Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Microb Drug Resist ; 27(5): 602-615, 2021 May.
Article in English | MEDLINE | ID: mdl-33983855

ABSTRACT

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.


Subject(s)
Antimicrobial Cationic Peptides/pharmacology , Bacterial Toxins/antagonists & inhibitors , Exotoxins/antagonists & inhibitors , Leukocidins/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/drug effects , Antimicrobial Cationic Peptides/pharmacokinetics , Humans , Microbial Sensitivity Tests , Molecular Docking Simulation , Cathelicidins
2.
J Pharm Pharmacol ; 73(4): 505-514, 2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33793838

ABSTRACT

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.


Subject(s)
Aggregatibacter actinomycetemcomitans , Catechin/analogs & derivatives , Exotoxins , Periodontitis , Aggregatibacter actinomycetemcomitans/drug effects , Aggregatibacter actinomycetemcomitans/metabolism , Aggregatibacter actinomycetemcomitans/pathogenicity , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Catechin/pharmacology , Coculture Techniques/methods , Dose-Response Relationship, Drug , Exotoxins/antagonists & inhibitors , Exotoxins/metabolism , Humans , Periodontitis/drug therapy , Periodontitis/microbiology , Virulence/drug effects
3.
J Am Chem Soc ; 142(25): 10899-10904, 2020 06 24.
Article in English | MEDLINE | ID: mdl-32479075

ABSTRACT

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.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Exotoxins/antagonists & inhibitors , Sulfur Compounds/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain , Click Chemistry , Crystallography, X-Ray , Cysteine Proteinase Inhibitors/metabolism , Cysteine Proteinase Inhibitors/toxicity , Drug Discovery , Exotoxins/chemistry , Exotoxins/metabolism , High-Throughput Screening Assays , Humans , Jurkat Cells , Microsomes, Liver/metabolism , Proof of Concept Study , Protein Binding
4.
J Leukoc Biol ; 108(3): 967-981, 2020 09.
Article in English | MEDLINE | ID: mdl-32531864

ABSTRACT

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.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Exotoxins/antagonists & inhibitors , Induced Pluripotent Stem Cells/cytology , Leukocidins/antagonists & inhibitors , Macrophages/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Receptor, Anaphylatoxin C5a/drug effects , Staphylococcus aureus , Animals , CD11b Antigen/immunology , CRISPR-Cas Systems , Cell Differentiation , Cells, Cultured , Exotoxins/deficiency , Gene Knock-In Techniques , Humans , Interleukin-1beta/metabolism , Leukocyte Common Antigens/physiology , Lung/immunology , Lung/microbiology , Macrophages/cytology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Monocytes/cytology , Peptide Fragments/immunology , Pneumonia, Staphylococcal/immunology , Protein Subunits , Receptor, Anaphylatoxin C5a/deficiency , Receptor, Anaphylatoxin C5a/genetics , Receptor, Anaphylatoxin C5a/physiology , Recombinant Proteins/metabolism , Staphylococcus aureus/physiology
5.
Toxins (Basel) ; 11(11)2019 11 14.
Article in English | MEDLINE | ID: mdl-31739483

ABSTRACT

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.


Subject(s)
Aggregatibacter actinomycetemcomitans/chemistry , Exotoxins/antagonists & inhibitors , Proanthocyanidins/chemistry , Vaccinium macrocarpon/chemistry , Exotoxins/pharmacology , Humans , U937 Cells
6.
Sci Rep ; 9(1): 14928, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624289

ABSTRACT

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.


Subject(s)
ADP Ribose Transferases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Exotoxins/antagonists & inhibitors , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Single-Chain Antibodies/pharmacology , Virulence Factors/antagonists & inhibitors , ADP Ribose Transferases/genetics , ADP Ribose Transferases/immunology , ADP Ribose Transferases/metabolism , Anti-Bacterial Agents/immunology , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Bacterial Toxins/genetics , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Catalytic Domain/genetics , Complementarity Determining Regions/immunology , Complementarity Determining Regions/pharmacology , Exotoxins/genetics , Exotoxins/immunology , Exotoxins/metabolism , HeLa Cells , Humans , Molecular Docking Simulation , Peptide Library , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/immunology , Pseudomonas aeruginosa/pathogenicity , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Single-Chain Antibodies/immunology , Single-Chain Antibodies/therapeutic use , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/metabolism , Pseudomonas aeruginosa Exotoxin A
7.
J Med Microbiol ; 68(2): 255-262, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30556803

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/biosynthesis , Methicillin/pharmacology , Oxazolidinones/pharmacology , Staphylococcus aureus/drug effects , Tetrazoles/pharmacology , Animals , Anti-Bacterial Agents/administration & dosage , Bacterial Toxins/analysis , Bacterial Toxins/antagonists & inhibitors , Dose-Response Relationship, Drug , Enterotoxins/analysis , Enterotoxins/antagonists & inhibitors , Enterotoxins/biosynthesis , Erythrocytes/drug effects , Erythrocytes/metabolism , Exotoxins/analysis , Exotoxins/antagonists & inhibitors , Exotoxins/biosynthesis , Hemolysin Proteins/analysis , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/biosynthesis , Humans , Leukocidins/analysis , Leukocidins/antagonists & inhibitors , Leukocidins/biosynthesis , Linezolid/administration & dosage , Linezolid/pharmacology , Methicillin Resistance , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Nafcillin/administration & dosage , Nafcillin/pharmacology , Oxazolidinones/administration & dosage , Rabbits , Sheep , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Superantigens/analysis , Superantigens/biosynthesis , Tetrazoles/administration & dosage
8.
ACS Infect Dis ; 4(7): 1073-1081, 2018 07 13.
Article in English | MEDLINE | ID: mdl-29742342

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Exotoxins/antagonists & inhibitors , Lymphocyte Function-Associated Antigen-1/chemistry , Peptides/pharmacology , Amino Acid Sequence , Anti-Bacterial Agents/chemistry , Exotoxins/chemistry , Exotoxins/toxicity , Humans , Lymphocyte Function-Associated Antigen-1/pharmacology , Models, Biological , Peptides/chemistry , Protein Binding , Structure-Activity Relationship , THP-1 Cells , Virulence Factors/antagonists & inhibitors , Virulence Factors/chemistry
9.
J Infect Chemother ; 24(5): 383-388, 2018 May.
Article in English | MEDLINE | ID: mdl-29409694

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/blood , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/blood , Community-Acquired Infections/drug therapy , Exotoxins/antagonists & inhibitors , Exotoxins/blood , Immunoglobulins, Intravenous/administration & dosage , Leukocidins/antagonists & inhibitors , Leukocidins/blood , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Infections/drug therapy , Antibodies, Neutralizing/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Toxins/immunology , Buffers , Community-Acquired Infections/immunology , Exotoxins/immunology , Humans , Leukocidins/immunology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Neutrophils/drug effects , Neutrophils/immunology , Staphylococcal Infections/immunology
10.
PLoS One ; 11(11): e0166477, 2016.
Article in English | MEDLINE | ID: mdl-27829022

ABSTRACT

Cholera toxin (CT) is an AB-type protein toxin that contains a catalytic A1 subunit, an A2 linker, and a cell-binding B homopentamer. The CT holotoxin is released into the extracellular environment, but CTA1 attacks a target within the cytosol of a host cell. We recently reported that grape extract confers substantial resistance to CT. Here, we used a cell culture system to identify twelve individual phenolic compounds from grape extract that inhibit CT. Additional studies determined the mechanism of inhibition for a subset of the compounds: two inhibited CT binding to the cell surface and even stripped CT from the plasma membrane of a target cell; two inhibited the enzymatic activity of CTA1; and four blocked cytosolic toxin activity without directly affecting the enzymatic function of CTA1. Individual polyphenolic compounds from grape extract could also generate cellular resistance to diphtheria toxin, exotoxin A, and ricin. We have thus identified individual toxin inhibitors from grape extract and some of their mechanisms of inhibition against CT.


Subject(s)
Biflavonoids/pharmacology , Catechin/analogs & derivatives , Cholera Toxin/antagonists & inhibitors , Phenols/pharmacology , Proanthocyanidins/pharmacology , ADP Ribose Transferases/antagonists & inhibitors , Animals , Bacterial Toxins/antagonists & inhibitors , Binding Sites/drug effects , CHO Cells , Catechin/pharmacology , Cell Membrane/metabolism , Cells, Cultured , Chlorocebus aethiops , Cholera Toxin/metabolism , Cricetulus , Diphtheria Toxin/antagonists & inhibitors , Exotoxins/antagonists & inhibitors , Fruit/chemistry , Grape Seed Extract/pharmacology , Molecular Docking Simulation , Plant Extracts/pharmacology , Ricin/antagonists & inhibitors , Vero Cells , Virulence Factors/antagonists & inhibitors , Vitis/chemistry , Pseudomonas aeruginosa Exotoxin A
11.
J Membr Biol ; 249(4): 503-11, 2016 08.
Article in English | MEDLINE | ID: mdl-27039399

ABSTRACT

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.


Subject(s)
Anthraquinones/pharmacology , Antitoxins/pharmacology , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/metabolism , Anthraquinones/metabolism , Antitoxins/metabolism , Bacterial Toxins/toxicity , Cell Line , Cell Membrane/chemistry , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Exotoxins/antagonists & inhibitors , Exotoxins/metabolism , Exotoxins/toxicity , Humans , Membrane Fluidity/drug effects , Temperature
12.
J Biomol Screen ; 21(9): 942-55, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27112173

ABSTRACT

Synthetic lethal screens are used to discover new combination treatments for cancer. In traditional high-throughput synthetic lethal screens, compounds are tested at a single dose, and hit selection is based on threshold activity values from the variance of the efficacy of the compounds tested. The limitation of the single-dose screening for synthetic lethal screens is that it does not allow for the robust detection of differential activities from compound collections with a broad range of potencies and efficacies. There is therefore a need to develop screening approaches that enable the identification of compounds with synthetic lethal effects based on changes in both potency and efficacy. Here we describe the implementation of a dose response-based synthetic lethal screen to find drugs that enhance or mitigate the cytotoxic effect of an immunotoxin protein (HA22). We developed a data analysis framework for the selection of compounds with enhancing or mitigating cytotoxic activities based on the use of dose-response parameters. The data analysis framework includes an ensemble ranking approach that allows the use of multiple dose-response parameters in a nonparametric fashion. Quantitative high-throughput screening (HTS) enables the identification of compounds with synthetic lethal activity not identified by single-dose HTS.


Subject(s)
High-Throughput Screening Assays/methods , Neoplasms/drug therapy , Small Molecule Libraries/pharmacology , Synthetic Lethal Mutations/genetics , Bacterial Toxins/antagonists & inhibitors , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Exotoxins/antagonists & inhibitors , Humans , Neoplasms/genetics , Small Molecule Libraries/isolation & purification , Small Molecule Libraries/therapeutic use
13.
J Biomol Screen ; 21(6): 590-5, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26850638

ABSTRACT

The gram-negative bacterium Pseudomonas aeruginosa is an opportunistic pathogen associated with drug resistance complications and, as such, an important object for drug discovery efforts. One attractive target for development of therapeutics is the ADP-ribosyltransferase Exotoxin-S (ExoS), an early effector of the type III secretion system that is delivered into host cells to affect their transcription pattern and cytoskeletal dynamics. The purpose of this study was to formulate a real-time assay of purified recombinant ExoS activity for high-throughput application. We characterized the turnover kinetics of the fluorescent dinucleotide 1,N(6)-etheno-NAD+ as co-substrate for ExoS. Further, we found that the toxin relied on any of five tested isoforms of human 14-3-3 to modify vH-Ras and the Rho-family GTPases Rac1, -2, and -3 and RhoC. We then used 14-3-3ß-stimulated ExoS modification of vH-Ras to screen a collection of low-molecular-weight compounds selected to target the poly-ADP ribose polymerase family and identified 3-(4-oxo-3,5,6,7-tetrahydro-4H-cyclopenta[4,5]thieno[2,3-d]pyrimidin-2-yl)propanoic acid as an ExoS inhibitor with micromolar potency. Thus, we present an optimized method to screen for inhibitors of ExoS activity that is amenable to high-throughput format and an intermediate affinity inhibitor that can serve both as assay control and as a starting point for further development.


Subject(s)
ADP Ribose Transferases/antagonists & inhibitors , Bacterial Toxins/antagonists & inhibitors , Drug Discovery , Host-Pathogen Interactions/genetics , Pseudomonas Infections/drug therapy , Small Molecule Libraries/pharmacology , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Drug Resistance, Bacterial , Exotoxins/antagonists & inhibitors , Exotoxins/genetics , Humans , Pseudomonas Infections/genetics , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/pathogenicity , Substrate Specificity , rac1 GTP-Binding Protein/chemistry , rac1 GTP-Binding Protein/genetics
14.
Mol Oral Microbiol ; 31(1): 94-105, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26352738

ABSTRACT

The leukotoxin (LtxA) produced by Aggregatibacter actinomycetemcomitans kills host immune cells, allowing the bacterium to establish an ecological niche in the upper aerodigestive tract of its human host. The interaction of LtxA with human immune cells is both complex and multifaceted, involving membrane lipids as well as cell-surface proteins. In the initial encounter with the host cell, LtxA associates with lymphocyte function-associated antigen-1, a cell surface adhesion glycoprotein. However, we have also demonstrated that the toxin associates strongly with the plasma membrane lipids, specifically cholesterol. This association with cholesterol is regulated by a cholesterol recognition amino acid consensus (CRAC) motif, with a sequence of (334) LEEYSKR(340), in the N-terminal region of the toxin. Here, we have demonstrated that removal of cholesterol from the plasma membrane or mutation of the LtxA CRAC motif inhibits the activity of the toxin in THP-1 cells. To inhibit LtxA activity, we designed a short peptide corresponding to the CRAC(336) motif of LtxA (CRAC(336WT)). This peptide binds to cholesterol and thereby inhibits the toxicity of LtxA in THP-1 cells. Previously, we showed that this peptide inhibits LtxA toxicity against Jn.9 (Jurkat) cells, indicating that peptides derived from the cholesterol-binding site of LtxA may have a potential clinical applicability in controlling infections of repeats-in-toxin-producing organisms.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Cholesterol/metabolism , Exotoxins/antagonists & inhibitors , Exotoxins/toxicity , Pancreatic Elastase/antagonists & inhibitors , Peptides/metabolism , Aggregatibacter actinomycetemcomitans/immunology , Aggregatibacter actinomycetemcomitans/metabolism , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Bacterial Toxins/metabolism , Carrier Proteins/metabolism , Cell Adhesion/drug effects , Cell Membrane/drug effects , Cell Membrane/metabolism , Exotoxins/immunology , Exotoxins/metabolism , Host-Pathogen Interactions , Humans , Jurkat Cells , Lipid Bilayers/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Mutation , Pancreatic Elastase/metabolism
15.
World J Microbiol Biotechnol ; 31(10): 1565-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26193949

ABSTRACT

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.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Diterpenes/metabolism , Enterotoxins/antagonists & inhibitors , Enzyme Inhibitors/metabolism , Exotoxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Staphylococcus aureus/metabolism , Abietanes , Animals , Blotting, Western , Cell Line , Hemolysis , Macrophages/drug effects , Macrophages/microbiology , Mice , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
16.
Biochemistry ; 54(28): 4365-73, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26132413

ABSTRACT

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.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Cysteine Proteinase Inhibitors/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Exotoxins/antagonists & inhibitors , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Streptococcus pyogenes/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Catalytic Domain/drug effects , Crystallography, X-Ray , Cysteine Endopeptidases/chemistry , Cysteine Endopeptidases/metabolism , Exotoxins/chemistry , Exotoxins/metabolism , Humans , Molecular Docking Simulation , Protein Conformation/drug effects , Streptococcal Infections/drug therapy , Streptococcal Infections/microbiology , Streptococcus pyogenes/drug effects
17.
Toxicon ; 99: 130-5, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25841345

ABSTRACT

Ectoine (ECT) is a bacterial compatible solute with documented protective action however no data are available on its effects on various cells against bacterial toxins. Therefore, we determined the in vitro influence of ECT on bovine erythrocytes subjected to staphylococcal α-haemolysin (HlyA). The cells exposed to HlyA alone showed a distinct haemolysis and reduced glutathione (GSH)/oxidised glutathione (GSSG) level, however the toxic effects were attenuated in the combinations of HlyA + ECT suggesting ECT-induced protection of erythrocytes from HlyA.


Subject(s)
Amino Acids, Diamino/pharmacology , Bacterial Toxins/antagonists & inhibitors , Cryoprotective Agents/pharmacology , Erythrocytes/drug effects , Exotoxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Hemolysis/drug effects , Hemolytic Agents/toxicity , Animals , Bacterial Toxins/toxicity , Cattle , Exotoxins/toxicity , Glutathione/chemistry , Glutathione/metabolism , Hemolysin Proteins/toxicity , Hemolytic Agents/chemistry , Kinetics , Oxidation-Reduction , Oxidative Stress/drug effects , Poland
18.
Microbiol Mol Biol Rev ; 78(2): 199-230, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24847020

ABSTRACT

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


Subject(s)
Exotoxins/metabolism , Leukocidins/metabolism , Staphylococcus aureus/metabolism , Animals , Exotoxins/antagonists & inhibitors , Exotoxins/chemistry , Exotoxins/genetics , Gene Expression Regulation, Bacterial , Gene Order , Genome, Bacterial , History, 19th Century , History, 20th Century , Host-Pathogen Interactions , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Leukocidins/antagonists & inhibitors , Leukocidins/chemistry , Leukocidins/genetics , Microbiology/history , Signal Transduction , Species Specificity , Staphylococcal Infections/drug therapy , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Virulence
19.
Biochem Biophys Res Commun ; 444(3): 433-8, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24472539

ABSTRACT

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.


Subject(s)
Aptamers, Nucleotide/pharmacology , Bacterial Toxins/antagonists & inhibitors , Exotoxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Staphylococcus aureus/chemistry , Humans , Jurkat Cells , SELEX Aptamer Technique
20.
Antimicrob Agents Chemother ; 57(11): 5432-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959313

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacillus anthracis/drug effects , Staphylococcus aureus/drug effects , Streptococcus agalactiae/drug effects , Streptococcus pyogenes/drug effects , Vitamin K 2/pharmacology , Administration, Topical , Animals , Bacillus anthracis/growth & development , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Cell Membrane/drug effects , Drug Synergism , Exotoxins/antagonists & inhibitors , Exotoxins/metabolism , Humans , Laurates/pharmacology , Monoglycerides/pharmacology , Rabbits , Reactive Oxygen Species/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/metabolism , Shock, Septic/drug therapy , Shock, Septic/microbiology , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Streptococcus agalactiae/growth & development , Streptococcus pyogenes/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...