Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 839
Filter
1.
BMC Med Genomics ; 17(1): 174, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951840

ABSTRACT

BACKGROUND: This study investigates the distribution and characteristics of linezolid and vancomycin susceptibilities among Enterococcus faecalis (E. faecalis) and Enterococcus faecium (E. faecium) and explores the underlying resistance mechanisms. METHODS: A total of 2842 Enterococcus clinical isolates from patients were retrospectively collected, and their clinical data were further analyzed. The minimum inhibitory concentrations (MICs) of vancomycin and linezolid were validated by broth dilution method. The resistance genes optrA, cfr, vanA, vanB and vanM were investigated using polymerase chain reaction (PCR). Housekeeping genes and resistance genes were obtianed through whole-genome sequencing (WGS). RESULTS: Of the 2842 Enterococcus isolates, 88.5% (2516) originated from urine, with E. faecium accounted for 60.1% of these. The vanA gene was identified in 27/28 vancomycin resistant Enterococcus (VRE) isolates, 4 of which carried both vanA and vanM genes. The remaining strain was vanM positive. The optrA gene was identified in all E. faecalis isolates among linezolid resistant Enterococcus (LRE). E. faecium showed a higher multiple antibiotic resistance index (MAR index) compared to E. faecalis. The multi-locus sequence typing (MLST) showed the sequence type of E. faecium mainly belongs to clonal complex (CC) 17, nearly E. faecalis isolates analyzed were differentiated into 7 characteristics of sequence types (STs), among which ST16 of CC16 were the major lineage. CONCLUSION: Urine was the primary source of VRE and LRE isolates in this study. E. faecium showed higher levels of resistance compared to E. faecalis. OptrA gene was detected in 91.6% of LRE, which could explain linezolid resistance, and van genes were detected in all vancomycin resistant Enterococcus strains, while vanA was a key resistance mechanism in VRE identified in this study.


Subject(s)
Enterococcus faecium , Gram-Positive Bacterial Infections , Linezolid , Microbial Sensitivity Tests , Linezolid/pharmacology , Humans , China/epidemiology , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/drug therapy , Male , Middle Aged , Enterococcus faecalis/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/isolation & purification , Female , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Molecular Epidemiology , Adult , Vancomycin Resistance/genetics , Aged , Retrospective Studies , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/drug effects , Vancomycin-Resistant Enterococci/isolation & purification , Young Adult , Enterococcus/genetics , Enterococcus/drug effects , Enterococcus/isolation & purification
2.
Int J Med Microbiol ; 315: 151624, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38838390

ABSTRACT

Staphylococcus aureus is a notorious pathogen responsible for various severe diseases. Due to the emergence of drug-resistant strains, the prevention and treatment of S. aureus infections have become increasingly challenging. Vancomycin is considered to be one of the last-resort drugs for treating most methicillin-resistant S. aureus (MRSA), so it is of great significance to further reveal the mechanism of vancomycin resistance. VraFG is one of the few important ABC (ATP-binding cassette) transporters in S. aureus that can form TCS (two-component systems)/ABC transporter modules. ABC transporters can couple the energy released from ATP hydrolysis to translocate solutes across the cell membrane. In this study, we obtained a strain with decreased vancomycin susceptibility after serial passaging and selection. Subsequently, whole-genome sequencing was performed on this laboratory-derived strain MWA2 and a novel single point mutation was discovered in vraF gene, leading to decreased sensitivity to vancomycin and daptomycin. Furthermore, the mutation reduces autolysis of S. aureus and downregulates the expression of lytM, isaA, and atlA. Additionally, we observed that the mutant has a less net negative surface charge than wild-type strain. We also noted an increase in the expression of the dlt operon and mprF gene, which are associated with cell surface charge and serve to hinder the binding of cationic peptides by promoting electrostatic repulsion. Moreover, this mutation has been shown to enhance hemolytic activity, expand subcutaneous abscesses, reflecting an increased virulence. This study confirms the impact of a point mutation of VraF on S. aureus antibiotic resistance and virulence, contributing to a broader understanding of ABC transporter function and providing new targets for treating S. aureus infections.


Subject(s)
ATP-Binding Cassette Transporters , Anti-Bacterial Agents , Bacterial Proteins , Staphylococcal Infections , Staphylococcus aureus , Vancomycin , Virulence/genetics , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Vancomycin/pharmacology , Animals , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Staphylococcus aureus/metabolism , Microbial Sensitivity Tests , Vancomycin Resistance/genetics , Whole Genome Sequencing , Daptomycin/pharmacology , Mice , Autolysis , Humans , Point Mutation , Mutation , Female
3.
Euro Surveill ; 29(23)2024 Jun.
Article in English | MEDLINE | ID: mdl-38847117

ABSTRACT

BackgroundVancomycin-resistant enterococci (VRE) are increasing in Denmark and Europe. Linezolid and vancomycin-resistant enterococci (LVRE) are of concern, as treatment options are limited. Vancomycin-variable enterococci (VVE) harbour the vanA gene complex but are phenotypically vancomycin-susceptible.AimThe aim was to describe clonal shifts for VRE and VVE in Denmark between 2015 and 2022 and to investigate genotypic linezolid resistance among the VRE and VVE.MethodsFrom 2015 to 2022, 4,090 Danish clinical VRE and VVE isolates were whole genome sequenced. We extracted vancomycin resistance genes and sequence types (STs) from the sequencing data and performed core genome multilocus sequence typing (cgMLST) analysis for Enterococcus faecium. All isolates were tested for the presence of mutations or genes encoding linezolid resistance.ResultsIn total 99% of the VRE and VVE isolates were E. faecium. From 2015 through 2019, 91.1% of the VRE and VVE were vanA E. faecium. During 2020, to the number of vanB E. faecium increased to 254 of 509 VRE and VVE isolates. Between 2015 and 2022, seven E. faecium clusters dominated: ST80-CT14 vanA, ST117-CT24 vanA, ST203-CT859 vanA, ST1421-CT1134 vanA (VVE cluster), ST80-CT1064 vanA/vanB, ST117-CT36 vanB and ST80-CT2406 vanB. We detected 35 linezolid vancomycin-resistant E. faecium and eight linezolid-resistant VVEfm.ConclusionFrom 2015 to 2022, the numbers of VRE and VVE increased. The spread of the VVE cluster ST1421-CT1134 vanA E. faecium in Denmark is a concern, especially since VVE diagnostics are challenging. The finding of LVRE, although in small numbers, ia also a concern, as treatment options are limited.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Carbon-Oxygen Ligases , Enterococcus faecium , Gram-Positive Bacterial Infections , Linezolid , Microbial Sensitivity Tests , Multilocus Sequence Typing , Vancomycin Resistance , Vancomycin-Resistant Enterococci , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Vancomycin-Resistant Enterococci/drug effects , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Humans , Denmark/epidemiology , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Carbon-Oxygen Ligases/genetics , Linezolid/pharmacology , Vancomycin Resistance/genetics , Whole Genome Sequencing , Vancomycin/pharmacology , Vancomycin/therapeutic use , Genotype
4.
J Antimicrob Chemother ; 79(7): 1628-1636, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38785365

ABSTRACT

BACKGROUND: The glycopeptide vancomycin is the antimicrobial agent-of-choice for the treatment of severe non-gastrointestinal infections with members of Bacillus cereus sensu lato (s.l.). Recently, sporadic detection of vancomycin-resistant phenotypes emerged, mostly for agar diffusion testing such as the disc diffusion method or gradient test (e.g. Etest®) method. RESULTS: In this work, we were able to disprove a preliminarily assumed high resistance to vancomycin in an isolate of B. cereus s.l. using broth microdilution and agar dilution. Microscopic imaging during vancomycin susceptibility testing showed spreading towards the inhibition zone, which strongly suggested sliding motility. Furthermore, transcriptomic analysis using RNA-Seq on the nanopore platform revealed several key genes of biofilm formation (e.g. calY, tasA, krsEABC) to be up-regulated in pseudo-resistant cells, substantiating that bacterial sliding is responsible for the observed mobility. Down-regulation of virulence (e.g. hblABCD, nheABC, plcR) and flagellar genes compared with swarming cells also confirmed the non-swarming phenotype of the pseudo-resistant isolate. CONCLUSIONS: The results highlight an insufficiency of agar diffusion testing for vancomycin susceptibility in the B. cereus group, and reference methods like broth microdilution are strongly recommended. As currently no guideline mentions interfering phenotypes in antimicrobial susceptibility testing of B. cereus s.l., this knowledge is essential to obtain reliable results on vancomycin susceptibility. In addition, this is the first report of sliding motility undermining accurate antimicrobial susceptibility testing in B. cereus s.l. and may serve as a basis for future studies on bacterial motility in susceptibility testing and its potential impact on treatment efficacy.


Subject(s)
Anti-Bacterial Agents , Bacillus cereus , Microbial Sensitivity Tests , Vancomycin Resistance , Vancomycin , Bacillus cereus/drug effects , Bacillus cereus/genetics , Microbial Sensitivity Tests/methods , Vancomycin/pharmacology , Anti-Bacterial Agents/pharmacology , Vancomycin Resistance/genetics , Biofilms/drug effects , Humans , Gene Expression Profiling
5.
Mol Microbiol ; 121(6): 1182-1199, 2024 06.
Article in English | MEDLINE | ID: mdl-38690761

ABSTRACT

The dipeptide D-Ala-D-Ala is an essential component of peptidoglycan and the target of vancomycin. Most Clostridioides difficile strains possess the vanG operon responsible for the synthesis of D-Ala-D-Ser, which can replace D-Ala-D-Ala in peptidoglycan. The C. difficile vanG operon is regulated by a two-component system, VanRS, but is not induced sufficiently by vancomycin to confer resistance to this antibiotic. Surprisingly, in the absence of the VanS histidine kinase (HK), the vanG operon is still induced by vancomycin and also by another antibiotic, ramoplanin, in a VanR-dependent manner. This suggested the cross-regulation of VanR by another HK or kinases that are activated in the presence of certain lipid II-targeting antibiotics. We identified these HKs as CD35990 and CD22880. However, mutations in either or both HKs did not affect the regulation of the vanG operon in wild-type cells suggesting that intact VanS prevents the cross-activation of VanR by non-cognate HKs. Overproduction of VanR in the absence of VanS, CD35990, and CD22880 led to high expression of the vanG operon indicating that VanR can potentially utilize at least one more phosphate donor for its activation. Candidate targets of CD35990- and CD22880-mediated regulation in the presence of vancomycin or ramoplanin were identified by RNA-Seq.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Clostridioides difficile , Gene Expression Regulation, Bacterial , Histidine Kinase , Operon , Vancomycin Resistance , Vancomycin , Operon/genetics , Clostridioides difficile/genetics , Clostridioides difficile/drug effects , Clostridioides difficile/metabolism , Histidine Kinase/metabolism , Histidine Kinase/genetics , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Vancomycin/pharmacology , Vancomycin Resistance/genetics , Anti-Bacterial Agents/pharmacology , Depsipeptides/pharmacology , Transcription Factors
6.
Iran J Med Sci ; 49(5): 302-312, 2024 May.
Article in English | MEDLINE | ID: mdl-38751872

ABSTRACT

Background: Antibiotic resistance is a global public health concern that has been exacerbated by the overuse and misuse of antibiotics, leading to the emergence of resistant bacteria. The gut microbiota, often influenced by antibiotic usage, plays a crucial role in overall health. Therefore, this study aimed to investigate the prevalence of antibiotic resistant genes in the gut microbiota of Indonesian coastal and highland populations, as well as to identify vancomycin-resistant bacteria and their resistant genes. Methods: Stool samples were collected from 22 individuals residing in Pacet, Mojokerto, and Kenjeran, Surabaya Indonesia in 2022. The read count of antibiotic resistant genes was analyzed in the collected samples, and the bacterium concentration was counted by plating on the antibiotic-containing agar plate. Vancomycin-resistant strains were further isolated, and the presence of vancomycin-resistant genes was detected using a multiplex polymerase chain reaction (PCR). Results: The antibiotic resistant genes for tetracycline, aminoglycosides, macrolides, beta-lactams, and vancomycin were found in high frequency in all stool samples (100%) of the gut microbiota. Meanwhile, those meant for chloramphenicol and sulfonamides were found in 86% and 16% of the samples, respectively. Notably, vancomycin-resistant genes were found in 16 intrinsically resistant Gram-negative bacterial strains. Among the detected vancomycin-resistant genes, vanG was the most prevalent (27.3%), while vanA was the least prevalent (4.5%). Conclusion: The presence of multiple vancomycin resistance genes in intrinsically resistant Gram-negative bacterial strains demonstrated the importance of the gut microbiota as a reservoir and hub for the horizontal transfer of antibiotic resistant genes.


Subject(s)
Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/drug effects , Indonesia , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Vancomycin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Feces/microbiology , Male , Female , Bacteria/drug effects , Bacteria/genetics , Bacteria/classification , Adult , Genes, Bacterial
7.
Protein Sci ; 33(6): e5002, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723146

ABSTRACT

Bacteria that have acquired resistance to most antibiotics, particularly those causing nosocomial infections, create serious problems. Among these, the emergence of vancomycin-resistant enterococci was a tremendous shock, considering that vancomycin is the last resort for controlling methicillin-resistant Staphylococcus aureus. Therefore, there is an urgent need to develop an inhibitor of VanX, a protein involved in vancomycin resistance. Although the crystal structure of VanX has been resolved, its asymmetric unit contains six molecules aligned in a row. We have developed a structural model of VanX as a stable dimer in solution, primarily utilizing nuclear magnetic resonance (NMR) residual dipolar coupling. Despite the 46 kDa molecular mass of the dimer, the analyses, which are typically not as straightforward as those of small proteins around 10 kDa, were successfully conducted. We assigned the main chain using an amino acid-selective unlabeling method. Because we found that the zinc ion-coordinating active sites in the dimer structure were situated in the opposite direction to the dimer interface, we generated an active monomer by replacing an amino acid at the dimer interface. The monomer consists of only 202 amino acids and is expected to be used in future studies to screen and improve inhibitors using NMR.


Subject(s)
Bacterial Proteins , Protein Multimerization , Serine-Type D-Ala-D-Ala Carboxypeptidase , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Catalytic Domain , Metalloendopeptidases/chemistry , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/metabolism , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Serine-Type D-Ala-D-Ala Carboxypeptidase/chemistry , Serine-Type D-Ala-D-Ala Carboxypeptidase/metabolism , Serine-Type D-Ala-D-Ala Carboxypeptidase/physiology , Vancomycin Resistance/genetics , Methicillin-Resistant Staphylococcus aureus/enzymology , Methicillin-Resistant Staphylococcus aureus/metabolism
8.
Int J Antimicrob Agents ; 63(6): 107154, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599552

ABSTRACT

BACKGROUND: Vancomycin is frequently used as a last line of defence against infections due to multidrug-resistant Staphylococcus aureus (S. aureus). A recent finding described the acquisition of vancomycin-resistant S. aureus strains by the integration of an enterococcal plasmid containing the vanA operon into the S. aureus chromosome via homologous recombination involving a specific integration site called locus L2. METHODS: To characterise all mechanisms of acquisition of vanA, this study analysed the 15 706 S. aureus genomes to look for vanA and described its genetic environment. RESULTS: A complete vanA operon was found in 25 S. aureus strains isolated from 12 patients, including nine co-isolated with vancomycin-resistant Enterococcus strains. VanA was found within transposon Tn1546-like elements on 17 plasmids and eight chromosomes. VanA might be acquired through conjugation of enterococcal and staphylococcal plasmids, transposition of Tn1546 carrying vanA and plasmid integration into the chromosome. Further, L2 was detected in 2087 genomes (13.3%) of S. aureus strains across different continents. Six potential chromosomal hotspots for integration of the entire vanA-containing enterococcal plasmid were identified by homologous recombination via L2. CONCLUSIONS: These findings suggest that the recently described scenario in a New York patient could be reproduced anywhere. Surveillance of this possibility is mandatory, especially in patients with vancomycin-resistant Enterococcus infection or colonisation.


Subject(s)
Bacterial Proteins , Carbon-Oxygen Ligases , DNA Transposable Elements , Genome, Bacterial , Operon , Plasmids , Staphylococcal Infections , Staphylococcus aureus , Vancomycin Resistance , Humans , Plasmids/genetics , Vancomycin Resistance/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/drug effects , DNA Transposable Elements/genetics , Bacterial Proteins/genetics , Carbon-Oxygen Ligases/genetics , Genome, Bacterial/genetics , Staphylococcal Infections/microbiology , Anti-Bacterial Agents/pharmacology , Vancomycin/pharmacology
9.
Int Immunopharmacol ; 132: 111780, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38603853

ABSTRACT

BACKGROUND: Glycopeptide antibiotic vancomycin is a bactericidal antibiotic available for the infection to Staphylococcus aureus (SA), however, SA has a strong adaptive capacity and thereby acquires resistance to vancomycin. This study aims to illuminate the possible molecular mechanism of vancomycin resistance of SA based on the 16S rRNA sequencing data and microarray profiling data. METHODS: 16S rRNA sequencing data of control samples and urinary tract infection samples were retrieved from the EMBL-EBI (European Molecular Biology Laboratory - European Bioinformatics Institute) database. Correlation of gut flora and clinical indicators was evaluated. The possible targets regulated by SA were predicted by microarray profiling and subjected to KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis. CXCL10 gene knockout and overexpression were introduced to evaluate the effect of CXCL10 on the virulence of SA and the resistance to vancomycin. SA strains were co-cultured with urethral epithelial cells in vitro. The presence of SA virulence factors was detected using PCR. Biofilm formation of SA strains was assessed using the microtiter plate method. Furthermore, the antibiotic sensitivity of SA strains was evaluated through vancomycin testing. RESULTS: Gut flora and its species abundance had significant difference between urinary tract infection and control samples. SA was significantly differentially expressed in urinary tract infection samples. Resistance of SA to vancomycin mainly linked to the D-alanine metabolism pathway. SA may participate in the occurrence of urinary tract infection by upregulating CXCL10. In addition, CXCL10 mainly affected the SA resistance to vancomycin through the TLR signaling pathway. In vitro experimental results further confirmed that the overexpression of CXCL10 in SA increased SA virulence and decreased its susceptibility to vancomycin. In vitro experimental validation demonstrated that the knockout of CXCL10 in urethral epithelial cells enhanced the sensitivity of Staphylococcus aureus (SA) to vancomycin. CONCLUSION: SA upregulates the expression of CXCL10 in urethral epithelial cells, thereby activating the TLR signaling pathway and promoting resistance to glycopeptide antibiotics in SA.


Subject(s)
Anti-Bacterial Agents , Chemokine CXCL10 , Staphylococcal Infections , Staphylococcus aureus , Urinary Tract Infections , Vancomycin Resistance , Vancomycin , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Vancomycin/pharmacology , Humans , Staphylococcal Infections/microbiology , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Chemokine CXCL10/metabolism , Chemokine CXCL10/genetics , Vancomycin Resistance/genetics , Urinary Tract Infections/microbiology , Urinary Tract Infections/drug therapy , Biofilms/drug effects , Gastrointestinal Microbiome/drug effects , RNA, Ribosomal, 16S/genetics , Epithelial Cells/microbiology , Epithelial Cells/drug effects , Female , Male
10.
Mikrobiyol Bul ; 58(2): 125-134, 2024 Apr.
Article in Turkish | MEDLINE | ID: mdl-38676581

ABSTRACT

The World Health Organization has included the problem of antibiotic resistance among the top 10 important health problems in the world. Treatment of infectious diseases has become more difficult due to the spread of antibiotic resistance between bacteria via transposable elements. Vancomycin-resistant enterococci (VRE) are of critical medical and public health importance due to their association with serious nosocomial infections and high risk of death. One of the most important features of VREs is that they have multiple antibiotic resistance and treatment options are reduced. Therefore, new treatment methods are needed. The vanA gene constitutes the building block of the vancomycin resistance mechanism and causes high resistance to vancomycin. In this study, it was aimed to investigate the neutralization of the vancomycin resistance mechanism by creating vanA antisense RNA (asRNA). The vanA positive VRE50 strain in our culture collection which was isolated from the clinical sample, was used to amplify the vanA gene by polymerase chain reaction (PCR). The amplified vanA amplicon was inserted inversely into the pUC19 plasmid by means of the enzyme cutting sites in the primers used. The resulting plasmid was combined with the pAT392 plasmid which can replicate in gram-positive bacteria and a fusion plasmid was created. The fusion plasmid whose orientation was confirmed, was transferred to the wild strain VRE50 by electroporation method. Minimum inhibitory concentration (MIC) values of transformed VRE (tVRE50) and wild type VRE50 strains used as control were determined by the E-Test method. The vancomycin MIC value of the wild type VRE50 strain was determined as 1024 µg/mL and that of the tVRE50 strain was 32 µg/mL and it was determined that the vancomycin resistance of the tVRE50 strain decreased with asRNA (antisense RNA). Antisense RNA technology is an important method for neutralizing the expression of genes. This study showed that neutralization of the vancomycin resistance gene may provide a lower MIC value in a vancomycin-resistant enterococcus strain and lead to increased susceptibility. This new approach provides a new method for VRE treatment by neutralizing the vancomycin resistance mechanism. The result obtained in this study needs to be supported by in vivo tests.


Subject(s)
Bacterial Proteins , Carbon-Oxygen Ligases , RNA, Antisense , Vancomycin-Resistant Enterococci , Vancomycin , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/drug effects , Carbon-Oxygen Ligases/genetics , RNA, Antisense/genetics , Bacterial Proteins/genetics , Humans , Vancomycin/pharmacology , Plasmids/genetics , Vancomycin Resistance/genetics , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Gene Silencing
11.
Ann Lab Med ; 44(5): 450-454, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38475872

ABSTRACT

Vancomycin variable Enterococcus (VVE) bacteria are phenotypically susceptible to vancomycin, but they harbor the vanA gene. We aimed to ascertain the prevalence of VVE among clinically isolated vancomycin-susceptible Enterococcus faecium (VSE) isolates, as well as elucidate the molecular characteristics of the vanA gene cluster within these isolates. Notably, we investigated the prevalence and structure of the vanA gene cluster of VVE. Between June 2021 and May 2022, we collected consecutive, non-duplicated vancomycin-susceptible Enterococcus faecium (VSE) samples. Real-time PCR was performed to detect the presence of vanA, vanB, and vanC. Overlapping PCR with sequencing and whole-genome sequencing were performed for structural analysis. Sequence types (STs) were determined by multilocus sequence typing. Exposure testing was performed to assess the ability of the isolates to acquire vancomycin resistance. Among 282 VSE isolates tested, 20 isolates (7.1%) were VVE. Among them, 17 isolates had partial deletions in the IS1216 or IS1542 sequences in vanS (N=10), vanR (N=5), or vanH (N=2). All VVE isolates belonged to the CC17 complex and comprised five STs, namely ST17 (40.0%), ST1421 (25.0%), ST80 (25.0%), ST787 (5.0%), and ST981 (5.0%). Most isolates were related to three hospital-associated clones (ST17, ST1421, and ST80). After vancomycin exposure, 18 of the 20 VVEs acquired vancomycin resistance. Considering the high reversion rate, detecting VVE by screening VSE for vanA is critical for appropriate treatment and infection control.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterococcus faecium , Gram-Positive Bacterial Infections , Microbial Sensitivity Tests , Multilocus Sequence Typing , Vancomycin Resistance , Vancomycin , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Humans , Vancomycin/pharmacology , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/diagnosis , Vancomycin Resistance/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Carbon-Oxygen Ligases/genetics , Whole Genome Sequencing , Real-Time Polymerase Chain Reaction , Prevalence , Multigene Family
12.
J Antimicrob Chemother ; 79(5): 997-1005, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38501366

ABSTRACT

BACKGROUND: VRE are increasingly described worldwide. Screening of hospitalized patients at risk for VRE carriage is mandatory to control their dissemination. Here, we have developed the Bfast [VRE Panel] PCR kit, a rapid and reliable quantitative PCR assay for detection of vanA, vanB, vanD and vanM genes, from solid and liquid cultures adaptable to classical and ultrafast real-time PCR platforms. METHODS: Validation was carried out on 133 well characterized bacterial strains, including 108 enterococci of which 64 were VRE. Analytical performances were determined on the CFX96 Touch (Bio-Rad) and Chronos Dx (BforCure), an ultrafast qPCR machine. Widely used culture plates and broths for enterococci selection/growth were tested. RESULTS: All targeted van alleles (A, B, D and M) were correctly detected without cross-reactivity with other van genes (C, E, G, L and N) and no interference with the different routinely used culture media. A specificity and sensitivity of 100% and 99.7%, respectively, were determined, with limits of detection ranging from 21 to 238 cfu/reaction depending on the targets. The Bfast [VRE Panel] PCR kit worked equally well on the CFX and Chronos Dx platforms, with differences in multiplexing capacities (five and four optical channels, respectively) and in turnaround time (45 and 16 minutes, respectively). CONCLUSIONS: The Bfast [VRE Panel] PCR kit is robust, easy to use, rapid and easily implementable in clinical microbiology laboratories for ultra-rapid confirmation of the four main acquired van genes. Its features, especially on Chronos Dx, seem to be unmatched compared to other tools for screening of VRE.


Subject(s)
Real-Time Polymerase Chain Reaction , Sensitivity and Specificity , Vancomycin Resistance , Vancomycin-Resistant Enterococci , Humans , Real-Time Polymerase Chain Reaction/methods , Vancomycin Resistance/genetics , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/isolation & purification , Vancomycin-Resistant Enterococci/drug effects , Enterococcus/genetics , Enterococcus/drug effects , Enterococcus/isolation & purification , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/diagnosis , Bacterial Proteins/genetics , Time Factors , Genes, Bacterial/genetics
13.
Antimicrob Agents Chemother ; 68(5): e0115923, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38506549

ABSTRACT

Vancomycin heteroresistance is prone to missed detection and poses a risk of clinical treatment failure. We encountered one clinical Enterococcus faecium strain, SRR12, that carried a complete vanM gene cluster but was determined as susceptible to vancomycin using the broth microdilution method. However, distinct subcolonies appeared within the clear zone of inhibition in the E-test assay, one of which, named SRR12-v1, showed high-level resistance to vancomycin. SRR12 was confirmed as heteroresistant to vancomycin using population analysis profiling and displayed "revive" growth curves with a lengthy lag phase of over 13 hours when exposed to 2-32 mg/L vancomycin. The resistant subcolony SRR12-v1 was found to carry an identical vanM gene cluster to that of SRR12 but a significantly increased vanM copy number in the genome. Long-read whole genome sequencing revealed that a one-copy vanM gene cluster was located on a pELF1-like linear plasmid in SRR12. In comparison, tandem amplification of the vanM gene cluster jointed with IS1216E was seated on a linear plasmid in the genome of SRR12-v1. These amplifications of the vanM gene cluster were demonstrated as unstable and would decrease accompanied by fitness reversion after serial passaging for 50 generations under increasing vancomycin pressure or without antibiotic pressure but were relatively stable under constant vancomycin pressure. Further, vanM resistance in resistant variants was verified to be carried by conjugative plasmids with variable sizes using conjugation assays and S1-pulsed field gel electrophoresis blotting, suggesting the instability/flexibility of vanM cluster amplification in the genome and an increased risk of vanM resistance dissemination.


Subject(s)
Anti-Bacterial Agents , Enterococcus faecium , Microbial Sensitivity Tests , Multigene Family , Plasmids , Vancomycin Resistance , Vancomycin , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Plasmids/genetics , Vancomycin/pharmacology , Vancomycin Resistance/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Humans , Whole Genome Sequencing
14.
Sci Rep ; 14(1): 1895, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38253607

ABSTRACT

Clonal transmission and horizontal gene transfer (HGT) contribute to the spread of vancomycin-resistant enterococci (VRE) in global healthcare. Our study investigated vesiduction, a HGT mechanism via membrane vesicles (MVs), for vanA and vanB genes that determine vancomycin resistance. We isolated MVs for VRE of different sequence types (STs) and analysed them by nanoparticle tracking analysis. Selected MV samples were subjected to DNA sequence analysis. In resistance transfer experiments, vancomycin-susceptible enterococci were exposed to MVs and bacterial supernatants of VRE. Compared to bacteria grown in lysogeny broth (MVs/LB), cultivation under vancomycin stress (MVs/VAN) resulted in increased particle concentrations of up to 139-fold (ST80). As a key finding, we could show that VRE isolates of ST80 and ST117 produced remarkably more vesicles at subinhibitory antibiotic concentrations (approx. 9.2 × 1011 particles/ml for ST80 and 2.4 × 1011 particles/ml for ST117) than enterococci of other STs (range between 1.8 × 1010 and 5.3 × 1010 particles/ml). In those MV samples, the respective resistance genes vanA and vanB were completely verifiable using sequence analysis. Nevertheless, no vancomycin resistance transfer via MVs to vancomycin-susceptible Enterococcus faecium was phenotypically detectable. However, our results outline the potential of future research on ST-specific MV properties, promising new insights into VRE mechanisms.


Subject(s)
Enterococcus faecium , Vancomycin-Resistant Enterococci , Enterococcus faecium/genetics , Vancomycin Resistance/genetics , Vancomycin/pharmacology , Vancomycin-Resistant Enterococci/genetics , Membranes
15.
Diagn Microbiol Infect Dis ; 108(3): 116180, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38183897

ABSTRACT

The use of glycopeptide medications may decline in line with the annual decline in methicillin-resistant Staphylococcus aureus (MRSA) detection rates in China. The rate of heterogeneous vancomycin-intermediate Staphylococcus aureus (hVISA)detection may be impacted by this. However, there is currently a dearth of information on the incidence of hVISA in China. This study aims to analyze the recent epidemiology and molecular characteristics of hVISA strains in Beijing, China. A total of 175 non-duplicate MRSA strains from various infection sites were collected from a medical center between January 2018 and May 2023 and underwent molecular typing and susceptibility testing (Vitek2). Vancomycin and teicoplanin MICs were also evaluated by standard broth microdilution method and agar dilution method, respectively. Isolates growing on screening agar (BHIV4 and BHIT5, brain heart infusion agar containing 4 µg/ml vancomycin and 5 µg/ml teicoplanin, respectively) were characterized further by analysis of macro-Etest (MET) and population analysis profiling with area under the curve (PAP-AUC). The proportion of hVISA among MRSA isolates was 8.6 %. BHIT5 could select all hVISA strains while BHIV4 and MET only selected two hVISA strains. Compared with vancomycin- susceptible Staphylococcus aureus (VSSA), hVISA isolates were less susceptible to erythromycin and clindamycin. In addition, hVISA frequency was MIC-independent despite using different detection methods. In total, 11 types of STs, 28 types of spa typing, four types of SCCmec typing, and two types of agr typing were identified and the predominant type in both MRSA and hVISA isolates was ST239-t030-SCCmecIII-agr I. The analysis of biofilm formation, growth, and virulence genes in hVISA strains revealed sparse information. The dataset presented in this study provided the prevalence and molecular characteristics of hVISA in hospital settings and the combination of BHIT5 and PAP-AUC may identify hVISA efficiently. The result of genotyping suggested the genotype of hVISA was mainly consistent with that of local MRSA. Additional studies on the characteristics of hVISA strains were necessary.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Staphylococcal Infections , Humans , Vancomycin/pharmacology , Vancomycin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Staphylococcus aureus , Vancomycin-Resistant Staphylococcus aureus , Teicoplanin/pharmacology , Teicoplanin/therapeutic use , Staphylococcal Infections/epidemiology , Staphylococcal Infections/drug therapy , Tertiary Care Centers , Prevalence , Agar , Vancomycin Resistance/genetics , Microbial Sensitivity Tests
16.
Eur J Clin Microbiol Infect Dis ; 43(4): 673-682, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38296911

ABSTRACT

PURPOSE: To investigate the occurrence of vancomycin-variable enterococci (VVE) in a hospital in central Italy. METHODS: vanA positive but vancomycin-susceptible Enterococcus faecium isolates (VVE-S) were characterized by antibiotic susceptibility tests, molecular typing (PFGE and MLST), and WGS approach. The reversion of VVE-S to a resistant phenotype was assessed by exposure to increasing vancomycin concentrations, and the revertant isolates were used in filter mating experiments. qPCR was used to analyze the plasmid copy number. RESULTS: Eleven putative VVE-S were selected. WGS revealed two categories of vanA cluster plasmid located: the first type showed the lack of vanR, the deletion of vanS, and an intact vanH/vanA/vanX cluster; the second type was devoid of both vanR and vanS and showed a deletion of 544-bp at the 5'-end of the vanH. Strains (n = 7) carrying the first type of vanA cluster were considered VVE-S and were able to regain a resistance phenotype (VVE-R) in the presence of vancomycin, due to a 44-bp deletion in the promoter region of vanH/vanA/vanX, causing its constitutive expression. VVE-R strains were not able to transfer resistance by conjugation, and the resistance phenotype was unstable: after 11 days of growth without selective pressure, the revertants were still resistant but showed a lower vancomycin MIC. A higher plasmid copy number in the revertant strains was probably related to the resistance phenotype. CONCLUSION: We highlight the importance of VVE transition to VRE under vancomycin therapy resulting in a potential failure treatment. We also report the first-time identification of VVE-S isolates pstS-null belonging to ST1478.


Subject(s)
Enterococcus faecium , Gram-Positive Bacterial Infections , Humans , Vancomycin/pharmacology , Vancomycin/therapeutic use , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Multilocus Sequence Typing , Vancomycin Resistance/genetics , Microbial Sensitivity Tests , Enterococcus , Bacterial Proteins/genetics , Gram-Positive Bacterial Infections/microbiology
17.
Mar Pollut Bull ; 198: 115844, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38056291

ABSTRACT

Mangrove ecosystems are hotspots of biodiversity, but have been threatened by anthropogenic activities. Vancomycin-resistant enterococci (VRE) are nosocomial bacteria classified as high priority by the World Health Organization (WHO). Herein, we describe the identification and genomic characteristics of a vancomycin-resistant Enterococcus faecalis strain isolated from a highly impacted mangrove ecosystem of the northeastern Brazilian, in 2021. Genomic analysis confirmed the existence of the transposon Tn1546-vanA and clinically relevant antimicrobial resistance genes, such as streptogramins, tetracycline, phenicols, and fluoroquinolones. Virulome analysis identified several genes associated to adherence, immune modulation, biofilm, and exoenzymes production. The UFSEfl strain was assigned to sequence type (ST9), whereas phylogenomic analysis with publicly available genomes from a worldwide confirmed clonal relatedness with a hospital-associated Brazilian clone. Our findings highlight the successful expansion of hospital-associated VRE in a mangrove area and shed light on the need for strengthening genomic surveillance of WHO priority pathogens in these vital ecosystems.


Subject(s)
Ecosystem , Enterococcus faecium , Gram-Positive Bacterial Infections , Vancomycin-Resistant Enterococci , Humans , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Brazil/epidemiology , Clone Cells , Enterococcus faecalis/genetics , Gram-Positive Bacterial Infections/drug therapy , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/microbiology , Microbial Sensitivity Tests , Vancomycin , Vancomycin Resistance/genetics , Vancomycin-Resistant Enterococci/genetics , Cross Infection/microbiology
18.
BMC Microbiol ; 23(1): 380, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38049763

ABSTRACT

OBJECTIVE: The purpose of this study is to reduce the spread of the vanA gene by curing the vanA-harboring plasmid of vancomycin-resistant using the CRISPR-Cas9 system. METHODS: Two specific spacer sequence (sgRNAs) specific was designed to target the vanA gene and cloned into plasmid CRISPR-Cas9. The role of the CRISPR-Cas system in the plasmid elimination of drug-resistance genes was verified by chemically transformation and conjugation delivery methods. Moreover, the elimination efficiency in strains was evaluated by plate counting, PCR, and quantitative real-time PCR (qPCR). Susceptibility testing was performed by broth microdilution assay and by Etest strips (bioMérieux, France) to detect changes in bacterial drug resistance phenotype after drug resistance plasmid clearance. RESULTS: In the study, we constructed a specific prokaryotic CRISPR-Cas9 system plasmid targeting cleavage of the vanA gene. PCR and qPCR results indicated that recombinant pCas9-sgRNA plasmid can efficiently clear vanA-harboring plasmids. There was no significant correlation between sgRNA lengths and curing efficiency. In addition, the drug susceptibility test results showed that the bacterial resistance to vancomycin was significantly reduced after the vanA-containing drug-resistant plasmid was specifically cleaved by the CRISPR-Cas system. The CRISPR-Cas9 system can block the horizontal transfer of the conjugated plasmid pUC19-vanA. CONCLUSION: In conclusion, our study demonstrated that CRISPR-Cas9 achieved plasmid clearance and reduced antimicrobial resistance. The CRISPR-Cas9 system could block the horizontal transfer of plasmid carrying vanA. This strategy provided a great potential to counteract the ever-worsening spread of the vanA gene among bacterial pathogens and laid the foundation for subsequent research using the CRISPR-Cas9 system as adjuvant antibiotic therapy.


Subject(s)
RNA, Guide, CRISPR-Cas Systems , Vancomycin , Vancomycin/pharmacology , Vancomycin Resistance/genetics , CRISPR-Cas Systems , Anti-Bacterial Agents/pharmacology , Plasmids/genetics , Bacterial Proteins/genetics
19.
Microbiol Spectr ; 11(6): e0296223, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37905844

ABSTRACT

IMPORTANCE: The proportion of VREfm among all Enterococcus faecium isolated from blood cultures in German hospitals has increased in the period 2015-2020 from 11.9% to 22.3% with a country-wide spread of the clonal lineage ST117/CT71 vanB. In this study, we provided useful information about the genetic diversity of invasive strains of E. faecium. Moreover, our findings confirm the nosocomial spread of novel ST1299 vanA lineages, which recently had a rapid expansion in Austria and the south-eastern part of Germany.


Subject(s)
Cross Infection , Enterococcus faecium , Gram-Positive Bacterial Infections , Vancomycin-Resistant Enterococci , Humans , Vancomycin Resistance/genetics , Enterococcus faecium/genetics , Hospitals, University , Multilocus Sequence Typing , Gram-Positive Bacterial Infections/epidemiology , Cross Infection/epidemiology , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology
20.
BMC Genomics ; 24(1): 610, 2023 Oct 12.
Article in English | MEDLINE | ID: mdl-37828442

ABSTRACT

BACKGROUND: Vancomycin-resistant enterococci (VRE) are among the most common causative pathogens for nosocomial infections worldwide. Moreover, strains of VRE have been isolated from several domestic livestock in Egypt. METHODS: This study examined if healthy dogs are a potential source of VRE infection by isolating and characterizing Enterococcus faecium strains from stool samples on a morphological basis and biochemical activities. Subsequently, it was confirmed by genotypic characterization using polymerase chain reaction (PCR), followed by the detection of antibiotic resistance genes, virulence determinants, and genes contributing to enterocin production by PCR. Furthermore, the phylogenetic relationships among vanB and tetL genes were analyzed. RESULTS: All ten fecal samples were identified as E. faecium and confirmed by PCR. In addition, 90% of the isolates tested were positive for the virulence genes gelE and esp, and all the isolates tested were positive for the antibiotic resistance genes tetL and vanB. Only three of the five enterocin genes examined were detected. Ent As-48, bacteriocin 31, and Ent L50 were identified in 100%, 80%, and 60% of the samples, respectively. CONCLUSION: Dogs should be regarded as a reservoir of E. faecium that carries vancomycin resistance and virulence determinants that may affect public health in Egypt, considering a "One Health" task force approach to restrict their spread.


Subject(s)
Enterococcus faecium , Gram-Positive Bacterial Infections , Vancomycin-Resistant Enterococci , Dogs , Animals , Vancomycin-Resistant Enterococci/genetics , Enterococcus faecium/genetics , Vancomycin/pharmacology , Public Health , Egypt/epidemiology , Phylogeny , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Vancomycin Resistance/genetics , Tetracycline/pharmacology , Virulence Factors/genetics , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/veterinary , Enterococcus faecalis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...