Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 142
Filter
1.
Sci Rep ; 14(1): 9469, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658583

ABSTRACT

Bovine mastitis caused by S. aureus has a major economic impact on the dairy sector. With the crucial need for new therapies, anti-virulence strategies have gained attention as alternatives to antibiotics. Here we aimed to identify novel compounds that inhibit the production/activity of hemolysins, a virulence factor of S. aureus associated with mastitis severity. We screened Bacillus strains obtained from diverse sources for compounds showing anti-hemolytic activity. Our results demonstrate that lipopeptides produced by Bacillus spp. completely prevented the hemolytic activity of S. aureus at certain concentrations. Following purification, both iturins, fengycins, and surfactins were able to reduce hemolysis caused by S. aureus, with iturins showing the highest anti-hemolytic activity (up to 76% reduction). The lipopeptides showed an effect at the post-translational level. Molecular docking simulations demonstrated that these compounds can bind to hemolysin, possibly interfering with enzyme action. Lastly, molecular dynamics analysis indicated general stability of important residues for hemolysin activity as well as the presence of hydrogen bonds between iturins and these residues, with longevous interactions. Our data reveals, for the first time, an anti-hemolytic activity of lipopeptides and highlights the potential application of iturins as an anti-virulence therapy to control bovine mastitis caused by S. aureus.


Subject(s)
Bacillus , Hemolysin Proteins , Hemolysis , Lipopeptides , Molecular Docking Simulation , Staphylococcus aureus , Bacillus/metabolism , Bacillus/chemistry , Staphylococcus aureus/drug effects , Hemolysis/drug effects , Animals , Cattle , Lipopeptides/pharmacology , Lipopeptides/chemistry , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Mastitis, Bovine/microbiology , Mastitis, Bovine/drug therapy , Female , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemistry , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Molecular Dynamics Simulation
2.
APMIS ; 130(9): 578-589, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35751523

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is resistant to almost all ß-lactam antibiotics. Hence, new ways to control MRSA infection, such as antibacterial antibodies, need to be explored. α-hemolysin is the most important virulence factor widely expressed in S. aureus. This study aimed to develop a new fully human antibody against α-hemolysin of S. aureus and research its neutralizing effect. The single-chain antibody fragments (scFvs) against S. aureus were screened from a fully human scFv library using phage display technology. The selected scFvs had good binding affinities to α-hemolysin and S. aureus. The IgG-like scFv-Fc inserted into the pcDNA3.1 or pMH3 vector was expressed in HEK293F suspension cells to extend the half-life and restore Fc function. The size of purified scFv-Fc was about 55 kDa. The functions of expressed scFv-Fcs against α-hemolysin were validated. The cytotoxicity assays showed that scFv555-Fc had better protective effects on A549 cells than other scFv-Fcs. The results of anti-rabbit erythrocyte lysis and A549 cell apoptosis assay confirmed that scFv555-Fc had a significant neutralizing effect on α-hemolysin. The scFv555-Fc was used to construct the docking model of antigen-antibody complexes using Discovery Studio software. It predicted that the key binding sites of α-hemolysin were TYR28, LYS37, PHE39, ARG56, and LYS58, which might be the key toxic sites of α-hemolysin. A novel fully human scFv-Fc antibody neutralizing the α-hemolysin toxin of S. aureus was successfully developed. The findings might provide a new theoretical basis and treatment method for preventing MRSA infection.


Subject(s)
Antibodies, Neutralizing , Hemolysin Proteins , Methicillin-Resistant Staphylococcus aureus , Single-Chain Antibodies , A549 Cells , Antibodies, Neutralizing/chemistry , Hemolysin Proteins/antagonists & inhibitors , Humans , Single-Chain Antibodies/chemistry , Staphylococcal Infections/prevention & control
3.
Molecules ; 26(24)2021 Dec 14.
Article in English | MEDLINE | ID: mdl-34946657

ABSTRACT

The rapid emergence of bacterial coinfection caused by cytosolic bacteria has become a huge threat to public health worldwide. Past efforts have been devoted to discover the broad-spectrum antibiotics, while the emergence of antibiotic resistance encourages the development of antibacterial agents. In essence, bacterial virulence is a factor in antibiotic tolerance. However, the discovery and development of new antibacterial drugs and special antitoxin drugs is much more difficult in the antibiotic resistance era. Herein, we hypothesize that antitoxin hemolytic activity can serve as a screening principle to select antibacterial drugs to combat coinfection from natural products. Being the most abundant natural drug of plant origins, flavonoids were selected to assess the ability of antibacterial coinfections in this paper. Firstly, we note that four flavonoids, namely, baicalin, catechin, kaempferol, and quercetin, have previously exhibited antibacterial abilities. Then, we found that baicalin, kaempferol, and quercetin have better inhibitions of hemolytic activity of Hla than catechin. In addition, kaempferol and quercetin, have therapeutic effectivity for the coinfections of Staphylococcus aureus and Pseudomonas aeruginosa in vitro and in vivo. Finally, our results indicated that kaempferol and quercetin therapied the bacterial coinfection by inhibiting S. aureus α-hemolysin (Hla) and reduced the host inflammatory response. These results suggest that antitoxins may play a promising role as a potential target for screening flavonoids to combat bacterial coinfection.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Flavonoids , Hemolysin Proteins , Pseudomonas aeruginosa/metabolism , Staphylococcus aureus/metabolism , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Drug Evaluation, Preclinical , Flavonoids/chemistry , Flavonoids/pharmacology , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/metabolism
4.
Theranostics ; 11(17): 8152-8171, 2021.
Article in English | MEDLINE | ID: mdl-34373734

ABSTRACT

Serious infection caused by multi-drug-resistant bacteria is a major threat to human health. Bacteria can invade the host tissue and produce various toxins to damage or kill host cells, which may induce life-threatening sepsis. Here, we aimed to explore whether fructose-coated Ångstrom-scale silver particles (F-AgÅPs), which were prepared by our self-developed evaporation-condensation system and optimized coating approach, could kill bacteria and sequester bacterial toxins to attenuate fatal bacterial infections. Methods: A series of in vitro assays were conducted to test the anti-bacterial efficacy of F-AgÅPs, and to investigate whether F-AgÅPs could protect against multi-drug resistant Staphylococcus aureus (S. aureus)- and Escherichia coli (E. coli)-induced cell death, and suppress their toxins (S. aureus hemolysin and E. coli lipopolysaccharide)-induced cell injury or inflammation. The mouse models of cecal ligation and puncture (CLP)- or E. coli bloodstream infection-induced lethal sepsis were established to assess whether the intravenous administration of F-AgÅPs could decrease bacterial burden, inhibit inflammation, and improve the survival rates of mice. The levels of silver in urine and feces of mice were examined to evaluate the excretion of F-AgÅPs. Results: F-AgÅPs efficiently killed various bacteria that can cause lethal infections and also competed with host cells to bind with S. aureus α-hemolysin, thus blocking its cytotoxic activity. F-AgÅPs inhibited E. coli lipopolysaccharide-induced endothelial injury and macrophage inflammation, but not by directly binding to lipopolysaccharide. F-AgÅPs potently reduced bacterial burden, reversed dysregulated inflammation, and enhanced survival in mice with CLP- or E. coli bloodstream infection-induced sepsis, either alone or combined with antibiotic therapy. After three times injections within 48 h, 79.18% of F-AgÅPs were excreted via feces at the end of the 14-day observation period. Conclusion: This study suggests the prospect of F-AgÅPs as a promising intravenous agent for treating severe bacterial infections.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Sepsis/drug therapy , Silver/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Disease Models, Animal , Escherichia coli/drug effects , Fructose/pharmacology , Hemolysin Proteins/antagonists & inhibitors , Inflammation/drug therapy , Lipopolysaccharides/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/drug effects , Mice , Nanoparticles/therapeutic use , Sepsis/microbiology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects
5.
Bioorg Chem ; 114: 105074, 2021 09.
Article in English | MEDLINE | ID: mdl-34174629

ABSTRACT

α-Hemolysin (Hla) is an extracellular protein secreted by methicillin-resistant Staphylococcus aureus (MRSA) strains that plays a critical role in the pathogenesis of pulmonary, intraperitoneal, intramammary, and corneal infections, rendering Hla a potential therapeutic target. In this study, 10 unreported polycyclic polyprenylated acylphloroglucinol (PPAP) derivatives, garciyunnanins C-L (1-10), with diverse skeletons, were isolated from Garcinia yunnanensis Hu. The structures of these new compounds were determined by HRMS, NMR, electronic circular dichroism (ECD) calculations, single-crystal X-ray diffraction, and biomimetic transformation. Garciyunnanins C and D (1 and 2) were found to be potent Hla inhibitors in the anti-virulence efficacy evaluation against MRSA strain.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Garcinia/chemistry , Hemolysin Proteins/antagonists & inhibitors , Phloroglucinol/pharmacology , Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Bacterial Toxins/biosynthesis , Dose-Response Relationship, Drug , Hemolysin Proteins/biosynthesis , Microbial Sensitivity Tests , Molecular Structure , Phloroglucinol/chemistry , Phloroglucinol/isolation & purification , Staphylococcus aureus/metabolism , Structure-Activity Relationship
6.
Cell Chem Biol ; 28(9): 1310-1320.e5, 2021 09 16.
Article in English | MEDLINE | ID: mdl-33852903

ABSTRACT

Biofilms are rigid and largely impenetrable three-dimensional matrices constituting virulence determinants of various pathogenic bacteria. Here, we demonstrate that molecular tweezers, unique supramolecular artificial receptors, modulate biofilm formation of Staphylococcus aureus. In particular, the tweezers affect the structural and assembly properties of phenol-soluble modulin α1 (PSMα1), a biofilm-scaffolding functional amyloid peptide secreted by S. aureus. The data reveal that CLR01, a diphosphate tweezer, exhibits significant S. aureus biofilm inhibition and disrupts PSMα1 self-assembly and fibrillation, likely through inclusion of lysine side chains of the peptide. In comparison, different peptide binding occurs in the case of CLR05, a tweezer containing methylenecarboxylate units, which exhibits lower affinity for the lysine residues yet disrupts S. aureus biofilm more strongly than CLR01. Our study points to a possible role for molecular tweezers as potent biofilm inhibitors and antibacterial agents, particularly against untreatable biofilm-forming and PSM-producing bacteria, such as methicillin-resistant S. aureus.


Subject(s)
Amyloid/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Biofilms/drug effects , Hemolysin Proteins/antagonists & inhibitors , Staphylococcus aureus/drug effects , Amyloid/metabolism , Anti-Bacterial Agents/chemistry , Bacterial Toxins/metabolism , Hemolysin Proteins/metabolism , Microbial Sensitivity Tests , Optical Tweezers , Staphylococcus aureus/metabolism
7.
Mol Immunol ; 135: 45-52, 2021 07.
Article in English | MEDLINE | ID: mdl-33873093

ABSTRACT

Staphylococcus aureus (SA), especially the methicillin-resistant variant (MRSA), is becoming a serious threat to human health in hospitals and communities, making the development of an effective vaccine urgent. Alpha-hemolysin (Hla) is a key virulence factor and also a good target for the development of SA vaccines. However, the epitopes in Hla recognized by human immunity are not characterized in detail, which hinders the design of epitope-based human vaccines against SA. In this study, we collected sera from volunteers in a phase 1b clinical trial of a novel recombinant five-antigen SA vaccine (NCT03966040). Using a Luminex-based assay, we characterized the human serologic response against Hla, and identified Hla121-138 as a neutralizing epitope. In addition, we successfully produced ferritin nanoparticles carrying the neutralizing Hla121-138 epitope (EpNP) in E. coli. EpNP presented as homogenous nanoparticles in aqueous solution. Immunization with EpNP elicited potent hemolysis-neutralizing antibodies and conferred significant protection in a mouse model of SA skin infection. Our data suggest that EpNP, carrying the neutralizing epitope Hla121-138, is a good candidate for a vaccine against SA.


Subject(s)
Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Bacterial Toxins/immunology , Hemolysin Proteins/immunology , Methicillin-Resistant Staphylococcus aureus/immunology , Staphylococcal Infections/prevention & control , Animals , Bacterial Toxins/antagonists & inhibitors , Bacterial Vaccines/immunology , Epitopes/immunology , Female , Hemolysin Proteins/antagonists & inhibitors , Humans , Metal Nanoparticles/chemistry , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred BALB C , Staphylococcal Infections/microbiology , Vaccination , Virulence Factors
8.
Molecules ; 26(7)2021 Apr 01.
Article in English | MEDLINE | ID: mdl-33915741

ABSTRACT

As an important zoonotic pathogen, Streptococcus suis (S. suis) can cause a variety of diseases both in human and animals, especially Streptococcal toxic shock-like syndrome (STSLS), which commonly appears in severe S. suis infection. STSLS is often accompanied by excessive production of inflammatory cytokines, which is the main cause of host death. Therefore, it is urgent to find a new strategy to relieve the damage caused by STSLS. In this study, we found, for the first time, that apigenin, as a flavonoid compound, could combine with ampicillin to treat severe S. suis infection. Studies found that apigenin did not affect the growth of S. suis and the secretion of suilysin (SLY), but it could significantly inhibit the hemolytic activity of SLY by directly binding to SLY and destroying its secondary structure. In cell assays, apigenin was found to have no significant toxic effects on effective concentrations, and have a good protective effect on S. suis-infected cells. More importantly, compared with the survival rate of S. suis-infected mice treated with only ampicillin, the survival rate of apigenin combined with an ampicillin-treated group significantly increased to 80%. In conclusion, all results indicate that apigenin in combination with conventional antibiotics can be a potential strategy for treating severe S. suis infection.


Subject(s)
Ampicillin/pharmacology , Anti-Bacterial Agents/pharmacology , Apigenin/pharmacology , Streptococcal Infections/drug therapy , Streptococcal Infections/microbiology , Streptococcus suis/drug effects , Ampicillin/chemistry , Ampicillin/therapeutic use , Animals , Anti-Bacterial Agents/chemistry , Apigenin/chemistry , Apigenin/therapeutic use , Binding Sites , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , Erythrocytes/drug effects , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/chemistry , Host-Pathogen Interactions , Humans , Inflammation Mediators/metabolism , Mice , Microbial Sensitivity Tests , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Binding , Streptococcal Infections/diagnosis , Streptococcal Infections/metabolism , Structure-Activity Relationship , Treatment Outcome
9.
J Appl Microbiol ; 130(3): 736-744, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32750224

ABSTRACT

AIMS: Streptococcus suis is a highly zoonotic pathogen that is a serious threat to human health and the development of the pig industry worldwide. The virulence factors produced during S. suis infection play an important role, and the pore-forming activity of suilysin is considered an important virulence-related factor, especially in meningitis. Treatment of S. suis infection with traditional antibiotics is becoming increasingly challenging due to bacterial resistance. The purpose of this study is to verify the role of cryptotanshinone in the process of S. suis infection and provide a new drug precursor for the treatment of S. suis infection. METHODS AND RESULTS: In this study, we used circular dichroism spectroscopy to demonstrate that cryptotanshinone alters the secondary structure of suilysin. The results of the antibacterial activity and haemolysis assays showed cryptotanshinone could inhibit the pore-forming activity of suilysin without affecting bacterial growth or its expression. We also showed that cryptotanshinone reduces bacterial damage and penetration in vitro, reduce the S. suis-induced inflammatory response and provide protection against bacterial infections in vivo and in vitro. CONCLUSIONS: Cryptotanshinone is a potential compound precursor for treating S. suis infection. SIGNIFICANCE AND IMPACT OF THE STUDY: Cryptotanshinone may be a promising leading compound for S. suis infection and related diseases.


Subject(s)
Anti-Bacterial Agents/pharmacology , Hemolysin Proteins/antagonists & inhibitors , Phenanthrenes/pharmacology , Streptococcal Infections/drug therapy , Streptococcus suis/drug effects , Animals , Anti-Bacterial Agents/therapeutic use , Hemolysin Proteins/chemistry , Hemolysis/drug effects , Humans , Inflammation/drug therapy , Mice , Phenanthrenes/therapeutic use , Protein Structure, Secondary/drug effects , Streptococcus suis/pathogenicity , Virulence/drug effects , Virulence Factors/metabolism
10.
Toxins (Basel) ; 12(12)2020 12 19.
Article in English | MEDLINE | ID: mdl-33352744

ABSTRACT

Bacillus cereus is the fourth most common cause of foodborne illnesses that produces a variety of pore-forming proteins as the main pathogenic factors. B. cereus hemolysin II (HlyII), belonging to pore-forming ß-barrel toxins, has a C-terminal extension of 94 amino acid residues designated as HlyIICTD. An analysis of a panel of monoclonal antibodies to the recombinant HlyIICTD protein revealed the ability of the antibody HlyIIC-20 to inhibit HlyII hemolysis. A conformational epitope recognized by HlyIIC-20 was found. by the method of peptide phage display and found that it is localized in the N-terminal part of HlyIICTD. The HlyIIC-20 interacted with a monomeric form of HlyII, thus suppressing maturation of the HlyII toxin. Protection efficiencies of various B. cereus strains against HlyII were different and depended on the epitope amino acid composition, as well as, insignificantly, on downstream amino acids. Substitution of L324P and P324L in the hemolysins ATCC14579T and B771, respectively, determined the role of leucine localized to the epitope in suppressing the hemolysis by the antibody. Pre-incubation of HlyIIC-20 with HlyII prevented the death of mice up to an equimolar ratio. A strategy of detecting and neutralizing the toxic activity of HlyII could provide a tool for monitoring and reducing B. cereus pathogenicity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Bacillus cereus/drug effects , Bacterial Proteins/antagonists & inhibitors , Erythrocytes/drug effects , Hemolysin Proteins/antagonists & inhibitors , Hemolysis/drug effects , Animals , Antibodies, Monoclonal/chemistry , Bacillus cereus/chemistry , Bacillus cereus/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Erythrocytes/metabolism , Female , Hemolysin Proteins/chemistry , Hemolysin Proteins/metabolism , Hemolysis/physiology , Mice , Mice, Inbred BALB C , Protein Domains/drug effects , Protein Domains/physiology , Protein Structure, Secondary , Rabbits
11.
Chem Commun (Camb) ; 56(91): 14207-14210, 2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33111731

ABSTRACT

There is great interest in developing strategies to deliver proteins into the cytoplasm of cells. We report here a PEG-poly-eosin block copolymer (PEG-pEosin) that can encapsulate proteins and release them in active form under mildly acidic conditions. A PEG-pEosin formulation composed of Cre and the endosomolytic protein LLO efficiently performed gene editing in cells and in the brains of mice after an intracranial injection.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Eosine Yellowish-(YS)/pharmacology , Heat-Shock Proteins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Polyethylene Glycols/pharmacology , Animals , Bacterial Toxins/genetics , Cell Survival/drug effects , Eosine Yellowish-(YS)/chemistry , Erythrocytes/drug effects , Gene Editing , Heat-Shock Proteins/genetics , Hemolysin Proteins/genetics , Hydrogen-Ion Concentration , Mice , Models, Molecular , Molecular Structure , Polyethylene Glycols/chemistry
12.
Arch Microbiol ; 202(10): 2751-2760, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32737541

ABSTRACT

Staphylococcus aureus is an opportunistic pathogen that has the ability to cause a wide range of diseases including superficial infection and severe invasive life threatening infections. The pathogenicity of S. aureus is mediated by a group of virulence factors that mediate the colonization and penetration. The antibiotic resistance of S. aureus has evolved due to the abuse of antibiotics rendering the cure of infection very difficult especially with the shortage in new antibiotic production. To combat this shortage, repurposing of FDA-approved drugs against the virulence factors is a new strategy. The analgesic drug Diclofenac was found to have anti-virulence activity against Pseudomonas aeruginosa and Proteus mirabilis. This study aimed to demonstrate the anti-virulence effect of diclofenac against clinical MRSA isolates phenotypically and genotypically using qRT-PCR. In this study, diclofenac showed significant reduction in biofilm formation when compared to controls, the inhibition ranged between 22.67% and 70%. Also, remarkable inhibition of hemolysin activity was found (5.4-66.34%). Additionally, diclofenac has inhibitory activity against the staphyloxanthin production (8-57.2%). The results were confirmed by qRT-PCR that showed significant down-regulation of tested virulence genes. The down-regulation ranged from 43 to 64.05% for SarA, 36.85-64.75% for AgrA, 50-63.2% for hla, 38.55-60.35% for FnbA, 46.75-61.05% for IcaA, 27.55-64% for SigB and 51.05-72.8% for CrtM. In conclusion, diclofenac can be used in combination with antibiotics as anti-virulence agent against MDR-MRSA which will enhance the ability of immune system to eradicate infection.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Diclofenac/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Drug Resistance, Multiple, Bacterial/physiology , Genotype , Hemolysin Proteins/antagonists & inhibitors , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Microbial Sensitivity Tests , Proteus mirabilis/drug effects , Pseudomonas aeruginosa/drug effects , Staphylococcal Infections/drug therapy , Virulence/genetics , Virulence Factors/antagonists & inhibitors , Virulence Factors/genetics , Xanthophylls/antagonists & inhibitors
13.
Arch Pharm (Weinheim) ; 353(5): e1900352, 2020 May.
Article in English | MEDLINE | ID: mdl-32134150

ABSTRACT

In an attempt to counteract bacterial pathogenicity, a set of novel imidazolidine-2,4-dione and 2-thioxoimidazolidin-4-one derivatives was synthesized and evaluated as inhibitors of bacterial virulence. The new compounds were characterized and screened for their effects on the expression of virulence factors of Pseudomonas aeruginosa, including protease, hemolysin, and pyocyanin. Imidazolidine-2,4-diones 4c, 4j, and 12a showed complete inhibition of the protease enzyme, and they almost completely inhibited the production of hemolysin at 1/4 MIC (1/4 minimum inhibitory concentration; 1, 0.5, and 0.5 mg/ml, respectively). 2-Thioxoimidazolidin-4-one derivative 7a exhibited the best inhibitory activity (96.4%) against pyocyanin production at 1 mg/ml (1/4 MIC). A docking study was preformed to explore the potential binding interactions with quorum-sensing receptors (LasR and RhlR), which are responsible for the expression of virulence genes.


Subject(s)
Imidazolidines/pharmacology , Protease Inhibitors/pharmacology , Virulence Factors/antagonists & inhibitors , Dose-Response Relationship, Drug , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/biosynthesis , Imidazolidines/chemical synthesis , Imidazolidines/chemistry , Molecular Docking Simulation , Molecular Structure , Peptide Hydrolases/biosynthesis , Protease Inhibitors/chemical synthesis , Protease Inhibitors/chemistry , Pseudomonas aeruginosa/chemistry , Pseudomonas aeruginosa/enzymology , Pyocyanine/antagonists & inhibitors , Pyocyanine/biosynthesis , Structure-Activity Relationship , Virulence Factors/biosynthesis
14.
J Infect Dis ; 222(9): 1505-1516, 2020 10 01.
Article in English | MEDLINE | ID: mdl-31970394

ABSTRACT

BACKGROUND: Bacterial toxins disrupt plasma membrane integrity with multitudinous effects on host cells. The secreted pore-forming toxin listeriolysin O (LLO) of the intracellular pathogen Listeria monocytogenes promotes egress of the bacteria from vacuolar compartments into the host cytosol often without overt destruction of the infected cell. Intracellular LLO activity is tightly controlled by host factors including compartmental pH, redox, proteolytic, and proteostatic factors, and inhibited by cholesterol. METHODS: Combining infection studies of L. monocytogenes wild type and isogenic mutants together with biochemical studies with purified phospholipases, we investigate the effect of their enzymatic activities on LLO. RESULTS: Here, we show that phosphocholine (ChoP), a reaction product of the phosphatidylcholine-specific phospholipase C (PC-PLC) of L. monocytogenes, is a potent inhibitor of intra- and extracellular LLO activities. Binding of ChoP to LLO is redox-independent and leads to the inhibition of LLO-dependent induction of calcium flux, mitochondrial damage, and apoptosis. ChoP also inhibits the hemolytic activities of the related cholesterol-dependent cytolysins (CDC), pneumolysin and streptolysin. CONCLUSIONS: Our study uncovers a strategy used by L. monocytogenes to modulate cytotoxic LLO activity through the enzymatic activity of its PC-PLC. This mechanism appears to be widespread and also used by other CDC pore-forming toxin-producing bacteria.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Listeria monocytogenes/drug effects , Phosphorylcholine/pharmacology , Apoptosis , Calcium/metabolism , Caspase 3/metabolism , HeLa Cells , Humans , Listeria monocytogenes/enzymology , Listeria monocytogenes/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism
15.
ACS Synth Biol ; 9(2): 316-328, 2020 02 21.
Article in English | MEDLINE | ID: mdl-31995709

ABSTRACT

Lipid membranes are becoming increasingly popular in synthetic biology due to their biophysical properties and crucial role in communication between different compartments. Several alluring protein-membrane sensors have already been developed, whereas protein logic gates designs on membrane-embedded proteins are very limited. Here we demonstrate the construction of a two-level protein-membrane logic gate with an OR-AND logic. The system consists of an engineered pH-dependent pore-forming protein listeriolysin O and its DARPin-based inhibitor, conjugated to a lipid vesicle membrane. The gate responds to low pH and removal of the inhibitor from the membrane either by switching to a reducing environment, protease cleavage, or any other signal depending on the conjugation chemistry used for inhibitor attachment to the membrane. This unique protein logic gate vesicle system advances generic sensing and actuator platforms used in synthetic biology and could be utilized in drug delivery.


Subject(s)
Bacterial Toxins/metabolism , Heat-Shock Proteins/metabolism , Hemolysin Proteins/metabolism , Unilamellar Liposomes/metabolism , Ankyrin Repeat/genetics , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/genetics , Calorimetry , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/genetics , Hemolysin Proteins/antagonists & inhibitors , Hemolysin Proteins/genetics , Hydrogen-Ion Concentration , Mutagenesis, Site-Directed , Permeability , Protein Binding , Synthetic Biology/methods
16.
Fitoterapia ; 139: 104409, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31698059

ABSTRACT

Listeria monocytogenes (Lm) is a widespread foodborne intracellular pathogen that invades a variety of cells, causing abortions and severe human diseases. After internalization into host cells, pore-forming cytolysin listeriolysin O (LLO) disrupts the phagosome, which allows the bacterium to survive and colonize the cytoplasm, providing the bacterium the chance to infect neighboring cells. Betulin is an extracted natural compound from birch bark with diverse pharmacological activities. Here, we showed that LLO-induced rabbit red blood cell lysis in vitro was inhibited by preincubation with betulin, which suppressed the oligomerization process. Infectious assays performed with human monocyte macrophages indicated that betulin significantly protected cells against Lm-induced cell injury. In addition, Balb/c mice were used to perform a general infection, and betulin administration obviously inhibited organ damage and bacterial burden in livers and spleens of infected mice. In conclusion, betulin obviously inhibited Lm-induced cell injury in vitro and protected against infection in vivo through an antivirulence effect. Our results showed betulin as a new candidate against listeriosis by targeting LLO and highlight the potential of natural product-based medicine to be applied in the treatment of pathogenic infections.


Subject(s)
Bacterial Toxins/antagonists & inhibitors , Heat-Shock Proteins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Listeriosis/drug therapy , Triterpenes/pharmacology , Animals , Erythrocytes/drug effects , Female , Hemolysis/drug effects , Humans , Listeria monocytogenes , Liver/microbiology , Liver/pathology , Macrophages/drug effects , Mice, Inbred BALB C , Rabbits , Spleen/microbiology , Spleen/pathology , THP-1 Cells
17.
Microb Pathog ; 137: 103744, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31521800

ABSTRACT

Listeriosis is a serious infection linked to the consumption of food contaminated with Listeria monocytogenes. Outbreaks and mortality rates associated with this infection make it a significant public health concern. As biocontrol agents, probiotics such as Lactobacillus plantarum had been of interest for the promotion of antilisterial activities. However, a recent bacteriocin from epidemic L. monocytogenes strains called listeriolysin S (LLS) has been identified with the ability to target the prokaryotic cells that may hinder the anti-listerial properties of L. plantarum. The present study was designed to investigate the interplay between serotypes 4b (lineage I, LLS-producing strain) and 1/2a (NCTC7973, lineage II, non LLS-producing strain) L. monocytogenes and L. plantarum ATCC13643. According to the results of the co-culture assay, L. plantarum significantly reduced the growth of LLS- L. monocytogenes. However, there was a significant reduction in the growth of L. plantarum when co-cultured with LLS + L. monocytogenes. Moreover, according to the results of the culture assay using Caco-2 cell line, there was a significant reduced intracellular count of LLS- L. monocytogenes after L. plantarum exposure, whereas, no major differences were observed in the intracellular count of LLS + L. monocytogenes. These results suggest that L. plantarum may be unable to inhibit infections caused by LLS-producing L. monocytogenes. Also, phylogenetic studies showed the presence of LLS-like proteins in several environmental isolates including L. innocua which suggests a role for LLS in survival and bacterial colonization in harsh conditions. In overall, the ability of LLS to target certain bacterial cells should be taken into consideration during the development of anti-listerial probiotics. Future experiments are required to elucidate the exact mechanisms by which LLS achieves bacterial killing.


Subject(s)
Hemolysin Proteins/antagonists & inhibitors , Lactobacillus plantarum/metabolism , Listeria monocytogenes/metabolism , Listeria/drug effects , Bacteriocins/metabolism , Caco-2 Cells , Coculture Techniques , Gene Expression Regulation, Bacterial , Hemolysin Proteins/chemistry , Hemolysin Proteins/classification , Hemolysin Proteins/genetics , Humans , Phylogeny , Probiotics , Sequence Alignment , Sequence Analysis, Protein , Virulence Factors/antagonists & inhibitors
18.
Article in English | MEDLINE | ID: mdl-31380296

ABSTRACT

α-Hemolysin (Hla) is a significant virulence factor in Staphylococcus aureus (S. aureus)-caused infectious diseases such as pneumonia. Thus, to prevent the production of Hla when treating S. aureus infection, it is necessary to choose an antibiotic with good antibacterial activity and effect. In our study, we observed that Fosfomycin (FOM) at a sub-inhibitory concentration inhibited expression of Hla. Molecular dynamics demonstrated that FOM bound to the binding sites LYS 154 and ASP 108 of Hla, potentially inhibiting Hla. Furthermore, we verified that staphylococcal membrane-derived vesicles (SMVs) contain Hla and that FOM treatment significantly reduced the production of SMVs and Hla. Based on our pharmacological inhibition analysis, ERK and p38 activated NLRP3 inflammasomes. Moreover, FOM inhibited expression of MAPKs and NLRP3 inflammasome-related proteins in S. aureus as well as SMV-infected human macrophages (MΦ) and alveolar epithelial cells. In vivo, SMVs isolated from S. aureus DU1090 (an isogenic Hla deletion mutant) or the strain itself caused weaker inflammation than that of its parent strain 8325-4. FOM also significantly reduced the phosphorylation levels of ERK and P38 and expression of NLRP3 inflammasome-related proteins. In addition, FOM decreased MPO activity, pulmonary vascular permeability and edema formation in the lungs of mice with S. aureus-caused pneumonia. Taken together, these data indicate that FOM exerts protective effects against S. aureus infection in vitro and in vivo by inhibiting Hla in SMVs and blocking ERK/P38-mediated NLRP3 inflammasome activation by Hla.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Fosfomycin/pharmacology , Hemolysin Proteins/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pneumonia, Staphylococcal/drug therapy , Staphylococcus aureus/drug effects , Virulence Factors/antagonists & inhibitors , Animals , Anti-Bacterial Agents/chemistry , Bacterial Toxins/chemistry , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Binding Sites , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/microbiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Vesicles , Fosfomycin/chemistry , Gene Expression Regulation , Hemolysin Proteins/chemistry , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Host-Pathogen Interactions/genetics , Humans , Inflammasomes/drug effects , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/pathology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development , Staphylococcus aureus/pathogenicity , THP-1 Cells , Virulence Factors/chemistry , Virulence Factors/genetics , Virulence Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Infect Immun ; 87(10)2019 10.
Article in English | MEDLINE | ID: mdl-31383747

ABSTRACT

Postinfluenza methicillin-resistant Staphylococcus aureus (MRSA) infection can quickly develop into severe, necrotizing pneumonia, causing over 50% mortality despite antibiotic treatments. In this study, we investigated the efficacy of antibiotic therapies and the impact of S. aureus alpha-toxin in a model of lethal influenza virus and MRSA coinfection. We demonstrate that antibiotics primarily attenuate alpha-toxin-induced acute lethality, even though both alpha-toxin-dependent and -independent mechanisms significantly contribute to animal mortality after coinfection. Furthermore, we found that the protein synthesis-suppressing antibiotic linezolid has an advantageous therapeutic effect on alpha-toxin-induced lung damage, as measured by protein leak and lactate dehydrogenase (LDH) activity. Importantly, using a Panton-Valentine leucocidin (PVL)-negative MRSA isolate from patient sputum, we show that linezolid therapy significantly improves animal survival from postinfluenza MRSA pneumonia compared with vancomycin treatment. Rather than improved viral or bacterial control, this advantageous therapeutic effect is associated with a significantly attenuated proinflammatory cytokine response and acute lung damage in linezolid-treated mice. Together, our findings not only establish a critical role of alpha-toxin in the extreme mortality of secondary MRSA pneumonia after influenza but also provide support for the possibility that linezolid could be a more effective treatment than vancomycin to improve disease outcomes.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Toxins/antagonists & inhibitors , Hemolysin Proteins/antagonists & inhibitors , Linezolid/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Orthomyxoviridae Infections/complications , Pneumonia, Staphylococcal/drug therapy , Animals , Bacterial Toxins/genetics , Bacterial Toxins/metabolism , Female , Gene Expression , Gentamicins/pharmacology , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , L-Lactate Dehydrogenase/metabolism , Lung/microbiology , Lung/pathology , Male , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Mice , Mice, Inbred C57BL , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Plasmids/chemistry , Plasmids/metabolism , Pneumonia, Staphylococcal/complications , Pneumonia, Staphylococcal/microbiology , Pneumonia, Staphylococcal/mortality , Survival Analysis , Vancomycin/pharmacology
20.
Carbohydr Polym ; 218: 136-144, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31221314

ABSTRACT

Aqueous and KCl-soluble polysaccharides were extracted from Laurencia dendroidea (Rhodomelaceae, Ceramiales) and their chemical profile was accessed by anion-exchange chromatography, chemical and spectroscopic analyses. The homogeneous agaran DHS-4 (181.3 × 103 g. mol-1, 21.3% of NaSO3) presents A units mostly 2-sulfated (18.9 mol%), nonsubstituted (15.3 mol%) and 6-O-methylated (10.1 mol%), while B units are l-sugars composed predominantly by galactose 6-sulfate precursor units (19.2 mol%) and 3,6-anhydrogalactose (13.8 mol%), besides non-precursor galactose 6-sulfate units bearing d-xylose substituents on C-3 (8.1 mol%). The crude KCl-soluble DHS agaran (20.5% of NaSO3) inhibited proteolysis and hemolysis induced by Lachesis muta and Bothrops jararaca venoms. DHS was able to inhibit up to 75% the L. muta venom hemorrhagic effect and to reduce the lethality displayed by B. jararaca venom, increasing the mice survival time up to 3 times. Therefore, this agaran has high potential to be used as an additional tool to treat snakebite envenomation.


Subject(s)
Hemolysin Proteins/antagonists & inhibitors , Hemostatics/therapeutic use , Laurencia/chemistry , Polysaccharides/therapeutic use , Snake Venoms/antagonists & inhibitors , Sulfuric Acid Esters/therapeutic use , Animals , Bothrops , Hemolysis/drug effects , Hemostatics/chemistry , Hemostatics/isolation & purification , Mice , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Proteolysis/drug effects , Sulfuric Acid Esters/chemistry , Sulfuric Acid Esters/isolation & purification , Viperidae
SELECTION OF CITATIONS
SEARCH DETAIL
...