Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 221
Filter
1.
FEMS Microbiol Rev ; 46(5)2022 10 28.
Article in English | MEDLINE | ID: mdl-35675307

ABSTRACT

Staphylococcus aureus is an important human and livestock pathogen that is well-protected against environmental insults by a thick cell wall. Accordingly, the wall is a major target of present-day antimicrobial therapy. Unfortunately, S. aureus has mastered the art of antimicrobial resistance, as underscored by the global spread of methicillin-resistant S. aureus (MRSA). The major cell wall component is peptidoglycan. Importantly, the peptidoglycan network is not only vital for cell wall function, but it also represents a bacterial Achilles' heel. In particular, this network is continuously opened by no less than 18 different peptidoglycan hydrolases (PGHs) encoded by the S. aureus core genome, which facilitate bacterial growth and division. This focuses attention on the specific functions executed by these enzymes, their subcellular localization, their control at the transcriptional and post-transcriptional levels, their contributions to staphylococcal virulence and their overall importance in bacterial homeostasis. As highlighted in the present review, our understanding of the different aspects of PGH function in S. aureus has been substantially increased over recent years. This is important because it opens up new possibilities to exploit PGHs as innovative targets for next-generation antimicrobials, passive or active immunization strategies, or even to engineer them into effective antimicrobial agents.


Subject(s)
Cell Wall , Methicillin-Resistant Staphylococcus aureus , N-Acetylmuramoyl-L-alanine Amidase , Anti-Bacterial Agents/therapeutic use , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , N-Acetylmuramoyl-L-alanine Amidase/genetics , Peptidoglycan , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics , Virulence
2.
Molecules ; 26(21)2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34771148

ABSTRACT

Methicillin-resistant Staphylococcus aureus (MRSA) is an important threat as it causes serious hospital and community acquired infections with deathly outcomes oftentimes, therefore, development of new treatments against this bacterium is a priority. Shikimate kinase, an enzyme in the shikimate pathway, is considered a good target for developing antimicrobial drugs; this is given because of its pathway, which is essential in bacteria whereas it is absent in mammals. In this work, a computer-assisted drug design strategy was used to report the first potentials inhibitors for Shikimate kinase from methicillin-resistant Staphylococcus aureus (SaSK), employing approximately 5 million compounds from ZINC15 database. Diverse filtering criteria, related to druglike characteristics and virtual docking screening in the shikimate binding site, were performed to select structurally diverse potential inhibitors from SaSK. Molecular dynamics simulations were performed to elucidate the dynamic behavior of each SaSK-ligand complex. The potential inhibitors formed important interactions with residues that are crucial for enzyme catalysis, such as Asp37, Arg61, Gly82, and Arg138. Therefore, the compounds reported provide valuable information and can be seen as the first step toward developing SaSK inhibitors in the search of new drugs against MRSA.


Subject(s)
Anti-Bacterial Agents/pharmacology , Computer-Aided Design , Enzyme Inhibitors/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Anti-Bacterial Agents/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Methicillin-Resistant Staphylococcus aureus/enzymology , Models, Molecular , Molecular Structure , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Small Molecule Libraries/chemistry
3.
Sci Rep ; 11(1): 21726, 2021 11 05.
Article in English | MEDLINE | ID: mdl-34741111

ABSTRACT

Cryptotanshinone (CT) is an extract from the traditional Chinese medicine Salvia miltiorrhiza, which inhibits the growth of methicillin-resistant Staphylococcus aureus (MRSA) in vitro. This study aims to determine the antibacterial mechanisms of CT by integrating bioinformatics analysis and microbiology assay. The microarray data of GSE13203 was retrieved from the Gene Expression Omnibus (GEO) database to screen the differentially expressed genes (DEGs) of S. aureus strains that were treated with CT treatment. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were used to identify the potential target of CT. Data mining on the microarray dataset indicated that pyruvate kinase (PK) might be involved in the antimicrobial activities of CT. The minimum inhibition concentrations (MICs) of CT or vancomycin against the MRSA strain ATCC43300 and seven other clinical strains were determined using the broth dilution method. The effects of CT on the activity of PK were further measured. In vitro tests verified that CT inhibited the growth of an MRSA reference strain and seven other clinical strains. CT hampered the activity of the PK of ATCC43300 and five clinical MRSA strains. CT might hinder bacterial energy metabolism by inhibiting the activity of PK.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/drug effects , Phenanthrenes/pharmacology , Pyruvate Kinase/antagonists & inhibitors , Computational Biology , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Gene Expression Profiling , Humans , Methicillin-Resistant Staphylococcus aureus/enzymology , Phenanthrenes/therapeutic use , Phytotherapy , Staphylococcal Infections/drug therapy
4.
Curr Top Med Chem ; 21(27): 2474-2482, 2021.
Article in English | MEDLINE | ID: mdl-34645377

ABSTRACT

BACKGROUND: Methicillin-resistant S. aureus (MRSA) has already tormented humanity and the environment for a long time and is responsible for many difficult-to-treat infections. Unfortunately, there are limited therapeutic options, and MRSA isolates with complete resistance to vancomycin, the first-line drug for the treatment of MRSA infections, have already emerged in recent years. Moxifloxacin retained activity against mutant bacterial strains with various levels of fluoroquinolones resistance and had a lower potential to select for resistant mutants. Isatin is a versatile structure, and its derivatives are potent inhibitors of many enzymes and receptors. The fluoroquinolone- isatin derivatives demonstrated excellent antibacterial activity against both drug-sensitive and drug-resistant organisms. The structure-activity relationship elucidated that incorporation of 1,2,3-triazole moiety into the C-7 position of fluoroquinolone skeleton was favorable to the antibacterial activity. Accordingly, fluoroquinolone derivatives with isatin and 1,2,3-triazole fragments at the side chain on the C-7 position are promising candidates to fight against drug-resistant bacteria. OBJECTIVE: To explore more active moxifloxacin derivatives to fight against MRSA and enrich the structure-activity relationships. METHODS: The synthesized moxifloxacin derivatives 7a-i and 14a-f were evaluated for their antibacterial activity against a panel of MRSA strains by means of standard two-fold serial dilution method. RESULTS: The majority of the synthesized moxifloxacin derivatives were active against most of the tested MRSA strains with MIC values in a range of 1 to 64 µg/mL. The mechanistic investigations revealed that topoisomerase IV was one of the targets for antibacterial activity. CONCLUSION: These derivatives are useful scaffolds for the development of novel topoisomerase IV inhibitors.


Subject(s)
Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , DNA Topoisomerase IV/antagonists & inhibitors , Methicillin-Resistant Staphylococcus aureus/drug effects , Moxifloxacin/analogs & derivatives , Moxifloxacin/pharmacology , Fluoroquinolones/chemistry , Fluoroquinolones/pharmacology , Isatin/analogs & derivatives , Isatin/pharmacology , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Triazoles/chemistry , Triazoles/pharmacology
5.
J Med Chem ; 64(18): 13540-13550, 2021 09 23.
Article in English | MEDLINE | ID: mdl-34473495

ABSTRACT

The polyprenyl lipid undecaprenyl phosphate (C55P) is the universal carrier lipid for the biosynthesis of bacterial cell wall polymers. C55P is synthesized in its pyrophosphate form by undecaprenyl pyrophosphate synthase (UppS), an essential cis-prenyltransferase that is an attractive target for antibiotic development. We previously identified a compound (MAC-0547630) that showed promise as a novel class of inhibitor and an ability to potentiate ß-lactam antibiotics. Here, we provide a structural model for MAC-0547630's inhibition of UppS and a structural rationale for its enhanced effect on UppS from Bacillus subtilis versus Staphylococcus aureus. We also describe the synthesis of a MAC-0547630 derivative (JPD447), show that it too can potentiate ß-lactam antibiotics, and provide a structural rationale for its improved potentiation. Finally, we present an improved structural model of clomiphene's inhibition of UppS. Taken together, our data provide a foundation for structure-guided drug design of more potent UppS inhibitors in the future.


Subject(s)
Alkyl and Aryl Transferases/metabolism , Bacterial Proteins/metabolism , Enzyme Inhibitors/metabolism , Alkyl and Aryl Transferases/chemistry , Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Catalytic Domain , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Molecular Structure , Protein Binding , Structure-Activity Relationship
6.
Vet Microbiol ; 259: 109160, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34197979

ABSTRACT

High-level mupirocin resistance (HLMR) is determined by the plasmid-located ileS2 gene flanked by two copies of the insertion sequence 257 (IS257). The molecular epidemiology of high-level mupirocin-resistant isolates could be assessed by the determination of their IS257-ileS2 spacer regions conformation. In this study, 188 isolates of methicillin-resistant staphylococci were subjected to the detection of HLMR, and analysis of the conformation of the IS257-ileS2 spacer regions. Mupirocin resistance was detected in five (2,6%) isolates, among which two were recognized as Staphylococcus pseudintermedius, two as Staphylococcus haemolyticus, and one as Staphylococcus aureus. High-level mupirocin resistance was revealed by the agar disk diffusion method, and MIC values, and was confirmed by the detection of the ileS2 gene. The conformations of the IS257-ileS2 spacer regions were homologous in two S. haemolyticus strains tested. The remaining three isolates showed diverse IS257-ileS2 conformations. The results of this study indicate that HLMR occasionally occurs in staphylococci isolated from companion animals. The heterogeneity and the homogeneity of the IS257-ileS2 spacer regions confirm that the ileS2 gene spread among staphylococci of animal origin by the transfer of different as well as the same plasmids. Surveillance of the occurrence of mupirocin resistance and molecular characterization of resistant isolates are strongly recommended due to the possibility of plasmid-located resistance gene transfer between staphylococci.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Mupirocin/pharmacology , Pets/microbiology , Staphylococcal Infections/veterinary , Animals , Cats/microbiology , Coagulase/biosynthesis , DNA Transposable Elements , Dogs/microbiology , Genes, Bacterial , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Microbial Sensitivity Tests , Plasmids/genetics , Staphylococcal Infections/microbiology , Staphylococcus/classification , Staphylococcus/drug effects , Staphylococcus/genetics
7.
mBio ; 12(3): e0053021, 2021 06 29.
Article in English | MEDLINE | ID: mdl-34182779

ABSTRACT

Penicillin binding protein 2a (PBP2a)-dependent resistance to ß-lactam antibiotics in methicillin-resistant Staphylococcus aureus (MRSA) is regulated by the activity of the tricarboxylic acid (TCA) cycle via a poorly understood mechanism. We report that mutations in sucC and sucD, but not other TCA cycle enzymes, negatively impact ß-lactam resistance without changing PBP2a expression. Increased intracellular levels of succinyl coenzyme A (succinyl-CoA) in the sucC mutant significantly perturbed lysine succinylation in the MRSA proteome. Suppressor mutations in sucA or sucB, responsible for succinyl-CoA biosynthesis, reversed sucC mutant phenotypes. The major autolysin (Atl) was the most succinylated protein in the proteome, and increased Atl succinylation in the sucC mutant was associated with loss of autolytic activity. Although PBP2a and PBP2 were also among the most succinylated proteins in the MRSA proteome, peptidoglycan architecture and cross-linking were unchanged in the sucC mutant. These data reveal that perturbation of the MRSA succinylome impacts two interconnected cell wall phenotypes, leading to repression of autolytic activity and increased susceptibility to ß-lactam antibiotics. IMPORTANCEmecA-dependent methicillin resistance in MRSA is subject to regulation by numerous accessory factors involved in cell wall biosynthesis, nucleotide signaling, and central metabolism. Here, we report that mutations in the TCA cycle gene, sucC, increased susceptibility to ß-lactam antibiotics and was accompanied by significant accumulation of succinyl-CoA, which in turn perturbed lysine succinylation in the proteome. Although cell wall structure and cross-linking were unchanged, significantly increased succinylation of the major autolysin Atl, which was the most succinylated protein in the proteome, was accompanied by near complete repression of autolytic activity. These findings link central metabolism and levels of succinyl-CoA to the regulation of ß-lactam antibiotic resistance in MRSA through succinylome-mediated control of two interlinked cell wall phenotypes. Drug-mediated interference of the SucCD-controlled succinylome may help overcome ß-lactam resistance.


Subject(s)
Acyl Coenzyme A/genetics , Acyl Coenzyme A/metabolism , Anti-Bacterial Agents/pharmacology , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , beta-Lactams/pharmacology , Acyl Coenzyme A/analysis , Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Microbial Sensitivity Tests , Mutation , Proteome , beta-Lactam Resistance
8.
BMC Microbiol ; 21(1): 192, 2021 06 25.
Article in English | MEDLINE | ID: mdl-34172010

ABSTRACT

BACKGROUND: Methicillin-resistant Staphylococcus aureus (MRSA)-bloodstream infections (BSI) are predominantly seen in the hospital or healthcare-associated host. Nevertheless, the interactions of virulence factor (VFs) regulators and ß-lactam resistance in MRSA-BSI are unclear. This study aims to characterize the molecular relationship of two-component systems of VFs and the expression of the ß-lactamase gene in MRSA-BSI isolates. In this study, 639 samples were collected from BSI and identified by phenotypic methods. We performed extensive molecular characterization, including SCCmec type, agr type, VFs gene profiles determinations, and MLST on isolates. Also, a quantitative real-time PCR (q-RT PCR) assay was developed for identifying the gene expressions. RESULTS: Ninety-one (91) S. aureus and 61 MRSA (67.0%) strains were detected in BSI samples. The presence of VFs and SCCmec genes in MRSA isolates were as follows: tst (31.4%), etA (18.0%), etB (8.19%), lukS-PVL (31.4%), lukF-PV (18.0%), lukE-lukD (16.3%), edin (3.2%), hla (16.3%), hlb (18.0%), hld (14.7%), hlg (22.9%), SCCmecI (16.3%), SCCmecII (22.9%), SCCmecIII (36.0%), SCCmecIV (21.3%), and SCCmecV (16.3%). Quantitative real-time PCR showed overexpression of mecRI and mecI in the toxigenic isolates. Moreover, RNAIII and sarA genes were the highest expressions of MRSA strains. The multi-locus sequence typing data confirmed a high prevalence of CC5, CC8, and CC30. However, ST30, ST22, and ST5 were the most prevalent in the resistant and toxigenic strains. CONCLUSION: We demonstrated that although regulation of ß-lactamase gene expressions is a significant contributor to resistance development, two-component systems also influence antibiotic resistance development in MRSA-BSI isolates. This indicates that resistant strains might have pathogenic potential. We also confirmed that some MLST types are more successful colonizers with a potential for MRSA-BSI.


Subject(s)
Gene Expression Regulation, Bacterial , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Virulence/genetics , beta-Lactamases/genetics , Drug Resistance, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/enzymology , Virulence Factors/genetics
9.
J Enzyme Inhib Med Chem ; 36(1): 1088-1092, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34056990

ABSTRACT

Pathogenic bacteria resistant to most antibiotics, including the methicillin-resistant Staphylococcus aureus (MRSA) represent a serious medical problem. The search for new antiinfectives, possessing a diverse mechanism of action compared to the clinically used antibiotics, has become an attractive research field. S. aureus DNA encodes a ß-class carbonic anhydrase, SauBCA. It is a druggable target that can be inhibited by certain aromatic and heterocyclic sulphonamides. Here we investigated inorganic anions and some other small molecules for their inhibition of SauBCA. The halides, nitrite, nitrate, bicarbonate, carbonate, bisulphite, sulphate, stannate, and N,N-diethyldithiocarbamate were submillimolar SauBCA inhibitors with KIs in the range of 0.26 - 0.91 mM. The most effective inhibitors were sulfamide, sulfamate, phenylboronic acid, and phenylarsonic acid with KIs of 7 - 43 µM. Several interesting inhibitors detected here may be considered lead compounds for the development of even more effective derivatives, which should be investigated for their bacteriostatic effects.


Subject(s)
Anti-Bacterial Agents/pharmacology , Carbonic Anhydrase Inhibitors/pharmacology , Carbonic Anhydrases/metabolism , Methicillin-Resistant Staphylococcus aureus/drug effects , Small Molecule Libraries/pharmacology , Anions/chemical synthesis , Anions/chemistry , Anions/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Carbonic Anhydrase Inhibitors/chemical synthesis , Carbonic Anhydrase Inhibitors/chemistry , Dose-Response Relationship, Drug , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Molecular Structure , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Structure-Activity Relationship
10.
Drug Discov Today ; 26(9): 2190-2197, 2021 09.
Article in English | MEDLINE | ID: mdl-34048895

ABSTRACT

The Clp protease is an AAA+ protease that executes abnormally folded or malfunctioning proteins, and has an important role in producing virulence factors, forming biofilms or persisters and developing methicillin-resistant Staphylococcus aureus (MRSA). Recent studies showed that Clp protease controls virulence via agr signaling and degrades antitoxins of the toxin-antitoxin system to modulate the formation of persisters and biofilms. In this review, we focus on recent developments concerning the virulence and persistence regulatory pathways and resistance-related mechanism of Clp protease in S. aureus, with an overview of the Clp modulators developed to treat MRSA infection.


Subject(s)
Drug Resistance, Bacterial , Endopeptidase Clp/metabolism , Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Animals , Anti-Bacterial Agents/therapeutic use , Biofilms , Humans , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Methicillin-Resistant Staphylococcus aureus/physiology , Staphylococcal Infections/drug therapy , Virulence
11.
J Am Chem Soc ; 143(3): 1278-1283, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33428381

ABSTRACT

Deferasirox, ExJade, is an FDA-approved iron chelator used for the treatment of iron overload. In this work, we report several fluorescent deferasirox derivatives that display unique photophysical properties, i.e., aggregation-induced emission (AIE), excited state intramolecular proton transfer, charge transfer, and through-bond and through-space conjugation characteristics in aqueous media. Functionalization of the phenol units on the deferasirox scaffold afforded the fluorescent responsive pro-chelator ExPhos, which enabled the detection of the disease-based biomarker alkaline phosphatase (ALP). The diagnostic potential of these deferasirox derivatives was supported by bacterial biofilm studies.


Subject(s)
Deferasirox/analogs & derivatives , Fluorescent Dyes/chemistry , Alkaline Phosphatase/analysis , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/analysis , Biofilms/drug effects , Biomarkers/analysis , Cefoperazone/pharmacology , Deferasirox/pharmacology , Deferasirox/radiation effects , Fluorescent Dyes/pharmacology , Fluorescent Dyes/radiation effects , Light , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/physiology , Microbial Sensitivity Tests , Microscopy, Confocal , Microscopy, Fluorescence , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/enzymology , Pseudomonas aeruginosa/physiology , Sulbactam/pharmacology
12.
J Enzyme Inhib Med Chem ; 36(1): 295-306, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33404277

ABSTRACT

Five series of novel carbazole derivatives containing an aminoguanidine, dihydrotriazine, thiosemicarbazide, semicarbazide or isonicotinic moiety were designed, synthesised and evaluated for their antimicrobial activities. Most of the compounds exhibited potent inhibitory activities towards different bacterial strains (including one multidrug-resistant clinical isolate) and one fungal strain with minimum inhibitory concentrations (MICs) between 0.5 and 16 µg/ml. Compounds 8f and 9d showed the most potent inhibitory activities (MICs of 0.5-2 µg/ml). Furthermore, compounds 8b, 8d, 8f, 8k, 9b and 9e with antimicrobial activities were not cytotoxic to human gastric cancer cell lines (SGC-7901 and AGS) or a normal human liver cell line (L-02). Structure-activity relationship analyses and docking studies implicated the dihydrotriazine group in increasing the antimicrobial potency and reducing the toxicity of the carbazole compounds. In vitro enzyme activity assays suggested that compound 8f binding to dihydrofolate reductase might account for the antimicrobial effect.


Subject(s)
Anti-Infective Agents/chemical synthesis , Bacterial Proteins/chemistry , Carbazoles/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Escherichia coli/drug effects , Tetrahydrofolate Dehydrogenase/chemistry , Anti-Infective Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Binding Sites , Candida albicans/drug effects , Candida albicans/enzymology , Candida albicans/growth & development , Carbazoles/pharmacology , Cell Line , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Epithelial Cells/drug effects , Escherichia coli/enzymology , Escherichia coli/growth & development , Guanidines/chemistry , Hepatocytes/drug effects , Humans , Inhibitory Concentration 50 , Isonicotinic Acids/chemistry , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/growth & development , Microbial Sensitivity Tests , Molecular Docking Simulation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Semicarbazides/chemistry , Staphylococcus aureus/drug effects , Staphylococcus aureus/enzymology , Staphylococcus aureus/growth & development , Streptococcus mutans/drug effects , Streptococcus mutans/enzymology , Streptococcus mutans/growth & development , Structure-Activity Relationship , Tetrahydrofolate Dehydrogenase/metabolism , Triazines/chemistry
13.
Int Urol Nephrol ; 53(2): 373-380, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32804344

ABSTRACT

PURPOSE: Peritonitis is a serious complication of peritoneal dialysis and coagulase-negative Staphylococcus (CNS) is the most frequent cause of peritoneal dialysis (PD)-infections in many centers. This study aimed to investigate the molecular epidemiology of CNS isolated from PD-peritonitis in a Brazilian single center, focusing on the genetic determinants conferring methicillin resistance. METHODS: Bacterial strains were isolated from peritoneal fluid of patients presenting PD-peritonitis, identified by phenotypic and molecular methods, and those identified as CNS were submitted to mecA detection, SCCmec, pulsed-field gel electrophoresis (PFGE) and multilocus sequence typing (MLST). RESULTS: Over the 18-year period of this study (1995-2011), a total of 878 peritonitis episodes were diagnosed in this unit, 115 were caused by coagulase-negative staphylococci of which 72 by Staphylococcus epidermidis. mecA gene was detected in 55 CNS (47.8%), more frequently on the more recent years. SCCmec type III was the most frequent cassette, followed by SCCmec type IV and SCCmec type II. A diverstity of pulsotypes was observed among the S. epidermidis isolates, but five clusters (based on the 80% cutoff) were identified. Diversified sequence types (ST02, ST05, ST06, ST09, ST23, ST59 and ST371) were detected. CONCLUSIONS: Detection of SCCmec type III among coagulase-negative Staphylococcus underscores the role of hospital environments as potential source of methicillin-resistant Staphylococcus causing peritonitis in PD patients.


Subject(s)
Methicillin-Resistant Staphylococcus aureus/genetics , Peritoneal Dialysis/adverse effects , Peritonitis/microbiology , Staphylococcal Infections/diagnosis , Staphylococcal Infections/epidemiology , Staphylococcus epidermidis/genetics , Coagulase , Humans , Incidence , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Molecular Diagnostic Techniques/methods , Molecular Epidemiology/methods , Retrospective Studies , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/isolation & purification
14.
ACS Appl Mater Interfaces ; 13(1): 257-265, 2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33378174

ABSTRACT

Controllable drug release is promising for fighting against antimicrobial resistance, which is a critical threat to human health worldwide. Herein, new hyaluronidase-responsive conjugated oligo(thiophene ethynylene) (OTE)-covalently modified hyaluronic acid (OTE-HA) nanoparticles for on-demand release of antimicrobial agents are reported. The synthesis of amphiphilic OTE-HA was carried out by esterification reaction. The resulting macromolecules were self-assembled in water to form nanoparticles, in which the hydrophobic OTE section, as bactericides, formed "cores" and the hydrophilic hyaluronic acid (HA) formed "shells". The OTE-HA nanoparticles avoid bactericide premature leakage and effectively block the dark cytotoxicity of the OTE section, possessing excellent biocompatibility. Using methicillin-resistant Staphylococcus aureus (MRSA) as an example, hyaluronidase, largely secreted by MRSA, can in situ trigger the release of OTE via hydrolyzing OTE-HA nanoparticles into fragments, even disaccharides linked with OTE. Importantly, the OTE section could effectively break cell membranes, leading to bacterial death. The half-maximal inhibitory concentration of the nanoparticles against MRSA is 3.3 µg/mL. The great antibacterial activity of OTE-HA nanoparticles against Gram-positive bacteria Streptococcus pneumoniae further confirms the controllable bactericide delivery mechanism. OTE-HA nanoparticles coated on a surface can also effectively inhibit the growth of bacteria, which holds a remarkable promise in biomedical applications. Therefore, this work provides a favorable strategy of on-demand and in situ drug release for sterilization and defeating antimicrobial resistance.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Nanoparticles/chemistry , Quaternary Ammonium Compounds/pharmacology , Thiophenes/pharmacology , A549 Cells , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Drug Carriers/chemical synthesis , Drug Carriers/toxicity , Drug Design , Drug Liberation , Drug Resistance, Bacterial/drug effects , Humans , Hyaluronic Acid/chemical synthesis , Hyaluronic Acid/toxicity , Hyaluronoglucosaminidase/metabolism , Hydrolysis , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Nanoparticles/toxicity , Quaternary Ammonium Compounds/chemical synthesis , Quaternary Ammonium Compounds/metabolism , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Thiophenes/chemical synthesis , Thiophenes/metabolism
15.
Int J Mol Sci ; 21(23)2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33297331

ABSTRACT

The treatment of Staphylococcus aureus infections is impeded by the prevalence of MRSA and the formation of persisters and biofilms. Previously, we identified two celecoxib derivatives, Cpd36 and Cpd46, to eradicate MRSA and other staphylococci. Through whole-genome resequencing, we obtained several lines of evidence that these compounds might act by targeting the membrane protein translocase YidC2. Our data showed that ectopic expression of YidC2 in S. aureus decreased the bacterial susceptibility to Cpd36 and Cpd46, and that the YidC2-mediated tolerance to environmental stresses was suppressed by both compounds. Moreover, the membrane translocation of ATP synthase subunit c, a substrate of YidC2, was blocked by Cpd46, leading to a reduction in bacterial ATP production. Furthermore, we found that the thermal stability of bacterial YidC2 was enhanced, and introducing point mutations into the substrate-interacting cavity of YidC2 had a dramatic effect on Cpd36 binding via surface plasmon resonance assays. Finally, we demonstrated that these YidC2 inhibitors could effectively eradicate MRSA persisters and biofilms. Our findings highlight the potential of impeding YidC2-mediated translocation of membrane proteins as a new strategy for the treatment of bacterial infections.


Subject(s)
Anti-Bacterial Agents/pharmacology , Biofilms/drug effects , Celecoxib/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Cyclooxygenase 2 Inhibitors/chemistry , Cyclooxygenase 2 Inhibitors/pharmacology , Enzyme Stability , Membrane Transport Proteins/chemistry , Membrane Transport Proteins/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Protein Binding
16.
Int J Biol Macromol ; 165(Pt B): 2349-2362, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33098904

ABSTRACT

NAD synthetase (NadE) catalyzes the last step in NAD biosynthesis, transforming deamido-NAD+ into NAD+ by a two-step reaction with co-substrates ATP and amide donor ammonia. In this study, we report the crystal structure of Staphylococcus aureus NAD synthetase enzyme (saNadE) at 2.3 Å resolution. We used this structure to perform molecular dynamics simulations of apo-enzyme, enzyme-substrate (NadE with ATP and NaAD) and enzyme-intermediate complexes (NadE with NaAD-AMP) to investigate key binding interactions and explore the conformational transitions and flexibility of the binding pocket. Our results show large shift of N-terminal region in substrate bound form which is important for ATP binding. Substrates drive the correlated movement of loop regions surrounding it as well as some regions distal to the active site and stabilize them at complex state. Principal component analysis of atomic projections distinguish feasible trajectories to delineate distinct motions in enzyme-substrate to enzyme-intermediate states. Our results suggest mixed binding involving dominant induced fit and conformational selection. MD simulation extracted ensembles of NadE could potentially be utilized for in silico screening and structure based design of more effective Methicillin Resistant Staphylococcus aureus (MRSA) inhibitors.


Subject(s)
Amide Synthases/chemistry , Crystallography, X-Ray , Methicillin-Resistant Staphylococcus aureus/enzymology , Molecular Dynamics Simulation , Apoenzymes/chemistry , Catalytic Domain , Enzyme Stability , Humans , Hydrogen Bonding , NAD/biosynthesis , Principal Component Analysis , Protein Conformation , Protein Subunits/chemistry , Substrate Specificity
17.
Bioorg Med Chem ; 28(22): 115776, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33032189

ABSTRACT

The global increase in multidrug-resistant pathogens has caused severe problems in the treatment of infections. To overcome these difficulties, the advent of a new chemical class of antibacterial drug is eagerly desired. We aimed at creating novel antibacterial agents against bacterial type II topoisomerases, which are well-validated targets. TP0480066 (compound 32) has been identified by using structure-based optimization originated from lead compound 1, which was obtained as a result of our previous lead identification studies. The MIC90 values of TP0480066 against methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococci (VRE), and genotype penicillin-resistant Streptococcus pneumoniae (gPRSP) were 0.25, 0.015, and 0.06 µg/mL, respectively. Hence, TP0480066 can be regarded as a promising antibacterial drug candidate of this chemical class.


Subject(s)
Anti-Bacterial Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , Quinolines/pharmacology , Topoisomerase II Inhibitors/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Resistance, Bacterial/drug effects , Hep G2 Cells , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/enzymology , Microbial Sensitivity Tests , Models, Molecular , Molecular Structure , Quinolines/chemical synthesis , Quinolines/chemistry , Streptococcus pneumoniae/drug effects , Streptococcus pneumoniae/enzymology , Structure-Activity Relationship , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry , Transcriptional Regulator ERG/antagonists & inhibitors , Transcriptional Regulator ERG/metabolism , Vancomycin-Resistant Enterococci/drug effects , Vancomycin-Resistant Enterococci/enzymology
18.
mBio ; 11(5)2020 09 22.
Article in English | MEDLINE | ID: mdl-32963004

ABSTRACT

Staphylococcus aureus is a human pathogen causing life-threatening diseases. The increasing prevalence of multidrug-resistant S. aureus infections is a global health concern, requiring development of novel therapeutic options. Peptidoglycan-degrading enzymes (peptidoglycan hydrolases, PGHs) have emerged as a highly effective class of antimicrobial proteins against S. aureus and other pathogens. When applied to Gram-positive bacteria, PGHs hydrolyze bonds within the peptidoglycan layer, leading to rapid bacterial death by lysis. This activity is highly specific and independent of the metabolic activity of the cell or its antibiotic resistance patterns. However, systemic application of PGHs is limited by their often low activity in vivo and by an insufficient serum circulation half-life. To address this problem, we aimed to extend the half-life of PGHs selected for high activity against S. aureus in human serum. Half-life extension and increased serum circulation were achieved through fusion of PGHs to an albumin-binding domain (ABD), resulting in high-affinity recruitment of human serum albumin and formation of large protein complexes. Importantly, the ABD-fused PGHs maintained high killing activity against multiple drug-resistant S. aureus strains, as determined by ex vivo testing in human blood. The top candidate, termed ABD_M23, was tested in vivo to treat S. aureus-induced murine bacteremia. Our findings demonstrate a significantly higher efficacy of ABD_M23 than of the parental M23 enzyme. We conclude that fusion with ABD represents a powerful approach for half-life extension of PGHs, expanding the therapeutic potential of these enzybiotics for treatment of multidrug-resistant bacterial infections.IMPORTANCE Life-threatening infections with Staphylococcus aureus are often difficult to treat due to the increasing prevalence of antibiotic-resistant bacteria and their ability to persist in protected niches in the body. Bacteriolytic enzymes are promising new antimicrobials because they rapidly kill bacteria, including drug-resistant and persisting cells, by destroying their cell wall. However, when injected into the bloodstream, these enzymes are not retained long enough to clear an infection. Here, we describe a modification to increase blood circulation time of the enzymes and enhance treatment efficacy against S. aureus-induced bloodstream infections. This was achieved by preselecting enzyme candidates for high activity in human blood and coupling them to serum albumin, thereby preventing their elimination by kidney filtration and blood vessel cells.


Subject(s)
Bacteremia/drug therapy , N-Acetylmuramoyl-L-alanine Amidase/therapeutic use , Staphylococcal Infections/drug therapy , Staphylococcus aureus/enzymology , Adult , Animals , Female , Humans , Male , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mice, Inbred C57BL , N-Acetylmuramoyl-L-alanine Amidase/genetics , Peptidoglycan/metabolism , Serum Albumin/genetics , Serum Albumin/metabolism , Staphylococcus aureus/genetics
19.
ACS Appl Mater Interfaces ; 12(41): 45859-45872, 2020 Oct 14.
Article in English | MEDLINE | ID: mdl-32967419

ABSTRACT

Bacteria responsive color-changing wound dressings offer a valuable platform for continuous monitoring of the wound bed facilitating early detection of bacterial infections. In this study, we present a highly sensitive electrospun nanofibrous polyurethane wound dressing incorporating a hemicyanine-based chromogenic probe with a labile ester linkage that can be enzymatically cleaved by bacterial lipase released from clinically relevant strains, such as Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA). A rapid chromogenic response was achieved by localizing the dye at the surface of core-shell fibers, resulting in a 5x faster response relative to conventional nanofibers. By incorporating polyvinylpyrrolidone (PVP) dopant in the shell, the sensitivity was boosted to enable detection of bacteria at clinically relevant concentrations after 2 h exposure: 2.5 × 105 CFU/cm2 P. aeruginosa and 1.0 × 106 CFU/cm2 MRSA. Introduction of PVP in the shell also boosted the degree of hydrolysis of the chromogenic probe by a factor of 1.2× after a 3 h exposure to a low concentration of P. aeruginosa (105 CFU/cm2). PVP was also found to improve the discernibility of the color change at high bacterial concentrations. The co-operativity between the chromogenic probe, fiber structure, and polymer composition is well-suited for timely in situ detection of wound infection.


Subject(s)
Bacterial Infections/diagnosis , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Nanofibers/chemistry , Polyurethanes/chemistry , Povidone/chemistry , Pseudomonas aeruginosa/isolation & purification , Colorimetry , Lipase/metabolism , Methicillin-Resistant Staphylococcus aureus/enzymology , Molecular Structure , Particle Size , Polyurethanes/metabolism , Povidone/metabolism , Pseudomonas aeruginosa/enzymology , Surface Properties
20.
Discov Med ; 29(156): 65-77, 2020.
Article in English | MEDLINE | ID: mdl-32598864

ABSTRACT

Staphylococcus aureus can cause both acute and recurrent persistent infections such as peritonitis, endocarditis, abscesses, osteomyelitis, and chronic wound infections. Effective therapies to treat persistent disease are paramount. However, the mechanisms of S. aureus persistence are poorly understood. In this study, we performed a comprehensive and unbiased high-throughput mutant screen against a transposon-insertion mutant library of S. aureus USA300 and focused on the role of argJ encoding an acetyltransferase in the arginine biosynthesis pathway, whose transposon insertion caused a significant defect in persister formation using multiple drugs and stresses. Genetic complementation and arginine supplementation restored persistence in the argJ transposon insertion mutant while generation of mutations on the active site of the ArgJ protein caused a defect in persistence. Quantitative RT-PCR analysis showed that the genes encoded in the arg operon were over-expressed under drug stressed conditions and in stationary phase cultures. In addition, the argJ mutant had attenuated virulence in both mouse and C. elegans. Our studies identify a new mechanism of persistence mediated by arginine metabolism in S. aureus. These findings provide not only novel insights about the mechanisms of S. aureus persistence but also offer novel therapeutic targets that may help to develop more effective treatment of persistent S. aureus infections.


Subject(s)
Acetyltransferases/genetics , Bacterial Proteins/genetics , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Staphylococcal Infections/microbiology , Virulence Factors/genetics , Acetyltransferases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Arginine/biosynthesis , Bacterial Proteins/metabolism , Biosynthetic Pathways/genetics , Caenorhabditis elegans , DNA Transposable Elements/genetics , Disease Models, Animal , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Female , Gene Library , Genes, Bacterial/drug effects , High-Throughput Screening Assays , Humans , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Mutation/drug effects , Virulence/drug effects , Virulence/genetics , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...