Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Microb Pathog ; 48(2): 74-84, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19900530

ABSTRACT

It was recently shown that, as in yeast, alcohols selectively increase the hemolytic properties of certain staphylococci strains. This phenomenon has been called 'microbial alcohol-conferred hemolysis'(MACH). Here we present the changes in gene expression by Staphylococcus aureus 8325-4, in response to ethanol. Ethanol upregulated the expression of multiple toxins and increase the pathogen potential of S. aureus strain 8325-4. Ethanol also increased the level of genes considered necessary for production and viability of biofilm, such as: icaAD, sdrDE, pyr, and ure. Increased urease activity appeared to be an important factor in the ethanol response along with macromolecule repair mechanisms. Oxidative-stress responses, such as increased expression of sodA1, sodA2 and upregulation of zinc-containing alcohol dehydrogenase, alcohol-acetaldehyde dehydrogenase (adhE) and two aldehyde dehydrogenases (aldA1, aldA2), which can generate more reducing power, were also induced. Upregulation of fatty acid metabolism appears to be important in enabling the bacteria to handle excess amounts of ethanol which ultimately may lead to synthesis of lytic lypids. The patterns of regulation were confirmed by quantitive reverse transcriptase PCR (QRT-PCR). These results, taken together, suggest that exposure to ethanol increases pathogenic traits and induce oxidative-stress responses.


Subject(s)
Ethanol/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Acetaldehyde/metabolism , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Amino Acids/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/drug effects , Biofilms/growth & development , Cell Line , Ethanol/metabolism , Gene Expression Profiling , Hemolysin Proteins/genetics , Hemolysin Proteins/metabolism , Hemolysis , Humans , Hydrogen-Ion Concentration , Keratinocytes/cytology , Keratinocytes/physiology , Oligonucleotide Array Sequence Analysis , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/genetics , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Up-Regulation/drug effects , Urease/genetics , Urease/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
2.
FEMS Microbiol Lett ; 269(1): 153-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17227454

ABSTRACT

It was recently found that alcohols can confer hemolytic properties on certain species of yeast. Here, it is reported that alcohol can promote hemolysis by various species of staphylococci, including strains of Staphylococcus aureus, Staphylococcus epidermidis and Staphylococcus hominis. In order to study this novel phenomenon in S. aureus and S. epidermidis, strains that exhibit this phenomenon (e.g. S. aureus 8325-4, COL, SH1000, S. epidermidis), as compared with strains that exhibited little alcohol-enhanced hemolysis (e.g. S. aureus 8325-4 DeltaTRAP, RN6911) were examined. Both ethanol and n-butanol caused upregulation of the virulence regulator-RNAIII, with a concomitant increase in the production of alpha, beta and gamma-hemolysins in strain 8325-4. In S. aureus COL and SH1000, there was an increase in RNAIII but no change in transcription levels of alpha, beta and gamma hemolysins. Staphylococcus epidermidis stain sofi exhibited increased RNAIII and beta hemolysin production. Staphylococcus aureus mutant strains (8325-4 DeltaTRAP and RN6911) showed no change in the transcription level of the RNAIII regulator and the above hemolysins. Increased hemolysis in S. aureus COL, SH1000 and mutant strains may be caused by other hemolysins (not regulated by RNAIII) or through other mechanisms such as hyperoxidation or cytotoxic lipids.


Subject(s)
1-Butanol/pharmacology , Ethanol/pharmacology , Hemolysis/drug effects , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Hemolysin Proteins/biosynthesis , Hemolysin Proteins/genetics , Hydrogen Peroxide/pharmacology , RNA, Messenger/metabolism , Staphylococcus aureus/pathogenicity , Staphylococcus epidermidis/pathogenicity , Up-Regulation , Virulence Factors/biosynthesis , Virulence Factors/genetics
3.
Infect Immun ; 73(10): 6220-8, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16177293

ABSTRACT

Staphylococcus aureus is a gram-positive bacterium that is part of the normal healthy flora but that can become virulent and cause infections by producing biofilms and toxins. The production of virulence factors is regulated by cell-cell communication (quorum sensing) through the histidine phosphorylation of target of RNAIII-activating protein (TRAP), which is a 21-kDa protein that is highly conserved among staphylococci. Using microarray analysis, we show here that the expression and phosphorylation of TRAP upregulate the expression of most, if not all, toxins known to date, as well as their global regulator agr. In addition, we show here that the expression and phosphorylation of TRAP are also necessary for the expression of genes known to be necessary for the survival of the bacteria in a biofilm, like arc, pyr, and ure. TRAP is thus demonstrated to be a master regulator of staphylococcal pathogenesis.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Phosphoproteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Adaptor Proteins, Signal Transducing , Bacterial Proteins/genetics , Bacterial Proteins/physiology , Biofilms/growth & development , Gene Expression Profiling , Monomeric GTP-Binding Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Phosphoproteins/physiology , Phosphorylation , RNA Polymerase III/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Virulence/genetics
4.
Antimicrob Agents Chemother ; 48(7): 2544-50, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15215107

ABSTRACT

Staphylococcal bacteria are a prevalent cause of infections associated with foreign bodies and indwelling medical devices. Bacteria are capable of escaping antibiotic treatment through encapsulation into biofilms. RNA III-inhibiting peptide (RIP) is a heptapeptide that inhibits staphylococcal biofilm formation by obstructing quorum-sensing mechanisms. K(4)-S4(1-13)(a) is a 13-residue dermaseptin derivative (DD(13)) believed to kill bacteria via membrane disruption. We tested each of these peptides as well as a hybrid construct, DD(13)-RIP, for their ability to inhibit bacterial proliferation and suppress quorum sensing in vitro and for their efficacy in preventing staphylococcal infection in a rat graft infection model with methicillin-resistant Staphylococcus aureus (MRSA) or S. epidermidis (MRSE). In vitro, proliferation assays demonstrated that RIP had no inhibitory effect, while DD(13)-RIP and DD(13) were equally effective, and that the chimeric peptide but not DD(13) was slightly more effective than RIP in inhibiting RNA III synthesis, a regulatory RNA molecule important for staphylococcal pathogenesis. In vivo, the three peptides reduced graft-associated bacterial load in a dose-dependent manner, but the hybrid peptide was most potent in totally preventing staphylococcal infections at the lowest dose. In addition, each of the peptides acted synergistically with antibiotics. The data indicate that RIP and DD(13) act in synergy by attacking bacteria simultaneously by two different mechanisms. Such a chimeric peptide may be useful for coating medical devices to prevent drug-resistant staphylococcal infections.


Subject(s)
Amphibian Proteins/pharmacology , Anti-Infective Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Oligopeptides/antagonists & inhibitors , Peptides/pharmacology , Recombinant Fusion Proteins/pharmacology , Staphylococcal Infections/prevention & control , Transplants/adverse effects , Animals , Circular Dichroism , Drug Resistance, Bacterial , Male , Microbial Sensitivity Tests , Oligopeptides/biosynthesis , Polyethylene Terephthalates , Rats , Rats, Wistar , Spectrophotometry, Ultraviolet , Staphylococcal Infections/microbiology
5.
J Infect Dis ; 190(2): 318-20, 2004 Jul 15.
Article in English | MEDLINE | ID: mdl-15216467

ABSTRACT

Staphylococcus aureus and S. epidermidis are major causes of infection related to biofilm formed on indwelling medical devices. Such infections are common causes of morbidity and mortality and, because of biofilm resistance to antibiotics, are difficult to treat. The RNAIII-inhibiting peptide (RIP) (YSPWTNF-NH2) inhibits the pathogenesis of staphylococci by disrupting bacterial cell-cell communication (known as "quorum sensing"). Using a vascular-graft rat model, we show that RIP, applied locally and systemically, can completely inhibit drug-resistant S. aureus and S. epidermidis biofilms. The present study provides the first direct demonstration that interfering with cell-cell communication by use of a quorum-sensing inhibitor can eliminate medical device-associated staphylococcal infections. We suggest that medical devices could be coated with RIP to prevent infections, including those by antibiotic-resistant staphylococcal strains.


Subject(s)
Oligopeptides/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcal Infections/prevention & control , Staphylococcus aureus/drug effects , Staphylococcus epidermidis/drug effects , Animals , Biofilms/drug effects , Biofilms/growth & development , Disease Models, Animal , Drug Resistance, Bacterial , Equipment and Supplies/microbiology , Oligopeptides/administration & dosage , Rats , Staphylococcal Infections/microbiology
6.
J Biol Chem ; 279(15): 14665-72, 2004 Apr 09.
Article in English | MEDLINE | ID: mdl-14726534

ABSTRACT

Staphylococcus aureus cause infections by producing toxins, a process regulated by cell-cell communication (quorum sensing) through the histidine-phosphorylation of the target of RNAIII-activating protein (TRAP). We show here that TRAP is highly conserved in staphylococci and contains three completely conserved histidine residues (His-66, His-79, His-154) that are phosphorylated and essential for its activity. This was tested by constructing a TRAP(-) strain with each of the conserved histidine residues changed to alanine by site-directed mutagenesis. All mutants were tested for pathogenesis in vitro (expression of RNAIII and hemolytic activity) and in vivo (murine cellulitis model). Results show that RNAIII is not expressed in the TRAP(-) strain, that it is non hemolytic, and that it does not cause disease in vivo. These pathogenic phenotypes could be rescued in the strain containing the recovered traP, confirming the importance of TRAP in S. aureus pathogenesis. The phosphorylation of TRAP mutated in any of the conserved histidine residues was significantly reduced, and mutants defective in any one of these residues were non-pathogenic in vitro or in vivo, whereas those mutated in a non-conserved histidine residue (His-124) were as pathogenic as the wild type. These results confirm the importance of the three conserved histidine residues in TRAP activity. The phosphorylation pattern, structure, and gene organization of TRAP deviates from signaling molecules known to date, suggesting that TRAP belongs to a novel class of signal transducers.


Subject(s)
Histidine/chemistry , Staphylococcus aureus/metabolism , Amino Acid Sequence , Animals , Blotting, Northern , Cell Communication , DNA/metabolism , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Mice , Molecular Sequence Data , Mutagenesis , Mutagenesis, Site-Directed , Mutation , Phenotype , Phosphorylation , Phylogeny , Plasmids/metabolism , Protein Structure, Tertiary , RNA, Antisense/chemistry , RNA, Bacterial/chemistry , Sequence Homology, Amino Acid , Signal Transduction , Time Factors
7.
Circulation ; 108(6): 767-71, 2003 Aug 12.
Article in English | MEDLINE | ID: mdl-12885754

ABSTRACT

BACKGROUND: Bacteria that adhere to implanted medical devices play an important role in industry and in modern medicine. Staphylococci are among the most common pathogens that cause biomaterial infections. Vascular prosthetic graft infection is one of the most feared complications that the vascular surgeon treats, frequently resulting in prolonged hospitalization, organ failure, amputation, and death. A rat model was used to investigate the topical efficacies of temporin A and the quorum-sensing inhibitor RNAIII-inhibiting protein (RIP) as prophylactic agents of vascular prosthetic graft infections caused by Staphylococcus aureus and Staphylococcus epidermidis with intermediate resistance to glycopeptides. METHODS AND RESULTS: Graft infections were established in the back subcutaneous tissue of adult male Wistar rats by implantation of Dacron prostheses 1 cm2 followed by topical inoculation with 2x10(7) colony-forming units of bacterial strains. The study included, for each staphylococcal strain, a control group (no graft contamination), a contaminated group that did not receive antibiotic prophylaxis, and 6 contaminated groups that received grafts soaked with temporin A, RIP, rifampin, temporin A plus RIP, RIP plus rifampin, or temporin A plus RIP. The infection was evaluated by quantitative agar culture. When tested alone, temporin A and RIP showed comparable efficacies, and their efficacies were significantly higher than that of rifampin against both strains. All combinations showed efficacies significantly higher than that of each single compound. The combinations of temporin A and RIP exerted the strongest antistaphylococcal efficacies, eliminating infection by 100%. CONCLUSIONS: The results of the present study make these molecules potentially useful for antimicrobial chemoprophylaxis in vascular surgery.


Subject(s)
Implants, Experimental/adverse effects , Oligopeptides/administration & dosage , Proteins/administration & dosage , Staphylococcal Skin Infections/drug therapy , Subcutaneous Tissue/pathology , Administration, Topical , Animals , Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Resistance, Microbial , Drug Therapy, Combination/administration & dosage , Glycopeptides/pharmacology , Implants, Experimental/microbiology , Male , Microbial Sensitivity Tests , Oligopeptides/chemistry , Polyethylene Terephthalates/chemistry , Proteins/chemistry , Rats , Rats, Wistar , Rifampin/pharmacology , Staphylococcal Skin Infections/microbiology , Staphylococcal Skin Infections/pathology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/pathogenicity , Subcutaneous Tissue/microbiology , Treatment Outcome , Vancomycin/pharmacology , Vancomycin Resistance
8.
FEMS Microbiol Lett ; 223(2): 167-75, 2003 Jun 27.
Article in English | MEDLINE | ID: mdl-12829282

ABSTRACT

Staphylococcus aureus are Gram-positive bacteria and cause diverse serious diseases in humans and animals through the production of toxins. The production of toxins is regulated by quorum sensing mechanisms, where proteins such as RNAIII activating protein (RAP) are secreted by the bacteria and induce virulence. Antibodies to RAP have been shown to protect mice from infection, but the molecular structure of RAP was not known and hindered vaccine development. To characterize RAP, recombinant protein was made and tested for its ability to induce genes important for pathogenesis (agr). In addition, monoclonal antibodies were produced to identify its cellular localization. Results shown here indicate that RAP is a 277-aa protein that is an ortholog of the ribosomal protein L2. Like the native molecule, recombinant RAP activates the production of RNAIII (encoded by agr). Using RAP specific monoclonal antibodies we demonstrate that RAP is continuously secreted and while RAP is expressed also in other bacteria (like Staphylococcus epidermidis, Staphylococcus xylosus and Escherichia coli), it is secreted to the culture medium only by S. aureus. Our results show that the ribosomal protein L2 has an extraribosomal function and that when secreted RAP acts as an autoinducer of virulence to regulate S. aureus pathogenesis.


Subject(s)
Bacterial Proteins , Carrier Proteins/genetics , Staphylococcal Infections/microbiology , Staphylococcus aureus/genetics , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Carrier Proteins/immunology , Carrier Proteins/metabolism , Female , Gene Expression Regulation, Bacterial , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Phosphoproteins/metabolism , RNA, Antisense/metabolism , RNA, Bacterial/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism , Staphylococcus aureus/metabolism , Staphylococcus aureus/pathogenicity , Virulence
9.
Antimicrob Agents Chemother ; 47(6): 1979-83, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12760879

ABSTRACT

Staphylococcus aureus is a prevalent cause of bacterial infections associated with indwelling medical devices. RNA III inhibiting peptide (RIP) is known to inhibit S. aureus pathogenesis by disrupting quorum-sensing mechanisms. RIP was tested in the present study for its ability to inhibit S. aureus biofilm formation in a rat Dacron graft model. The activity of RIP was synergistic with those of antibiotics for the complete prevention of drug-resistant S. aureus infections.


Subject(s)
Biofilms/growth & development , Oligopeptides/pharmacology , Prostheses and Implants/microbiology , Staphylococcal Infections/drug therapy , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Animals , Anti-Bacterial Agents/pharmacology , Antibiotic Prophylaxis , Biofilms/drug effects , Cell Communication/drug effects , Colony Count, Microbial , Drug Synergism , Male , Microbial Sensitivity Tests , Polyethylene Terephthalates , Rats , Rats, Wistar
10.
J Infect Dis ; 187(4): 625-30, 2003 Feb 15.
Article in English | MEDLINE | ID: mdl-12599079

ABSTRACT

Staphylococcus epidermidis is a frequent cause of infections associated with foreign bodies and indwelling medical devices. The bacteria are capable of surviving antibiotic treatment through encapsulation into biofilms. RNAIII-inhibiting peptide (RIP) is a heptapeptide that inhibits S. aureus pathogenesis by disrupting quorum-sensing mechanisms. In this study, RIP inhibited drug-resistant S. epidermidis biofilm formation through a mechanism similar to that evidenced for S. aureus. RIP is synergistic with antibiotics in eliminating 100% of graft-associated in vivo S. epidermidis infections, which suggests that RIP may be used to coat medical devices to prevent staphylococcal infections. Disruption of cell-cell communication can prevent infections associated with antibiotic-resistant strains.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins , Oligopeptides/pharmacology , Staphylococcus epidermidis/drug effects , Adaptor Proteins, Signal Transducing , Animals , Bacterial Adhesion , Biofilms/growth & development , Cell Line , Drug Resistance , Humans , Male , Phosphoproteins/metabolism , Phosphorylation/drug effects , Rats , Staphylococcal Infections/drug therapy , Staphylococcus epidermidis/metabolism
11.
Kidney Int ; 63(1): 340-5, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12472801

ABSTRACT

BACKGROUND: Dialysis patients, often carriers of Staphylococcus aureus in their nares, are at high risk of S. aureus infections. METHODS: We examined whether RNAIII inhibiting peptide (RIP), which interferes with quorum sensing mechanisms, reduces adherence of S. aureus to host cells and to dialysis catheter polymers in vitro. Adherence was tested by spectroscopy using safranin staining, by confocal scanning laser microscopy and by atomic force microscopy. RESULTS: RIP inhibited bacterial adherence to HaCat and HEp-2 cells and reduced adherence and biofilm formation not only on polystyrene, but also on both polyurethane- and silicone-made dialysis catheters, with a preponderant effect on silicone, to which bacteria were more adherent. CONCLUSION: RIP opens a new perspective in anti-S. aureus prophylaxis, particularly in dialysis patients.


Subject(s)
Biofilms/growth & development , Catheters, Indwelling/microbiology , Oligopeptides/pharmacology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/growth & development , Bacterial Adhesion/drug effects , Cell Line, Tumor , Humans , In Vitro Techniques , Keratinocytes/microbiology , Kidney Failure, Chronic/microbiology , Kidney Failure, Chronic/therapy , Nose/cytology , Renal Dialysis , Skin/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...