Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
J Ethnopharmacol ; 275: 114133, 2021 Jul 15.
Article in English | MEDLINE | ID: mdl-33892068

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , A549 Cells , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Hemolysis/drug effects , Humans , Lung/drug effects , Lung/pathology , Medicine, Chinese Traditional , Mice, Inbred BALB C , Sheep , Streptococcus pneumoniae/pathogenicity , Streptolysins/metabolism , Virulence/drug effects
2.
J Appl Microbiol ; 130(3): 843-851, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32621771

ABSTRACT

AIMS: Pneumolysin, a pore-forming toxin, is an important virulence factor of Streptococcus pneumoniae with multiple biological activity, such as cell lysis and DNA damage. Thus, targeting this toxin is alternative strategy for the treatment of S. pneumoniae infection. METHODS AND RESULTS: Haemolysin assay was performed to identify the potential PLY inhibitor. The mechanism by which betulin, a natural compound from birch bark, against PLY was determined via MICs determination, western blot analysis and oligomerization analysis. Cytotoxicity and Immunofluorescence assays were further used to evaluate the protection of betulin against PLY-induced cell injury and DNA damage. Here, betulin, a natural compound from birch bark, was indentified as an effective inhibitor of PLY. Importantly, at the concentrations required for such inhibition, betulin has no influence on S. pneumoniae viability or PLY production. The interaction of betulin with PLY restrict the olgomerizaiton of this toxin and, thus, directly neutralizing the activity of PLY. Additionally, betulin treatment alleviate PLY induced cells injury and DNA damage in the co-culture system of PLY and A549 cells. CONCLUSIONS: Betulin could be used as a promising leading compound against S. pneumoniae virulence by directly targeting PLY without antibacterial activity. SIGNIFICANCE AND IMPACT OF THE STUDY: The results presented in this work provided a novel strategy and candidate for S. pneumoniae infection.


Subject(s)
Anti-Infective Agents/pharmacology , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , Triterpenes/pharmacology , Virulence Factors/antagonists & inhibitors , A549 Cells , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , DNA Damage , Hemolysis , Humans , Microbial Sensitivity Tests , Streptococcus pneumoniae/pathogenicity , Streptolysins/metabolism , Virulence Factors/metabolism
3.
PLoS One ; 15(6): e0235139, 2020.
Article in English | MEDLINE | ID: mdl-32574205

ABSTRACT

Viral infections complicated by a bacterial infection are typically referred to as coinfections or superinfections. Streptococcus pyogenes, the group A streptococcus (GAS), is not the most common bacteria associated with influenza A virus (IAV) superinfections but did cause significant mortality during the 2009 influenza pandemic even though all isolates are susceptible to penicillin. One approach to improve the outcome of these infections is to use passive immunization targeting GAS. To test this idea, we assessed the efficacy of passive immunotherapy using antisera against either the streptococcal M protein or streptolysin O (SLO) in a murine model of IAV-GAS superinfection. Prophylactic treatment of mice with antiserum to either SLO or the M protein decreased morbidity compared to mice treated with non-immune sera; however, neither significantly decreased mortality. Therapeutic use of antisera to SLO decreased morbidity compared to mice treated with non-immune sera but neither antisera significantly reduced mortality. Overall, the results suggest that further development of antibodies targeting the M protein or SLO may be a useful adjunct in the treatment of invasive GAS diseases, including IAV-GAS superinfections, which may be particularly important during influenza pandemics.


Subject(s)
Antigens, Bacterial/immunology , Bacterial Outer Membrane Proteins/immunology , Carrier Proteins/immunology , Immunotherapy/methods , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Streptolysins/immunology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Outer Membrane Proteins/antagonists & inhibitors , Bacterial Outer Membrane Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Coinfection/microbiology , Coinfection/therapy , Coinfection/virology , Female , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/immunology , Humans , Immune Sera/immunology , Immune Sera/pharmacology , Influenza A virus/physiology , Mice, Inbred BALB C , Orthomyxoviridae Infections/therapy , Orthomyxoviridae Infections/virology , Rabbits , Streptococcal Infections/microbiology , Streptococcal Infections/therapy , Streptococcus pyogenes/metabolism , Streptococcus pyogenes/physiology , Streptolysins/antagonists & inhibitors , Streptolysins/metabolism , Superinfection/microbiology , Superinfection/therapy , Superinfection/virology
4.
Biol Pharm Bull ; 43(6): 994-999, 2020.
Article in English | MEDLINE | ID: mdl-32475921

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Ephedra sinica , Plant Preparations/therapeutic use , Pneumococcal Infections/drug therapy , Streptolysins/antagonists & inhibitors , A549 Cells , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bronchoalveolar Lavage Fluid/immunology , Female , Humans , Interleukin-6/immunology , Lung/drug effects , Lung/pathology , Medicine, Chinese Traditional , Mice, Inbred BALB C , Plant Preparations/pharmacology , Pneumococcal Infections/immunology , Pneumococcal Infections/pathology , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/growth & development , Streptolysins/metabolism , Tumor Necrosis Factor-alpha/immunology
5.
J Pharm Pharmacol ; 72(8): 1092-1100, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32390150

ABSTRACT

OBJECTIVES: Streptococcus pneumoniae (S. pneumoniae) is an important commensal and pathogenic bacterium responsible for pneumonia, meningitis and other invasive diseases. Pneumolysin (PLY) is the major virulence factor that contributes significantly to the interaction between S. pneumoniae and the host. KEY FINDINGS: In this study, the results of antibacterial analysis, the haemolysis test and the Western blotting assay showed that acacetin inhibited PLY-mediated pore-forming activity caused by S. pneumoniae culture precipitates and purified PLY without anti-S. pneumoniae activity. In addition, acacetin treatment inhibited PLY oligomerization without affecting the expression of PLY in S. pneumoniae culture supernatants. Live/dead cells and cytotoxicity assays suggested that acacetin significantly enhanced the survival rate of injured cells by inhibiting the biological toxicity of PLY without cytotoxicity in the coculture system. The in vivo mouse model of S. pneumoniae infection further demonstrated that acacetin treatment could significantly reduce the levels of inflammatory factors (INF-γ and IL-ß) in bronchoalveolar lavage fluid (BALF) and alleviate the pathological damage of lung injury. CONCLUSIONS: Taken together, the results presented in this study indicated that acacetin inhibited the pore-forming activity of PLY and reduced the virulence of S. pneumoniae in vivo and in vitro, which may provide a leading compound for the treatment of S. pneumoniae infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Flavones/pharmacology , Lung/drug effects , Pneumonia, Pneumococcal/drug therapy , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , A549 Cells , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Female , Hemolysis/drug effects , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Interleukin-1beta/metabolism , Lung/immunology , Lung/metabolism , Lung/microbiology , Mice, Inbred BALB C , Microbial Viability/drug effects , Pneumonia, Pneumococcal/immunology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/metabolism , Streptococcus pneumoniae/pathogenicity , Streptolysins/metabolism , Virulence
6.
Nano Lett ; 19(9): 5904-5914, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31389707

ABSTRACT

Sensory neurons promote profound suppressive effects on neutrophils during Streptococcus pyogenes infection and contribute to the pathogenesis of necrotizing infection ("flesh-eating disease"). Thus, the development of new antibacterial agents for necrotizing infection is promising because of the clear streptococcal neuro-immune communication. Herein, based on the immune escape membrane exterior and competitive membrane functions of the glioma cell membrane, a novel nano neuro-immune blocker capsule was designed to prevent neuronal activation and improve neutrophil immune responses for necrotizing infection. These nano neuro-immune blockers could neutralize streptolysin S, suppress neuron pain conduction and calcitonin gene-related peptide release, and recruit neutrophils to the infection site, providing a strong therapeutic effect against necrotizing infection. Furthermore, nano neuro-immune blockers could serve as an effective inflammatory regulator and antibacterial agent via photothermal effects under near-infrared irradiation. In the Streptococcus pyogenes-induced necrotizing fasciitis mouse model, nano neuro-immune blockers showed significant therapeutic efficacy by ameliorating sensitivity to pain and promoting the antibacterial effect of neutrophils.


Subject(s)
Anti-Bacterial Agents/pharmacology , Inflammation/drug therapy , Necrosis/drug therapy , Pain/drug therapy , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/radiation effects , Bacterial Proteins/antagonists & inhibitors , Humans , Immunity, Innate/drug effects , Immunity, Innate/radiation effects , Inflammation/microbiology , Light , Mice , Necrosis/microbiology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/radiation effects , Neurons/drug effects , Neurons/microbiology , Neutrophils/drug effects , Neutrophils/microbiology , Pain/microbiology , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/pathogenicity , Streptolysins/antagonists & inhibitors
7.
Biocontrol Sci ; 23(2): 77-80, 2018.
Article in English | MEDLINE | ID: mdl-29910212

ABSTRACT

 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.


Subject(s)
Anti-Infective Agents/metabolism , Blood Cells/drug effects , Hemolysis/drug effects , Phenylethyl Alcohol/analogs & derivatives , Streptolysins/antagonists & inhibitors , Animals , Anti-Infective Agents/isolation & purification , Bacterial Proteins/antagonists & inhibitors , Inhibitory Concentration 50 , Olea/chemistry , Phenylethyl Alcohol/isolation & purification , Phenylethyl Alcohol/metabolism , Plant Extracts/chemistry , Rabbits
8.
Int J Mol Sci ; 19(4)2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29641429

ABSTRACT

Pneumolysin (PLY), a member of the family of Gram-positive bacterial, cholesterol-dependent, ß-barrel pore-forming cytolysins, is the major protein virulence factor of the dangerous respiratory pathogen, Streptococcus pneumoniae (pneumococcus). PLY plays a major role in the pathogenesis of community-acquired pneumonia (CAP), promoting colonization and invasion of the upper and lower respiratory tracts respectively, as well as extra-pulmonary dissemination of the pneumococcus. Notwithstanding its role in causing acute lung injury in severe CAP, PLY has also been implicated in the development of potentially fatal acute and delayed-onset cardiovascular events, which are now recognized as being fairly common complications of this condition. This review is focused firstly on updating mechanisms involved in the immunopathogenesis of PLY-mediated myocardial damage, specifically the direct cardiotoxic and immunosuppressive activities, as well as the indirect pro-inflammatory/pro-thrombotic activities of the toxin. Secondly, on PLY-targeted therapeutic strategies including, among others, macrolide antibiotics, natural product antagonists, cholesterol-containing liposomes, and fully humanized monoclonal antibodies, as well as on vaccine-based preventive strategies. These sections are preceded by overviews of CAP in general, the role of the pneumococcus as the causative pathogen, the occurrence and types of CAP-associated cardiac complication, and the structure and biological activities of PLY.


Subject(s)
Cardiovascular Diseases/etiology , Community-Acquired Infections/complications , Pneumonia, Pneumococcal/complications , Streptolysins/toxicity , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/toxicity , Cardiovascular Diseases/drug therapy , Community-Acquired Infections/drug therapy , Community-Acquired Infections/etiology , Humans , Pneumonia, Pneumococcal/drug therapy , Pneumonia, Pneumococcal/etiology , Streptolysins/antagonists & inhibitors
9.
Life Sci ; 177: 1-7, 2017 May 15.
Article in English | MEDLINE | ID: mdl-28385613

ABSTRACT

AIMS: Streptococcus pneumoniae (S. pneumoniae) is a common pathogen that can cause severe infections in humans. Pneumolysin (PLY) is an important virulence trait of S. pneumoniae and has cytotoxicity, genotoxicity and pro-inflammatory activity; it is essential for the pathogenesis of S. pneumoniae pneumonia and is an anti-virulence target of small molecule drug development. The treatment options for this microbe were limit due to the ubiquitous antibiotic resistance; therefore, new drugs and treatment strategies are needed. METHODS: Shikonin was selected by drug screening based on haemolysis assays, and its mechanism of suppressing PLY toxicity was determined by oligomerization assay. Meanwhile, the in vitro cell viability assays and in vivo experiments were performed to explore the capability of shikonin to protect cells and tissue from S. pneumoniae-mediated damage. KEY FINDINGS: Shikonin was found to significantly decrease PLY-induced haemolytic activity, cytotoxicity and genotoxicity via lessening the formation of oligomers; moreover, the agent can reduce the mortality of mice caused by lethal pneumonia and mitigate the injury of target organs as well. SIGNIFICANCE: We suggest that shikonin could be a potent candidate for a novel therapeutic or auxiliary substance in the treatment of infections encountering insufficient vaccines and antimicrobial resistance to traditional antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Naphthoquinones/pharmacology , Pneumonia, Pneumococcal/drug therapy , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , Animals , Bacterial Proteins/antagonists & inhibitors , Cell Survival/drug effects , Drug Design , Female , Hemolysis/drug effects , Humans , Mice , Mice, Inbred C57BL , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/pathogenicity
10.
J Cell Mol Med ; 21(10): 2586-2598, 2017 10.
Article in English | MEDLINE | ID: mdl-28402019

ABSTRACT

Streptococcus pneumoniae (pneumococcus), the causative agent of several human diseases, possesses numerous virulence factors associated with pneumococcal infection and pathogenesis. Pneumolysin (PLY), an important virulence factor, is a member of the cholesterol-dependent cytolysin family and has cytolytic activity. Sortase A (SrtA), another crucial pneumococcal virulence determinate, contributes greatly to the anchoring of many virulence-associated surface proteins to the cell wall. In this study, epigallocatechin gallate (EGCG), a natural compound with little known antipneumococcal activity, was shown to directly inhibit PLY-mediated haemolysis and cytolysis by blocking the oligomerization of PLY and simultaneously reduce the peptidase activity of SrtA. The biofilm formation, production of neuraminidase A (NanA, the pneumococcal surface protein anchored by SrtA), and bacterial adhesion to human epithelial cells (Hep2) were inhibited effectively when S. pneumoniae D39 was cocultured with EGCG. The results from molecular dynamics simulations and mutational analysis confirmed the interaction of EGCG with PLY and SrtA, and EGCG binds to Glu277, Tyr358, and Arg359 in PLY and Thr169, Lys171, and Phe239 in SrtA. In vivo studies further demonstrated that EGCG protected mice against S. pneumoniae pneumonia. Our results imply that EGCG is an effective inhibitor of both PLY and SrtA and that an antivirulence strategy that directly targets PLY and SrtA using EGCG is a promising therapeutic option for S. pneumoniae pneumonia.


Subject(s)
Aminoacyltransferases/antagonists & inhibitors , Bacterial Proteins/antagonists & inhibitors , Catechin/analogs & derivatives , Pneumonia, Pneumococcal/prevention & control , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , A549 Cells , Aminoacyltransferases/genetics , Aminoacyltransferases/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/drug effects , Catechin/pharmacology , Cell Line, Tumor , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/metabolism , Female , Gene Expression Regulation, Bacterial/drug effects , Humans , Lung/drug effects , Lung/microbiology , Lung/pathology , Mice, Inbred BALB C , Microbial Sensitivity Tests , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/pathogenicity , Streptolysins/genetics , Streptolysins/metabolism , Virulence
11.
J Infect ; 74(6): 527-544, 2017 06.
Article in English | MEDLINE | ID: mdl-28322888

ABSTRACT

Acute pulmonary and cardiac injury remain significant causes of morbidity and mortality in those afflicted with severe pneumococcal disease, with the risk for early mortality often persisting several years beyond clinical recovery. Although remaining to be firmly established in the clinical setting, a considerable body of evidence, mostly derived from murine models of experimental infection, has implicated the pneumococcal, cholesterol-binding, pore-forming toxin, pneumolysin (Ply), in the pathogenesis of lung and myocardial dysfunction. Topics covered in this review include the burden of pneumococcal disease, risk factors, virulence determinants of the pneumococcus, complications of severe disease, antibiotic and adjuvant therapies, as well as the structure of Ply and the role of the toxin in disease pathogenesis. Given the increasing recognition of the clinical potential of Ply-neutralisation strategies, the remaining sections of the review are focused on updates of the types, benefits and limitations of currently available therapies which may attenuate, directly and/or indirectly, the injurious actions of Ply. These include recently described experimental therapies such as various phytochemicals and lipids, and a second group of more conventional agents the members of which remain the subject of ongoing clinical evaluation. This latter group, which is covered more extensively, encompasses macrolides, statins, corticosteroids, and platelet-targeted therapies, particularly aspirin.


Subject(s)
Pneumococcal Infections/drug therapy , Pneumonia, Pneumococcal/drug therapy , Streptolysins/antagonists & inhibitors , Streptolysins/metabolism , Adrenal Cortex Hormones/therapeutic use , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chemotherapy, Adjuvant , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Inflammation/drug therapy , Lactones/therapeutic use , Lung/microbiology , Lung/pathology , Macrolides/therapeutic use , Mice , Platelet Aggregation Inhibitors , Pneumococcal Infections/complications , Pneumococcal Infections/microbiology , Pneumococcal Infections/therapy , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/microbiology , Pyridines/therapeutic use , Risk Factors , Streptococcus pneumoniae/pathogenicity , Streptolysins/chemistry
12.
Fitoterapia ; 115: 31-36, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27693741

ABSTRACT

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.


Subject(s)
Apigenin/pharmacology , Pneumonia, Pneumococcal/drug therapy , Streptolysins/antagonists & inhibitors , A549 Cells , Animals , Bacterial Proteins/antagonists & inhibitors , Female , Hemolysis , Humans , Mice , Mice, Inbred BALB C
13.
Mol Pharmacol ; 89(3): 376-87, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26700563

ABSTRACT

Pneumolysin (PLY), an essential virulence factor of Streptococcus pneumoniae (pneumococcus), can penetrate the physical defenses of the host and possesses inflammatory properties. The vital role PLY plays in pneumococcus pathogenesis makes this virulence factor one of the most promising targets for the treatment of pneumococcal infection. Verbascoside (VBS) is an agent that does not exhibit bacteriostatic activity but has been shown to inhibit PLY-mediated cytotoxicity. The results from molecular dynamics simulations and mutational analysis indicated that VBS binds to the cleft between domains 3 and 4 of PLY, thereby blocking PLY's oligomerization and counteracting its hemolytic activity. Moreover, VBS can effectively alleviate PLY-mediated human alveolar epithelial (A549) cell injury, and treatment with VBS provides significant protection against lung damage and reduces mortality in a pneumococcal pneumonia murine model. Our results demonstrate that VBS is a strong candidate as a novel therapeutic in the treatment of Streptococcus pneumoniae infection.


Subject(s)
Glucosides/metabolism , Glucosides/therapeutic use , Phenols/metabolism , Phenols/therapeutic use , Pneumonia, Pneumococcal/drug therapy , Streptolysins/antagonists & inhibitors , Streptolysins/metabolism , Animals , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Anti-Infective Agents/therapeutic use , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites/drug effects , Binding Sites/physiology , Female , Glucosides/pharmacology , Humans , Mice , Mice, Inbred C57BL , Phenols/pharmacology , Pneumonia, Pneumococcal/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sheep , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/metabolism
14.
Am J Respir Cell Mol Biol ; 53(5): 689-702, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25874372

ABSTRACT

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.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Epithelial Cells/drug effects , Hemolysin Proteins/antagonists & inhibitors , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunity, Innate/drug effects , Simvastatin/pharmacology , Streptolysins/antagonists & inhibitors , Animals , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/toxicity , Bacterial Toxins/toxicity , Cell Line, Transformed , Epithelial Cells/immunology , Epithelial Cells/pathology , Hemolysin Proteins/toxicity , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/immunology , Injections, Intraperitoneal , Lung/drug effects , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Pravastatin/immunology , Pravastatin/pharmacology , Primary Cell Culture , Respiratory Mucosa/drug effects , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Simvastatin/immunology , Staphylococcus aureus/chemistry , Streptococcus pneumoniae/chemistry , Streptolysins/toxicity
15.
ACS Chem Biol ; 10(5): 1217-26, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25668590

ABSTRACT

Streptolysin S (SLS) is a post-translationally modified peptide cytolysin that is produced by the human pathogen Streptococcus pyogenes. SLS belongs to a large family of azole-containing natural products that are biosynthesized via an evolutionarily conserved pathway. SLS is an important virulence factor during S. pyogenes infections, but despite an extensive history of study, further investigations are needed to clarify several steps of its biosynthesis. To this end, chemical inhibitors of SLS biosynthesis would be valuable tools to interrogate the various maturation steps of both SLS and biosynthetically related natural products. Such chemical inhibitors could also potentially serve as antivirulence therapeutics, which in theory may alleviate the spread of antibiotic resistance. In this work, we demonstrate that FDA-approved HIV protease inhibitors, especially nelfinavir, block a key proteolytic processing step during SLS production. This inhibition was demonstrated in live S. pyogenes cells and through in vitro protease inhibition assays. A panel of 57 nelfinavir analogs was synthesized, leading to a series of compounds with improved anti-SLS activity while illuminating structure-activity relationships. Nelfinavir was also found to inhibit the maturation of other azole-containing natural products, namely those involved in listeriolysin S, clostridiolysin S, and plantazolicin production. The use of nelfinavir analogs as inhibitors of SLS production has allowed us to begin examining the proteolysis event in SLS maturation and will aid in further investigations of the biosynthesis of SLS and related natural products.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , HIV Protease Inhibitors/pharmacology , Streptolysins/antagonists & inhibitors , Amino Acid Sequence , Aspartic Acid Proteases/antagonists & inhibitors , Bacterial Proteins/biosynthesis , Molecular Sequence Data , Protease Inhibitors/pharmacology , Proteolysis , Sequence Homology, Amino Acid , Streptococcus pyogenes/drug effects , Streptococcus pyogenes/metabolism , Streptolysins/biosynthesis
16.
FASEB J ; 29(4): 1516-28, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25550455

ABSTRACT

The virulence of many Gram-positive bacteria depends on cholesterol-dependent cytolysins (CDCs), which form pores in eukaryotic cell plasma membranes. Pyolysin (PLO) from Trueperella pyogenes provided a unique opportunity to explore cellular responses to CDCs because it does not require thiol activation. Sublytic concentrations of PLO stimulated phosphorylation of MAPK ERK and p38 in primary stromal cells, and induced autophagy as determined by protein light-chain 3B cleavage. Although, inhibitors of MAPK or autophagy did not affect PLO-induced cytolysis. However, 10 µM 3-hydroxynaphthalene-2-carboxylic acid-(3,4-dihydroxybenzylidene)-hydrazide (Dynasore), a dynamin guanosine 5'-triphosphatase inhibitor, protected stromal cells against PLO-induced cytolysis as determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay (85 ± 17% versus 50 ± 9% cell viability), measuring extracellular ATP, and kinetic assays. This was a generalized mechanism because Dynasore also protected HeLa cells against streptolysin O. Furthermore, the effect was reversible, with stromal cell sensitivity to PLO restored within 30 minutes of Dynasore removal. The protective effect of Dynasore was not conferred by dynamin inhibition, induction of ERK phosphorylation, or Dynasore binding to PLO. Rather, Dynasore reduced cellular cholesterol and disrupted plasma membrane lipid rafts, similar to positive control methyl-ß-cyclodextrin. Dynasore is a tractable tool to explore the complexity of cholesterol homeostasis in eukaryotic cells and to develop strategies to counter CDCs.


Subject(s)
Actinomycetaceae/pathogenicity , Cytotoxins/antagonists & inhibitors , Cytotoxins/toxicity , Dynamins/antagonists & inhibitors , Hydrazones/pharmacology , Animals , Autophagy/drug effects , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/toxicity , Cattle , Cell Survival/drug effects , Cells, Cultured , Cholesterol/metabolism , Endometrium/drug effects , Endometrium/metabolism , Endometrium/microbiology , Female , HeLa Cells , Humans , MAP Kinase Signaling System/drug effects , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Models, Biological , Streptolysins/antagonists & inhibitors , Streptolysins/toxicity , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/microbiology
17.
PLoS Pathog ; 10(9): e1004353, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25188225

ABSTRACT

Streptococcus pneumoniae produces the pore-forming toxin pneumolysin (PLY), which is a member of the cholesterol-dependent cytolysin (CDC) family of toxins. The CDCs recognize and bind the 3ß-hydroxyl group of cholesterol at the cell surface, which initiates membrane pore formation. The cholesterol transport lipoproteins, which carry cholesterol in their outer monolayer, are potential off-pathway binding targets for the CDCs and are present at significant levels in the serum and the interstitial spaces of cells. Herein we show that cholesterol carried specifically by the ApoB-100-containing lipoprotein particles (CH-ApoB-100) in the mouse, but not that carried by human or guinea pig particles, is a potent inhibitor of the PLY pore-forming mechanism. Cholesterol present in the outer monolayer of mouse ApoB-100 particles is recognized and bound by PLY, which stimulates premature assembly of the PLY oligomeric complex thereby inactivating PLY. These studies further suggest that the vast difference in the inhibitory capacity of mouse CH-ApoB-100 and that of the human and the guinea pig is due to differences in the presentation of cholesterol in the outer monolayer of their ApoB-100 particles. Therefore mouse CH-ApoB-100 represents a significant innate CDC inhibitor that is absent in humans, which may underestimate the contribution of CDCs to human disease when utilizing mouse models of disease.


Subject(s)
Apolipoprotein B-100/metabolism , Cholesterol/metabolism , Hemolysis/drug effects , Lipoproteins/metabolism , Streptolysins/antagonists & inhibitors , Streptolysins/pharmacology , Animals , Antibodies, Neutralizing/blood , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/pharmacology , Cell Membrane/metabolism , Guinea Pigs , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
18.
Antimicrob Agents Chemother ; 58(9): 5164-80, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24957840

ABSTRACT

In this study, our objective was to determine whether a synergistic antimicrobial combination in vitro would be beneficial in the downregulation of pneumococcal virulence genes and whether the associated inflammation of the lung tissue induced by multidrug-resistant Streptococcus pneumoniae infection in vivo needs to be elucidated in order to consider this mode of therapy in case of severe pneumococcal infection. We investigated in vivo changes in the expression of these virulence determinants using an efficacious combination determined in previous studies. BALB/c mice were infected with 10(6) CFU of bacteria. Intravenous levofloxacin at 150 mg/kg and/or ceftriaxone at 50 mg/kg were initiated 18 h postinfection; the animals were sacrificed 0 to 24 h after the initiation of treatment. The levels of cytokines, chemokines, and C-reactive protein (CRP) in the serum and lungs, along with the levels of myeloperoxidase and nitric oxide the inflammatory cell count in bronchoalveolar lavage fluid (BALF), changes in pneumolysin and autolysin gene expression and COX-2 and inducible nitric oxide synthase (iNOS) protein expression in the lungs were estimated. Combination therapy downregulated inflammation and promoted bacterial clearance. Pneumolysin and autolysin expression was downregulated, with a concomitant decrease in the expression of COX-2 and iNOS in lung tissue. Thus, the combination of levofloxacin and ceftriaxone can be considered for therapeutic use even in cases of pneumonia caused by drug-resistant isolates.


Subject(s)
Ceftriaxone/pharmacology , Levofloxacin/pharmacology , N-Acetylmuramoyl-L-alanine Amidase/antagonists & inhibitors , Pneumococcal Infections/drug therapy , Pneumonia, Pneumococcal/drug therapy , Pneumonia/drug therapy , Streptococcus pneumoniae/drug effects , Streptolysins/antagonists & inhibitors , Animals , Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Bacteremia/metabolism , Bacteremia/microbiology , Bacterial Proteins/antagonists & inhibitors , Bronchoalveolar Lavage Fluid/microbiology , Disease Models, Animal , Drug Resistance, Multiple, Bacterial/drug effects , Drug Therapy, Combination/methods , Male , Mice , Mice, Inbred BALB C , Pneumococcal Infections/metabolism , Pneumococcal Infections/microbiology , Pneumonia/metabolism , Pneumonia/microbiology , Pneumonia, Pneumococcal/metabolism , Pneumonia, Pneumococcal/microbiology , Streptococcus pneumoniae/metabolism , Virulence/drug effects , Virulence Factors/metabolism
19.
Biochem Biophys Res Commun ; 430(1): 14-9, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23159621

ABSTRACT

A previous study showed that the minimal epitope recognised by the PLY-5 mAb in the conserved undecapeptide Trp-rich loop of bacterial CDCs should consist of WEWWRT (Jacobs et al., 1999) [5]. Now, through immunoscreening of amino acid substitution analogues, it is concluded that the second Trp and the Arg residues are essential in the PLY-5 epitope. The E residue is an auxiliary epitope contributor. Antibody modelling and docking simulations provided support for these findings. For recognition by the antibody, the Trp-rich loop flipped out, mimicking the mechanism of membrane insertion. The displaced second Trp was seen to establish aromatic stacking interactions with aromatic residues of the antibody paratope and the notably extruded guanidium tip of the arginine residue mediated electrostatic interactions with well-exposed carboxylic groups of glutamic residues on the surface of the paratope. Thus, the epitope/paratope interaction is mainly mediated by aromatic and by ionic interactions.


Subject(s)
Antibodies, Blocking/immunology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Cholesterol/immunology , Cytotoxins/immunology , Epitopes/immunology , Streptolysins/immunology , Amino Acid Sequence , Amino Acid Substitution , Animals , Antibodies, Blocking/genetics , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Neutralizing/genetics , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/immunology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Conserved Sequence , Cytotoxins/antagonists & inhibitors , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/immunology , Hemolysis/immunology , Mice , Molecular Sequence Data , Oligopeptides/immunology , Streptolysins/antagonists & inhibitors , Tryptophan/immunology
20.
J Immunol ; 187(9): 4890-9, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21957143

ABSTRACT

Streptococcus pneumoniae is a Gram-positive, extracellular bacterium that is responsible for significant mortality and morbidity worldwide. Pneumolysin (PLY), a cytolysin produced by all clinical isolates of the pneumococcus, is one of the most important virulence factors of this pathogen. We have previously reported that PLY is an essential factor for activation of caspase-1 and consequent secretion of IL-1ß and IL-18 in macrophages infected with S. pneumoniae. However, the host molecular factors involved in caspase-1 activation are still unclear. To further elucidate the mechanism of caspase-1 activation in macrophages infected with S. pneumoniae, we examined the involvement of inflammasomes in inducing this cellular response. Our study revealed that apoptosis-associated specklike protein containing a caspase recruitment domain (ASC), an adaptor protein for inflammasome receptors such as nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) and absent in melanoma 2 (AIM2), is essentially required for the induction of caspase-1 activation by S. pneumoniae. Caspase-1 activation was partially impaired in NLRP3(-/-) macrophages, whereas knockdown and knockout of AIM2 resulted in a clear decrease in caspase-1 activation in response to S. pneumoniae. These results suggest that ASC inflammasomes, including AIM2 and NLRP3, are critical for caspase-1 activation induced by S. pneumoniae. Furthermore, ASC(-/-) mice were more susceptible than wild-type mice to S. pneumoniae, with impaired secretion of IL-1ß and IL-18 into the bronchoalveolar lavage after intranasal infection, suggesting that ASC inflammasomes contribute to the protection of host from infection with PLY-producing S. pneumoniae.


Subject(s)
Caspase 1/metabolism , Cytoskeletal Proteins/physiology , Immunity, Innate , Inflammasomes/physiology , Pneumococcal Infections/immunology , Pneumococcal Infections/metabolism , Animals , Apoptosis Regulatory Proteins , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/biosynthesis , CARD Signaling Adaptor Proteins , Carrier Proteins/physiology , Caspase 1/deficiency , Caspase 1/genetics , Cell Line , Cell Line, Transformed , Cells, Cultured , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , DNA-Binding Proteins , Disease Resistance/immunology , Enzyme Activation/immunology , Female , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Nuclear Proteins/physiology , Pneumococcal Infections/enzymology , Streptolysins/antagonists & inhibitors , Streptolysins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...