Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Medicinas Complementárias
Métodos Terapéuticos y Terapias MTCI
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Fitoterapia ; 162: 105261, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35944753

RESUMEN

Streptococcus pneumoniae (S. pneumoniae) is a major Gram-positive opportunistic pathogen that causes pneumonia, bacteremia, and other fatal infections. This bacterium is responsible for more deaths than any other single pathogen in the world. Inexplicably, these symptoms persist despite the administration of effective antibiotics. Targeting pneumolysin (PLY) and sortase A (SrtA), the major virulence factors of S. pneumoniae, this study uncovered a novel resistance mechanism to S. pneumoniae infection. Using protein phenotype assays, we determined that the small molecule inhibitor alnustone is a potent drug that inhibits both PLY and SrtA. As essential virulence factors of S. pneumoniae, PLY and SrtA play a significant role in the occurrence of infection. Furthermore, evaluation using PLY-mediated hemolysis assay demonstrated alunstone had the potential to interrupt the haemolytic activity of PLY with treatment alunstone (4 µg/ml). Co-incubation of S. pneumoniae D39 SrtA with small-molecule inhibitors decreases cell wall-bound Nan A (pneumococcal-anchored surface protein SrtA), inhibits biofilm formation, and reduces biomass significantly. The protective effect of invasive pneumococcal disease (IPD) on murine S. pneumoniae was demonstrated further. Our study proposes a comprehensive bacteriostatic mechanism for S. pneumoniae and highlights the significant translational potential of targeting both PLY and SrtA to prevent pneumococcal infections. Our findings indicate that the antibacterial strategy of directly targeting PLY and SrtA with alnustone is a promising treatment option for S. pneumoniae and that alnustone is a potent inhibitor of PLY and SrtA.


Asunto(s)
Infecciones Neumocócicas , Streptococcus pneumoniae , Aminoaciltransferasas , Animales , Antibacterianos/farmacología , Proteínas Bacterianas , Cisteína Endopeptidasas , Hemólisis , Ratones , Estructura Molecular , Infecciones Neumocócicas/tratamiento farmacológico , Infecciones Neumocócicas/microbiología , Estreptolisinas , Virulencia , Factores de Virulencia/farmacología , Factores de Virulencia/uso terapéutico
2.
Nat Commun ; 13(1): 5032, 2022 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-36028511

RESUMEN

In pneumococcal meningitis, bacterial growth in the cerebrospinal fluid results in lysis, the release of toxic factors, and subsequent neuroinflammation. Exposure of primary murine glia to Streptococcus pneumoniae lysates leads to strong proinflammatory cytokine and chemokine production, blocked by inhibition of the intracellular innate receptor Nod1. Lysates enhance dynamin-dependent endocytosis, and dynamin inhibition reduces neuroinflammation, blocking ligand internalization. Here we identify the cholesterol-dependent cytolysin pneumolysin as a pro-endocytotic factor in lysates, its elimination reduces their proinflammatory effect. Only pore-competent pneumolysin enhances endocytosis in a dynamin-, phosphatidylinositol-3-kinase- and potassium-dependent manner. Endocytic enhancement is limited to toxin-exposed parts of the membrane, the effect is rapid and pneumolysin permanently alters membrane dynamics. In a murine model of pneumococcal meningitis, mice treated with chlorpromazine, a neuroleptic with a complementary endocytosis inhibitory effect show reduced neuroinflammation. Thus, the dynamin-dependent endocytosis emerges as a factor in pneumococcal neuroinflammation, and its enhancement by a cytolysin represents a proinflammatory control mechanism.


Asunto(s)
Meningitis Neumocócica , Streptococcus pneumoniae , Animales , Proteínas Bacterianas , Citotoxinas , Endocitosis , Inflamación , Ratones , Estreptolisinas
3.
Front Immunol ; 13: 878244, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35529870

RESUMEN

Pneumolysin (PLY) is a pore-forming toxin produced by the human pathobiont Streptococcus pneumoniae, the major cause of pneumonia worldwide. PLY, a key pneumococcal virulence factor, can form transmembrane pores in host cells, disrupting plasma membrane integrity and deregulating cellular homeostasis. At lytic concentrations, PLY causes cell death. At sub-lytic concentrations, PLY triggers host cell survival pathways that cooperate to reseal the damaged plasma membrane and restore cell homeostasis. While PLY is generally considered a pivotal factor promoting S. pneumoniae colonization and survival, it is also a powerful trigger of the innate and adaptive host immune response against bacterial infection. The dichotomy of PLY as both a key bacterial virulence factor and a trigger for host immune modulation allows the toxin to display both "Yin" and "Yang" properties during infection, promoting disease by membrane perforation and activating inflammatory pathways, while also mitigating damage by triggering host cell repair and initiating anti-inflammatory responses. Due to its cytolytic activity and diverse immunomodulatory properties, PLY is integral to every stage of S. pneumoniae pathogenesis and may tip the balance towards either the pathogen or the host depending on the context of infection.


Asunto(s)
Infecciones Neumocócicas , Estreptolisinas , Proteínas Bacterianas/metabolismo , Humanos , Infecciones Neumocócicas/microbiología , Streptococcus pneumoniae , Estreptolisinas/metabolismo , Factores de Virulencia/metabolismo
4.
STAR Protoc ; 2(3): 100757, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34458872

RESUMEN

The pore-forming toxin streptolysin-O (SLO) enables intracellular delivery of molecules up to 100 kDa and has been used for short-term delivery of membrane-impermeable substances to assess their effects on cellular activities. A limitation of this technique is the loss of intracellular components and the potential unpredicted alterations of cellular metabolism and signaling. This protocol, optimized for primary mouse T lymphocytes, describes steps for SLO-mediated cell membrane permeabilization and substance supplementation, followed by immunoblotting and immunofluorescent microscopy for assessing cellular effects. For complete details on the use and execution of this protocol, please refer to Xu et al., 2021a, Xu et al., 2021b.


Asunto(s)
Permeabilidad de la Membrana Celular/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Biología Molecular/métodos , Estreptolisinas/farmacocinética , Linfocitos T/efectos de los fármacos , Adenosina Trifosfato/administración & dosificación , Adenosina Trifosfato/farmacocinética , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacocinética , Separación Celular , Técnica del Anticuerpo Fluorescente , Immunoblotting , Activación de Linfocitos , Ratones , Biología Molecular/instrumentación , Receptores de Antígenos de Linfocitos T/metabolismo , Bazo/citología , Estreptolisinas/química , Linfocitos T/metabolismo
5.
J Ethnopharmacol ; 275: 114133, 2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-33892068

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Ma-xing-shi-gan-tang (MXSGT), which is documented in the Treatise on Febrile Diseases and is a therapeutic drug, is a well-known classic prescription in China and has been widely studied. Previous studies have shown that MXSGT has various pharmacological activities, including anti-influenza virus activity, and ameliorates microvascular hyperpermeability and inflammatory reactions. However, no study has reported the effect of MXSGT in the treatment of bacterial pneumonia. AIM OF THE STUDY: In this study, the potential inhibition of MXSGT against the virulence of S. pneumoniae by targeting PLY was investigated. MATERIALS AND METHODS: First, HPLC analysis was used to determine the main components of MXSGT. Then PLY protein was constructed and used for hemolysis assay and western blot to test the ability of MXSGT to inhibit PLY activity, production and widowed characteristics. The growth curve of S. pneumoniae was drawled with or without MXSGT treatment. In addition, the inhibition of MXSGT against PLY-mediated A549 cell death was examined by cytotoxicity assay. Finally, the mouse experiment was used to verify the effect of MXSGT on mouse lungs. RESULTS: This work has discovered that MXSGT, a TCM prescription, is an effective inhibitor of PLY, an important virulence factor that is essential for S. pneumoniae pathogenicity. MXSGT inhibits the oligomerization of PLY without affecting S. pneumoniae growth and PLY production. In addition, experimental MXSGT treatment was effective against S. pneumoniae infection both in vitro and in vivo. CONCLUSION: These findings directly demonstrate the potential mechanism of the Chinese herbal formula MXSGT in the treatment of pneumococcal disease and provide additional evidence for promotion of the wide use of MXSGT in the clinic.


Asunto(s)
Antibacterianos/farmacología , Medicamentos Herbarios Chinos/farmacología , Streptococcus pneumoniae/efectos de los fármacos , Estreptolisinas/antagonistas & inhibidores , Células A549 , Animales , Antibacterianos/química , Antibacterianos/uso terapéutico , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Modelos Animales de Enfermedad , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Hemólisis/efectos de los fármacos , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Medicina Tradicional China , Ratones Endogámicos BALB C , Ovinos , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/metabolismo , Virulencia/efectos de los fármacos
6.
Biol Pharm Bull ; 43(6): 994-999, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32475921

RESUMEN

Streptococcus pneumoniae (S. pneumoniae) is an opportunistic pathogen that causes pneumonia, meningitis and bacteremia in humans and animals. Pneumolysin (PLY), a major pore-forming toxin that is important for S. pneumoniae pathogenicity, is a promising target for the development of anti-infective agents. Ephedra sinica granules (ESG) is one of the oldest medical preparation with multiple biological activities (such as a divergent wind and cold effect); however, the detailed mechanism remains unknown. In this study, we found that ESG treatment significantly inhibited the oligomerization of PLY and then reduced the activity of PLY without affecting S. pneumoniae growth and PLY production. In a PLY and A549 cell co-incubation system, the addition of ESG resulted in significant protection against PLY-mediated cell injury. Furthermore, S. pneumoniae-infected mice showed decreased mortality, and alleviated tissue damage and inflammatory reactions following treatment with ESG. Our results indicate that ESG is a potential candidate treatment for S. pneumoniae infection that targets PLY. This finding partially elucidates the mechanism of the Chinese herbal formula ESG in the treatment of pneumococcal disease.


Asunto(s)
Antibacterianos/uso terapéutico , Ephedra sinica , Preparaciones de Plantas/uso terapéutico , Infecciones Neumocócicas/tratamiento farmacológico , Estreptolisinas/antagonistas & inhibidores , Células A549 , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Humanos , Interleucina-6/inmunología , Pulmón/efectos de los fármacos , Pulmón/patología , Medicina Tradicional China , Ratones Endogámicos BALB C , Preparaciones de Plantas/farmacología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Streptococcus pneumoniae/efectos de los fármacos , Streptococcus pneumoniae/crecimiento & desarrollo , Estreptolisinas/metabolismo , Factor de Necrosis Tumoral alfa/inmunología
7.
Cells ; 9(5)2020 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-32397494

RESUMEN

Microbial proteins have recently been found to have more benefits in clinical disease treatment because of their better-developed strategy and properties than traditional medicine. In this study, we investigated the effectiveness of a truncated peptide synthesized from the C-terminal sequence of pneumolysin, i.e., C70PLY4, in Streptococcus pneumoniae, in treating chronic inflammatory conditions. It has been shown that C70PLY4 significantly blocks the transendothelial migration of neutrophils and attenuates the formation of atherosclerotic plaque and the secretion of soluble forms of the intercellular adhesion molecule-1 (ICAM-1), the vascular cell adhesion molecule 1 (VCAM-1), and E-selectin in high-fat-diet/streptozotocin-induced inflammatory rats. The mechanism and the docking simulation analysis further indicated that C70PLY4 might serve as a Toll-like receptor 4 (TLR4) antagonist by competing for the binding site of MD2, an indispensable protein for lipopolysaccharide (LPS)-TLR4 interaction signaling, on the TLR4 structure. Moreover, compared to the full-length PLY, C70PLY4 seems to have no cytotoxicity in human vascular endothelial cells. Our study elucidated a possible therapeutic efficacy of C70PLY4 in reducing chronic inflammatory conditions and clarified the underlying mechanism. Thus, our findings identify a new drug candidate that, by blocking TLR4 activity, could be an effective treatment for patients with chronic inflammatory diseases.


Asunto(s)
Inflamación/tratamiento farmacológico , Proteínas Mutantes/farmacología , Proteínas Mutantes/uso terapéutico , Streptococcus pneumoniae/metabolismo , Estreptolisinas/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Apoptosis/efectos de los fármacos , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacología , Sitios de Unión , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Dieta Alta en Grasa , Selectina E/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos , Ratones , Simulación del Acoplamiento Molecular , Proteínas Mutantes/química , FN-kappa B/metabolismo , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Fosforilación/efectos de los fármacos , Solubilidad , Estreptolisinas/química , Estreptozocina , Receptor Toll-Like 4/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Molécula 1 de Adhesión Celular Vascular/metabolismo
8.
Infect Immun ; 87(12)2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31527126

RESUMEN

Severe manifestations of group A Streptococcus (GAS) infections are associated with massive tissue destruction and high mortality. Clindamycin (CLI), a bacterial protein synthesis inhibitor, is recommended for treating patients with severe invasive GAS infection. Nonetheless, the subinhibitory concentration of CLI induces the production of GAS virulent exoproteins, such as streptolysin O (SLO) and NADase, which would enhance bacterial virulence and invasiveness. A better understanding of the molecular mechanism of how CLI triggers GAS virulence factor expression will be critical to develop appropriate therapeutic approaches. The present study shows that CLI activates SLO and NADase expressions in the emm1-type CLI-susceptible wild-type strain but not in covS or control of virulence sensor (CovS) phosphatase-inactivated mutants. Supplementation with Mg2+, which is a CovS phosphatase inhibitor, inhibits the CLI-mediated SLO upregulation in a dose-dependent manner in CLI-susceptible and CLI-resistant strains. These results not only reveal that the phosphorylation of response regulator CovR is essential for responding to CLI stimuli, but also suggest that inhibiting the phosphatase activity of CovS could be a potential strategy for the treatment of invasive GAS infection with CLI.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Clindamicina/farmacología , Histidina Quinasa/metabolismo , Proteínas Represoras/metabolismo , Streptococcus pyogenes/metabolismo , Estreptolisinas/biosíntesis , Proteínas Bacterianas/biosíntesis , Histidina Quinasa/antagonistas & inhibidores , Histidina Quinasa/genética , Magnesio/farmacología , Monoéster Fosfórico Hidrolasas/metabolismo , Streptococcus pyogenes/patogenicidad
9.
Biocontrol Sci ; 23(2): 77-80, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29910212

RESUMEN

 Group A streptococcus is a bacterium that resides in the throat and skin and causes respiratory infection and occasionally glomerulonephritis and rheumatic fever. Streptolysin O (SLO) produced by Streptococcus pyogenes (S. pyogenes) binds to the cell membrane, particularly to that of white and red blood cells, and is toxic to the cells and tissue. In this study, we evaluated the inhibitory activity of water-soluble polyphenols in olives (Olea europaea) against SLO-induced hemolysis. Hydroxytyrosol inhibited SLO-induced hemolytic activity, and the amount required for 50% inhibition of hemolysis was 1.30 µg. These findings suggest that the water-soluble polyphenols contained in olives have inhibitory activity against SLO-induced hemolysis.


Asunto(s)
Antiinfecciosos/metabolismo , Células Sanguíneas/efectos de los fármacos , Hemólisis/efectos de los fármacos , Alcohol Feniletílico/análogos & derivados , Estreptolisinas/antagonistas & inhibidores , Animales , Antiinfecciosos/aislamiento & purificación , Proteínas Bacterianas/antagonistas & inhibidores , Concentración 50 Inhibidora , Olea/química , Alcohol Feniletílico/aislamiento & purificación , Alcohol Feniletílico/metabolismo , Extractos Vegetales/química , Conejos
10.
Biomaterials ; 155: 152-164, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-29179131

RESUMEN

Vaccine design ushered in the era of nanotechnology, as the vaccine is being developed toward particulate formulation. We have previously shown that the attenuated pneumolysin mutant (ΔA146PLY) was a safe and effective pneumococcal vaccine candidate. Here, to further optimize the formulation, we fused calcium phosphate (CaP) binding domains with ΔA146PLY so that the biocompatible CaP can mineralize with the protein automatically, allowing simple production of nanoparticle antigen during preparation. We fabricated four different nanoparticles, and then we compared the characteristics of different CaP-ΔA146PLY nanoparticles and demonstrated the influence of CaP binding domains on the size, shape and surface calcium content of the nanoparticles. It was found that these self-biomineralized CaP-ΔA146PLY nanoparticles varied in their capacity to induce BMDCs and splenocytes production of cytokines. We further demonstrated that, compared to free proteins, nanoparticle antigens induced more efficient humoral and cellular immune responses which was strong enough to protect mice from both pneumonia and sepsis infection. Also, the integration of CaP to protein has no significant impairment on body weight of animals, and subcutaneous injection of ΔA146PLY-peptides@CaP nanoparticles did not lead to permanent formation of nodules in the skin relative to Alum adjuvant formulated antigens. Together, our data sufficiently suggest that soluble ΔA146PLY vaccine candidate could be processed into nanoparticles by self-biomineralization of CaP, the immunogenicity of which could be efficiently improved by the CaP binding domains and biomineralization.


Asunto(s)
Fosfatos de Calcio/química , Fosfatos de Calcio/metabolismo , Nanopartículas/química , Estreptolisinas/química , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Ratones , Infecciones Neumocócicas/metabolismo , Streptococcus pneumoniae/metabolismo , Streptococcus pneumoniae/patogenicidad
11.
Br J Pharmacol ; 174(23): 4295-4307, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28888095

RESUMEN

BACKGROUND AND PURPOSE: Streptococcus pneumoniae is the most common cause of bacterial meningitis in adults and is characterized by high lethality and substantial cognitive disabilities in survivors. Here, we have studied the capacity of an established therapeutic agent, magnesium, to improve survival in pneumococcal meningitis by modulating the neurological effects of the major pneumococcal pathogenic factor, pneumolysin. EXPERIMENTAL APPROACH: We used mixed primary glial and acute brain slice cultures, pneumolysin injection in infant rats, a mouse meningitis model and complementary approaches such as Western blot, a black lipid bilayer conductance assay and live imaging of primary glial cells. KEY RESULTS: Treatment with therapeutic concentrations of magnesium chloride (500 mg·kg-1 in animals and 2 mM in cultures) prevented pneumolysin-induced brain swelling and tissue remodelling both in brain slices and in animal models. In contrast to other divalent ions, which diminish the membrane binding of pneumolysin in non-therapeutic concentrations, magnesium delayed toxin-driven pore formation without affecting its membrane binding or the conductance profile of its pores. Finally, magnesium prolonged the survival and improved clinical condition of mice with pneumococcal meningitis, in the absence of antibiotic treatment. CONCLUSIONS AND IMPLICATIONS: Magnesium is a well-established and safe therapeutic agent that has demonstrated capacity for attenuating pneumolysin-triggered pathogenic effects on the brain. The improved animal survival and clinical condition in the meningitis model identifies magnesium as a promising candidate for adjunctive treatment of pneumococcal meningitis, together with antibiotic therapy.


Asunto(s)
Cloruro de Magnesio/administración & dosificación , Meningitis Neumocócica/tratamiento farmacológico , Streptococcus pneumoniae/efectos de los fármacos , Estreptolisinas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/microbiología , Modelos Animales de Enfermedad , Femenino , Cloruro de Magnesio/farmacología , Meningitis Neumocócica/microbiología , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Neuroglía/microbiología , Ratas , Ratas Sprague-Dawley , Streptococcus pneumoniae/aislamiento & purificación , Tasa de Supervivencia
12.
BMC Microbiol ; 16(1): 271, 2016 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-27829373

RESUMEN

BACKGROUND: Streptococcus pneumoniae is one of the leading causes of community acquired pneumonia and acute otitis media. Certain aspects of S. pneumoniae's virulence are dependent upon expression and release of the protein toxin pneumolysin (PLY) and upon the activity of the peroxide-producing enzyme, pyruvate oxidase (SpxB). We investigated the possible synergy of these two proteins and identified that release of PLY is enhanced by expression of SpxB prior to stationary phase growth. RESULTS: Mutants lacking the spxB gene were defective in PLY release and complementation of spxB restored PLY release. This was demonstrated by cytotoxic effects of sterile filtered supernatants upon epithelial cells and red blood cells. Additionally, peroxide production appeared to contribute to the mechanism of PLY release since a significant correlation was found between peroxide production and PLY release among a panel of clinical isolates. Exogenous addition of H2O2 failed to induce PLY release and catalase supplementation prevented PLY release in some strains, indicating peroxide may exert its effect intracellularly or in a strain-dependent manner. SpxB expression did not trigger bacterial cell death or LytA-dependent autolysis, but did predispose cells to deoxycholate lysis. CONCLUSIONS: Here we demonstrate a novel link between spxB expression and PLY release. These findings link liberation of PLY toxin to oxygen availability and pneumococcal metabolism.


Asunto(s)
Piruvato Oxidasa/metabolismo , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo , Autólisis , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Catalasa , ADN Bacteriano/genética , Células Epiteliales/microbiología , Eritrocitos/microbiología , Genes Bacterianos , Peróxido de Hidrógeno/metabolismo , Oxígeno , Piruvato Oxidasa/genética , Eliminación de Secuencia , Streptococcus pneumoniae/enzimología , Streptococcus pneumoniae/genética , Estreptolisinas/genética , Virulencia
13.
Fitoterapia ; 115: 31-36, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27693741

RESUMEN

Streptococcus pneumoniae is an important human pathogenic bacterium that can cause various life-threatening infections. Pneumolysin (PLY), the pore-forming toxin that forms large pores in the cell membrane, is a key virulence factor secreted by S. pneumoniae that penetrates the physical defenses of the host and plays an important role in the pathogenesis of pneumococcal diseases, such as pneumonia, meningitis, bacteremia and otitis media. This study showed that apigenin, one of the bioflavonoids widely found in herbs, inhibits PLY-induced hemolysis by inhibiting the oligomerization of PLY and has no anti-S. pneumoniae activity. In addition, when PLY was incubated with human alveolar epithelial (A549) cells, apigenin could effectively alleviate PLY-mediated cell injury. In vivo studies further demonstrated that apigenin could protect mice against S. pneumoniae pneumonia. These results imply that apigenin could directly interact with PLY to decrease the pathogenicity of S. pneumoniae and that novel therapeutics against S. pneumoniae PLY might provide greater effectiveness in combatting S. pneumoniae pneumonia.


Asunto(s)
Apigenina/farmacología , Neumonía Neumocócica/tratamiento farmacológico , Estreptolisinas/antagonistas & inhibidores , Células A549 , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Femenino , Hemólisis , Humanos , Ratones , Ratones Endogámicos BALB C
14.
Clin Vaccine Immunol ; 22(10): 1079-89, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26245351

RESUMEN

Immunization with the pneumococcal proteins pneumolysin (Ply), choline binding protein A (CbpA), or pneumococcal surface protein A (PspA) elicits protective responses against invasive pneumococcal disease in animal models. In this study, we used different mouse models to test the efficacy of a variety of multivalent protein-based vaccines that comprised various combinations of full-length or peptide regions of the immunogens Ply, CbpA, or PspA: Ply toxoid with the L460D substitution (referred to herein as L460D); L460D fused with protective peptide epitopes from CbpA (YPT-L460D-NEEK [YLN]); L460D fused with the CD2 peptide containing the proline-rich region (PRR) of PspA (CD2-L460D); a combination of L460D and H70 (L460D+H70), a slightly larger PspA-derived peptide containing the PRR and the SM1 region; H70+YLN; and other combinations. Each mouse was immunized either intraperitoneally (i.p.) or subcutaneously (s.c.) with three doses (at 2-week intervals) of the various antigen combinations in alum adjuvant and then challenged in mouse models featuring different infection routes with multiple Streptococcus pneumoniae strains. In the i.p. infection sepsis model, H70+YLN consistently provided significant protection against three different challenge strains (serotypes 1, 2, and 6A); the CD2+YLN and H70+L460D combinations also elicited significant protection. Protection against intravenous (i.v.) sepsis (type 3 and 6A challenge strains) was largely dependent on PspA-derived antigen components, and the most protection was elicited by H70 with or without L460D or YLN. In a type 4 intratracheal (i.t.) challenge model that results in progression to meningitis, antigen combinations that contained YLN elicited the strongest protection. Thus, the trivalent antigen combination of H70+YLN elicited the strongest and broadest protection in diverse pneumococcal challenge models.


Asunto(s)
Proteínas Bacterianas/inmunología , Infecciones Neumocócicas/prevención & control , Vacunas Neumococicas/inmunología , Sepsis/prevención & control , Streptococcus pneumoniae/inmunología , Estreptolisinas/inmunología , Animales , Proteínas Bacterianas/genética , Modelos Animales de Enfermedad , Epítopos/genética , Epítopos/inmunología , Esquemas de Inmunización , Inmunoglobulina G/sangre , Meningitis Neumocócica/inmunología , Meningitis Neumocócica/microbiología , Meningitis Neumocócica/prevención & control , Ratones Endogámicos BALB C , Infecciones Neumocócicas/inmunología , Vacunas Neumococicas/administración & dosificación , Vacunas Neumococicas/genética , Neumonía Neumocócica/inmunología , Neumonía Neumocócica/microbiología , Neumonía Neumocócica/prevención & control , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Sepsis/microbiología , Streptococcus pneumoniae/clasificación , Toxoides/inmunología , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
15.
Am J Respir Cell Mol Biol ; 53(5): 689-702, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25874372

RESUMEN

Statins are widely used to prevent cardiovascular disease. In addition to their inhibitory effects on cholesterol synthesis, statins have beneficial effects in patients with sepsis and pneumonia, although molecular mechanisms have mostly remained unclear. Using human airway epithelial cells as a proper in vitro model, we show that prior exposure to physiological nanomolar serum concentrations of simvastatin (ranging from 10-1,000 nM) confers significant cellular resistance to the cytotoxicity of pneumolysin, a pore-forming toxin and the main virulence factor of Streptococcus pneumoniae. This protection could be demonstrated with a different statin, pravastatin, or on a different toxin, α-hemolysin. Furthermore, through the use of gene silencing, pharmacological inhibitors, immunofluorescence microscopy, and biochemical and metabolic rescue approaches, we demonstrate that the mechanism of protection conferred by simvastatin at physiological nanomolar concentrations could be different from the canonical mevalonate pathways seen in most other mechanistic studies conducted with statins at micromolar levels. All of these data are integrated into a protein synthesis-dependent, calcium-dependent model showing the interconnected pathways used by statins in airway epithelial cells to elicit an increased resistance to pore-forming toxins. This research fills large gaps in our understanding of how statins may confer host cellular protection against bacterial infections in the context of airway epithelial cells without the confounding effect from the presence of immune cells. In addition, our discovery could be potentially developed into a host-centric strategy for the adjuvant treatment of pore-forming toxin associated bacterial infections.


Asunto(s)
Toxinas Bacterianas/antagonistas & inhibidores , Células Epiteliales/efectos de los fármacos , Proteínas Hemolisinas/antagonistas & inhibidores , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inmunidad Innata/efectos de los fármacos , Simvastatina/farmacología , Estreptolisinas/antagonistas & inhibidores , Animales , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Línea Celular Transformada , Células Epiteliales/inmunología , Células Epiteliales/patología , Proteínas Hemolisinas/toxicidad , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/inmunología , Inyecciones Intraperitoneales , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Pravastatina/inmunología , Pravastatina/farmacología , Cultivo Primario de Células , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Simvastatina/inmunología , Staphylococcus aureus/química , Streptococcus pneumoniae/química , Estreptolisinas/toxicidad
16.
EMBO J ; 34(5): 669-88, 2015 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-25595798

RESUMEN

P4-ATPases translocate aminophospholipids, such as phosphatidylserine (PS), to the cytosolic leaflet of membranes. PS is highly enriched in recycling endosomes (REs) and is essential for endosomal membrane traffic. Here, we show that PS flipping by an RE-localized P4-ATPase is required for the recruitment of the membrane fission protein EHD1. Depletion of ATP8A1 impaired the asymmetric transbilayer distribution of PS in REs, dissociated EHD1 from REs, and generated aberrant endosomal tubules that appear resistant to fission. EHD1 did not show membrane localization in cells defective in PS synthesis. ATP8A2, a tissue-specific ATP8A1 paralogue, is associated with a neurodegenerative disease (CAMRQ). ATP8A2, but not the disease-causative ATP8A2 mutant, rescued the endosomal defects in ATP8A1-depleted cells. Primary neurons from Atp8a2-/- mice showed a reduced level of transferrin receptors at the cell surface compared to Atp8a2+/+ mice. These findings demonstrate the role of P4-ATPase in membrane fission and give insight into the molecular basis of CAMRQ.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Endosomas/metabolismo , Modelos Biológicos , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Adenosina Trifosfatasas/genética , Análisis de Varianza , Animales , Proteínas Bacterianas , Transporte Biológico/fisiología , Western Blotting , Células COS , Chlorocebus aethiops , Cartilla de ADN/genética , ADN Complementario/genética , Células HeLa , Humanos , Inmunohistoquímica , Ratones , Ratones Noqueados , Microscopía Confocal , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/genética , Reacción en Cadena de la Polimerasa , Interferencia de ARN , Estreptolisinas
17.
Curr Comput Aided Drug Des ; 10(1): 59-74, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24694051

RESUMEN

Streptococcus pyogenes is a notorious pathogenic bacterium which causes various human diseases ranging from localized infections to life threatening invasive diseases. Streptolysin-O (SLO), pore-forming thiol-activated cytolysin, is the major virulent factor for streptococcal infections. Present therapies against streptococcal infections are limited as most of the strains have developed multi-drug resistance to present generation of drugs. Hence, there is a need for alternative therapeutic substances. Structure based virtual screening is a novel platform to select lead molecules with better pharmacokinetic properties. The 3D structure of SLO (not available in native form), essential for such studies, was computationally generated and this homology model was used as probable drug target. Based on literature survey, several phytoligands from 25 medicinal plants were selected. Out of these, leads from 11 plants showed better pharmacokinetic properties. The best lead molecules were screened based on computer aided drug likeness and pharmacokinetic predictions. The inhibitory properties of selected herbal leads against SLO were studied by molecular docking. An in vitro assay was further carried out and variations observed were found to be significant (p<0.05). Antibiotic sensitivity testing was also performed with the clinical strain of Streptococcus pyogenes with conventional drugs. The clinical strain showed multi-drug resistance to conventional drugs. Our study revealed that numerous phytoligands have better inhibitory properties towards the toxin. We noticed that incorporation of selected herbal extracts in blood agar medium showed significant reduction in hemolysis (MIC 300µl/plate), indicating inhibition of SLO. Furthermore, the butanol extracts of selected herbal preparation based on computer aided screening showed significant inhibitory properties at 250 mcg/disc concentration. We also noticed that selected herbal formulations have better antimicrobial properties at MIC range of 300- 400µl. Hence, our study suggests that these herbal extracts have better inhibitory properties against the toxin as well as drug resistant Streptococcus pyogenes.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Preparaciones de Plantas/química , Preparaciones de Plantas/farmacología , Streptococcus pyogenes/efectos de los fármacos , Estreptolisinas/química , Estreptolisinas/farmacología , Secuencia de Aminoácidos , Proteínas Bacterianas/química , Proteínas Bacterianas/farmacología , Simulación por Computador , Diseño Asistido por Computadora , Diseño de Fármacos , Ligandos , Modelos Moleculares , Datos de Secuencia Molecular
18.
PLoS Pathog ; 9(6): e1003380, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785278

RESUMEN

Streptococcus pneumoniae (pneumococcal) meningitis is a common bacterial infection of the brain. The cholesterol-dependent cytolysin pneumolysin represents a key factor, determining the neuropathogenic potential of the pneumococci. Here, we demonstrate selective synaptic loss within the superficial layers of the frontal neocortex of post-mortem brain samples from individuals with pneumococcal meningitis. A similar effect was observed in mice with pneumococcal meningitis only when the bacteria expressed the pore-forming cholesterol-dependent cytolysin pneumolysin. Exposure of acute mouse brain slices to only pore-competent pneumolysin at disease-relevant, non-lytic concentrations caused permanent dendritic swelling, dendritic spine elimination and synaptic loss. The NMDA glutamate receptor antagonists MK801 and D-AP5 reduced this pathology. Pneumolysin increased glutamate levels within the mouse brain slices. In mouse astrocytes, pneumolysin initiated the release of glutamate in a calcium-dependent manner. We propose that pneumolysin plays a significant synapto- and dendritotoxic role in pneumococcal meningitis by initiating glutamate release from astrocytes, leading to subsequent glutamate-dependent synaptic damage. We outline for the first time the occurrence of synaptic pathology in pneumococcal meningitis and demonstrate that a bacterial cytolysin can dysregulate the control of glutamate in the brain, inducing excitotoxic damage.


Asunto(s)
Lóbulo Frontal/metabolismo , Ácido Glutámico/metabolismo , Meningitis Neumocócica/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Streptococcus pneumoniae/metabolismo , Estreptolisinas/metabolismo , Sinapsis/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Astrocitos/metabolismo , Astrocitos/microbiología , Astrocitos/patología , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Dendritas/metabolismo , Dendritas/microbiología , Dendritas/patología , Maleato de Dizocilpina/farmacología , Lóbulo Frontal/microbiología , Lóbulo Frontal/patología , Humanos , Meningitis Neumocócica/genética , Meningitis Neumocócica/microbiología , Meningitis Neumocócica/patología , Ratones , Persona de Mediana Edad , Receptores de N-Metil-D-Aspartato/antagonistas & inhibidores , Streptococcus pneumoniae/genética , Streptococcus pneumoniae/patogenicidad , Estreptolisinas/genética , Sinapsis/microbiología , Sinapsis/patología
19.
Infect Immun ; 79(2): 867-78, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21098102

RESUMEN

Zinc metalloprotease B (ZmpB) is present in all isolated pneumococcal strains and contributes to the pathogenesis of pneumococcal infection. In this study, recombinant ZmpB was cloned and expressed in Escherichia coli. The expression of ZmpB by different pneumococcal strains was detectable by Western blotting with antisera raised to recombinant ZmpB. Flow cytometry analysis demonstrated that anti-ZmpB polyclonal antibodies could bind to the cell surface of the pneumococcal strains analyzed. Both recombinant ZmpB protein and anti-ZmpB polyclonal antibodies significantly inhibited the adhesion of Streptococcus pneumoniae D39 to A549 cells. In mouse models, mucosal immunization with recombinant ZmpB could significantly reduce pneumococcal lung colonization caused by S. pneumoniae serotypes 19F and 14 and significantly increase mice survival times following invasive pneumococcal challenge with different pneumococcal strains, including serotypes 2, 3, 6B, and 14. Furthermore, intraperitoneal immunization with recombinant ZmpB in combination with the recombinant pneumolysin mutant (DeltaA146 Ply) and heat shock protein 40 (DnaJ) could enhance the protection against pneumococcal infection compared to protection provided by single-protein antigens. Passive immunization with hyperimmune antisera against these three antigens also demonstrated that the combination of three hyperimmune antisera could provide better protection than single antisera. Taken together, our results suggest that ZmpB is a good candidate pneumococcal vaccine antigen.


Asunto(s)
Proteínas Bacterianas/inmunología , Vacunas Bacterianas/inmunología , Metaloendopeptidasas/inmunología , Infecciones Neumocócicas/prevención & control , Adyuvantes Inmunológicos , Administración a través de la Mucosa , Animales , Proteínas Bacterianas/administración & dosificación , Vacunas Bacterianas/administración & dosificación , Ratones , Ratones Endogámicos BALB C , Proteínas Recombinantes , Estreptolisinas/administración & dosificación , Estreptolisinas/inmunología
20.
Toxicon ; 57(4): 540-5, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21184771

RESUMEN

Pneumolysin (PLY) is a key virulence factor contributes to the pathogenesis of Streptococcus pneumoniae. In this study we investigated the effect of allicin and aqueous garlic extracts on hemolytic activity of PLY both in prelysed and intact cells. Additionally the antimicrobial activity of allicin was tested against the bacteria. All tested materials potently inhibited the PLY hemolytic activity. Allicin neutralizes PLY in a concentration- and time-dependent manner. Twenty five minute incubation of PLY (2 HU/mL) with 0.61 µM/mL concentration of allicin, totally inhibited hemolytic activity of PLY (IC50 = 0.28 µM/mL). The inhibitory activity of old extract of garlic was similar to pure allicin (IC50 = 50.46 µL/mL; 0.31 µM/mL; P < 0.05). In contrast fresh extract of garlic inhibits the PLY hemolytic activity at lower concentrations (IC50 = 13.96 µL/mL; 0.08 µM/mL allicin). Exposure of intact cells to allicin (1.8 µM) completely inhibited hemolytic activity of PLY inside bacterial cells. The inhibitory effect of the allicin was restored by addition of reducing agent DTT at 5 mM, proposing that allicin likely inhibits the PLY by binding to cysteinyl residue in the binding site. The MIC value of allicin was determined to be 512 µg/mL (3.15 µM/mL). These results indicate that PLY is a novel target for allicin and may provide a new line of investigation on pneumococcal diseases in the future.


Asunto(s)
Antiinfecciosos/toxicidad , Ajo/química , Estreptolisinas/antagonistas & inhibidores , Ácidos Sulfínicos/toxicidad , Proteínas Bacterianas/antagonistas & inhibidores , Proteínas Bacterianas/metabolismo , Disulfuros , Ditiotreitol/farmacología , Antagonismo de Drogas , Eritrocitos/química , Eritrocitos/efectos de los fármacos , Hemoglobinas/análisis , Hemólisis/efectos de los fármacos , Humanos , Pruebas de Sensibilidad Microbiana , Oxidación-Reducción , Streptococcus pneumoniae/efectos de los fármacos , Estreptolisinas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA