Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
J Bone Jt Infect ; 6(7): 241-253, 2021.
Article in English | MEDLINE | ID: mdl-34262845

ABSTRACT

The high antibiotic tolerance of Staphylococcus aureus biofilms is associated with challenges for treating periprosthetic joint infection. The toxin-antitoxin system, YefM-YoeB, is thought to be a regulator for antibiotic tolerance, but its physiological role is unknown. The objective of this study was to determine the biofilm and antibiotic susceptibility phenotypes associated with S. aureus yoeB homologs. We hypothesized the toxin-antitoxin yoeB homologs contribute to biofilm formation and antibiotic susceptibility. Disruption of yoeB1 and yoeB2 resulted in decreased biofilm formation in comparison to Newman and JE2 wild-type (WT) S. aureus strains. In comparison to yoeB mutants, both Newman and JE2 WT strains had higher polysaccharide intercellular adhesin (PIA) production. Treatment with sodium metaperiodate increased biofilm formation in Newman WT, indicating biofilm formation may be increased under conditions of oxidative stress. DNase I treatment decreased biofilm formation in Newman WT but not in the absence of yoeB1 or yoeB2. Additionally, WT strains had a higher extracellular DNA (eDNA) content in comparison to yoeB mutants but no differences in biofilm protein content. Moreover, loss of yoeB1 and yoeB2 decreased biofilm survival in both Newman and JE2 strains. Finally, in a neutropenic mouse abscess model, deletion of yoeB1 and yoeB2 resulted in reduced bacterial burden. In conclusion, our data suggest that yoeB1 and yoeB2 are associated with S. aureus planktonic growth, extracellular dependent biofilm formation, antibiotic tolerance, and virulence.

2.
Infect Immun ; 89(10): e0034721, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34227840

ABSTRACT

GraS is a membrane sensor in Staphylococcus aureus that induces mprF and dltABCD expression to alter the surface positive charge upon exposure to cationic human defense peptides (HDPs). The sensing domain of GraS likely resides in the 9-residue extracellular loop (EL). In this study, we assessed a hospital-acquired methicillin-resistant S. aureus (HA-MRSA) strain (COL) for the specific role of two distinct EL mutations: F38G (bulk) and D/35/37/41K (charged inversion). Activation of mprF by polymyxin B (PMB) was reduced in the D35/37/41K mutant versus the D35/37/41G mutant, correlating with reduced surface positive charge; in contrast, these effects were less prominent in the F38G mutant but still lower than those in the parent. These data indicated that both electrostatic charge and steric bulk of the EL of GraS influence induction of genes impacting HDP resistance. Using mprF expression as a readout, we confirmed GraS signaling was pH dependent, increasing as pH was lowered (from pH 7.5 down to pH 5.5). In contrast to PMB activation, reduction of mprF was comparable at pH 5.5 between the P38G and D35/37/41K point mutants, indicating a mechanistic divergence between GraS activation by acidic pH versus cationic peptides. Survival assays in human blood and purified polymorphonuclear leukocytes (PMNs) revealed lower survival of the D35/37/41K mutant versus the F38G mutant, with both being lower than that of the parent. Virulence studies in the rabbit endocarditis model mirrored whole blood and PMN killing assay data described above. Collectively, these data confirmed the importance of specific residues within the EL of GraS in conferring essential bacterial responses for MRSA survival in infections.


Subject(s)
Bacterial Proteins/genetics , Cardiovascular Infections/metabolism , Cardiovascular Infections/microbiology , Drug Resistance, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Neutrophils/metabolism , Staphylococcal Infections/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Endocarditis/metabolism , Endocarditis/microbiology , Female , Gene Expression Regulation, Bacterial/genetics , Humans , Microbial Sensitivity Tests/methods , Microbial Viability/genetics , Neutrophils/microbiology , Rabbits , Staphylococcal Infections/microbiology
3.
J Infect Dis ; 222(7): 1188-1198, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32333768

ABSTRACT

Persistent methicillin-resistant Staphylococcus aureus (MRSA) endovascular infections represent a significant clinical-therapeutic challenge. Of particular concern is antibiotic treatment failure in infections caused by MRSA that are "susceptible" to antibiotic in vitro. In the current study, we investigate specific purine biosynthetic pathways and stringent response mechanism(s) related to this life-threatening syndrome using genetic matched persistent and resolving MRSA clinical bacteremia isolates (PB and RB, respectively), and isogenic MRSA strain sets. We demonstrate that PB isolates (vs RB isolates) have significantly higher (p)ppGpp production, phenol-soluble-modulin expression, polymorphonuclear leukocyte lysis and survival, fibronectin/endothelial cell (EC) adherence, and EC damage. Importantly, an isogenic strain set, including JE2 parental, relP-mutant and relP-complemented strains, translated the above findings into significant outcome differences in an experimental endocarditis model. These observations indicate a significant regulation of purine biosynthesis on stringent response, and suggest the existence of a previously unknown adaptive genetic mechanism in persistent MRSA infection.


Subject(s)
Endocarditis/microbiology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Purines/biosynthesis , Staphylococcal Infections/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Bacteremia/metabolism , Bacteremia/microbiology , Biosynthetic Pathways , Disease Models, Animal , Endocarditis/metabolism , Humans , Methicillin/pharmacology , Rabbits
4.
Elife ; 82019 11 12.
Article in English | MEDLINE | ID: mdl-31713513

ABSTRACT

Microbes often live in multispecies communities where interactions among community members impact both the individual constituents and the surrounding environment. Here, we developed a system to visualize interspecies behaviors at initial encounters. By imaging two prevalent pathogens known to be coisolated from chronic illnesses, Pseudomonas aeruginosa and Staphylococcus aureus, we observed P. aeruginosa can modify surface motility in response to secreted factors from S. aureus. Upon sensing S. aureus, P. aeruginosa transitioned from collective to single-cell motility with an associated increase in speed and directedness - a behavior we refer to as 'exploratory motility'. Explorer cells moved preferentially towards S. aureus and invaded S. aureus colonies through the action of the type IV pili. These studies reveal previously undescribed motility behaviors and lend insight into how P. aeruginosa senses and responds to other species. Identifying strategies to harness these interactions may open avenues for new antimicrobial strategies.


Subject(s)
Fimbriae, Bacterial/physiology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Bacterial Physiological Phenomena , Cystic Fibrosis/microbiology , Humans , Microbial Interactions , Movement/physiology , Species Specificity , Staphylococcus aureus/metabolism
5.
mBio ; 10(6)2019 11 26.
Article in English | MEDLINE | ID: mdl-31772059

ABSTRACT

Staphylococcus aureus is the major organism responsible for surgical implant infections. Antimicrobial treatment of these infections often fails, leading to expensive surgical intervention and increased risk of mortality to the patient. The challenge in treating these infections is associated with the high tolerance of S. aureus biofilm to antibiotics. MazEF, a toxin-antitoxin system, is thought to be an important regulator of this phenotype, but its physiological function in S. aureus is controversial. Here, we examined the role of MazEF in developing chronic infections by comparing growth and antibiotic tolerance phenotypes in three S. aureus strains to their corresponding strains with disruption of mazF expression. Strains lacking mazF production showed increased biofilm growth and decreased biofilm antibiotic tolerance. Deletion of icaADBC in the mazF::Tn background suppressed the growth phenotype observed with mazF-disrupted strains, suggesting the phenotype was ica dependent. We confirmed these phenotypes in our murine animal model. Loss of mazF resulted in increased bacterial burden and decreased survival rate of mice compared to its wild-type strain demonstrating that loss of the mazF gene caused an increase in S. aureus virulence. Although lack of mazF gene expression increased S. aureus virulence, it was more susceptible to antibiotics in vivo Combined, the ability of mazF to inhibit biofilm formation and promote biofilm antibiotic tolerance plays a critical role in transitioning from an acute to chronic infection that is difficult to eradicate with antibiotics alone.IMPORTANCE Surgical infections are one of the most common types of infections encountered in a hospital. Staphylococcus aureus is the most common pathogen associated with this infection. These infections are resilient and difficult to eradicate, as the bacteria form biofilm, a community of bacteria held together by an extracellular matrix. Compared to bacteria that are planktonic, bacteria in a biofilm are more resistant to antibiotics. The mechanism behind how bacteria develop this resistance and establish a chronic infection is unknown. We demonstrate that mazEF, a toxin-antitoxin gene, inhibits biofilm formation and promotes biofilm antibiotic tolerance which allows S. aureus to transition from an acute to chronic infection that cannot be eradicated with antibiotics but is less virulent. This gene not only makes the bacteria more tolerant to antibiotics but makes the bacteria more tolerant to the host.


Subject(s)
Antitoxins/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Biofilms , Drug Resistance, Bacterial , Staphylococcal Infections/microbiology , Staphylococcus aureus/physiology , Animals , Anti-Bacterial Agents/pharmacology , Antitoxins/genetics , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Biofilms/drug effects , Chronic Disease , Female , Humans , Male , Mice , Mice, Inbred C57BL , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Toxin-Antitoxin Systems
6.
Mol Microbiol ; 112(2): 532-551, 2019 08.
Article in English | MEDLINE | ID: mdl-31074903

ABSTRACT

Staphyloxanthin, a carotenoid in S. aureus, is a powerful antioxidant against oxidative stresses. The crtOPQMN operon driving pigment synthesis is under the control of σB . CspA, a cold shock protein, is known to control σB activity. To ascertain genes that regulate cspA, we screened a transposon library that exhibited reduced cspA expression and pigmentation. We found that the adaptor protein YjbH activates cspA expression. Spx, the redox-sensitive transcriptional regulator and a proteolytic target for YjbH and ClpXP, complexes with αCTD of RNAP prior to binding the cspA promoter to repress cspA activity. Increased cspA expression in trans in the inactive spx C10A mutant of JE2 did not enhance pigment production while it did in JE2, suggesting that cspA is downstream to Spx in pigmentation control. As the staphyloxanthin pigment is critical to S. aureus survival in human hosts, we demonstrated that the cspA and yjbH mutants survived less well than the parent in whole blood killing assay. Collectively, our studies suggest a pathway wherein YjbH and ClpXP proteolytically cleave Spx, a repressor of cspA transcription, to affect σB -dependent carotenoid expression, thus providing a critical link between intracellular redox sensing by Spx and carotenoid production to improve S. aureus survival during infections.


Subject(s)
Bacterial Proteins/metabolism , Carotenoids/metabolism , Cold Shock Proteins and Peptides/metabolism , Gene Expression Regulation, Bacterial , Sigma Factor/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Cold Shock Proteins and Peptides/genetics , Endopeptidase Clp/genetics , Endopeptidase Clp/metabolism , Operon , Oxidation-Reduction , Proteolysis , Sigma Factor/genetics , Staphylococcus aureus/genetics
7.
J Infect Dis ; 218(9): 1367-1377, 2018 09 22.
Article in English | MEDLINE | ID: mdl-29868791

ABSTRACT

Persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia (PB) represents an important subset of S. aureus endovascular infections. In this study, we investigated potential genetic mechanisms underlying the persistent outcomes. Compared with resolving bacteremia (RB) isolates (defined as isolates associated with negative results of blood cultures 2-4 days after initiation of therapy), PB strains (defined as isolates associated with positive results of blood cultures ≥7 days after initiation of therapy) had significantly earlier onset activation of key virulence regulons and structural genes (eg, sigB, sarA, sae, and cap5), higher expression of purine biosynthesis genes (eg, purF), and faster growth rates, with earlier entrance into stationary phase. Importantly, an isogenic strain set featuring a wild-type MRSA isolate, a purF mutant strain, and a purF-complemented strain and use of strategic purine biosynthesis inhibitors implicated a causal relationship between purine biosynthesis and the in vivo persistent outcomes. These observations suggest that purine biosynthesis plays a key role in the outcome of PB and may represent a new target for enhanced efficacy in treating life-threatening MRSA infections.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/pathogenicity , Purines/biosynthesis , Staphylococcal Infections/metabolism , Animals , Anti-Bacterial Agents/therapeutic use , Bacteremia/metabolism , Bacteremia/microbiology , Disease Models, Animal , Humans , Methicillin/pharmacology , Rabbits
8.
J Bacteriol ; 199(16)2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28559294

ABSTRACT

Staphylococcus epidermidis is the leading cause of infections on indwelling medical devices worldwide. Intrinsic antibiotic resistance and vigorous biofilm production have rendered these infections difficult to treat and, in some cases, require the removal of the offending medical prosthesis. With the exception of two widely passaged isolates, RP62A and 1457, the pathogenesis of infections caused by clinical S. epidermidis strains is poorly understood due to the strong genetic barrier that precludes the efficient transformation of foreign DNA into clinical isolates. The difficulty in transforming clinical S. epidermidis isolates is primarily due to the type I and IV restriction-modification systems, which act as genetic barriers. Here, we show that efficient plasmid transformation of clinical S. epidermidis isolates from clonal complexes 2, 10, and 89 can be realized by employing a plasmid artificial modification (PAM) in Escherichia coli DC10B containing a Δdcm mutation. This transformative technique should facilitate our ability to genetically modify clinical isolates of S. epidermidis and hence improve our understanding of their pathogenesis in human infections.IMPORTANCE Staphylococcus epidermidis is a source of considerable morbidity worldwide. The underlying mechanisms contributing to the commensal and pathogenic lifestyles of S. epidermidis are poorly understood. Genetic manipulations of clinically relevant strains of S. epidermidis are largely prohibited due to the presence of a strong restriction barrier. With the introductions of the tools presented here, genetic manipulation of clinically relevant S. epidermidis isolates has now become possible, thus improving our understanding of S. epidermidis as a pathogen.

9.
Nat Microbiol ; 1: 16051, 2016 Apr 18.
Article in English | MEDLINE | ID: mdl-27572649

ABSTRACT

Persisters are dormant phenotypic variants of bacterial cells that are tolerant to killing by antibiotics(1). Persisters are associated with chronic infections and antibiotic treatment failure(1-3). In Escherichia coli, toxin-antitoxin modules have been linked to persister formation(4-6). The mechanism of persister formation in Gram-positive bacteria is unknown. Staphylococcus aureus is a major human pathogen, responsible for a variety of chronic and relapsing infections such as osteomyelitis, endocarditis and infections of implanted devices. Deleting toxin-antitoxin modules in S. aureus did not affect the level of persisters. Here, we show that S. aureus persisters are produced due to a stochastic entrance into the stationary phase accompanied by a drop in intracellular adenosine triphosphate. Cells expressing stationary-state markers are present throughout the growth phase, and increase in frequency with cell density. Cell sorting revealed that the expression of stationary markers is associated with a 100-1,000-fold increase in the likelihood of survival to antibiotic challenge. The adenosine triphosphate level of the cell is predictive of bactericidal antibiotic efficacy and explains bacterial tolerance to antibiotics.


Subject(s)
Adenosine Triphosphate/metabolism , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Anti-Bacterial Agents/pharmacology , Drug Tolerance , Staphylococcus aureus/drug effects
10.
Article in English | MEDLINE | ID: mdl-27398229

ABSTRACT

Persisters are dormant phenotypic variants of bacterial cells that are tolerant to killing by antibiotics1. Persisters are associated with chronic infections and antibiotic treatment failure1-3. In Escherichia coli, toxin/antitoxin (TA) modules have been linked to persister formation4-6. The mechanism of persister formation in Gram-positive bacteria is unknown. Staphylococcus aureus is a major human pathogen, responsible for a variety of chronic and relapsing infections such as osteomyelitis, endocarditis and infections of implanted devices. Deleting TA modules in S. aureus did not affect the level of persisters. Here we show that S. aureus persisters are produced due to a stochastic entrance into stationary phase accompanied by a drop in intracellular ATP. Cells expressing stationary state markers are present throughout the growth phase, increasing in frequency with cell density. Cell sorting revealed that expression of stationary markers is associated with a 100-1000 fold increase in the likelihood of survival to antibiotic challenge. The ATP level of the cell is predictive of bactericidal antibiotic efficacy and explains bacterial tolerance to antibiotics.

11.
J Med Microbiol ; 65(8): 848-857, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27375177

ABSTRACT

Staphylococcus aureus is responsible for a wide variety of infections that include superficial skin and soft tissue infections, septicaemia, central nervous system infections, endocarditis, osteomyelitis and pneumonia. Others have demonstrated the importance of toxin-antitoxin (TA) modules in the formation of persisters and the role of the Clp proteolytic system in the regulation of these TA modules. This study was conducted to determine the effect of clpP and clpC deletion on S. aureus persister cell numbers following antibiotic treatment. Deletion of clpP resulted in a significant decrease in persister cells following treatment with oxacillin and erythromycin but not with levofloxacin and daptomycin. Deletion of clpC resulted in a decrease in persister cells following treatment with oxacillin. These differences were dependent on the antibiotic class and the CFU ml-1 in which the cells were treated. Persister revival assays for all the bacterial strains in these studies demonstrated a significant delay in resumption of growth characteristic of persister cells, indicating that the surviving organisms in this study were not likely due to spontaneous antibiotic resistance. Based on our results, ClpP and possibly ClpC play a role in persister cell formation or maintenance, and this effect is dependent on antibiotic class and the CFU ml-1 or the growth phase of the cells.


Subject(s)
Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Gene Deletion , Heat-Shock Proteins/metabolism , Microbial Viability/drug effects , Staphylococcus aureus/drug effects , Staphylococcus aureus/physiology , Bacterial Proteins/genetics , Heat-Shock Proteins/genetics , Staphylococcus aureus/genetics
12.
PLoS One ; 10(3): e0119487, 2015.
Article in English | MEDLINE | ID: mdl-25807379

ABSTRACT

Methicillin resistant Staphylococcus aureus (MRSA) is an opportunistic pathogen found in hospital and community environments that can cause serious infections. A major barrier to genetic manipulations of clinical isolates has been the considerable difficulty in transforming these strains with foreign plasmids, such as those from E. coli, in part due to the type I and IV Restriction Modification (R-M) barriers. Here we combine a Plasmid Artificial Modification (PAM) system with DC10B E. coli cells (dcm mutants) to bypass the barriers of both type I and IV R-M of S. aureus, thus allowing E. coli plasmid DNA to be transformed directly into clinical MRSA strains MW2, N315 and LAC, representing three of the most common clonal complexes. Successful transformation of clinical S. aureus isolates with E. coli-derived plasmids should greatly increase the ability to genetically modify relevant S. aureus strains and advance our understanding of S. aureus pathogenesis.


Subject(s)
DNA, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Staphylococcal Infections/genetics , Escherichia coli/genetics , Humans , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Microbial Sensitivity Tests
13.
J Bacteriol ; 196(23): 4140-51, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25225270

ABSTRACT

Staphylococcus aureus responds to changing extracellular environments in part by adjusting its proteome through alterations of transcriptional priorities and selective degradation of the preexisting pool of proteins. In Bacillus subtilis, the proteolytic adaptor protein MecA has been shown to play a role in assisting with the proteolytic degradation of proteins involved in competence and the oxidative stress response. However, the targets of TrfA, the MecA homolog in S. aureus, have not been well characterized. In this work, we investigated how TrfA assists chaperones and proteases to regulate the proteolysis of several classes of proteins in S. aureus. By fusing the last 3 amino acids of the SsrA degradation tag to Venus, a rapidly folding yellow fluorescent protein, we obtained both fluorescence-based and Western blot assay-based evidence that TrfA and ClpCP are the adaptor and protease, respectively, responsible for the degradation of the SsrA-tagged protein in S. aureus. Notably, the impact of TrfA on degradation was most prominent during late log phase and early stationary phase, due in part to a combination of transcriptional regulation and proteolytic degradation of TrfA by ClpCP. We also characterized the temporal transcriptional regulation governing TrfA activity, wherein Spx, a redox-sensitive transcriptional regulator degraded by ClpXP, activates trfA transcription while repressing its own promoter. Finally, the scope of TrfA-mediated proteolysis was expanded by identifying TrfA as the adaptor that works with ClpCP to degrade antitoxins in S. aureus. Together, these results indicate that the adaptor TrfA adds temporal nuance to protein degradation by ClpCP in S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Endopeptidase Clp/metabolism , Gene Expression Regulation, Bacterial , Heat-Shock Proteins/metabolism , Staphylococcus aureus/metabolism , Gene Expression Profiling , Proteolysis
14.
Antimicrob Agents Chemother ; 57(2): 855-63, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23208713

ABSTRACT

Community-acquired methicillin-resistant Staphylococcus aureus (CA-MRSA) frequently causes skin and soft tissue infections, including impetigo, cellulitis, folliculitis, and infected wounds and ulcers. Uncomplicated CA-MRSA skin infections are typically managed in an outpatient setting with oral and topical antibiotics and/or incision and drainage, whereas complicated skin infections often require hospitalization, intravenous antibiotics, and sometimes surgery. The aim of this study was to develop a mouse model of CA-MRSA wound infection to compare the efficacy of commonly used systemic and topical antibiotics. A bioluminescent USA300 CA-MRSA strain was inoculated into full-thickness scalpel wounds on the backs of mice and digital photography/image analysis and in vivo bioluminescence imaging were used to measure wound healing and the bacterial burden. Subcutaneous vancomycin, daptomycin, and linezolid similarly reduced the lesion sizes and bacterial burden. Oral linezolid, clindamycin, and doxycycline all decreased the lesion sizes and bacterial burden. Oral trimethoprim-sulfamethoxazole decreased the bacterial burden but did not decrease the lesion size. Topical mupirocin and retapamulin ointments both reduced the bacterial burden. However, the petrolatum vehicle ointment for retapamulin, but not the polyethylene glycol vehicle ointment for mupirocin, promoted wound healing and initially increased the bacterial burden. Finally, in type 2 diabetic mice, subcutaneous linezolid and daptomycin had the most rapid therapeutic effect compared with vancomycin. Taken together, this mouse model of CA-MRSA wound infection, which utilizes in vivo bioluminescence imaging to monitor the bacterial burden, represents an alternative method to evaluate the preclinical in vivo efficacy of systemic and topical antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Luminescent Measurements , Methicillin-Resistant Staphylococcus aureus/drug effects , Staphylococcal Infections/drug therapy , Acetamides/administration & dosage , Acetamides/therapeutic use , Administration, Oral , Administration, Topical , Animals , Bacterial Load , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Clindamycin/administration & dosage , Clindamycin/therapeutic use , Community-Acquired Infections/drug therapy , Community-Acquired Infections/microbiology , Daptomycin/administration & dosage , Daptomycin/therapeutic use , Diabetes Mellitus, Type 2 , Disease Models, Animal , Diterpenes , Doxycycline/administration & dosage , Doxycycline/therapeutic use , Linezolid , Male , Mice , Mice, Inbred C57BL , Mupirocin/administration & dosage , Mupirocin/therapeutic use , Oxazolidinones/administration & dosage , Oxazolidinones/therapeutic use , Skin/injuries , Skin/microbiology , Soft Tissue Infections/drug therapy , Soft Tissue Infections/microbiology , Staphylococcal Infections/microbiology , Staphylococcal Skin Infections/drug therapy , Staphylococcal Skin Infections/microbiology , Trimethoprim, Sulfamethoxazole Drug Combination/administration & dosage , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use , Vancomycin/administration & dosage , Vancomycin/therapeutic use , Wound Healing/drug effects
15.
Article in English | MEDLINE | ID: mdl-21393849

ABSTRACT

mazEF modules encode toxin-antitoxin pairs that are involved in the bacterial stress response through controlled and specific degradation of mRNA. Staphylococcus aureus MazF and MazE constitute a unique toxin-antitoxin module under regulation of the sigB operon. A MazF-type mRNA interferase is combined with an antitoxin of unknown fold. Crystals of S. aureus MazF (SaMazF) were grown in space group P2(1)2(1)2(1). The crystals diffracted to 2.1 Šresolution and are likely to contain two SaMazF dimers in the asymmetric unit.


Subject(s)
Bacterial Proteins/chemistry , DNA-Binding Proteins/chemistry , Endoribonucleases/chemistry , RNA, Messenger/metabolism , Staphylococcus aureus/enzymology , Crystallization , Crystallography, X-Ray , Molecular Sequence Data
16.
J Invest Dermatol ; 131(4): 907-15, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21191403

ABSTRACT

Staphylococcus aureus skin infections represent a significant public health threat because of the emergence of antibiotic-resistant strains such as methicillin-resistant S. aureus (MRSA). As greater understanding of protective immune responses and more effective antimicrobial therapies are needed, a S. aureus skin wound infection model was developed in which full-thickness scalpel cuts on the backs of mice were infected with a bioluminescent S. aureus (methicillin sensitive) or USA300 community-acquired MRSA strain and in vivo imaging was used to noninvasively monitor the bacterial burden. In addition, the infection-induced inflammatory response was quantified using in vivo fluorescence imaging of LysEGFP mice. Using this model, we found that both IL-1α and IL-1ß contributed to host defense during a wound infection, whereas IL-1ß was more critical during an intradermal S. aureus infection. Furthermore, treatment of a USA300 MRSA skin infection with retapamulin ointment resulted in up to 85-fold reduction in bacterial burden and a 53% decrease in infection-induced inflammation. In contrast, mupirocin ointment had minimal clinical activity against this USA300 strain, resulting in only a 2-fold reduction in bacterial burden. Taken together, this S. aureus wound infection model provides a valuable preclinical screening method to investigate cutaneous immune responses and the efficacy of topical antimicrobial therapies.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dermoscopy/methods , Methicillin-Resistant Staphylococcus aureus/immunology , Microscopy, Fluorescence/methods , Mupirocin/pharmacology , Staphylococcal Skin Infections , Administration, Topical , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Community-Acquired Infections/drug therapy , Community-Acquired Infections/immunology , Community-Acquired Infections/pathology , Dermatitis/drug therapy , Dermatitis/immunology , Dermatitis/pathology , Disease Models, Animal , Diterpenes , Drug Monitoring/methods , Green Fluorescent Proteins/genetics , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Staphylococcal Skin Infections/drug therapy , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/pathology
17.
Infect Immun ; 78(10): 4384-91, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20696829

ABSTRACT

The expression of virulence factors in Staphylococcus aureus is tightly coordinated by a vast network of regulatory molecules. In this report, we characterize a genetic locus unique to staphylococci called rsr that has a role in repressing two key virulence regulators, sarR and agr. Using strain SH1000, we showed that the transcription of virulence effectors, such as hla, sspA, and spa, is altered in an rsr mutant in a way consistent with agr upregulation. Analysis of RNAIII expression of the agr locus in rsr and rsr-sarR mutants indicated that rsr likely contributes to agr expression independently of SarR. We also provide evidence using a murine model of S. aureus skin infection that the effects mediated by rsr reduce disease progression.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial/physiology , Staphylococcus aureus/genetics , Staphylococcus aureus/pathogenicity , Trans-Activators/metabolism , Animals , Bacterial Proteins/genetics , Genetic Linkage , Male , Mice , Mice, Inbred C57BL , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/metabolism , Trans-Activators/genetics , Transcription, Genetic , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
18.
J Bacteriol ; 192(5): 1416-22, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20038589

ABSTRACT

Bacterial toxin-antitoxin (TA) systems typically consist of a small, labile antitoxin that inactivates a specific longer-lived toxin. In Escherichia coli, such antitoxins are proteolytically regulated by the ATP-dependent proteases Lon and ClpP. Under normal conditions, antitoxin synthesis is sufficient to replace this loss from proteolysis, and the bacterium remains protected from the toxin. However, if TA production is interrupted, antitoxin levels decrease, and the cognate toxin is free to inhibit the specific cellular component, such as mRNA, DnaB, or gyrase. To date, antitoxin degradation has been studied only in E. coli, so it remains unclear whether similar mechanisms of regulation exist in other organisms. To address this, we followed antitoxin levels over time for the three known TA systems of the major human pathogen Staphylococcus aureus, mazEF, axe1-txe1, and axe2-txe2. We observed that the antitoxins of these systems, MazE(sa), Axe1, and Axe2, respectively, were all degraded rapidly (half-life [t(1/2)], approximately 18 min) at rates notably higher than those of their E. coli counterparts, such as MazE (t(1/2), approximately 30 to 60 min). Furthermore, when S. aureus strains deficient for various proteolytic systems were examined for changes in the half-lives of these antitoxins, only strains with clpC or clpP deletions showed increased stability of the molecules. From these studies, we concluded that ClpPC serves as the functional unit for the degradation of all known antitoxins in S. aureus.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Endopeptidase Clp/metabolism , Gene Expression Regulation, Enzymologic , Heat-Shock Proteins/metabolism , Staphylococcus aureus/physiology , Endopeptidase Clp/deficiency , Gene Knockout Techniques , Half-Life , Heat-Shock Proteins/deficiency
19.
Nat Chem Biol ; 6(1): 41-5, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19935660

ABSTRACT

It is postulated that in addition to cell density, other factors such as the dimensions and diffusional characteristics of the environment could influence quorum sensing (QS) and induction of genetic reprogramming. Modeling studies predict that QS may operate at the level of a single cell, but, owing to experimental challenges, the potential benefits of QS by individual cells remain virtually unexplored. Here we report a physical system that mimics isolation of a bacterium, such as within an endosome or phagosome during infection, and maintains cell viability under conditions of complete chemical and physical isolation. For Staphylococcus aureus, we show that quorum sensing and genetic reprogramming can occur in a single isolated organism. Quorum sensing allows S. aureus to sense confinement and to activate virulence and metabolic pathways needed for survival. To demonstrate the benefit of confinement-induced quorum sensing to individuals, we showed that quorum-sensing bacteria have significantly greater viability over non-QS bacteria.


Subject(s)
Quorum Sensing/genetics , Staphylococcus aureus/genetics , Endosomes/metabolism , Gene Deletion , Gene Expression Regulation, Bacterial , Green Fluorescent Proteins/chemistry , Kinetics , Models, Biological , Models, Genetic , Mutation , Nanotechnology/methods , Phagocytosis , Phagosomes/metabolism , Virulence/genetics , Virulence Factors/genetics
20.
J Bacteriol ; 191(8): 2795-805, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19181798

ABSTRACT

In Staphylococcus aureus, the sigB operon codes for the alternative sigma factor sigma(B) and its regulators that enable the bacteria to rapidly respond to environmental stresses via redirection of transcriptional priorities. However, a full model of sigma(B) regulation in S. aureus has not yet emerged. Earlier data has suggested that mazEF, a toxin-antitoxin (TA) module immediately upstream of the sigB operon, was transcribed with the sigB operon. Here we demonstrate that the promoter P(mazE) upstream of mazEF is essential for full sigma(B) activity and that instead of utilizing autorepression typical of TA systems, sigB downregulates this promoter, providing a negative-feedback loop for sigB to repress its own transcription. We have also found that the transcriptional regulator SarA binds and activates P(mazE). In addition, P(mazE) was shown to respond to environmental and antibiotic stresses in a way that provides an additional layer of control over sigB expression. The antibiotic response also appears to occur in two other TA systems in S. aureus, indicating a shared mechanism of regulation.


Subject(s)
Bacterial Proteins/biosynthesis , Bacterial Toxins/biosynthesis , Gene Expression Regulation, Bacterial , Sigma Factor/biosynthesis , Staphylococcus aureus/physiology , Transcription Factors/biosynthesis , Bacterial Proteins/metabolism , DNA, Bacterial/metabolism , Gene Order , Genes, Bacterial , Operon , Promoter Regions, Genetic , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL