Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.257
Filter
1.
Ann Clin Microbiol Antimicrob ; 23(1): 41, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38704577

ABSTRACT

BACKGROUND: Infections caused by linezolid-resistant enterococci (LRE) are clinically difficult to treat and threaten patient health. However, there is a lack of studies on long time-span LRE strains in China. For this reason, our study comprehensively revealed the resistance mechanisms of LRE strains collected in a Chinese tertiary care hospital from 2011 to 2022. METHODS: Enterococcal strains were screened and verified after retrospective analysis of microbial data. Subsequently, 65 LRE strains (61 Enterococcus faecalis and 4 Enterococcus faecium, MIC ≥ 8 µg/ml), 1 linezolid-intermediate Enterococcus faecium (MIC = 4 µg/ml) and 1 linezolid-susceptible Enterococcus faecium (MIC = 1.5 µg/ml) were submitted for whole-genome sequencing (WGS) analysis and bioinformatics analysis. RESULTS: The optrA gene was found to be the most common linezolid resistance mechanism in our study. We identified the wild-type OptrA and various OptrA variants in 98.5% of LRE strains (61 Enterococcus faecalis and 3 Enterococcus faecium). We also found one linezolid-resistant Enterococcus faecium strain carried both optrA and cfr(D) gene, while one linezolid-resistant Enterococcus faecium only harbored the poxtA gene. Most optrA genes (55/64) were located on plasmids, with impB-fexA-optrA, impB-fexA-optrA-erm(A), fexA-optrA-erm(A), and fexA-optrA segments. A minority of optrA genes (9/64) were found on chromosomes with the Tn6674-like platform. Besides, other possible linezolid resistance-associated mechanisms (mutations in the rplC and rplD genes) were also found in 26 enterococcal strains. CONCLUSIONS: Our study suggested that multiple mechanisms of linezolid resistance exist among clinical LRE strains in China.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Enterococcus faecalis , Enterococcus faecium , Gram-Positive Bacterial Infections , Linezolid , Microbial Sensitivity Tests , Whole Genome Sequencing , Linezolid/pharmacology , China/epidemiology , Humans , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Drug Resistance, Bacterial/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Anti-Bacterial Agents/pharmacology , Retrospective Studies , Enterococcus/drug effects , Enterococcus/genetics , Bacterial Proteins/genetics , Genome, Bacterial , Molecular Epidemiology , Tertiary Care Centers , Genomics
2.
Vet Microbiol ; 293: 110103, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718528

ABSTRACT

Oxazolidinones are potent antimicrobial agents used to treat human infections caused by multidrug-resistant Gram-positive bacteria. The growing resistance to oxazolidinones poses a significant threat to public health. In August 2021, a linezolid-resistant Enterococcus faecium BN83 was isolated from a raw milk sample of cow in Inner Mongolia, China. This isolate exhibited a multidrug resistance phenotype and was resistant to most of drugs tested including linezolid and tedizolid. PCR detection showed that two mobile oxazolidinones resistance genes, optrA and poxtA, were present in this isolate. Whole genome sequencing analysis revealed that the genes optrA and poxtA were located on two different plasmids, designated as pBN83-1 and pBN83-2, belonging to RepA_N and Inc18 families respectively. Genetic context analysis suggested that optrA gene on plasmid pBN83-1 was located in transposon Tn6261 initially found in E. faecalis. Comprehensive analysis revealed that Tn6261 act as an important horizontal transmission vector for the spread of optrA in E. faecium. Additionally, poxtA-bearing pBN83-2 displayed high similarity to numerous plasmids from Enterococcus of different origin and pBN83-2-like plasmid represented a key mobile genetic element involved in movement of poxtA in enterococcal species. The presence of optrA- and poxtA-carrying E. faecium in raw bovine milk represents a public health concern and active surveillance is urgently warranted to investigate the prevalence of oxazolidinone resistance genes in animal-derived food products.


Subject(s)
Anti-Bacterial Agents , Enterococcus faecium , Milk , Oxazolidinones , Animals , Cattle , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Milk/microbiology , China/epidemiology , Oxazolidinones/pharmacology , Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests , Drug Resistance, Multiple, Bacterial/genetics , Plasmids/genetics , Linezolid/pharmacology , Whole Genome Sequencing , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/veterinary , Gram-Positive Bacterial Infections/epidemiology , Genes, Bacterial/genetics
3.
mBio ; 15(5): e0017024, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38564699

ABSTRACT

Penicillin-binding protein 5 (PBP5) of Enterococcus faecium (Efm) is vital for ampicillin resistance (AMP-R). We previously designated three forms of PBP5, namely, PBP5-S in Efm clade B strains [ampicillin susceptible (AMP-S)], PBP5-S/R (AMP-S or R), and PBP5-R (AMP-R) in clade A strains. Here, pbp5 deletion resulted in a marked reduction in AMP minimum inhibitory concentrations (MICs) to 0.01-0.09 µg/mL for clade B and 0.12-0.19 µg/mL for clade A strains; in situ complementation restored parental AMP MICs. Using D344SRF (lacking ftsW/psr/pbp5), constructs with ftsWA/psrA (from a clade A1 strain) cloned upstream of pbp5-S and pbp5-S/R alleles resulted in modest increases in MICs to 3-8 µg/mL, while high MICs (>64 µg/mL) were seen using pbp5 from A1 strains. Next, using ftsW ± psr from clade B and clade A/B and B/A hybrid constructs, the presence of psrB, even alone or in trans, resulted in much lower AMP MICs (3-8 µg/mL) than when psrA was present (MICs >64 µg/mL). qRT PCR showed relatively greater pbp5 expression (P = 0.007) with pbp5 cloned downstream of clade A1 ftsW/psr (MIC >128 µg/mL) vs when cloned downstream of clade B ftsW/psr (MIC 4-16 µg/mL), consistent with results in western blots. In conclusion, we report the effect of clade A vs B psr on AMP MICs as well as the impact of pbp5 alleles from different clades. While previously, Psr was not thought to contribute to AMP MICs in Efm, our results showed that the presence of psrB resulted in a major decrease in Efm AMP MICs. IMPORTANCE: The findings of this study shed light on ampicillin resistance in Enterococcus faecium clade A strains. They underscore the significance of alterations in the amino acid sequence of penicillin-binding protein 5 (PBP5) and the pivotal role of the psr region in PBP5 expression and ampicillin resistance. Notably, the presence of a full-length psrB leads to reduced PBP5 expression and lower minimum inhibitory concentrations (MICs) of ampicillin compared to the presence of a shorter psrA, regardless of the pbp5 allele involved. Additionally, clade B E. faecium strains exhibit lower AMP MICs when both psr alleles from clades A and B are present, although it is important to consider other distinctions between clade A and B strains that may contribute to this effect. It is intriguing to note that the divergence between clade A and clade B E. faecium and the subsequent evolution of heightened AMP MICs in hospital-associated strains appear to coincide with changes in Pbp5 and psr. These changes in psr may have resulted in an inactive Psr, facilitating increased PBP5 expression and greater ampicillin resistance. These results raise the possibility that a mimicker of PsrB, if one could be designed, might be able to lower MICs of ampicillin-resistant E. faecium, thus potentially resorting ampicillin to our therapeutic armamentarium for this species.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Enterococcus faecium , Microbial Sensitivity Tests , Penicillin-Binding Proteins , beta-Lactam Resistance , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , beta-Lactam Resistance/genetics , Ampicillin/pharmacology , Genome, Bacterial
4.
Antimicrob Agents Chemother ; 68(5): e0143923, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38591854

ABSTRACT

Phage therapy has (re)emerged as a serious possibility for combating multidrug-resistant bacterial infections, including those caused by vancomycin-resistant Enterococcus faecium strains. These opportunistic pathogens belong to a specific clonal complex 17, against which relatively few phages have been screened. We isolated a collection of 21 virulent phages growing on these vancomycin-resistant isolates. Each of these phages harbored a typical narrow plaquing host range, lysing at most 5 strains and covering together 10 strains of our panel of 14 clinical isolates. To enlarge the host spectrum of our phages, the Appelmans protocol was used. We mixed four out of our most complementary phages in a cocktail that we iteratively grew on eight naive strains from our panel, of which six were initially refractory to at least three of the combined phages. Fifteen successive passages permitted to significantly improve the lytic activity of the cocktail, from which phages with extended host ranges within the E. faecium species could be isolated. A single evolved phage able to kill up to 10 of the 14 initial E. faecium strains was obtained, and it barely infected nearby species. All evolved phages had acquired point mutations or a recombination event in the tail fiber genetic region, suggesting these genes might have driven phage evolution by contributing to their extended host spectra.


Subject(s)
Bacteriophages , Enterococcus faecium , Host Specificity , Vancomycin-Resistant Enterococci , Enterococcus faecium/drug effects , Bacteriophages/genetics , Vancomycin-Resistant Enterococci/drug effects , Phage Therapy/methods , Gram-Positive Bacterial Infections/microbiology , Vancomycin Resistance , Vancomycin/pharmacology , Humans , Anti-Bacterial Agents/pharmacology
5.
J Nat Prod ; 87(4): 831-836, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38551509

ABSTRACT

Two novel polyketides, accraspiroketides A (1) and B (2), which feature unprecedented [6 + 6+6 + 6] + [5 + 5] spiro chemical architectures, were isolated from Streptomyces sp. MA37 ΔaccJ mutant strain. Compounds 1-2 exhibit excellent activity against Gram-positive bacteria (MIC = 1.5-6.3 µg/mL). Notably, 1 and 2 have superior activity against clinically isolated Enterococcus faecium K60-39 (MIC = 4.0 µg/mL and 4.7 µg/mL, respectively) than ampicillin (MIC = 25 µg/mL).


Subject(s)
Anti-Bacterial Agents , Enterococcus faecium , Microbial Sensitivity Tests , Polyketides , Streptomyces , Polyketides/pharmacology , Polyketides/chemistry , Polyketides/isolation & purification , Streptomyces/chemistry , Molecular Structure , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Enterococcus faecium/drug effects , Gram-Positive Bacteria/drug effects , Spiro Compounds/chemistry , Spiro Compounds/pharmacology , Spiro Compounds/isolation & purification , Naphthacenes/chemistry , Naphthacenes/pharmacology
6.
J Food Prot ; 87(5): 100267, 2024 May.
Article in English | MEDLINE | ID: mdl-38492644

ABSTRACT

Bacitracin is an antimicrobial used in the feed or water of poultry in the U.S. for the prevention, treatment, and control of clostridial diseases such as necrotic enteritis. Concern has been raised that bacitracin can select for antimicrobial-resistant bacteria that can be transmitted to humans and subsequently cause disease that is more difficult to treat because of the resistance. The objective of the present study was to perform a quantitative risk assessment (QRA) to estimate the potential risk in the U.S. of human infection with antimicrobial-resistant Enterococcus faecalis and E. faecium derived from chicken and turkey products as a result of bacitracin usage in U.S. poultry. The modeling approach estimated the annual number of healthcare-associated enterococcal infections in the U.S. that would be resistant to antimicrobial therapy and that would be derived from poultry sources because of bacitracin use in poultry. Parameter estimates were developed to be "maximum risk" to overestimate the risk to humans. While approximately 60% of E. faecalis and E. faecium derived from poultry were predicted to possess bacitracin resistance based on the presence of the bcrABDR gene locus, very few human-derived isolates possessed this trait. Furthermore, no vancomycin or linezolid-resistant strains of E. faecalis or E. faecium were detected in poultry sources between the years 2002 and 2019. The model estimated the number of antimicrobial-resistant E. faecalis and E. faecium cases per year that might resist therapy due to bacitracin use in poultry as 0.86 and 0.14, respectively, which translates to an annual risk estimate for E. faecalis of less than 1 in 350 million and for E. faecium of less than 1 in 2 billion for members of the U.S. population. Even with the use of risk-maximizing assumptions, the results indicate that there is a high probability that the use of bacitracin according to label instructions in U.S. poultry presents a negligible risk to human health.


Subject(s)
Anti-Bacterial Agents , Bacitracin , Drug Resistance, Bacterial , Bacitracin/pharmacology , Animals , Humans , Anti-Bacterial Agents/pharmacology , Risk Assessment , Enterococcus faecalis/drug effects , Gram-Positive Bacterial Infections/drug therapy , Poultry , Chickens , Enterococcus faecium/drug effects
7.
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
8.
Antimicrob Agents Chemother ; 68(5): e0171623, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38506550

ABSTRACT

Outbreaks caused by vancomycin-resistant enterococci that transcend jurisdictional boundaries are occurring worldwide. This study focused on a vancomycin-resistant enterococcus outbreak that occurred between 2018 and 2021 across two cities in Hiroshima, Japan. The study involved genetic and phylogenetic analyses using whole-genome sequencing of 103 isolates of vancomycin-resistant enterococci to identify the source and transmission routes of the outbreak. Phylogenetic analysis was performed using core genome multilocus sequence typing and core single-nucleotide polymorphisms; infection routes between hospitals were inferred using BadTrIP. The outbreak was caused by Enterococcus faecium sequence type (ST) 80 carrying the vanA plasmid, which was derived from strain A10290 isolated in India. Of the 103 isolates, 93 were E. faecium ST80 transmitted across hospitals. The circular vanA plasmid of the Hiroshima isolates was similar to the vanA plasmid of strain A10290 and transferred from E. faecium ST80 to other STs of E. faecium and other Enterococcus species by conjugation. The inferred transmission routes across hospitals suggest the existence of a central hospital serving as a hub, propagating vancomycin-resistant enterococci to multiple hospitals. Our study highlights the importance of early intervention at the key central hospital to prevent the spread of the infection to small medical facilities, such as nursing homes, with limited medical resources and a high number of vulnerable individuals.


Subject(s)
Disease Outbreaks , Enterococcus faecium , Gram-Positive Bacterial Infections , Multilocus Sequence Typing , Phylogeny , Plasmids , Vancomycin-Resistant Enterococci , Whole Genome Sequencing , Enterococcus faecium/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Japan/epidemiology , Humans , Vancomycin-Resistant Enterococci/genetics , Vancomycin-Resistant Enterococci/drug effects , Vancomycin-Resistant Enterococci/isolation & purification , Plasmids/genetics , Gram-Positive Bacterial Infections/transmission , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/epidemiology , Cross Infection/microbiology , Cross Infection/transmission , Cross Infection/epidemiology , Bacterial Proteins/genetics , Anti-Bacterial Agents/pharmacology , Carbon-Oxygen Ligases/genetics , Microbial Sensitivity Tests , Polymorphism, Single Nucleotide , Hospitals , Vancomycin/pharmacology , Genome, Bacterial/genetics
9.
Int J Antimicrob Agents ; 63(5): 107125, 2024 May.
Article in English | MEDLINE | ID: mdl-38431109

ABSTRACT

RATIONALE AND OBJECTIVES: ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) constitute a threat to humans worldwide. India is now the most populous country. The goal was to investigate the evolution of the rates of antimicrobial resistance in ESKAPE pathogens across India over the 2010-20 decade. METHODS: The data (89 studies) were retrieved from the Medline PubMed repository using specific keywords. RESULTS: The study of 20 177 ESKAPE isolates showed that A. baumannii isolates were the most represented (35.9%, n = 7238), followed by P. aeruginosa (25.3%, n = 5113), K. pneumoniae (19.5%, n = 3934), S. aureus (16.3%, n = 3286), E. faecium (2.6%, n = 517) and Enterobacter spp. (0.4%, n = 89). A notable increase in the resistance rates to antimicrobial agents occurred over the 2010-20 decade. The most important levels of resistance were observed in 2016-20 for A. baumannii (90% of resistance to the amoxicillin-clavulanate combination) and K. pneumoniae (81.6% of resistance to gentamycin). The rise in ß-lactamase activities was correlated with an increase in the positivity of Gram-negative isolates for ß-lactamase genes. CONCLUSIONS: This review highlighted that, in contrast to developed countries that kept resistance levels under control, a considerable increase in resistance to various classes of antibiotics occurred in ESKAPE pathogens in India over the 2010-2020 decade.


Subject(s)
Acinetobacter baumannii , Anti-Bacterial Agents , Klebsiella pneumoniae , India/epidemiology , Humans , Anti-Bacterial Agents/pharmacology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Acinetobacter baumannii/isolation & purification , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/isolation & purification , Microbial Sensitivity Tests , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/isolation & purification , Drug Resistance, Bacterial , Staphylococcus aureus/drug effects , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Enterococcus faecium/isolation & purification , Drug Resistance, Multiple, Bacterial/genetics , Enterobacter/drug effects , Enterobacter/genetics , Enterobacter/isolation & purification
10.
J Biol Chem ; 299(3): 102912, 2023 03.
Article in English | MEDLINE | ID: mdl-36649910

ABSTRACT

Daptomycin (DAP) is an antibiotic frequently used as a drug of last resort against vancomycin-resistant enterococci. One of the major challenges when using DAP against vancomycin-resistant enterococci is the emergence of resistance, which is mediated by the cell-envelope stress system LiaFSR. Indeed, inhibition of LiaFSR signaling has been suggested as a strategy to "resensitize" enterococci to DAP. In the absence of LiaFSR, alternative pathways mediating DAP resistance have been identified, including adaptive mutations in the enolpyruvate transferase MurAA (MurAAA149E), which catalyzes the first committed step in peptidoglycan biosynthesis; however, how these mutations confer resistance is unclear. Here, we investigated the biochemical basis for MurAAA149E-mediated adaptation to DAP to determine whether such an alternative pathway would undermine the potential efficacy of therapies that target the LiaFSR pathway. We found cells expressing MurAAA149E had increased susceptibility to glycoside hydrolases, consistent with decreased cell wall integrity. Furthermore, structure-function studies of MurAA and MurAAA149E using X-ray crystallography and biochemical analyses indicated only a modest decrease in MurAAA149E activity, but a 16-fold increase in affinity for MurG, which performs the last intracellular step of peptidoglycan synthesis. Exposure to DAP leads to mislocalization of cell division proteins including MurG. In Bacillus subtilis, MurAA and MurG colocalize at division septa and, thus, we propose MurAAA149E may contribute to DAP nonsusceptibility by increasing the stability of MurAA-MurG interactions to reduce DAP-induced mislocalization of these essential protein complexes.


Subject(s)
Daptomycin , Enterococcus faecium , Transferases , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Daptomycin/metabolism , Daptomycin/pharmacology , Drug Resistance, Bacterial , Enterococcus faecium/drug effects , Enterococcus faecium/metabolism , Microbial Sensitivity Tests , Peptidoglycan/metabolism , Transferases/metabolism
11.
J Glob Antimicrob Resist ; 30: 50-55, 2022 09.
Article in English | MEDLINE | ID: mdl-35660662

ABSTRACT

OBJECTIVES: Enterococcus faecium is one of the important pathogens causing nosocomial infection, which can be resistant to fosfomycin by obtaining the plasmid-encoded fosfomycin resistance genes, and the mutation of MurA protein encoded by chromosome is a newly discovered fosfomycin resistance mechanism in recent years. METHODS: In this study, we found a fosfomycin-resistant clinical isolate of E. faecium Efm_1415 with fosfomycin MIC of 512 mg/L, carrying Asp50Glu mutant of MurA protein, which was never reported before. To study the role and mechanism of this mutant protein in fosfomycin resistance, we used gene cloning, protein expression, and purification, steady-state kinetic, fosfomycin inhibition assay, and next-generation sequencing (NGS) to investigate the functions, characters, and enzymatic kinetic properties of MurA protein. RESULTS: The results revealed that the Asp50Glu MurA can mediate a 4-fold increase in the fosfomycin MIC of the host bacteria. Compared with the wild-type MurA, the affinity of the Asp50Glu MurA to the substrates was increased, and the enzyme activity cannot be inhibited by the concentration of fosfomycin less than 100 mg/L. CONCLUSIONS: The research on the mutant MurA had gained a new understanding of the fosfomycin resistance mechanisms and helped to find new antibiotics with MurA enzyme as the target of action.


Subject(s)
Alkyl and Aryl Transferases , Anti-Bacterial Agents , Bacterial Proteins , Drug Resistance, Bacterial , Enterococcus faecium , Fosfomycin , Alkyl and Aryl Transferases/genetics , Amino Acid Substitution , Anti-Bacterial Agents/pharmacology , Aspartic Acid/genetics , Bacterial Proteins/genetics , Drug Resistance, Bacterial/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Fosfomycin/pharmacology , Glutamic Acid/genetics , Mutation
12.
Nat Commun ; 13(1): 586, 2022 01 31.
Article in English | MEDLINE | ID: mdl-35102136

ABSTRACT

Bacterial bloodstream infections are a major cause of morbidity and mortality among patients undergoing hematopoietic cell transplantation (HCT). Although previous research has demonstrated that pathogens may translocate from the gut microbiome into the bloodstream to cause infections, the mechanisms by which HCT patients acquire pathogens in their microbiome have not yet been described. Here, we use linked-read and short-read metagenomic sequencing to analyze 401 stool samples collected from 149 adults undergoing HCT and hospitalized in the same unit over three years, many of whom were roommates. We use metagenomic assembly and strain-specific comparison methods to search for high-identity bacterial strains, which may indicate transmission between the gut microbiomes of patients. Overall, the microbiomes of patients who share time and space in the hospital do not converge in taxonomic composition. However, we do observe six pairs of patients who harbor identical or nearly identical strains of the pathogen Enterococcus faecium, or the gut commensals Akkermansia muciniphila and Hungatella hathewayi. These shared strains may result from direct transmission between patients who shared a room and bathroom, acquisition from a common hospital source, or transmission from an unsampled intermediate. We also identify multiple patients with identical strains of species commonly found in commercial probiotics, including Lactobacillus rhamnosus and Streptococcus thermophilus. In summary, our findings indicate that sharing of identical pathogens between the gut microbiomes of multiple patients is a rare phenomenon. Furthermore, the observed potential transmission of commensal, immunomodulatory microbes suggests that exposure to other humans may contribute to microbiome reassembly post-HCT.


Subject(s)
Bacteria/metabolism , Bacterial Infections/transmission , Gastrointestinal Microbiome , Hospitalization , Adult , Aged , Anti-Bacterial Agents/pharmacology , Cross Infection/microbiology , Cross Infection/transmission , Drug Resistance, Microbial/drug effects , Drug Resistance, Microbial/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Escherichia coli/drug effects , Escherichia coli/isolation & purification , Female , Gastrointestinal Microbiome/drug effects , Hematopoietic Stem Cell Transplantation , Hospitals , Humans , Length of Stay , Male , Metagenome/genetics , Metagenomics , Middle Aged , Phylogeny , Probiotics , Sequence Analysis, DNA , Time Factors
13.
BMC Microbiol ; 22(1): 16, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34996348

ABSTRACT

BACKGROUND: Previous reports have demonstrated two thiazolidione derivatives (H2-60 and H2-81) can robustly inhibit the planktonic growth and biofilm formation of S. epidermidis and S. aureus by targeting the histidine kinase YycG. Whereas the antibacterial and anti-biofilm activity of these two thiazolidione derivatives (H2-60 and H2-81) against Enterococcus faecium remains elusive. Here, the pET28a-YycG recombinant plasmid were in vitro expressed in E. coli competent cell BL21 (DE3) and induced to express YycG' protein (conding HisKA and HATPase_c domain) by 0.5 mM IPTG and was purified by Ni - NTA agarose and then for the autophosphorylation test. Antimicrobial testing and time-killing assay were also be determined. Anti-biofilm activity of two derivatives with sub-MIC concentration towards positive biofilm producers of clinical E. faecium were detected using polystyrene microtiter plate and CLSM. RESULTS: The MICs of H2-60 and H2-81 in the clinical isolates of E. faecium were in the range from 3.125 mg/L to 25 mg/L. Moreover, either H2-60 or H2-81 showed the excellent bactericidal activity against E. faecium with monotherapy or its combination with daptomycin by time-killing assay. E. faecium planktonic cells can be decreased by H2-60 or H2-81 for more than 3 × log10 CFU/mL after 24 h treatment when combined with daptomycin. Furthermore, over 90% of E. faecium biofilm formation could markedly be inhibited by H2-60 and H2-81 at 1/4 × MIC value. In addition, the frequency of the eradicated viable cells embedded in mature biofilm were evaluated by the confocal laser microscopy, suggesting that of H2-60 combined with ampicillin or daptomycin was significantly high when compared with single treatment (78.17 and 74.48% vs. 41.59%, respectively, P < 0.01). CONCLUSION: These two thiazolidione derivatives (H2-60 and H2-81) could directly impact the kinase phosphoration activity of YycG of E. faecium. H2-60 combined with daptomycin exhibit the excellent antibacterial and anti-biofilm activity against E. faecium by targeting YycG.


Subject(s)
Anti-Bacterial Agents/pharmacology , Daptomycin/pharmacology , Enterococcus faecium/drug effects , Thiazoles/pharmacology , Ampicillin/pharmacology , Anti-Bacterial Agents/chemistry , Biofilms/drug effects , Biofilms/growth & development , Drug Synergism , Enterococcus faecium/enzymology , Enterococcus faecium/growth & development , Gram-Positive Bacterial Infections/microbiology , Histidine Kinase/antagonists & inhibitors , Histidine Kinase/metabolism , Humans , Microbial Sensitivity Tests , Recombinant Proteins/metabolism , Thiazoles/chemistry
14.
Microb Drug Resist ; 28(2): 217-221, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34705570

ABSTRACT

Vancomycin-resistant Enterococcus faecium (VREfm) is one of the most important nosocomial pathogens with limited therapeutic alternatives. In this study, we followed the trends of VREfm and E. faecium causing bloodstream infections (BSIs) in a Spanish hospital, from 2011 to 2020. During this period, 832 E. faecium strains were isolated and 121 (14.5%) were vancomycin resistant. Nineteen of 101 BSIs (18.8%) caused by E. faecium were due to VREfm. The number of BSI-producing E. faecium isolates increased significantly over the past 5 years, with the percentage of invasive VREfm isolates being substantially higher than the average values in Europe and especially in Spain (<3%). VREfm isolates recovered in 2018 (28) and BSI-producing isolates from 2019 (3) and 2020 (2) were molecularly characterized. All were positive for vanA and belonged to sequence type (ST) 80 (28) or ST117 (5), within clonal complex 17. The isolates were only susceptible to linezolid, although most of them were also susceptible (dose dependent) to daptomycin. We report for the first time the establishment and persistence of the VREfm ST80 and ST117 clones in a Spanish hospital. The spread and establishment of hospital-adapted, multidrug-resistant VREfm clones in health care settings are cause for concern and may precede an increment in the BSIs caused by them.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteremia/microbiology , Enterococcus faecium/drug effects , Glycopeptides/pharmacology , Gram-Positive Bacterial Infections/microbiology , Vancomycin-Resistant Enterococci/drug effects , Clone Cells , Cross Infection/microbiology , Health Facilities , Humans , Microbial Sensitivity Tests
15.
Microb Drug Resist ; 28(1): 73-80, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34491861

ABSTRACT

Vancomycin-resistant Enterococcus faecium (VREfm) is a globally significant nosocomial pathogen with a rapidly increasing prevalence. The objectives were to investigate VREfm outbreak duration and study the additional impact that infection control bundle strategies (ICBSs) set up to curb coronavirus disease 2019 (COVID-19) spreading had on VREfm outbreaks. Outbreak data set were collected prospectively from April 2, 2014 to August 13, 2020 at Copenhagen University Hospital Bispebjerg, Denmark. All VREfm samples had polymerase chain reaction performed for vanA/vanB genes before whole genome sequencing using the Illumina MiSeq platform. The relatedness of isolates was studied by core genome multilocus sequence typing (cgMLST) using Ridom SeqSphere. Eighty-one outbreaks had a median outbreak duration of 32.5 days (range 5-204 days) and 1,161 VREfm isolates were sequenced. The same cgMLST cluster types reappeared after outbreaks were terminated. When comparing the first 5 months of the COVID-19 pandemic with the corresponding period in 2019, we found a 10-fold decrease in VREfm outbreak patients and median outbreak duration decreased from 56 to 7 days (88%). Several COVID-19 ICBSs were implemented from March 13 through summer 2020. VREfm outbreaks lasted up to 204 days, but our findings suggest that outbreaks might last longer since the same cgMLST persisted in the same wards for years implying an endemic situation with recurrent outbreaks caused by hospital reservoirs or readmittance of unknown VREfm carriers. The sharp decline in VREfm outbreaks during the COVID-19 pandemic was most likely due to the ICBSs, resulting in a decrease in VREfm transmission.


Subject(s)
COVID-19 , Enterococcus faecium/genetics , Pandemics , Quarantine , Streptococcal Infections/epidemiology , Vancomycin Resistance/genetics , Aged , Carrier State/microbiology , Denmark/epidemiology , Enterococcus faecium/drug effects , Female , Hospitalization , Humans , Male , Streptococcal Infections/microbiology , Whole Genome Sequencing
16.
Microbiol Spectr ; 9(3): e0091321, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34756065

ABSTRACT

Enterococcus faecium is a clinically important pathogen that can cause significant morbidity and death. In this study, we aimed to develop a machine learning (ML) algorithm-based rapid susceptibility method to distinguish vancomycin-resistant E. faecium (VREfm) and vancomycin-susceptible E. faecium (VSEfm) strains. A predictive model was developed and validated to distinguish VREfm and VSEfm strains by analyzing the matrix-assisted laser desorption ionization-time of flight (MALDI-TOF) mass spectrometry (MS) spectra of unique E. faecium isolates from different specimen types. The algorithm used 5,717 mass spectra, including 2,795 VREfm and 2,922 VSEfm mass spectra, and was externally validated with 2,280 mass spectra of isolates (1,222 VREfm and 1,058 VSEfm strains). A random forest-based algorithm demonstrated overall good classification performances for the isolates from the specimens, with mean accuracy, sensitivity, and specificity of 0.78, 0.79, and 0.77, respectively, with 10-fold cross-validation, timewise validation, and external validation. Furthermore, the algorithm provided rapid results, which would allow susceptibility prediction prior to the availability of phenotypic susceptibility results. In conclusion, an ML algorithm designed using mass spectra obtained from the routine workflow may be able to rapidly differentiate VREfm strains from VSEfm strains; however, susceptibility results must be confirmed by routine methods, given the demonstrated performance of the assay. IMPORTANCE A modified binning method was incorporated to cluster MS shifting ions into a set of representative peaks based on a large-scale MS data set of clinical VREfm and VSEfm isolates, including 2,795 VREfm and 2,922 VSEfm isolates. Predictions with the algorithm were significantly more accurate than empirical antibiotic use, the accuracy of which was 0.50, based on the local epidemiology. The algorithm improved the accuracy of antibiotic administration, compared to empirical antibiotic prescription. An ML algorithm designed using MALDI-TOF MS spectra obtained from the routine workflow accurately differentiated VREfm strains from VSEfm strains, especially in blood and sterile body fluid samples, and can be applied to facilitate the rapid and accurate clinical testing of pathogens.


Subject(s)
Anti-Bacterial Agents/pharmacology , Enterococcus faecium/drug effects , Gram-Positive Bacterial Infections/drug therapy , Microbial Sensitivity Tests/methods , Vancomycin/pharmacology , Algorithms , Enterococcus faecium/genetics , Gram-Positive Bacterial Infections/microbiology , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Vancomycin Resistance/genetics , Vancomycin-Resistant Enterococci/drug effects
17.
Chem Biodivers ; 18(12): e2100741, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34786854

ABSTRACT

Three new hydroxyphenylacetic acid derivatives, stachylines E-G (1-3), and a new alkaloid, mortieridinone (4), along with six known compounds (5-10), were isolated from endophytic fungus Mortierella sp. in Epimedium acuminatum Franch. Their structures were determined by their spectroscopic analyses and by comparison with the literature data. Compounds 7 and 10 showed selective antibacterial activity against tested multidrug-resistant bacteria with minimum inhibitory concentration (MIC) values ranging from 25 to 3.13 µg/mL.


Subject(s)
Alkaloids/pharmacology , Anti-Bacterial Agents/pharmacology , Epimedium/microbiology , Mortierella/chemistry , Phenylacetates/pharmacology , Alkaloids/chemistry , Alkaloids/isolation & purification , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Drug Resistance, Multiple, Bacterial/drug effects , Enterococcus faecium/drug effects , Microbial Sensitivity Tests , Molecular Structure , Phenylacetates/chemistry , Phenylacetates/isolation & purification , Staphylococcus aureus/drug effects
18.
Int J Mol Sci ; 22(21)2021 Oct 20.
Article in English | MEDLINE | ID: mdl-34768762

ABSTRACT

The use of phenicol antibiotics in animals has increased. In recent years, it has been reported that the transferable gene mediates phenicol-oxazolidinone resistance. This study analyzed the prevalence and characteristics of phenicol-oxazolidinone resistance genes in Enterococcus faecalis and Enterococcus faecium isolated from food-producing animals and meat in Korea in 2018. Furthermore, for the first time, we reported the genome sequence of E. faecalis strain, which possesses the phenicol-oxazolidinone resistance gene on both the chromosome and plasmid. Among the 327 isolates, optrA, poxtA, and fexA genes were found in 15 (4.6%), 8 (2.5%), and 17 isolates (5.2%), respectively. Twenty E. faecalis strains carrying resistance genes belonged to eight sequence types (STs), and transferability was found in 17 isolates. The genome sequences revealed that resistant genes were present in the chromosome or plasmid, or both. In strains EFS17 and EFS108, optrA was located downstream of the ermA and ant(9)-1 genes. The strains EFS36 and EFS108 harboring poxtA-encoding plasmid cocarried fexA and cfr(D). These islands also contained IS1216E or the transposon Tn554, enabling the horizontal transfer of the phenicol-oxazolidinone resistance with other antimicrobial-resistant genes. Our results suggest that it is necessary to promote the prudent use of antibiotics through continuous monitoring and reevaluation.


Subject(s)
Anti-Infective Agents/pharmacology , Chloramphenicol/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Enterococcus faecalis/genetics , Enterococcus faecium/genetics , Meat/microbiology , Oxazolidinones/pharmacology , Animals , Cattle/microbiology , Computational Biology , Enterococcus faecalis/drug effects , Enterococcus faecalis/isolation & purification , Enterococcus faecium/drug effects , Enterococcus faecium/isolation & purification , Food Analysis , Gene Transfer, Horizontal , Genes, Bacterial/drug effects , Genome, Bacterial , Multilocus Sequence Typing , Plasmids , Republic of Korea , Swine/microbiology , Whole Genome Sequencing
19.
Molecules ; 26(19)2021 Oct 06.
Article in English | MEDLINE | ID: mdl-34641589

ABSTRACT

Pristine high-density bulk disks of MgB2 with added hexagonal BN (10 wt.%) were prepared using spark plasma sintering. The BN-added samples are machinable by chipping them into desired geometries. Complex shapes of different sizes can also be obtained by the 3D printing of polylactic acid filaments embedded with MgB2 powder particles (10 wt.%). Our present work aims to assess antimicrobial activity quantified as viable cells (CFU/mL) vs. time of sintered and 3D-printed materials. In vitro antimicrobial tests were performed against the bacterial strains Escherichia coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, Staphylococcus aureus ATCC 25923, Enterococcus faecium DSM 13590, and Enterococcus faecalis ATCC 29212; and the yeast strain Candida parapsilosis ATCC 22019. The antimicrobial effects were found to depend on the tested samples and microbes, with E. faecium being the most resistant and E. coli the most susceptible.


Subject(s)
Anti-Infective Agents/pharmacology , Bacteria/drug effects , Boron Compounds/pharmacology , Fungi/drug effects , Magnesium Compounds/pharmacology , Candida parapsilosis/drug effects , Enterococcus faecalis/drug effects , Enterococcus faecium/drug effects , Escherichia coli/drug effects , Microbial Sensitivity Tests , Polyesters/pharmacology , Printing, Three-Dimensional , Pseudomonas aeruginosa/drug effects , Staphylococcus aureus/drug effects
20.
Vet Microbiol ; 262: 109219, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34500344

ABSTRACT

The emergence of the phenicol-oxazolidinone-tetracycline resistance gene poxtA becomes a significant challenge for public health, since it confers a decreased susceptibility not only to the last resort drug linezolid, but also to florfenicol and doxycycline widely used in veterinary medicine. To determine the dissemination mechanism of poxtA in enterococci isolates from different healthy pigs in the swine farm, a total of 178 florfenicol-resistant enterococci isolates were collected from 400 fresh faecal swabs in a swine farm in China. The poxtA gene was detected in 11 (6.18 %) enterococci isolates, including 8 E. faecium, 2 E. hirae and 1 E. casseliflavus isolates. Whole genome sequencing indicated that the eight poxtA-harbouring E. faecium strains belonged to four different sequence types, including ST156 and three new STs, ST1818, ST1819 and ST1820. Five out of the 11 poxtA-positive enterococci isolates also harboured optrA gene. Moreover, E. casseliflavus strain DY31 co-harboured poxtA, optrA and cfr. Seven different poxtA-harbouring plasmids were obtained through Nanopore combined with Illumina sequencing. The poxtA-harbouring plasmids exhibited high genetic variation, six out of which belonged to rep2 plasmid of Inc18 family. The poxtA gene was flanked by IS1216E in the left and/or right ends.The optrA and cfr genes were located on different plasmids, respectively, but those genes could be co-transferred with poxtA gene into the recipient E. faecalis strain by electrotransformation. Our study highlights that both clonal spread and horizontal transfer mediated by Inc18 plasmid and IS1216E promote the dissemination of poxtA in enterococci isolates from different healthy pigs in the swine farm.


Subject(s)
Enterococcaceae , Enterococcus faecium , Gene Transfer, Horizontal , Gram-Positive Bacterial Infections , Oxazolidinones , Swine Diseases , Tetracycline Resistance , Animals , Anti-Bacterial Agents/pharmacology , China , Drug Resistance, Bacterial/genetics , Enterococcaceae/drug effects , Enterococcaceae/genetics , Enterococcus faecalis/drug effects , Enterococcus faecalis/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Farms , Gene Transfer, Horizontal/genetics , Gram-Positive Bacterial Infections/epidemiology , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/veterinary , Microbial Sensitivity Tests/veterinary , Oxazolidinones/pharmacology , Swine , Swine Diseases/epidemiology , Tetracycline Resistance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...