Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Microbiol ; 92(6): 1299-312, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24779437

ABSTRACT

We investigated the in vivo relevance of the impact of sarA and saeRS on protease production using derivatives of the USA300 strain LAC. The results confirmed that mutation of saeRS or sarA reduces virulence in a bacteremia model to a comparable degree. However, while eliminating protease production restored virulence in the sarA mutant, it had little impact in the saeRS mutant. Additionally, constitutive activation of saeRS (saeRS(C)) enhanced the virulence of LAC and largely restored virulence in the isogenic sarA mutant. Based on these results, together with our analysis of the representative virulence factors alpha toxin, protein A (Spa), and extracellular nucleases, we propose a model in which the attenuation of saeRS mutants is defined primarily by decreased production of such factors, while constitutive activation of saeRS increases virulence, and reverses the attenuation of sarA mutants, because it results in both increased production and decreased protease-mediated degradation of these same factors. This regulatory balance was also apparent in a murine model of catheter-associated infection, with the results suggesting that the impact of saeRS on nuclease production plays an important role during the early stages of these infections that is partially offset by increased protease production in sarA mutants.


Subject(s)
Bacterial Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Peptide Hydrolases/metabolism , Protein Kinases/metabolism , Animals , Bacteremia/microbiology , Bacteremia/pathology , Bacterial Proteins/genetics , Catheter-Related Infections/microbiology , Catheter-Related Infections/pathology , Disease Models, Animal , Mice , Transcription Factors , Virulence
2.
Cell Host Microbe ; 13(6): 759-72, 2013 Jun 12.
Article in English | MEDLINE | ID: mdl-23768499

ABSTRACT

Osteomyelitis is a common manifestation of invasive Staphylococcus aureus infection. Pathogen-induced bone destruction limits antimicrobial penetration to the infectious focus and compromises treatment of osteomyelitis. To investigate mechanisms of S. aureus-induced bone destruction, we developed a murine model of osteomyelitis. Microcomputed tomography of infected femurs revealed that S. aureus triggers profound alterations in bone turnover. The bacterial regulatory locus sae was found to be critical for osteomyelitis pathogenesis, as Sae-regulated factors promote pathologic bone remodeling and intraosseous bacterial survival. Exoproteome analyses revealed the Sae-regulated protease aureolysin as a major determinant of the S. aureus secretome and identified the phenol-soluble modulins as aureolysin-degraded, osteolytic peptides that trigger osteoblast cell death and bone destruction. These studies establish a murine model for pathogen-induced bone remodeling, define Sae as critical for osteomyelitis pathogenesis, and identify protease-dependent exoproteome remodeling as a major determinant of the staphylococcal virulence repertoire.


Subject(s)
Bacterial Proteins/metabolism , Bone Remodeling , Metalloendopeptidases/metabolism , Osteomyelitis/pathology , Staphylococcal Infections/pathology , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Animals , Disease Models, Animal , Femur/diagnostic imaging , Femur/pathology , Mice , Osteomyelitis/microbiology , Staphylococcal Infections/microbiology , Staphylococcus aureus/enzymology , Tomography, X-Ray Computed
3.
Mol Microbiol ; 86(5): 1183-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23075270

ABSTRACT

Mutation of staphylococcal accessory regulator (sarA) results in increased production of extracellular proteases in Staphylococcus aureus, which has been correlated with decreased biofilm formation and decreased accumulation of extracellular toxins. We used murine models of implant-associated biofilm infection and S. aureus bacteraemia (SAB) to compare virulence of USA300 strain LAC, its isogenic sarA mutant, and derivatives of each of these strains with mutations in all 10 of the genes encoding recognized extracellular proteases. The sarA mutant was attenuated in both models, and this was reversed by eliminating production of extracellular proteases. To examine the mechanistic basis, we identified proteins impacted by sarA in a protease-dependent manner. We identified 253 proteins where accumulation was reduced in the sarA mutant compared with the parent strain, and was restored in the sarA/protease mutant. Additionally, in SAB, the LAC protease mutant exhibited a hypervirulent phenotype by comparison with the isogenic parent strain, demonstrating that sarA also positively regulates production of virulence factors, some of which are subject to protease-mediated degradation. We propose a model in which attenuation of sarA mutants is defined by their inability to produce critical factors and simultaneously repress production of extracellular proteases that would otherwise limit accumulation of virulence factors.


Subject(s)
Bacterial Proteins/pharmacology , Gene Expression Regulation, Bacterial , Peptide Hydrolases/drug effects , Staphylococcus aureus/pathogenicity , Virulence Factors/metabolism , Animals , Animals, Outbred Strains , Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Biofilms/drug effects , Biofilms/growth & development , Catheter-Related Infections/microbiology , Daptomycin/pharmacology , Disease Models, Animal , Female , Humans , Mice , Microbial Sensitivity Tests , Peptide Hydrolases/genetics , Peptide Hydrolases/metabolism , Staphylococcal Infections/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Virulence , Virulence Factors/genetics
4.
PLoS One ; 7(6): e38453, 2012.
Article in English | MEDLINE | ID: mdl-22685571

ABSTRACT

Mutation of the staphylococcal accessory regulator (sarA) limits biofilm formation in diverse strains of Staphylococcus aureus, but there are exceptions. One of these is the commonly studied strain Newman. This strain has two defects of potential relevance, the first being mutations that preclude anchoring of the fibronectin-binding proteins FnbA and FnbB to the cell wall, and the second being a point mutation in saeS that results in constitutive activation of the saePQRS regulatory system. We repaired these defects to determine whether either plays a role in biofilm formation and, if so, whether this could account for the reduced impact of sarA in Newman. Restoration of surface-anchored FnbA enhanced biofilm formation, but mutation of sarA in this fnbA-positive strain increased rather than decreased biofilm formation. Mutation of sarA in an saeS-repaired derivative of Newman (P18L) or a Newman saeRS mutant (ΔsaeRS) resulted in a biofilm-deficient phenotype like that observed in clinical isolates, even in the absence of surface-anchored FnbA. These phenotypes were correlated with increased production of extracellular proteases and decreased accumulation of FnbA and/or Spa in the P18L and ΔsaeRS sarA mutants by comparison to the Newman sarA mutant. The reduced accumulation of Spa was reversed by mutation of the gene encoding aureolysin, while the reduced accumulation of FnbA was reversed by mutation of the sspABC operon. These results demonstrate that saeRS and sarA act synergistically to repress the production of extracellular proteases that would otherwise limit accumulation of critical proteins that contribute to biofilm formation, with constitutive activation of saeRS limiting protease production, even in a sarA mutant, to a degree that can be correlated with increased enhanced capacity to form a biofilm. Although it remains unclear whether these effects are mediated directly or indirectly, studies done with an sspA::lux reporter suggest they are mediated at a transcriptional level.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Peptide Hydrolases/metabolism , Protein Kinases/metabolism , Staphylococcus aureus/physiology , Trans-Activators/metabolism , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Proteins/genetics , Blotting, Western , Extracellular Space/enzymology , Gene Expression Regulation, Bacterial , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mutation , Peptide Hydrolases/genetics , Phenotype , Protein Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Trans-Activators/genetics , Transcription Factors
5.
J Bacteriol ; 193(12): 2948-58, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21478342

ABSTRACT

We demonstrate that mutation of the staphylococcal accessory regulator (sarA) limits the accumulation of alpha-toxin and phenol-soluble modulins (PSMs) in Staphylococcus aureus isolates of the USA300 clonal lineage. Degradation assays and experiments done with protease inhibitors suggested that this was due to the increased production of extracellular proteases rather than differences associated with the impact of sarA on transcription of the target gene (hla) or the accessory gene regulator (agr). This was confirmed by demonstrating that concomitant mutation of the gene encoding aureolysin (aur) reversed the alpha-toxin and PSM-deficient phenotypes of a USA300 sarA mutant. Mutation of sarA had little impact on the alpha-toxin or PSM phenotypes of the commonly studied strain Newman, which is known to have a mutation in saeS that results in constitutive activation of the saeRS regulatory system, and we also demonstrate that repair of this defect resulted in the increased production of extracellular proteases and reversed both the alpha-toxin and PSM-positive phenotypes of a Newman sarA mutant.


Subject(s)
Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Gene Expression Regulation, Bacterial/physiology , Hemolysin Proteins/metabolism , Staphylococcus aureus/metabolism , Bacterial Proteins/genetics , Bacterial Toxins/genetics , Genetic Variation , Hemolysin Proteins/genetics , Mutation , Phenols/chemistry , Phenotype , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Staphylococcus aureus/genetics
6.
PLoS One ; 5(5): e10790, 2010 May 24.
Article in English | MEDLINE | ID: mdl-20520723

ABSTRACT

BACKGROUND: The accessory gene regulator (agr) and staphylococcal accessory regulator (sarA) play opposing roles in Staphylococcus aureus biofilm formation. There is mounting evidence to suggest that these opposing roles are therapeutically relevant in that mutation of agr results in increased biofilm formation and decreased antibiotic susceptibility while mutation of sarA has the opposite effect. To the extent that induction of agr or inhibition of sarA could potentially be used to limit biofilm formation, this makes it important to understand the epistatic relationships between these two loci. METHODOLOGY/PRINCIPAL FINDINGS: We generated isogenic sarA and agr mutants in clinical isolates of S. aureus and assessed the relative impact on biofilm formation. Mutation of agr resulted in an increased capacity to form a biofilm in the 8325-4 laboratory strain RN6390 but had little impact in clinical isolates S. aureus. In contrast, mutation of sarA resulted in a reduced capacity to form a biofilm in all clinical isolates irrespective of the functional status of agr. This suggests that the regulatory role of sarA in biofilm formation is independent of the interaction between sarA and agr and that sarA is epistatic to agr in this context. This was confirmed by demonstrating that restoration of sarA function restored the ability to form a biofilm even in the corresponding agr mutants. Mutation of sarA in clinical isolates also resulted in increased production of extracellular proteases and extracellular nucleases, both of which contributed to the biofilm-deficient phenotype of sarA mutants. However, studies comparing different strains with and without proteases inhibitors and/or mutation of the nuclease genes demonstrated that the agr-independent, sarA-mediated repression of extracellular proteases plays a primary role in this regard. CONCLUSIONS AND SIGNIFICANCE: The results we report suggest that inhibitors of sarA-mediated regulation could be used to limit biofilm formation in S. aureus and that the efficacy of such inhibitors would not be limited by spontaneous mutation of agr in the human host.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Epistasis, Genetic , Staphylococcus aureus/genetics , Staphylococcus aureus/physiology , Trans-Activators/metabolism , Biofilms/drug effects , Deoxyribonucleases/genetics , Epistasis, Genetic/drug effects , Extracellular Space/drug effects , Extracellular Space/enzymology , Mutation/genetics , Peptide Hydrolases/metabolism , Polysaccharides, Bacterial/biosynthesis , Protease Inhibitors/pharmacology , RNA, Bacterial/metabolism , Staphylococcus aureus/drug effects , Staphylococcus aureus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...