azufre/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico , Química Clic , Cristalografía por Rayos X , Inhibidores de Cisteína Proteinasa/metabolismo , Inhibidores de Cisteína Proteinasa/toxicidad , Descubrimiento de Drogas , Exotoxinas/química , Exotoxinas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Jurkat , Microsomas Hepáticos/metabolismo , Prueba de Estudio Conceptual , Unión Proteica
2.
J Infect Chemother ; 24(5): 383-388, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29409694

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/sangre , Infecciones Comunitarias Adquiridas/tratamiento farmacológico , Exotoxinas/antagonistas & inhibidores , Exotoxinas/sangre , Inmunoglobulinas Intravenosas/administración & dosificación , Leucocidinas/antagonistas & inhibidores , Leucocidinas/sangre , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Infecciones Estafilocócicas/tratamiento farmacológico , Anticuerpos Neutralizantes/inmunología , Proteínas Bacterianas/genética , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Tampones (Química) , Infecciones Comunitarias Adquiridas/inmunología , Exotoxinas/inmunología , Humanos , Leucocidinas/inmunología , Staphylococcus aureus Resistente a Meticilina/genética , Staphylococcus aureus Resistente a Meticilina/aislamiento & purificación , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Infecciones Estafilocócicas/inmunología
3.
J Membr Biol ; 249(4): 503-11, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27039399

RESUMEN

The repeats-in-toxin family of toxins includes proteins produced by Gram negative bacteria such as Escherichia coli (α-hemolysin), Bordetella pertussis (adenylate cyclase toxin), and Aggregatibacter actinomycetemcomitans (LtxA), which contribute to the pathogenesis of these organisms by killing host cells. In the case of LtxA produced by A. actinomycetemcomitans, white blood cells are targeted, allowing the bacteria to avoid clearance by the host immune system. In its association with target cells, LtxA binds to a receptor, lymphocyte function-associated antigen-1, as well as membrane lipids and cholesterol, before being internalized via a lysosomal-mediated pathway. The motivation for this project comes from our discovery that DRAQ5™, a membrane-permeable nuclear stain, prevents the internalization of LtxA in a Jurkat T cell line. We hypothesized that DRAQ5™, in crossing the plasma membrane, alters the properties of the membrane to inhibit LtxA internalization. To investigate how DRAQ5™ interacts with the lipid membrane to prevent LtxA internalization, we used studied DRAQ5™-mediated membrane changes in model membranes using a variety of techniques, including differential scanning calorimetry and fluorescence spectroscopy. Our results suggest that DRAQ5™ inhibits the activity of LtxA by decreasing the fluidity of the cellular lipid membrane, which decreases LtxA binding. These results present an interesting possible anti-virulence strategy; by altering bacterial toxin activity by modifying membrane fluidity, it may be possible to inhibit the pathogenicity of A. actinomycetemcomitans.


Asunto(s)
Antraquinonas/farmacología , Antitoxinas/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/metabolismo , Antraquinonas/metabolismo , Antitoxinas/metabolismo , Toxinas Bacterianas/toxicidad , Línea Celular , Membrana Celular/química , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Exotoxinas/toxicidad , Humanos , Fluidez de la Membrana/efectos de los fármacos , Temperatura
4.
Biochemistry ; 54(28): 4365-73, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26132413

RESUMEN

The secreted Streptococcus pyogenes cysteine protease SpeB is implicated in host immune system evasion and bacterial virulence. We present a small molecule inhibitor of SpeB 2477 identified from a high-throughput screen based on the hydrolysis of a fluorogenic peptide substrate Ac-AIK-AMC. 2477 inhibits other SpeB-related proteases but not human caspase-3, suggesting that the molecule targets proteases with the papain-like structural fold. A 1.59 Å X-ray crystal structure of 2477 bound to the SpeB active site reveals the mechanism of inhibition and the essential constituents of 2477 necessary for binding. An assessment against a panel of 2477 derivatives confirms our structural findings and shows that a carbamate and nitrile on 2477 are required for SpeB inhibition, as these moieties provide an extensive network of electrostatic and hydrogen-bonding interactions with SpeB active site residues. Surprisingly, despite 2477 having a reduced inhibitory potential against papain, the majority of 2477-related compounds inhibit papain to a much greater and broader extent than SpeB. These findings indicate that SpeB is more stringently selective than papain for this panel of small molecule inhibitors. On the basis of our structural and biochemical characterization, we propose modifications to 2477 for subsequent rounds of inhibitor design that will impart specificity to SpeB over other papain-like proteases, including alterations of the compound to exploit the differences in CA protease active site pocket sizes and electrostatics.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/química , Inhibidores de Cisteína Proteinasa/farmacología , Exotoxinas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología , Streptococcus pyogenes/enzimología , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Dominio Catalítico/efectos de los fármacos , Cristalografía por Rayos X , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/metabolismo , Exotoxinas/química , Exotoxinas/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Conformación Proteica/efectos de los fármacos , Infecciones Estreptocócicas/tratamiento farmacológico , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/efectos de los fármacos
5.
World J Microbiol Biotechnol ; 31(10): 1565-73, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26193949

RESUMEN

Staphylococcus aureus (S. aureus) causes a wide variety of infections, which are of major concern worldwide. S. aureus produces multiple virulence factors, resulting in food infection and poisoning. These virulence factors include hyaluronidases, proteases, coagulases, lipases, deoxyribonucleases and enterotoxins. Among the extracellular proteins produced by S. aureus that contribute to pathogenicity, the exotoxins α-hemolysin, staphylococcal enterotoxin A (SEA) and staphylococcal enterotoxin B (SEB) are thought to be of major significance. Totarol, a plant extract, has been revealed to inhibit the proliferation of several pathogens effectively. However, there are no reports on the effects of totarol on the production of α-hemolysin, SEA or SEB secreted by S. aureus. The aim of this study was to evaluate the effects of totarol on these three exotoxins. Hemolysis assay, western blotting and real-time reverse transcriptase-PCR assay were performed to identify the influence of graded subinhibitory concentrations of totarol on the production of α-hemolysin and the two major enterotoxins, SEA and SEB, by S. aureus in a dose-dependent manner. Moreover, an enzyme linked immunosorbent assay showed that the TNF-α production of RAW264.7 cells stimulated by S. aureus supernatants was inhibited by subinhibitory concentrations of totarol. Form the data, we propose that totarol could potentially be used as a promising natural compound in the food and pharmaceutical industries.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Diterpenos/metabolismo , Enterotoxinas/antagonistas & inhibidores , Inhibidores Enzimáticos/metabolismo , Exotoxinas/antagonistas & inhibidores , Proteínas Hemolisinas/antagonistas & inhibidores , Staphylococcus aureus/metabolismo , Abietanos , Animales , Western Blotting , Línea Celular , Hemólisis , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo
6.
Biochem Biophys Res Commun ; 444(3): 433-8, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24472539

RESUMEN

Staphylococcus aureus is a versatile pathogen capable of causing a broad spectrum of diseases ranging from superficial skin infections to life threatening conditions such as endocarditis, septicemia, pneumonia and toxic shock syndrome. In vitro and in vivo studies identified an exotoxin, α-toxin, as a major cause of S. aureus toxicity. Because S. aureus has rapidly evolved resistance to a number of antibiotics, including methicillin, it is important to identify new therapeutic strategies, other than antibiotics, for inhibiting the harmful effects of this pathogen. Aptamers are single-stranded DNA or RNA oligonucleotides with three-dimensional folded conformations that bind with high affinity and selectivity to targets and modulate their biological functions. The goal of this study was to isolate DNA aptamers that specifically inhibit the cytotoxic activity of α-toxin. After 10 rounds of Systematic Evolution of Ligands by EXponential Enrichment (SELEX), 49 potential anti-α-toxin aptamers were identified. In vitro neutralization assays demonstrated that 4 of these 49 aptamers, AT-27, AT-33, AT-36, and AT-49, significantly inhibited α-toxin-mediated cell death in Jurkat T cells. Furthermore, RT-PCR analysis revealed that α-toxin increased the transcription of the inflammatory cytokines TNF-α and IL-17 and that anti-α-toxin aptamers AT-33 and AT-36 inhibited the upregulation of these genes. Collectively, the data suggest the feasibility of generating functionally effective aptamers against α-toxin for treatment of S. aureus infections.


Asunto(s)
Aptámeros de Nucleótidos/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Exotoxinas/antagonistas & inhibidores , Proteínas Hemolisinas/antagonistas & inhibidores , Staphylococcus aureus/química , Humanos , Células Jurkat , Técnica SELEX de Producción de Aptámeros
7.
Biochem J ; 450(3): 559-71, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23282185

RESUMEN

PVL (Panton-Valentine leukocidin) and other Staphylococcus aureus ß-stranded pore-forming toxins are important virulence factors involved in various pathologies that are often necrotizing. The present study characterized leukotoxin inhibition by selected SCns (p-sulfonato-calix[n]arenes): SC4, SC6 and SC8. These chemicals have no toxic effects on human erythrocytes or neutrophils, and some are able to inhibit both the activity of and the cell lysis by leukotoxins in a dose-dependent manner. Depending on the type of leukotoxins and SCns, flow cytometry revealed IC50 values of 6-22 µM for Ca2+ activation and of 2-50 µM for cell lysis. SCns were observed to affect membrane binding of class S proteins responsible for cell specificity. Electrospray MS and surface plasmon resonance established supramolecular interactions (1:1 stoichiometry) between SCns and class S proteins in solution, but not class F proteins. The membrane-binding affinity of S proteins was Kd=0.07-6.2 nM. The binding ability was completely abolished by SCns at different concentrations according to the number of benzenes (30-300 µM; SC8>SC6≫SC4). The inhibitory properties of SCns were also observed in vivo in a rabbit model of PVL-induced endophthalmitis. These calixarenes may represent new therapeutic avenues aimed at minimizing inflammatory reactions and necrosis due to certain virulence factors.


Asunto(s)
Calixarenos/farmacología , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Staphylococcus aureus/metabolismo , Animales , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/metabolismo , Calixarenos/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Proteínas Hemolisinas/antagonistas & inhibidores , Proteínas Hemolisinas/metabolismo , Humanos , Sustancias Macromoleculares/metabolismo , Modelos Biológicos , Fenoles/metabolismo , Fenoles/farmacología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Conejos , Esfingomielina Fosfodiesterasa/antagonistas & inhibidores , Esfingomielina Fosfodiesterasa/metabolismo , Staphylococcus aureus/patogenicidad , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/metabolismo
8.
J Infect Dis ; 207(12): 1869-77, 2013 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-23493729

RESUMEN

Staphylococcus aureus and group A Streptococcus pyogenes (GAS) express superantigen (SAg) exotoxin proteins capable of inducing lethal shock. To induce toxicity, SAgs must bind not only to the major histocompatibility complex II molecule of antigen-presenting cells and the variable ß chain of the T-cell receptor but also to the dimer interface of the T-cell costimulatory receptor CD28. Here, we show that the CD28-mimetic peptide AB103 (originally designated "p2TA") protects mice from lethal challenge with streptococcal exotoxin A, as well as from lethal GAS bacterial infection in a murine model of necrotizing soft-tissue infection. Administration of a single dose of AB103 increased survival when given up to 5 hours after infection, reduced inflammatory cytokine expression and bacterial burden at the site of infection, and improved muscle inflammation in a dose-dependent manner, without compromising cellular and humoral immunity. Thus, AB103 merits further investigation as a potential therapeutic in SAg-mediated necrotizing soft-tissue infection.


Asunto(s)
Antígenos CD28/inmunología , Péptidos/uso terapéutico , Choque Séptico/tratamiento farmacológico , Infecciones Estreptocócicas/tratamiento farmacológico , Streptococcus pyogenes/inmunología , Superantígenos/toxicidad , Animales , Anticuerpos Antibacterianos/inmunología , Antígenos CD28/antagonistas & inhibidores , Antígenos CD28/metabolismo , Proliferación Celular , Recuento de Colonia Microbiana , Citocinas/sangre , Citocinas/inmunología , Relación Dosis-Respuesta a Droga , Exotoxinas/antagonistas & inhibidores , Exotoxinas/inmunología , Exotoxinas/toxicidad , Femenino , Inmunidad Celular , Ratones , Ratones Endogámicos BALB C , Péptidos/farmacología , Choque Séptico/inmunología , Choque Séptico/microbiología , Transducción de Señal , Infecciones de los Tejidos Blandos/tratamiento farmacológico , Infecciones de los Tejidos Blandos/microbiología , Organismos Libres de Patógenos Específicos , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología , Streptococcus pyogenes/metabolismo , Superantígenos/inmunología , Factores de Virulencia
9.
Antimicrob Agents Chemother ; 57(11): 5432-7, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23959313

RESUMEN

Gram-positive bacteria cause serious human illnesses through combinations of cell surface and secreted virulence factors. We initiated studies with four of these organisms to develop novel topical antibacterial agents that interfere with growth and exotoxin production, focusing on menaquinone analogs. Menadione, 1,4-naphthoquinone, and coenzymes Q1 to Q3 but not menaquinone, phylloquinone, or coenzyme Q10 inhibited the growth and to a greater extent exotoxin production of Staphylococcus aureus, Bacillus anthracis, Streptococcus pyogenes, and Streptococcus agalactiae at concentrations of 10 to 200 µg/ml. Coenzyme Q1 reduced the ability of S. aureus to cause toxic shock syndrome in a rabbit model, inhibited the growth of four Gram-negative bacteria, and synergized with another antimicrobial agent, glycerol monolaurate, to inhibit S. aureus growth. The staphylococcal two-component system SrrA/B was shown to be an antibacterial target of coenzyme Q1. We hypothesize that menaquinone analogs both induce toxic reactive oxygen species and affect bacterial plasma membranes and biosynthetic machinery to interfere with two-component systems, respiration, and macromolecular synthesis. These compounds represent a novel class of potential topical therapeutic agents.


Asunto(s)
Antibacterianos/farmacología , Bacillus anthracis/efectos de los fármacos , Staphylococcus aureus/efectos de los fármacos , Streptococcus agalactiae/efectos de los fármacos , Streptococcus pyogenes/efectos de los fármacos , Vitamina K 2/farmacología , Administración Tópica , Animales , Bacillus anthracis/crecimiento & desarrollo , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Membrana Celular/efectos de los fármacos , Sinergismo Farmacológico , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Humanos , Lauratos/farmacología , Monoglicéridos/farmacología , Conejos , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/metabolismo , Choque Séptico/tratamiento farmacológico , Choque Séptico/microbiología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Streptococcus agalactiae/crecimiento & desarrollo , Streptococcus pyogenes/crecimiento & desarrollo
10.
Immunopharmacol Immunotoxicol ; 35(2): 296-303, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23384342

RESUMEN

CONTEXT: Liver injury can be induced by various hepatotoxicants, including Pseudomonas aeruginosa exotoxin A (PEA). Our previous study indicated that PEA-induced rat hepatotoxicity was T cells and Kupffer cells dependent. Several reports have demonstrated that non-toxic doses of bacterial lipopolysaccharide (LPS) can protect liver against the chemicals-induced toxicity such as acetaminophen and concanavalin-A. OBJECTIVE: This study aimed to investigate the protecting mechanisms of LPS on PEA-induced hepatotoxicity. RESULTS: Rats pretreated with LPS (40 µg/kg, 12 h before PEA admission) significantly decreased animal mortality, serum enzyme (ALT, AST and T-bil) activities, histopathological changes and hepatocytes apoptosis following challenge with PEA. The concentrations of tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ) and interleukin-2 (IL-2) were reduced, but IL-6 and IL-10 were increased in the serum. In addition, prior treatment of these LPS-pretreated rats with gadolinium chloride (GdCl3), a selective Kupffer cell depletion agent, markedly enhanced liver injury after PEA administration. In contrast, the pretreatment of LPS to T-cell deficient athymic nude rats still display significant attenuation of PEA-induced liver injury. This observation further confirmed our hypothesis that LPS ameliorate PEA-hepatotoxicity was through Kupffer cells but not T cells. Moreover, LPS-induced hepatoprotection ability was neutralized by co-treatment with anti-TNF-α antibodies, but not with anti-IFN-γ antibodies. Finally, replacement of LPS with RS-LPS (Rhodobacter sphaeroides LPS), a Toll like receptor-4 (TLR-4) antagonist, resulted in severe hepatotoxicity. CONCLUSION: These results suggested that Kupffer cells, TNF-α and TLR-4 play central mediator roles during the hepatoprotection against PEA-induced hepatotoxicity conferred by LPS.


Asunto(s)
ADP Ribosa Transferasas/antagonistas & inhibidores , ADP Ribosa Transferasas/toxicidad , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Exotoxinas/antagonistas & inhibidores , Exotoxinas/toxicidad , Lipopolisacáridos/farmacología , Hígado/efectos de los fármacos , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/toxicidad , Alanina Transaminasa/metabolismo , Animales , Apoptosis/efectos de los fármacos , Aspartato Aminotransferasas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Interacciones Farmacológicas , Gadolinio/farmacología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Interferón gamma/metabolismo , Interleucinas/metabolismo , Macrófagos del Hígado/efectos de los fármacos , Macrófagos del Hígado/metabolismo , Hígado/enzimología , Masculino , Pseudomonas/metabolismo , Ratas , Ratas Desnudas , Ratas Wistar , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Exotoxina A de Pseudomonas aeruginosa
11.
Microb Drug Resist ; 27(5): 602-615, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33983855

RESUMEN

Incidence of drug resistance in clinical isolates of methicillin-resistant Staphylococcus aureus (MRSA) is attributed to its diverse repertoire of virulence factors. Of these virulence determinants, Panton-Valentine Leukocidin (PVL) has been experimentally validated as a prospective drug target due to its conspicuous and comprehensive role in nosocomial infections. This study encompassed an in silico approach to elucidate the antimicrobial potentiality of human cathelicidin LL-37 against PVL toxin of MRSA. Molecular docking studies of LL-37 and its segments with the PVL toxin subunits LukS and LukF were carried out using PatchDock server and the results were refined using FireDock server. The paramount ligand-receptor combination was selected and analyzed based on diverse parametric attributes and compared with the commercial inhibitors of PVL viz. Andrimid, Beclobrate, Beta-sitosterol, Diathymosulfone, and Probucol to determine the most potent inhibitor among them. Our results elucidated that the interaction of LL-37 with the LukS subunit of PVL toxin (minimum global energy of -61.82 kcal/mol) depicted 34 molecular interactions, while the commercial PVL inhibitors depicted fewer and insubstantial interactions. SWISS-ADME (Absorption, Distribution, Metabolism, and Excretion) and ToxinPred analysis of LL-37 further corroborated its null potency of toxicity in systemic milieu. The results obtained may credit this study as basis for the development of LL-37 as a potential inhibitor against virulent MRSA toxins, thereby exalting the treatment regimes for nosocomial infections in health care facilities worldwide.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Exotoxinas/antagonistas & inhibidores , Leucocidinas/antagonistas & inhibidores , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Péptidos Catiónicos Antimicrobianos/farmacocinética , Humanos , Pruebas de Sensibilidad Microbiana , Simulación del Acoplamiento Molecular , Catelicidinas
12.
J Pharm Pharmacol ; 73(4): 505-514, 2021 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-33793838

RESUMEN

OBJECTIVES: We and others have previously shown that epigallocatechin gallate (EGCg) inhibits the activity of an important virulence factor, leukotoxin (LtxA), produced by the oral bacterium Aggregatibacter actinomycetemcomitans, suggesting the potential use of this molecule as an anti-virulence strategy to treat periodontal infections. Here, we sought to better understand the effects of EGCg on toxin secretion and A. actinomycetemcomitans pathogenicity in a co-culture model. METHODS: We used a quantitative immunoblot assay to determine the concentrations of LtxA in the bacterial supernatant and on the bacterial cell surface. Using a co-culture model, consisting of A. actinomycetemcomitans and THP-1 cells, we studied the impact of EGCg-mediated changes in LtxA secretion on the toxicity of A. actinomycetemcomitans. KEY FINDINGS: EGCg increased production of LtxA and changed the localization of secreted LtxA from the supernatant to the surface of the bacterial cells. In the co-culture model, a single low dose of EGCg did not protect host THP-1 cells from A. actinomycetemcomitans-mediated cytotoxicity, but a multiple dosing strategy had improved effects. CONCLUSIONS: Together, these results demonstrate that EGCg has important, but complicated, effects on toxin secretion and activity; new dosing strategies and comprehensive model systems may be required to properly develop these anti-virulence activities.


Asunto(s)
Aggregatibacter actinomycetemcomitans , Catequina/análogos & derivados , Exotoxinas , Periodontitis , Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Aggregatibacter actinomycetemcomitans/metabolismo , Aggregatibacter actinomycetemcomitans/patogenicidad , Antibacterianos/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/metabolismo , Catequina/farmacología , Técnicas de Cocultivo/métodos , Relación Dosis-Respuesta a Droga , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Humanos , Periodontitis/tratamiento farmacológico , Periodontitis/microbiología , Virulencia/efectos de los fármacos
13.
J Clin Microbiol ; 48(5): 1952-5, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20129956
14.
J Periodontal Res ; 45(1): 123-8, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19602106

RESUMEN

BACKGROUND AND OBJECTIVE: Quinol peroxidase (QPO) catalyzes peroxidase activity using quinol in the respiratory chain as a substrate. Quinol peroxidase is essential for the secretion of leukotoxin (LtxA), which destroys leukocytes and erythrocytes in humans and is one of the major virulence factors of Aggregatibacter actinomycetemcomitans, which is associated with localized aggressive periodontitis. In the present study, we aimed to find a highly potent QPO inhibitor to attenuate the virulence of A. actinomycetemcomitans. MATERIAL AND METHODS: For screening of QPO inhibitors, QPO activity was measured kinetically by SpectraMax Plus with 96-well UV plates. Three hundred compounds in the Kitasato Institute for Life Sciences Chemical Library were screened. Secretion of LtxA in the culture supernatant was examined by sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Cytotoxicity against human promyelocytic leukemia cell line (HL-60) cells from the culture supernatant was measured by Trypan Blue exclusion test. RESULTS: The present study characterized ascofuranone as a highly potent inhibitor of QPO (K(i) = 9.557 +/- 0.865 nm). Ascofuranone inhibited secretion of LtxA by A. actinomycetemcomitans in a dose-dependent manner, making A. actinomycetemcomitans less pathogenic to HL-60 cells. CONCLUSION: Quinol peroxidase inhibitors are promising candidates as alternative drugs for the treatment and prevention of the onset of localized aggressive periodontitis. Using ascofuranone as a seed compound, further study of QPO inhibitors could provide novel chemotherapeutic strategies for controlling localized aggressive periodontitis.


Asunto(s)
Aggregatibacter actinomycetemcomitans/efectos de los fármacos , Toxinas Bacterianas/antagonistas & inhibidores , Citotoxinas/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Exotoxinas/antagonistas & inhibidores , Hidroquinonas/antagonistas & inhibidores , Peroxidasas/antagonistas & inhibidores , Sesquiterpenos/farmacología , Aggregatibacter actinomycetemcomitans/crecimiento & desarrollo , Aggregatibacter actinomycetemcomitans/patogenicidad , Técnicas Bacteriológicas , Recuento de Colonia Microbiana , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/administración & dosificación , Escherichia coli/efectos de los fármacos , Células HL-60 , Humanos , Pruebas de Sensibilidad Microbiana , Estrés Oxidativo/efectos de los fármacos , Sesquiterpenos/administración & dosificación , Streptococcus gordonii/efectos de los fármacos , Virulencia/efectos de los fármacos , Factores de Virulencia/antagonistas & inhibidores
15.
J Leukoc Biol ; 108(3): 967-981, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32531864

RESUMEN

Staphylococcus aureus causes necrotizing pneumonia by secreting toxins such as leukocidins that target front-line immune cells. The mechanism by which leukocidins kill innate immune cells and trigger inflammation during S. aureus lung infection, however, remains unresolved. Here, we explored human-induced pluripotent stem cell-derived macrophages (hiPSC-dMs) to study the interaction of the leukocidins Panton-Valentine leukocidin (PVL) and LukAB with lung macrophages, which are the initial leukocidin targets during S. aureus lung invasion. hiPSC-dMs were susceptible to the leukocidins PVL and LukAB and both leukocidins triggered NLPR3 inflammasome activation resulting in IL-1ß secretion. hiPSC-dM cell death after LukAB exposure, however, was only temporarily dependent of NLRP3, although NLRP3 triggered marked cell death after PVL treatment. CRISPR/Cas9-mediated deletion of the PVL receptor, C5aR1, protected hiPSC-dMs from PVL cytotoxicity, despite the expression of other leukocidin receptors, such as CD45. PVL-deficient S. aureus had reduced ability to induce lung IL-1ß levels in human C5aR1 knock-in mice. Unexpectedly, inhibiting NLRP3 activity resulted in increased wild-type S. aureus lung burdens. Our findings suggest that NLRP3 induces macrophage death and IL-1ß secretion after PVL exposure and controls S. aureus lung burdens.


Asunto(s)
Proteínas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/antagonistas & inhibidores , Exotoxinas/antagonistas & inhibidores , Células Madre Pluripotentes Inducidas/citología , Leucocidinas/antagonistas & inhibidores , Macrófagos/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/efectos de los fármacos , Staphylococcus aureus , Animales , Antígeno CD11b/inmunología , Sistemas CRISPR-Cas , Diferenciación Celular , Células Cultivadas , Exotoxinas/deficiencia , Técnicas de Sustitución del Gen , Humanos , Interleucina-1beta/metabolismo , Antígenos Comunes de Leucocito/fisiología , Pulmón/inmunología , Pulmón/microbiología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Monocitos/citología , Fragmentos de Péptidos/inmunología , Neumonía Estafilocócica/inmunología , Subunidades de Proteína , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptor de Anafilatoxina C5a/fisiología , Proteínas Recombinantes/metabolismo , Staphylococcus aureus/fisiología
16.
Bioorg Med Chem ; 17(14): 5139-45, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19540764

RESUMEN

Here, we report the results of a quantitative high-throughput screen (qHTS) measuring the endocytosis and translocation of a beta-lactamase-fused-lethal factor and the identification of small molecules capable of obstructing the process of anthrax toxin internalization. Several small molecules protect RAW264.7 macrophages and CHO cells from anthrax lethal toxin and protected cells from an LF-Pseudomonas exotoxin fusion protein and diphtheria toxin. Further efforts demonstrated that these compounds impaired the PA heptamer pre-pore to pore conversion in cells expressing the CMG2 receptor, but not the related TEM8 receptor, indicating that these compounds likely interfere with toxin internalization.


Asunto(s)
Carbunco/tratamiento farmacológico , Antibacterianos/farmacología , Antígenos Bacterianos/metabolismo , Bacillus anthracis/metabolismo , Toxinas Bacterianas/metabolismo , Endocitosis/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antibacterianos/química , Bacillus anthracis/efectos de los fármacos , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/antagonistas & inhibidores , Células CHO , Muerte Celular/efectos de los fármacos , Corynebacterium diphtheriae/metabolismo , Cricetinae , Cricetulus , Toxina Diftérica/antagonistas & inhibidores , Toxina Diftérica/metabolismo , Exotoxinas/antagonistas & inhibidores , Exotoxinas/metabolismo , Macrófagos/citología , Macrófagos/efectos de los fármacos , Ratones , Pseudomonas/metabolismo , Bibliotecas de Moléculas Pequeñas/química
17.
Toxins (Basel) ; 11(11)2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31739483

RESUMEN

Aggregatibacter actinomycetemcomitans is a Gram-negative bacterium that has been strongly associated with localized aggressive periodontitis. The capacity of A. actinomycetemcomitans to produce a leukotoxin (LtxA) that activates pyroptosis in macrophages and induces the release of endogenous danger signals is thought to play a key role in the disease process. The aim of the present study was to investigate the effects of cranberry proanthocyanidins (PACs) on gene expression and cytotoxic activities of LtxA. We showed that cranberry PACs dose-dependently attenuate the expression of genes making up the leukotoxin operon, including ltxB and ltxC, in the two strains of A. actinomycetemcomitans tested. Cranberry PACs (≥62.5 µg/mL) protected macrophages against the cytotoxic effect of purified LtxA. Moreover, cranberry PACs reduced caspase-1 activation in LtxA-treated macrophages and consequently decreased the release of both IL-1ß and IL-18, which are known as damage-associated molecular patterns (DAMPs) and contribute to the progression of periodontitis by increasing cell migration and osteoclastogenesis. In addition, cranberry PACs reduced the expression of genes encoding the P2X7 receptor and NALP3 (NACHT, LRR and PYD domains-containing protein 3), which play key roles in pore formation and cell death. Lastly, cranberry PACs blocked the binding of LtxA to macrophages and consequently reduced the LtxA-mediated cytotoxicity. In summary, the present study showed that cranberry PACs reduced LtxA gene expression in A. actinomycetemcomitans and neutralized the cytolytic and pro-inflammatory responses of human macrophages treated with LtxA. Given these properties, cranberry PACs may represent promising molecules for prevention and treatment of the aggressive form of periodontitis caused by A. actinomycetemcomitans.


Asunto(s)
Aggregatibacter actinomycetemcomitans/química , Exotoxinas/antagonistas & inhibidores , Proantocianidinas/química , Vaccinium macrocarpon/química , Exotoxinas/farmacología , Humanos , Células U937
18.
Sci Rep ; 9(1): 14928, 2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31624289

RESUMEN

Targeting bacterial virulence factors directly provides a new paradigm for the intervention and treatment of bacterial diseases. Pseudomonas aeruginosa produces a myriad of virulence factors to cause fatal diseases in humans. In this study, human single-chain antibodies (HuscFvs) that bound to P. aeruginosa exotoxin A (ETA) were generated by phage display technology using recombinant ETA, ETA-subdomains and the synthetic peptide of the ETA-catalytic site as baits for selecting ETA-bound-phages from the human-scFv phage display library. ETA-bound HuscFvs derived from three phage-transfected E. coli clones neutralized the ETA-induced mammalian cell apoptosis. Computerized simulation demonstrated that these HuscFvs used several residues in their complementarity-determining regions (CDRs) to form contact interfaces with the critical residues in ETA-catalytic domain essential for ADP-ribosylation of eukaryotic elongation factor 2, which should consequently rescue ETA-exposed-cells from apoptosis. The HuscFv-treated ETA-exposed cells also showed decremented apoptosis-related genes, i.e., cas3 and p53. The effective HuscFvs have high potential for future evaluation in animal models and clinical trials as a safe, novel remedy for the amelioration of exotoxin A-mediated pathogenesis. HuscFvs may be used either singly or in combination with the HuscFv cognates that target other P. aeruginosa virulence factors as an alternative therapeutic regime for difficult-to-treat infections.


Asunto(s)
ADP Ribosa Transferasas/antagonistas & inhibidores , Antibacterianos/farmacología , Toxinas Bacterianas/antagonistas & inhibidores , Exotoxinas/antagonistas & inhibidores , Infecciones por Pseudomonas/tratamiento farmacológico , Pseudomonas aeruginosa/efectos de los fármacos , Anticuerpos de Cadena Única/farmacología , Factores de Virulencia/antagonistas & inhibidores , ADP Ribosa Transferasas/genética , ADP Ribosa Transferasas/inmunología , ADP Ribosa Transferasas/metabolismo , Antibacterianos/inmunología , Antibacterianos/uso terapéutico , Apoptosis/efectos de los fármacos , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Dominio Catalítico/genética , Regiones Determinantes de Complementariedad/inmunología , Regiones Determinantes de Complementariedad/farmacología , Exotoxinas/genética , Exotoxinas/inmunología , Exotoxinas/metabolismo , Células HeLa , Humanos , Simulación del Acoplamiento Molecular , Biblioteca de Péptidos , Infecciones por Pseudomonas/microbiología , Pseudomonas aeruginosa/inmunología , Pseudomonas aeruginosa/patogenicidad , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/uso terapéutico , Factores de Virulencia/genética , Factores de Virulencia/inmunología , Factores de Virulencia/metabolismo , Exotoxina A de Pseudomonas aeruginosa
19.
J Med Microbiol ; 68(2): 255-262, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30556803

RESUMEN

PURPOSE: Potent extracellular toxins including alpha-haemolysin, Panton-Valentine leukocidin (PVL) and toxic-shock syndrome toxin 1 (TSST-1) significantly contribute to Staphylococcus aureus pathogenesis, thus, toxin suppression is a primary focus in treatment of staphylococcal disease. S. aureus maintains complex strategies to regulate toxin expression and previous data have demonstrated that subinhibitory concentrations of beta-lactam antibiotics can adversely increase S. aureus exotoxin production. The current study evaluates the effects of subinhibitory concentrations of tedizolid, a second-generation oxazolidinone derivative, on expression of staphylococcal exotoxins in both methicillin-resistant and methicillin-sensitive S. aureus. METHODOLOGY: S. aureus exotoxin expression levels were compared at 12 and 24 h following treatment with tedizolid, linezolid, nafcillin or vehicle control. RESULTS: Our findings show that the level of antibiotic required to alter toxin production was strain-dependent and corresponds with the quantity of toxin produced, but both tedizolid and linezolid could effectively reduce expression of alpha-haemolysin, PVL and TSST-1 toxin at subinhibitory concentrations. In contrast, nafcillin showed less attenuation and, in some S. aureus strains, led to an increase in toxin expression. Tedizolid consistently inhibited toxin production at a lower overall drug concentration than comparator agents. CONCLUSION: Together, our data support that tedizolid has the potential to improve outcomes of infection due to its superior ability to inhibit S. aureus growth and attenuate exotoxin production.


Asunto(s)
Antibacterianos/farmacología , Toxinas Bacterianas/biosíntesis , Meticilina/farmacología , Oxazolidinonas/farmacología , Staphylococcus aureus/efectos de los fármacos , Tetrazoles/farmacología , Animales , Antibacterianos/administración & dosificación , Toxinas Bacterianas/análisis , Toxinas Bacterianas/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Enterotoxinas/análisis , Enterotoxinas/antagonistas & inhibidores , Enterotoxinas/biosíntesis , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Exotoxinas/análisis , Exotoxinas/antagonistas & inhibidores , Exotoxinas/biosíntesis , Proteínas Hemolisinas/análisis , Proteínas Hemolisinas/antagonistas & inhibidores , Proteínas Hemolisinas/biosíntesis , Humanos , Leucocidinas/análisis , Leucocidinas/antagonistas & inhibidores , Leucocidinas/biosíntesis , Linezolid/administración & dosificación , Linezolid/farmacología , Resistencia a la Meticilina , Staphylococcus aureus Resistente a Meticilina/efectos de los fármacos , Staphylococcus aureus Resistente a Meticilina/crecimiento & desarrollo , Staphylococcus aureus Resistente a Meticilina/metabolismo , Pruebas de Sensibilidad Microbiana , Nafcilina/administración & dosificación , Nafcilina/farmacología , Oxazolidinonas/administración & dosificación , Conejos , Ovinos , Staphylococcus aureus/crecimiento & desarrollo , Staphylococcus aureus/metabolismo , Superantígenos/análisis , Superantígenos/biosíntesis , Tetrazoles/administración & dosificación
20.
ACS Infect Dis ; 4(7): 1073-1081, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29742342

RESUMEN

The Gram-negative bacterium Aggregatibacter actinomycetemcomitans, commonly associated with localized aggressive periodontitis (LAP), secretes an RTX (repeats-in-toxin) protein leukotoxin (LtxA) that targets human white blood cells, an interaction that is driven by its recognition of the lymphocyte function-associated antigen-1 (LFA-1) integrin. In this study, we report on the inhibition of LtxA-LFA-1 binding as an antivirulence strategy to inhibit LtxA-mediated cytotoxicity. Specifically, we designed and synthesized peptides corresponding to the reported LtxA binding domain on LFA-1 and characterized their capability to inhibit LtxA binding to LFA-1 and subsequent cytotoxic activity in human immune cells. We found that several of these peptides, corresponding to sequential ß-strands in the LtxA-binding domain of LFA-1, inhibit LtxA activity, demonstrating the effectiveness of this approach. Further investigations into the mechanism by which these peptides inhibit LtxA binding to LFA-1 reveal a correlation between toxin-peptide affinity and LtxA-mediated cytotoxicity, leading to a diminished association between LtxA and LFA-1 on the cell membrane. Our results demonstrate the possibility of using target-based peptides to inhibit LtxA activity, and we expect that a similar approach could be used to hinder the activity of other RTX toxins.


Asunto(s)
Antibacterianos/farmacología , Exotoxinas/antagonistas & inhibidores , Antígeno-1 Asociado a Función de Linfocito/química , Péptidos/farmacología , Secuencia de Aminoácidos , Antibacterianos/química , Exotoxinas/química , Exotoxinas/toxicidad , Humanos , Antígeno-1 Asociado a Función de Linfocito/farmacología , Modelos Biológicos , Péptidos/química , Unión Proteica , Relación Estructura-Actividad , Células THP-1 , Factores de Virulencia/antagonistas & inhibidores , Factores de Virulencia/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA