Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 275
Filter
1.
Mol Cell ; 73(2): 278-290.e4, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30503774

ABSTRACT

Adaptive immune systems must accurately distinguish between self and non-self in order to defend against invading pathogens while avoiding autoimmunity. Type III CRISPR-Cas systems employ guide RNA to recognize complementary RNA targets, which triggers the degradation of both the invader's transcripts and their template DNA. These systems can broadly eliminate foreign targets with multiple mutations but circumvent damage to the host genome. To explore the molecular basis for these features, we use single-molecule fluorescence microscopy to study the interaction between a type III-A ribonucleoprotein complex and various RNA substrates. We find that Cas10-the DNase effector of the complex-displays rapid conformational fluctuations on foreign RNA targets, but is locked in a static configuration on self RNA. Target mutations differentially modulate Cas10 dynamics and tune the CRISPR interference activity in vivo. These findings highlight the central role of the internal dynamics of CRISPR-Cas complexes in self versus non-self discrimination and target specificity.


Subject(s)
Autoimmunity , Bacterial Proteins/immunology , CRISPR-Associated Proteins/immunology , CRISPR-Cas Systems/immunology , Clustered Regularly Interspaced Short Palindromic Repeats/immunology , RNA, Bacterial/immunology , Self Tolerance , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli/immunology , Kinetics , Microscopy, Fluorescence , Mutation , Nucleic Acid Conformation , Protein Conformation , RNA, Bacterial/chemistry , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Signal Transduction , Single Molecule Imaging/methods , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/immunology , Structure-Activity Relationship
2.
Molecules ; 29(16)2024 Aug 10.
Article in English | MEDLINE | ID: mdl-39202871

ABSTRACT

Staphylococcus aureus (S. aureus) is a major bacterial infection in humans, leading to severe disease and causing death. The stagnation of antibiotic development in recent decades has made it difficult to combat drug-resistant infections. In this study, we performed an in silico structure-based drug screening (SBDS) targeting the S. aureus MurE (saMurE) enzyme involved in cell wall synthesis of S. aureus. saMurE is an enzyme that is essential for the survival of S. aureus but not present in humans. SBDS identified nine saMurE inhibitor candidates, Compounds 1-9, from a structural library of 154,118 compounds. Among them, Compound 2 showed strong antibacterial activity against Staphylococcus epidermidis (S. epidermidis) used as a model bacterium. Amino acid sequence homology between saMurE and S. epidermidis MurE is 87.4%, suggesting that Compound 2 has a similar inhibitory effect on S. aureus. Compound 2 showed an IC50 value of 301 nM for S. epidermidis in the dose-dependent growth inhibition assay. Molecular dynamics simulation showed that Compound 2 binds stably to both S. aureus MurD and S. aureus MurF, suggesting that it is a potential multi-pharmacological pharmacological inhibitor. The structural and bioactivity information of Compound 2, as well as its potential multiple-target activity, could contribute to developing new antimicrobial agents based on MurE inhibition.


Subject(s)
Anti-Bacterial Agents , Drug Evaluation, Preclinical , Staphylococcus aureus , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/enzymology , Microbial Sensitivity Tests , Molecular Docking Simulation , Computer Simulation , Drug Discovery , Structure-Activity Relationship , Peptide Synthases/antagonists & inhibitors , Peptide Synthases/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Humans , Molecular Dynamics Simulation
3.
Development ; 147(22)2020 11 30.
Article in English | MEDLINE | ID: mdl-33093152

ABSTRACT

Mini-III RNase (mR3), a member of RNase III endonuclease family, can bind to and cleave double-stranded RNAs (dsRNAs). Inactive mR3 protein without the α5ß-α6 loop loses the dsRNA cleavage activity, but retains dsRNA binding activity. Here, we establish an inactive mR3-based non-engineered mR3/dsRNA system for RNA tracking in zebrafish embryos. In vitro binding experiments show that inactive Staphylococcus epidermidis mR3 (dSmR3) protein possesses the highest binding affinity with dsRNAs among mR3s from other related species, and its binding property is retained in zebrafish embryos. Combined with a fluorescein-labeled antisense RNA probe recognizing the target mRNAs, dSmR3 tagged with a nuclear localization sequence and a fluorescent protein could allow visualization of the dynamics of endogenous target mRNAs. The dSmR3/antisense probe dual-color system provides a new approach for tracking non-engineered RNAs in real-time, which will help understand how endogenous RNAs dynamically move during embryonic development.


Subject(s)
Bacterial Proteins/metabolism , Fluorescein , RNA, Antisense , RNA, Messenger/metabolism , Ribonuclease III/metabolism , Staphylococcus epidermidis , Zebrafish/metabolism , Animals , Bacterial Proteins/genetics , Fluorescein/chemistry , Fluorescein/pharmacology , Microscopy, Fluorescence , RNA, Antisense/chemistry , RNA, Antisense/pharmacology , RNA, Messenger/genetics , Ribonuclease III/genetics , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Zebrafish/genetics
4.
J Allergy Clin Immunol ; 147(3): 955-966.e16, 2021 03.
Article in English | MEDLINE | ID: mdl-32634452

ABSTRACT

BACKGROUND: Staphylococcus aureus and Staphylococcus epidermidis are the most abundant bacteria found on the skin of patients with atopic dermatitis (AD). S aureus is known to exacerbate AD, whereas S epidermidis has been considered a beneficial commensal organism. OBJECTIVE: In this study, we hypothesized that S epidermidis could promote skin damage in AD by the production of a protease that damages the epidermal barrier. METHODS: The protease activity of S epidermidis isolates was compared with that of other staphylococcal species. The capacity of S epidermidis to degrade the barrier and induce inflammation was examined by using human keratinocyte tissue culture and mouse models. Skin swabs from atopic and healthy adult subjects were analyzed for the presence of S epidermidis genomic DNA and mRNA. RESULTS: S epidermidis strains were observed to produce strong cysteine protease activity when grown at high density. The enzyme responsible for this activity was identified as EcpA, a cysteine protease under quorum sensing control. EcpA was shown to degrade desmoglein-1 and LL-37 in vitro, disrupt the physical barrier, and induce skin inflammation in mice. The abundance of S epidermidis and expression of ecpA mRNA were increased on the skin of some patients with AD, and this correlated with disease severity. Another commensal skin bacterial species, Staphylococcus hominis, can inhibit EcpA production by S epidermidis. CONCLUSION: S epidermidis has commonly been regarded as a beneficial skin microbe, whereas S aureus has been considered deleterious. This study suggests that the overabundance of S epidermidis found on some atopic patients can act similarly to S aureus and damage the skin by expression of a cysteine protease.


Subject(s)
Bacterial Proteins/metabolism , Cysteine Proteases/metabolism , Dermatitis, Atopic/microbiology , Microbiota , Skin/microbiology , Staphylococcal Skin Infections/microbiology , Staphylococcus epidermidis/enzymology , Animals , Antimicrobial Cationic Peptides/metabolism , Cells, Cultured , DNA, Bacterial/genetics , Dermatitis, Atopic/pathology , Desmoglein 1/metabolism , Humans , Keratinocytes/microbiology , Keratinocytes/pathology , Mice , Mice, Inbred C57BL , Severity of Illness Index , Skin/pathology , Staphylococcal Skin Infections/pathology , Cathelicidins
5.
J Bacteriol ; 203(17): e0017821, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34096781

ABSTRACT

Antimicrobial peptides (AMPs) are one of the key immune responses that can eliminate pathogenic bacteria through membrane perturbation. As a successful skin commensal, Staphylococcus epidermidis can sense and respond to AMPs through the GraXRS two-component system and an efflux system comprising the VraG permease and VraF ATPase. GraS is a membrane sensor known to function in AMP resistance through a negatively charged, 9-residue extracellular loop, which is predicted to be linear without any secondary structure. An important question is how GraS can impart effective sensing of AMPs through such a small unstructured sequence. In this study, we verified the role of graS and vraG in AMP sensing in S. epidermidis, as demonstrated by the failure of the ΔgraS or ΔvraG mutants to sense. Deletion of the extracellular loop of VraG did not affect sensing but reduced survival with polymyxin B. Importantly, a specific region within the extracellular loop, termed the guard loop (GL), has inhibitory activity since sensing of polymyxin B was enhanced in the ΔGL mutant, indicating that the GL may act as a gatekeeper for sensing. Bacterial two-hybrid analysis demonstrated that the extracellular regions of GraS and VraG interact, but interaction appears dispensable to sensing activity. Mutation of the extracellular loop of VraG, the GL, and the active site of VraF suggested that an active detoxification function of VraG is necessary for AMP resistance. Altogether, we provide evidence for a unique sensory scheme that relies on the function of a permease to impart effective information processing. IMPORTANCE Staphylococcus epidermidis has become an important opportunistic pathogen that is responsible for nosocomial and device-related infections that account for considerable morbidity worldwide. A thorough understanding of the mechanisms that enable S. epidermidis to colonize human skin successfully is essential for the development of alternative treatment strategies and prophylaxis. Here, we demonstrate the importance of an AMP response system in a clinically relevant S. epidermidis strain. Furthermore, we provide evidence for a unique sensory scheme that would rely on the detoxification function of a permease to effect information processing.


Subject(s)
Adenosine Triphosphatases/metabolism , Bacterial Proteins/metabolism , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/enzymology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Antimicrobial Cationic Peptides/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Humans , Membrane Transport Proteins/genetics , Protein Conformation, alpha-Helical , Staphylococcal Infections/metabolism , Staphylococcus epidermidis/chemistry , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics
6.
J Biol Chem ; 295(49): 16863-16876, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32994223

ABSTRACT

RNase J enzymes are metallohydrolases that are involved in RNA maturation and RNA recycling, govern gene expression in bacteria, and catalyze both exonuclease and endonuclease activity. The catalytic activity of RNase J is regulated by multiple mechanisms which include oligomerization, conformational changes to aid substrate recognition, and the metal cofactor at the active site. However, little is known of how RNase J paralogs differ in expression and activity. Here we describe structural and biochemical features of two Staphylococcus epidermidis RNase J paralogs, RNase J1 and RNase J2. RNase J1 is a homodimer with exonuclease activity aided by two metal cofactors at the active site. RNase J2, on the other hand, has endonuclease activity and one metal ion at the active site and is predominantly a monomer. We note that the expression levels of these enzymes vary across Staphylococcal strains. Together, these observations suggest that multiple interacting RNase J paralogs could provide a strategy for functional improvisation utilizing differences in intracellular concentration, quaternary structure, and distinct active site architecture despite overall structural similarity.


Subject(s)
Bacterial Proteins/metabolism , Ribonucleases/metabolism , Staphylococcus epidermidis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/classification , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Coenzymes/chemistry , Coenzymes/metabolism , Crystallography, X-Ray , Dimerization , Gene Expression Regulation, Bacterial , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Phylogeny , Protein Structure, Quaternary , RNA/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Ribonucleases/chemistry , Ribonucleases/classification , Ribonucleases/genetics , Substrate Specificity
7.
Biochem Biophys Res Commun ; 534: 1064-1068, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33092791

ABSTRACT

In this work, the impact of APTES-modified TiO2 photocatalysts on antioxidant enzymes (catalase and superoxide dismutase) activity secreted by bacteria was presented. Microbial tests has been examined using Escherichia coli (ATCC 29425) and Staphylococcus epidermidis (ATCC 49461) as model organisms. It was found that APTES-TiO2 affected the activity of antioxidant enzymes. Additionally, obtained APTES-TiO2 photocatalysts were capable of total E. coli and S. epidermidis inactivation under artificial solar light irradiation. The sample modified with the concentration of APTES equals 300 mM (TiO2-4h-120°C-300mM) showed the strongest photocatalytic activity toward both bacteria species. The two-stage photocatalytic mechanism of bacteria response to photocatalysts was proposed.


Subject(s)
Catalase/metabolism , Escherichia coli/enzymology , Propylamines/chemistry , Silanes/chemistry , Staphylococcus epidermidis/enzymology , Superoxide Dismutase/metabolism , Titanium/chemistry , Catalysis/radiation effects , Disinfection , Enzyme Activation/radiation effects , Escherichia coli/cytology , Escherichia coli/radiation effects , Light , Microbial Viability/radiation effects , Oxidative Stress/radiation effects , Photochemical Processes/radiation effects , Staphylococcus epidermidis/cytology , Staphylococcus epidermidis/radiation effects
8.
BMC Microbiol ; 20(1): 240, 2020 08 05.
Article in English | MEDLINE | ID: mdl-32758127

ABSTRACT

BACKGROUND: Staphylococcus epidermidis is the leading coagulase negative staphylococci (CoNS) species associated with healthcare associated infections. In order to de-escalate antimicrobial therapy, isolates of S. epidermidis lacking the blaZ gene should be eligible for targeted antimicrobial therapy. However, testing the susceptibility of coagulase negative staphylococci (CoNS) to penicillin G is no longer recommended by EUCAST, given the low performances for penicillinase detection in CoNS. The objective of this work was to determine a phenotypic method with high performance for detecting penicillinase production in S. epidermidis. RESULTS: Four techniques for the detection of penicillinase production (disk diffusion, zone edge test, nitrocefin test, Minimal Inhibitory Concentration (MIC) by automated system Vitek2®) were evaluated on 182 S. epidermidis isolates, using identification of blaZ gene by PCR as the reference method. The performance of the methods for penicillinase detection was compared by the sensitivity, the specificity, the negative predictive value and the positive predictive value, and with Cohen's kappa statistical test. Among the 182 S. epidermidis included in this study, 55 carried the blaZ gene. The nitrocefin test, characterized by a poor sensitivity (91%), was therefore excluded from S. epidermidis penicillinase detection. The algorithm proposed here for the penicillinase detection in S. epidermidis involved two common antimicrobial susceptibility techniques: disk diffusion method and MIC by Vitek2® system. Disk diffusion method, interpreted with a 26 mm breakpoint for penicillin G, was associated with a high sensitivity (98%) and specificity (100%). This method was completed with zone edge test for S. epidermidis with penicillin G diameter from 26 to 35 mm (sensitivity of 98%). The Vitek2® system is associated with a low sensitivity (93%) and a high specificity (99%) This low sensitivity is associated with false negative results, in isolates with 0.12 mg/L Penicillin G MIC values and blaZ positive. Thus for penicillin G MIC of 0.06 mg/L or 0.12 mg/L, a second step with disc diffusion method is suggested. CONCLUSIONS: According to our results, the strategy proposed here allows the interpretation of penicillin G susceptibility in S. epidermidis isolates, with an efficient detection of penicillin G resistance.


Subject(s)
Microbial Sensitivity Tests/methods , Penicillinase/isolation & purification , Staphylococcus epidermidis/enzymology , Algorithms , Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Genes, Bacterial/genetics , Humans , Penicillin G/pharmacology , Penicillin Resistance/drug effects , Penicillin Resistance/genetics , Penicillinase/metabolism , Phenotype , Polymerase Chain Reaction , Sensitivity and Specificity , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/drug effects , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/isolation & purification
9.
Org Biomol Chem ; 18(3): 518-529, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31854421

ABSTRACT

Mevalonate diphosphate decarboxylase (MDD) catalyses a crucial step of the mevalonate pathway via Mg2+-ATP-dependent phosphorylation and decarboxylation reactions to ultimately produce isopentenyl diphosphate, the precursor of isoprenoids, which is essential to bacterial functions and provides ideal building blocks for the biosynthesis of isopentenols. However, the metal ion(s) in MDD has not been unambiguously resolved, which limits the understanding of the catalytic mechanism and the exploitation of enzymes for the development of antibacterial therapies or the mevalonate metabolic pathway for the biosynthesis of biofuels. Here by analogizing structurally related kinases and molecular dynamics simulations, we constructed a model of the MDD-substrate-ATP-Mg2+ complex and proposed that MDD requires two Mg2+ ions for maintaining a catalytically active conformation. Subsequent QM/MM studies indicate that MDD catalyses the phosphorylation of its substrate mevalonate diphosphate (MVAPP) via a direct phosphorylation reaction, instead of the previously assumed catalytic base mechanism. The results here would shed light on the active conformation of MDD-related enzymes and their catalytic mechanisms and therefore be useful for developing novel antimicrobial therapies or reconstructing mevalonate metabolic pathways for the biosynthesis of biofuels.


Subject(s)
Bacterial Proteins/chemistry , Carboxy-Lyases/chemistry , Mevalonic Acid/analogs & derivatives , Adenosine Triphosphate/chemistry , Amino Acid Sequence , Catalytic Domain , Density Functional Theory , Magnesium/chemistry , Mevalonic Acid/chemistry , Models, Chemical , Molecular Dynamics Simulation , Phosphorylation , Staphylococcus epidermidis/enzymology
10.
J Clin Microbiol ; 57(12)2019 12.
Article in English | MEDLINE | ID: mdl-31462553

ABSTRACT

Methicillin (ß-lactam) resistance in Staphylococcus epidermidis is mediated by the mecA gene, with resistance reported to be as high as 90%. The goal of this study was to evaluate oxacillin and cefoxitin disk diffusion (DD) and broth microdilution (BMD) methods for the detection of mecA-mediated ß-lactam resistance in 100 human isolates of S. epidermidis (48 mecA-positive isolates and 52 mecA negative isolates). Oxacillin DD tests using the Clinical and Laboratory Standards Institute (CLSI) M100-S28 breakpoints for S. pseudintermedius/S. schleiferi accurately differentiated mecA-positive and -negative S. epidermidis isolates, with categorical agreement (CA) of 100% and no very major errors (VMEs) or major errors (MEs) identified. Likewise, oxacillin BMD and cefoxitin DD tests using the coagulase-negative Staphylococcus species (CoNS) breakpoints were highly reliable for detecting mecA-mediated ß-lactam resistance in S. epidermidis isolates. For cefoxitin DD and BMD results interpreted using S. aureus/S. lugdunensis breakpoints, the CA was 97.6% and 96.2%, respectively. There were 4.9% VMEs for cefoxitin DD with 0% MEs, and 3.6% VMEs and 3.9% MEs for cefoxitin BMD. Oxacillin BMD using S. aureus/S. lugdunensis breakpoints yielded the highest VMEs at 17.4% and 90% CA. Our findings demonstrate that oxacillin DD tests using the CLSI M100-S28 breakpoints for S. pseudintermedius/S. schleiferi and oxacillin BMD and cefoxitin DD tests using the CoNS breakpoints reliably identified mecA-mediated ß-lactam resistance in S. epidermidis Using mecA PCR as the gold standard, the PBP2a SA culture colony test (Abbott Diagnostics) exhibited 100% sensitivity and specificity whereas 2 false negatives were identified using the PBP2' latex agglutination test kit (Thermo Fisher Scientific) with sensitivity and specificity of 95.8% and 100%, respectively.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cefoxitin/pharmacology , Microbial Sensitivity Tests/methods , Oxacillin/pharmacology , Staphylococcus epidermidis/drug effects , beta-Lactam Resistance , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Humans , Polymerase Chain Reaction , Sensitivity and Specificity , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/isolation & purification
11.
Nucleic Acids Res ; 45(4): 2112-2123, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28204542

ABSTRACT

CRISPR­Cas (Clustered regularly interspaced short palindromic repeats-CRISPR-associated proteins) is a prokaryotic immune system that destroys foreign nucleic acids in a sequence-specific manner using Cas nucleases guided by short RNAs (crRNAs). Staphylococcus epidermidis harbours a Type III-A CRISPR­Cas system that encodes the Cas10­Csm interference complex and crRNAs that are subjected to multiple processing steps. The final step, called maturation, involves a concerted effort between Csm3, a ruler protein in Cas10­Csm that measures six-nucleotide increments, and the activity of a nuclease(s) that remains unknown. Here, we elucidate the contributions of the Cas10­Csm complex toward maturation and explore roles of non-Cas nucleases in this process. Using genetic and biochemical approaches, we show that charged residues in Csm3 facilitate its self-assembly and dictate the extent of maturation cleavage. Additionally, acidic residues in Csm5 are required for efficient maturation, but recombinant Csm5 fails to cleave crRNAs in vitro. However, we detected cellular nucleases that co-purify with Cas10­Csm, and show that Csm5 regulates their activities through distinct mechanisms. Altogether, our results support roles for non-Cas nuclease(s) during crRNA maturation and establish a link between Type III-A CRISPR­Cas immunity and central nucleic acid metabolism.


Subject(s)
CRISPR-Associated Proteins/metabolism , CRISPR-Cas Systems , RNA, Bacterial/metabolism , Ribonucleases/metabolism , CRISPR-Associated Proteins/chemistry , Polyribonucleotide Nucleotidyltransferase , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism
12.
Mol Microbiol ; 103(5): 860-874, 2017 03.
Article in English | MEDLINE | ID: mdl-27997732

ABSTRACT

The otherwise harmless skin inhabitant Staphylococcus epidermidis is a major cause of healthcare-associated medical device infections. The species' selective pathogenic potential depends on its production of surface adherent biofilms. The Cell wall-anchored protein Aap promotes biofilm formation in S. epidermidis, independently from the polysaccharide intercellular adhesin PIA. Aap requires proteolytic cleavage to act as an intercellular adhesin. Whether and which staphylococcal proteases account for Aap processing is yet unknown. Here, evidence is provided that in PIA-negative S. epidermidis 1457Δica, the metalloprotease SepA is required for Aap-dependent S. epidermidis biofilm formation in static and dynamic biofilm models. qRT-PCR and protease activity assays demonstrated that under standard growth conditions, sepA is repressed by the global regulator SarA. Inactivation of sarA increased SepA production, and in turn augmented biofilm formation. Genetic and biochemical analyses demonstrated that SepA-related induction of biofilm accumulation resulted from enhanced Aap processing. Studies using recombinant proteins demonstrated that SepA is able to cleave the A domain of Aap at residue 335 and between the A and B domains at residue 601. This study identifies the mechanism behind Aap-mediated biofilm maturation, and also demonstrates a novel role for a secreted staphylococcal protease as a requirement for the development of a biofilm.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Metalloendopeptidases/metabolism , Protein Processing, Post-Translational , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/physiology , Adhesins, Bacterial/metabolism , Bacterial Adhesion , Polysaccharides, Bacterial/metabolism , Protein Binding , Staphylococcus epidermidis/chemistry , Staphylococcus epidermidis/genetics
13.
Biochem Biophys Res Commun ; 495(2): 2078-2084, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29242153

ABSTRACT

Polynucleotide phosphorylase catalyzes both 3'-5' exoribonuclease and polyadenylation reactions. The crystal structure of Staphylococcus epidermidis PNPase revealed a bound phosphate in the PH2 domain of each protomer coordinated by three adjacent serine residues. Mutational analysis suggests that phosphate coordination by these serine residues is essential to maintain the catalytic center in an active conformation. We note that PNPase forms a complex with RNase J1 and RNase J2 without substantially altering either exo-ribonuclease or polyadenylation activity of this enzyme. This decoupling of catalytic activity from protein-protein interactions suggests that association of these endo- or exo-ribonucleases with PNPase could be more relevant for cellular localization or concerted targeting of structured RNA for recycling.


Subject(s)
Molecular Docking Simulation , Nucleotidyltransferases/chemistry , Nucleotidyltransferases/ultrastructure , Ribonucleases/chemistry , Ribonucleases/ultrastructure , Staphylococcus epidermidis/enzymology , Binding Sites , Enzyme Activation , Enzyme Stability , Models, Chemical , Multienzyme Complexes , Protein Binding , Protein Conformation , Structure-Activity Relationship , Substrate Specificity
14.
BMC Oral Health ; 18(1): 89, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29776416

ABSTRACT

BACKGROUND: Urease is an enzyme produced by plaque bacteria hydrolysing urea from saliva and gingival exudate into ammonia in order to regulate the pH in the dental biofilm. The aim of this study was to assess the urease activity among oral bacterial species by using the rapid urease test (RUT) in a micro-plate format and to examine whether this test could be used for measuring the urease activity in site-specific supragingival dental plaque samples ex vivo. METHODS: The RUT test is based on 2% urea in peptone broth solution and with phenol red at pH 6.0. Oral bacterial species were tested for their urease activity using 100 µl of RUT test solution in the well of a micro-plate to which a 1 µl amount of cells collected after growth on blood agar plates or in broth, were added. The color change was determined after 15, 30 min, and 1 and 2 h. The reaction was graded in a 4-graded scale (none, weak, medium, strong). Ex vivo evaluation of dental plaque urease activity was tested in supragingival 1 µl plaque samples collected from 4 interproximal sites of front teeth and molars in 18 adult volunteers. The color reaction was read after 1 h in room temperature and scored as in the in vitro test. RESULTS: The strongest activity was registered for Staphylococcus epidermidis, Helicobacter pylori, Campylobacter ureolyticus and some strains of Haemophilus parainfluenzae, while known ureolytic species such as Streptococcus salivarius and Actinomyces naeslundii showed a weaker, variable and strain-dependent activity. Temperature had minor influence on the RUT reaction. The interproximal supragingival dental plaque between the lower central incisors (site 31/41) showed significantly higher scores compared to between the upper central incisors (site 11/21), between the upper left first molar and second premolar (site 26/25) and between the lower right second premolar and molar (site 45/46). CONCLUSION: The rapid urease test (RUT) in a micro-plate format can be used as a simple and rapid method to test urease activity in bacterial strains in vitro and as a chair-side method for testing urease activity in site-specific supragingival plaque samples ex vivo.


Subject(s)
Bacteria/enzymology , Bacteriological Techniques/methods , Dental Plaque/microbiology , Urease/analysis , Actinomyces/enzymology , Campylobacter/enzymology , Haemophilus parainfluenzae/enzymology , Helicobacter pylori/enzymology , Humans , Staphylococcus epidermidis/enzymology , Streptococcus salivarius/enzymology
15.
Article in English | MEDLINE | ID: mdl-28559263

ABSTRACT

Coagulase-negative staphylococci (CoNS) are the major causative agents of foreign-body-related infections, including catheter-related bloodstream infections. Because of the involvement of biofilms, foreign-body-related infections are difficult to treat. P128, a chimeric recombinant phage-derived ectolysin, has been shown to possess bactericidal activity on strains of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA). We tested the killing potential of P128 on three clinically significant species of CoNS, S. epidermidis, S. haemolyticus, and S. lugdunensis, under a variety of physiological conditions representing growing and nongrowing states. The MIC90 and minimum bactericidal concentration at which 90% of strains tested are killed (MBC90) of P128 on 62 clinical strains of CoNS were found to be 16 and 32 µg/ml (0.58 and 1.16 µM), respectively, demonstrating the bactericidal nature of P128 on CoNS strains. Serum showed a potentiating effect on P128 inhibition, as indicated by 4- to 32-fold lower MIC values observed in serum. P128 caused a rapid loss of viability in all CoNS strains tested. Persisters of CoNS that were enriched in the presence of vancomycin or daptomycin were killed by P128 at 1× the MIC in a rapid manner. Low concentrations of P128 caused a 2- to 5-log reduction in CFU in stationary-phase or poorly metabolizing CoNS cultures. P128 at low concentrations eliminated CoNS biofilms in microtiter plates and on the surface of catheters. Combinations of P128 and standard-of-care (SoC) antibiotics were highly synergistic in inhibiting growth in preformed biofilms. Potent activity on planktonic cells, persisters, and biofilms of CoNS suggests that P128 is a promising candidate for the clinical development of treatments for foreign-body-related and other CoNS infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Recombinant Fusion Proteins/pharmacology , Staphylococcal Infections/drug therapy , Staphylococcus epidermidis/drug effects , Staphylococcus haemolyticus/drug effects , Staphylococcus lugdunensis/drug effects , Catheter-Related Infections/drug therapy , Catheter-Related Infections/microbiology , Coagulase/metabolism , Daptomycin/pharmacology , Drug Synergism , Drug Therapy, Combination , Foreign-Body Reaction/drug therapy , Foreign-Body Reaction/microbiology , Humans , Microbial Sensitivity Tests , Staphylococcus epidermidis/enzymology , Staphylococcus haemolyticus/enzymology , Staphylococcus lugdunensis/enzymology , Vancomycin/pharmacology
16.
Microb Pathog ; 105: 264-271, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259672

ABSTRACT

The second messenger cyclic di-guanylate (c-di-GMP) plays an important role in controlling the switch between planktonic and biofilm lifestyles. The synthesis of c-di-GMP is catalyzed by di-guanylate cyclases (DGCs) and the enzymes are characterized by the presence of a conserved GGDEF domain. In the sequenced staphylococcal genomes, gdpS is the only gene encoding a GGDEF domain-containing protein. Previous studies have shown that gdpS contributes to staphylococcal biofilm formation, but its effect remains under debate. In the present study, we deleted gdpS in Staphylococcus epidermidis strain RP62A. Disruption of gdpS in this strain impaired biofilm formation under both static and dynamic flow conditions, suggesting that gdpS act as a positive regulator of biofilm development in this high-biofilm-forming isolate. The predicted translational start site of gdpS in S. epidermidis differs between the Refseq database and the Genbank database. By using site-directed mutagenesis and Western blot analysis, we determined GdpS is translated from the start codon annotated in the Refseq database. In addition, mutation in the GGDEF domain did not affect the ability of gdpS to complement the biofilm defect of the gdpS mutant. Heterologous di-guanylate cyclases expressed in trans failed to complement the gdpS mutant. These results confirmed that gdpS modulates staphylococcal biofilm independently of c-di-GMP signaling pathway. Furthermore, mutations of the start codon did not abolish the capacity of gdpS to enhance biofilm formation. Taken together, these findings indicated that the S. epidermidis gdpS regulates biofilm formation independently of its protein-coding function.


Subject(s)
Biofilms/growth & development , Cyclic GMP/analogs & derivatives , Staphylococcus epidermidis/physiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Blotting, Western/methods , Cyclic GMP/genetics , Cyclic GMP/metabolism , DNA, Bacterial/genetics , DNA, Bacterial/isolation & purification , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Genetic Complementation Test , Mutagenesis, Site-Directed/methods , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
17.
Antonie Van Leeuwenhoek ; 110(1): 133-143, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27757703

ABSTRACT

Staphylococcus aureus and Staphylococcus epidermidis are two of the most significant opportunistic human pathogens, causing medical implant and nosocomial infections worldwide. These bacteria contain surface proteins that play crucial roles in multiple biological processes. It has become apparent that they have evolved a number of unique mechanisms by which they can immobilise proteins on their surface. Notably, a conserved cell membrane-anchored enzyme, sortase A (SrtA), can catalyse the covalent attachment of precursor bacterial cell wall-attached proteins to peptidoglycan. Considering its indispensable role in anchoring substrates to the cell wall and its effects on virulence, SrtA has attracted great attention. In this study, a 549-bp gene was cloned from a pathogenic S. epidermidis strain, YC-1, which shared high identity with srtA from other Staphylococcus spp. A mutant strain, YC-1ΔsrtA, was then constructed by allelic exchange mutagenesis. The direct survival rate assay suggested that YC-1ΔsrtA had a lower survival capacity in healthy mice blood compare with the wild-type strain, indicating that the deletion of srtA affects the virulence and infectious capacity of S. epidermidis YC-1. YC-1ΔsrtA was then administered via intraperitoneal injection and it provided a relative percent survival value of 72.7 % in mice against S. aureus TC-1 challenge. These findings demonstrate the possbility that YC-1ΔsrtA might be used as a live attenuated vaccine to produce cross-protection against S. aureus.


Subject(s)
Aminoacyltransferases/immunology , Bacterial Proteins/immunology , Bacterial Vaccines/immunology , Cysteine Endopeptidases/immunology , Staphylococcal Infections/immunology , Staphylococcal Infections/prevention & control , Staphylococcus aureus/physiology , Staphylococcus epidermidis/enzymology , Aminoacyltransferases/administration & dosage , Aminoacyltransferases/genetics , Animals , Antibodies, Bacterial/immunology , Bacterial Proteins/administration & dosage , Bacterial Proteins/genetics , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/genetics , Cross Protection , Cysteine Endopeptidases/administration & dosage , Cysteine Endopeptidases/genetics , Humans , Male , Mice , Staphylococcal Infections/microbiology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/immunology
18.
Int J Mol Sci ; 18(11)2017 Nov 04.
Article in English | MEDLINE | ID: mdl-29113034

ABSTRACT

In the industrial processes, lipases are expected to operate at temperatures above 45 °C and could retain activity in organic solvents. Hence, a C-terminal truncated lipase from Staphylococcus epidermis AT2 (rT-M386) was engineered by directed evolution. A mutant with glycine-to-cysteine substitution (G210C) demonstrated a remarkable improvement of thermostability, whereby the mutation enhanced the activity five-fold when compared to the rT-M386 at 50 °C. The rT-M386 and G210C lipases were purified concurrently using GST-affinity chromatography. The biochemical and biophysical properties of both enzymes were investigated. The G210C lipase showed a higher optimum temperature (45 °C) and displayed a more prolonged half-life in the range of 40-60 °C as compared to rT-M386. Both lipases exhibited optimal activity and stability at pH 8. The G210C showed the highest stability in the presence of polar organic solvents at 50 °C compared to the rT-M386. Denatured protein analysis presented a significant change in the molecular ellipticity value above 60 °C, which verified the experimental result on the temperature and thermostability profile of G210C.


Subject(s)
Bacterial Proteins/metabolism , Evolution, Molecular , Lipase/metabolism , Protein Denaturation , Staphylococcus epidermidis/enzymology , Amino Acid Substitution , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Enzyme Stability , Lipase/chemistry , Lipase/genetics , Protein Domains , Staphylococcus epidermidis/genetics
19.
Proteins ; 84(9): 1328-32, 2016 09.
Article in English | MEDLINE | ID: mdl-27213893

ABSTRACT

The negatively charged bacterial polysaccharides-wall teichoic acids (WTAs) are synthesized intracellularly and exported by a two-component transporter, TagGH, comprising a transmembrane subunit TagG and an ATPase subunit TagH. We determined the crystal structure of the C-terminal domain of TagH (TagH-C) to investigate its function. The structure shows an N-terminal SH3-like subdomain wrapped by a C-terminal subdomain with an anti-parallel ß-sheet and an outer shell of α-helices. A stretch of positively charged surface across the subdomain interface is flanked by two negatively charged regions, suggesting a potential binding site for negatively charged polymers, such as WTAs or acidic peptide chains. Proteins 2016; 84:1328-1332. © 2016 Wiley Periodicals, Inc.


Subject(s)
ATP-Binding Cassette Transporters/chemistry , Bacterial Proteins/chemistry , Hydrolases/chemistry , Protein Subunits/chemistry , Staphylococcus epidermidis/chemistry , Teichoic Acids/chemistry , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Amino Acid Motifs , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Binding Sites , Biological Transport , Cell Wall/chemistry , Cloning, Molecular , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrolases/genetics , Hydrolases/metabolism , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Staphylococcus epidermidis/enzymology , Static Electricity , Teichoic Acids/metabolism
20.
Nature ; 465(7296): 346-9, 2010 May 20.
Article in English | MEDLINE | ID: mdl-20485435

ABSTRACT

Commensal bacteria are known to inhibit pathogen colonization; however, complex host-microbe and microbe-microbe interactions have made it difficult to gain a detailed understanding of the mechanisms involved in the inhibition of colonization. Here we show that the serine protease Esp secreted by a subset of Staphylococcus epidermidis, a commensal bacterium, inhibits biofilm formation and nasal colonization by Staphylococcus aureus, a human pathogen. Epidemiological studies have demonstrated that the presence of Esp-secreting S. epidermidis in the nasal cavities of human volunteers correlates with the absence of S. aureus. Purified Esp inhibits biofilm formation and destroys pre-existing S. aureus biofilms. Furthermore, Esp enhances the susceptibility of S. aureus in biofilms to immune system components. In vivo studies have shown that Esp-secreting S. epidermidis eliminates S. aureus nasal colonization. These findings indicate that Esp hinders S. aureus colonization in vivo through a novel mechanism of bacterial interference, which could lead to the development of novel therapeutics to prevent S. aureus colonization and infection.


Subject(s)
Bacterial Proteins/metabolism , Biofilms/growth & development , Nose/microbiology , Serine Proteases/metabolism , Staphylococcus aureus/growth & development , Staphylococcus epidermidis/enzymology , Staphylococcus epidermidis/physiology , Bacterial Proteins/chemistry , Bacterial Proteins/isolation & purification , Bacterial Proteins/pharmacology , Female , Humans , Male , Odds Ratio , Serine Proteases/chemistry , Serine Proteases/deficiency , Serine Proteases/isolation & purification , Staphylococcal Infections/microbiology , Staphylococcal Infections/prevention & control , Staphylococcal Infections/therapy , Staphylococcus aureus/immunology , Staphylococcus epidermidis/genetics , Superinfection/immunology , Superinfection/microbiology , Superinfection/prevention & control , Superinfection/therapy , Young Adult , beta-Defensins/immunology , beta-Defensins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL