Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.293
Filtrer
1.
Nat Microbiol ; 9(10): 2570-2582, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39242817

RÉSUMÉ

Human microbiota assembly commences at birth, seeded by both maternal and environmental microorganisms. Ecological theory postulates that primary colonizers dictate microbial community assembly outcomes, yet such microbial priority effects in the human gut remain underexplored. Here using longitudinal faecal metagenomics, we characterized neonatal microbiota assembly for a cohort of 1,288 neonates from the UK. We show that the pioneering neonatal gut microbiota can be stratified into one of three distinct community states, each dominated by a single microbial species and influenced by clinical and host factors, such as maternal age, ethnicity and parity. A community state dominated by Enterococcus faecalis displayed stochastic microbiota assembly with persistent high pathogen loads into infancy. In contrast, community states dominated by Bifidobacterium, specifically B. longum and particularly B. breve, exhibited a stable assembly trajectory and long-term pathogen colonization resistance, probably due to strain-specific functional adaptions to a breast milk-rich neonatal diet. Consistent with our human cohort observation, B. breve demonstrated priority effects and conferred pathogen colonization resistance in a germ-free mouse model. Our findings solidify the crucial role of Bifidobacteria as primary colonizers in shaping the microbiota assembly and functions in early life.


Sujet(s)
Bifidobacterium , Fèces , Microbiome gastro-intestinal , Humains , Fèces/microbiologie , Animaux , Nouveau-né , Bifidobacterium/génétique , Bifidobacterium/isolement et purification , Souris , Femelle , Royaume-Uni , Métagénomique , Enterococcus faecalis/génétique , Enterococcus faecalis/isolement et purification , Lait humain/microbiologie , Mâle
2.
Pol J Microbiol ; 73(3): 349-362, 2024 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-39268957

RÉSUMÉ

The aim of this work was to provide a theoretical and scientific basis for the treatment, prevention, and control of clinical drug-resistant bacterial infections by studying the molecular epidemiology and horizontal transfer mechanism of optrA-carrying linezolid-resistant Enterococcus faecalis strains (LREfs) that were clinically isolated in a tertiary hospital in Kunming, China. Non-repetitive LREfs retained in a tertiary A hospital in Kunming, China. The strains were identified by Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS). The transferability and horizontal transfer mechanism of optrA gene were analyzed using polymerase chain reaction (PCR), whole-genome sequencing (WGS), and conjugation experiments. A total of 39 LREfs strains were collected, and all of them were multi-drug resistant. There were 30 LREfs strains (76.9%) carrying the optrA gene, The cfr, poxtA genes and mutations in the 23S rRNA gene were not detected. The conjugation experiments showed that only three of 10 randomly selected optrA-carrying LREfs were successfully conjugated with JH2-2. Further analysis of one successfully conjugated strain revealed that the optrA gene, located in the donor bacterium, formed the IS1216E-erm(A)-optrA-fexA-IS1216E transferable fragment under the mediation of the mobile genetic element (MGE) IS1216E, which was then transferred to the recipient bacterium via horizontal plasmid transfer. Carrying the optrA gene is the primary resistance mechanism of LREfs strains. The optrA gene could carry the erm(A) and fexA genes to co-transfer among E. faecalis. MGEs such as insertion sequence IS1216E play an important role in the horizontal transfer of the optrA gene.


Sujet(s)
Antibactériens , Enterococcus faecalis , Transfert horizontal de gène , Infections bactériennes à Gram positif , Linézolide , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Linézolide/pharmacologie , Antibactériens/pharmacologie , Humains , Infections bactériennes à Gram positif/microbiologie , Infections bactériennes à Gram positif/épidémiologie , Chine/épidémiologie , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Épidémiologie moléculaire , Résistance bactérienne aux médicaments/génétique , Tests de sensibilité microbienne , Multirésistance bactérienne aux médicaments/génétique , Séquençage du génome entier , Conjugaison génétique
3.
J Water Health ; 22(9): 1628-1640, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39340376

RÉSUMÉ

Coastal water quality is facing increasing threats due to human activities. Their contamination by sewage discharges poses significant risks to the environment and public health. We aimed to investigate the presence of antibiotic-resistant Enterococcus in beach waters. Over a 10-month period, samples were collected from four beaches in the State of São Paulo (Brazil). Enterococcus isolates underwent matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF/MS) and molecular analysis for accurate genus and species identification. The antimicrobial susceptibility for 14 antibiotics was evaluated using the disc diffusion method followed by a multidrug-resistance (MDR) classification. PCR amplification method was used to detect antimicrobial resistance genes (ARGs). Our findings revealed the prevalence of Enterococcus faecalis, E. faecium and E. hirae. Out of 130 isolates, 118 were resistant to multiple antibiotics. The detection of resistance genes provided evidence of the potential transfer of antibiotic resistance within the environment. Our findings underscore the necessity for continuous research and surveillance to enhance understanding of the pathogenicity and antimicrobial resistance mechanisms of Enterococcus, which is crucial to implement effective measures to preserve the integrity of coastal ecosystems.


Sujet(s)
Antibactériens , Enterococcus faecalis , Enterococcus faecium , Enterococcus hirae , Brésil , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/génétique , Enterococcus faecalis/isolement et purification , Enterococcus faecium/effets des médicaments et des substances chimiques , Enterococcus faecium/génétique , Enterococcus faecium/isolement et purification , Antibactériens/pharmacologie , Enterococcus hirae/effets des médicaments et des substances chimiques , Enterococcus hirae/génétique , Enterococcus hirae/isolement et purification , Résistance bactérienne aux médicaments , Plage pour la baignade , Tests de sensibilité microbienne , Eau de mer/microbiologie
4.
Sci Rep ; 14(1): 21741, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39289457

RÉSUMÉ

Enterococci, common hospital-acquired infections in immunocompromised patients, have garnered attention in clinical microbiology. To determine the clinical relevance of enterococci as food-borne pathogens, 116 fish, 90 vegetables, and 120 human diarrheal samples were tested for E. faecalis and E. faecium pathogenicity. Conventionally, 69 of 326 (21.17%) samples were positive for Enterococcus species, 52 (15.95%) of which were molecularly classified as E. faecalis and 13 (3.99%) as E. faecium. The E. faecalis contamination percentage of fresh fish (19.70%) was higher than frozen fish (4%). Cauliflower had the highest E. faecalis percentage (16.67%) when fish and vegetable samples didn't harbor the E. faecium atpA gene. 23.33% and 10.83% of participants' samples were molecularly confirmed as E. faecalis and E. faecium positive, respectively. E. faecalis isolates had all virulence genes, with gels being the most common (65.38%), while cylA and asa1 genes couldn't be detected in E. faecium isolates. E. faecalis showed the highest resistance against vancomycin and tetracycline (69.23%), whereas E. faecium extremely resisted tetracycline (76.92%) and erythromycin (69.23%) with the recognition of MDR among 44.2% of E. faecalis and 38.5% of E. faecium isolates. The great similarity of our isolates showed the clinical importance of food-borne antibiotic-resistant enterococci.


Sujet(s)
Enterococcus faecalis , Enterococcus faecium , Poissons , Génotype , Légumes , Enterococcus faecium/génétique , Enterococcus faecium/isolement et purification , Enterococcus faecium/pathogénicité , Enterococcus faecium/classification , Enterococcus faecalis/génétique , Enterococcus faecalis/isolement et purification , Enterococcus faecalis/pathogénicité , Légumes/microbiologie , Humains , Animaux , Poissons/microbiologie , Phénotype , Infections bactériennes à Gram positif/microbiologie , Microbiologie alimentaire , Tests de sensibilité microbienne , Antibactériens/pharmacologie , Facteurs de virulence/génétique
5.
PLoS Genet ; 20(8): e1011071, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39102428

RÉSUMÉ

Sortase-assembled pili contribute to virulence in many Gram-positive bacteria. In Enterococcus faecalis, the endocarditis and biofilm-associated pilus (Ebp) is polymerized on the membrane by sortase C (SrtC) and attached to the cell wall by sortase A (SrtA). In the absence of SrtA, polymerized pili remain anchored to the membrane (i.e. off-pathway). Here we show that the high temperature requirement A (HtrA) bifunctional chaperone/protease of E. faecalis is a quality control system that clears aberrant off-pathway pili from the cell membrane. In the absence of HtrA and SrtA, accumulation of membrane-bound pili leads to cell envelope stress and partially induces the regulon of the ceftriaxone resistance-associated CroRS two-component system, which in turn causes hyper-piliation and cell morphology alterations. Inactivation of croR in the OG1RF ΔsrtAΔhtrA background partially restores the observed defects of the ΔsrtAΔhtrA strain, supporting a role for CroRS in the response to membrane perturbations. Moreover, absence of SrtA and HtrA decreases basal resistance of E. faecalis against cephalosporins and daptomycin. The link between HtrA, pilus biogenesis and the CroRS two-component system provides new insights into the E. faecalis response to endogenous membrane perturbations.


Sujet(s)
Aminoacyltransferases , Protéines bactériennes , Biofilms , Cysteine endopeptidases , Enterococcus faecalis , Fimbriae bactériens , Chaperons moléculaires , Fimbriae bactériens/génétique , Fimbriae bactériens/métabolisme , Aminoacyltransferases/génétique , Aminoacyltransferases/métabolisme , Enterococcus faecalis/génétique , Cysteine endopeptidases/génétique , Cysteine endopeptidases/métabolisme , Protéines bactériennes/génétique , Protéines bactériennes/métabolisme , Chaperons moléculaires/génétique , Chaperons moléculaires/métabolisme , Biofilms/croissance et développement , Membrane cellulaire/métabolisme , Régulation de l'expression des gènes bactériens , Virulence/génétique , Antibactériens/pharmacologie , Ceftriaxone/pharmacologie
6.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 15-21, 2024 Jul 28.
Article de Anglais | MEDLINE | ID: mdl-39097900

RÉSUMÉ

A biofilm is a complex microbial structure that promotes the progression of persistent infections, particularly in nosocomial settings via indwelling medical devices. Conventional antibiotics are often ineffective treatments for biofilms; hence, it is crucial to investigate or design non-antibiotic antibiofilm compounds that can successfully reduce and eradicate biofilm-related infections. This study was an attempt to repurpose chronic disease medications of the antihypertensive and antilipidemic drug classes, including candesartan cilexetil (CC) and ursodeoxycholic acid (UDCA), respectively, to be used as antibiofilm agents against the two infectious pathogens Staphylococcus aureus and Enterococcus faecalis. Crystal violet (CV) staining assay was used to evaluate the antibiofilm activity of the drugs. Real-time polymerase chain reaction (RT-PCR) was performed to determine the transcription levels of the biofilm-related genes (icaA and icaR in S. aureus and fsrC and gelE in E. faecalis) following treatment with different concentrations of CC and UDCA. we found that a concentration of greater than 1.5 µg/ml of CC significantly (p < 0.005) inhibited the biofilm formation of both bacterial isolates, and a concentration of greater than 50 µg/ml of UDCA significantly (p < 0.005) inhibited the biofilm formation of both bacterial isolates. Interestingly, the mRNA expression levels of biofilm-related genes were decreased in the two bacterial isolates at concentrations that were lower than the human pharmaceutical daily doses.


Sujet(s)
Biofilms , Enterococcus faecalis , Staphylococcus aureus , Acide ursodésoxycholique , Biofilms/effets des médicaments et des substances chimiques , Biofilms/croissance et développement , Staphylococcus aureus/effets des médicaments et des substances chimiques , Staphylococcus aureus/génétique , Staphylococcus aureus/physiologie , Humains , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/génétique , Enterococcus faecalis/physiologie , Acide ursodésoxycholique/pharmacologie , Antibactériens/pharmacologie , Maladie chronique , Tests de sensibilité microbienne , Régulation de l'expression des gènes bactériens/effets des médicaments et des substances chimiques , Benzimidazoles/pharmacologie , Tétrazoles/pharmacologie , Dérivés du biphényle/pharmacologie
7.
Arch Microbiol ; 206(9): 369, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39110213

RÉSUMÉ

The RNA-based study provides an excellent indication of an organism's gene expression profile. Obtaining high-yield and high-purity RNA from Gram-positive and acid-fast bacteria is difficult without high-end kits and facilities. We optimised effective and simple protocol for RNA isolation that is a combination of enzymatic, physical and chemical treatment to disrupt cells. We successfully isolated high quality intact total RNA with yields ranging from 23.13 ± 0.40 to 61.51 ± 0.27 µg and the 260/280 purity ratio of 1.95 ± 0.01 to 2.05 ± 0.01 from Staphylococcus aureus, Staphylococcus epidermidis, Enterococcus faecalis, and Mycobacterium smegmatis. These results represents a significantly enhanced yield and purity compared to other combination of techniques which we performed. Compared to previous studies the yield obtained by this method is high for the studied organisms. Furthermore the yielded RNA was successfully used for downstream applications such as quantitative real time PCR. The described method can be easily optimised and used for various bacteria.


Sujet(s)
ARN bactérien , ARN bactérien/génétique , ARN bactérien/isolement et purification , Staphylococcus aureus/génétique , Staphylococcus aureus/isolement et purification , Bactéries à Gram positif/génétique , Bactéries à Gram positif/isolement et purification , Staphylococcus epidermidis/génétique , Staphylococcus epidermidis/isolement et purification , Enterococcus faecalis/génétique , Enterococcus faecalis/isolement et purification , Mycobacterium smegmatis/génétique
8.
Appl Environ Microbiol ; 90(9): e0087624, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39158272

RÉSUMÉ

The horizontal transfer of antibiotic resistance genes among bacteria is a pressing global issue. The bacterial defense system clustered regularly interspaced short palindromic repeats (CRISPR)-Cas acts as a barrier to the spread of antibiotic resistance plasmids, and CRISPR-Cas-based antimicrobials can be effective to selectively deplete antibiotic-resistant bacteria. While significant surveillance efforts monitor the spread of antibiotic-resistant bacteria in the clinical context, a major, often overlooked aspect of the issue is resistance emergence in agriculture. Farm animals are commonly treated with antibiotics, and antibiotic resistance in agriculture is on the rise. Yet, CRISPR-Cas efficacy has not been investigated in this setting. Here, we evaluate the prevalence of CRISPR-Cas in agricultural Enterococcus faecalis strains and its antiplasmid efficacy in an agricultural niche: manure. Analyzing 1,986 E. faecalis genomes from human and animal hosts, we show that the prevalence of CRISPR-Cas subtypes is similar between clinical and agricultural E. faecalis strains. Using plasmid conjugation assays, we found that CRISPR-Cas is a significant barrier against resistance plasmid transfer in manure. Finally, we used a CRISPR-based antimicrobial approach to cure resistant E. faecalis of erythromycin resistance, but this was limited by delivery efficiency of the CRISPR antimicrobial in manure. However, immunization of bacteria against resistance gene acquisition in manure was highly effective. Together, our results show that E. faecalis CRISPR-Cas is prevalent and effective in an agricultural setting and has the potential to be utilized for depleting antibiotic-resistant populations. Our work has broad implications for tackling antibiotic resistance in the increasingly relevant agricultural setting, in line with a One Health approach.IMPORTANCEAntibiotic resistance is a growing global health crisis in human and veterinary medicine. Previous work has shown technologies based on CRISPR-Cas-a bacterial defense system-to be effective in tackling antibiotic resistance. Here we test if CRISPR-Cas is present and effective in agricultural niches, specifically in the ubiquitously present bacterium, Enterococcus faecalis. We show that CRISPR-Cas is both prevalent and functional in manure and has the potential to be used to specifically kill bacteria carrying antibiotic resistance genes. This study demonstrates the utility of CRISPR-Cas-based strategies for control of antibiotic resistance in agricultural settings.


Sujet(s)
Antibactériens , Systèmes CRISPR-Cas , Résistance bactérienne aux médicaments , Enterococcus faecalis , Transfert horizontal de gène , Fumier , Plasmides , Plasmides/génétique , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Animaux , Fumier/microbiologie , Antibactériens/pharmacologie , Résistance bactérienne aux médicaments/génétique , Humains , Suidae , Bovins
9.
Genome Res ; 34(7): 1081-1088, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39134411

RÉSUMÉ

Studies of bacterial adaptation and evolution are hampered by the difficulty of measuring traits such as virulence, drug resistance, and transmissibility in large populations. In contrast, it is now feasible to obtain high-quality complete assemblies of many bacterial genomes thanks to scalable high-accuracy long-read sequencing technologies. To exploit this opportunity, we introduce a phenotype- and alignment-free method for discovering coselected and epistatically interacting genomic variation from genome assemblies covering both core and accessory parts of genomes. Our approach uses a compact colored de Bruijn graph to approximate the intragenome distances between pairs of loci for a collection of bacterial genomes to account for the impacts of linkage disequilibrium (LD). We demonstrate the versatility of our approach to efficiently identify associations between loci linked with drug resistance and adaptation to the hospital niche in the major human bacterial pathogens Streptococcus pneumoniae and Enterococcus faecalis.


Sujet(s)
Enterococcus faecalis , Épistasie , Génome bactérien , Streptococcus pneumoniae , Streptococcus pneumoniae/génétique , Enterococcus faecalis/génétique , Déséquilibre de liaison , Humains , Génomique/méthodes
10.
Nat Commun ; 15(1): 6955, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39138193

RÉSUMÉ

The prevalence of multidrug resistant (MDR) bacterial infections continues to rise as the development of antibiotics needed to combat these infections remains stagnant. MDR enterococci are a major contributor to this crisis. A potential therapeutic approach for combating MDR enterococci is bacteriophage (phage) therapy, which uses lytic viruses to infect and kill pathogenic bacteria. While phages that lyse some strains of MDR enterococci have been identified, other strains display high levels of resistance and the mechanisms underlying this resistance are poorly defined. Here, we use a CRISPR interference (CRISPRi) screen to identify a genetic locus found on a mobilizable plasmid from Enterococcus faecalis involved in phage resistance. This locus encodes a putative serine recombinase followed by a Type IV restriction enzyme (TIV-RE) that we show restricts the replication of phage phi47 in vancomycin-resistant E. faecalis. We further find that phi47 evolves to overcome restriction by acquiring a missense mutation in a TIV-RE inhibitor protein. We show that this inhibitor, termed type IV restriction inhibiting factor A (tifA), binds and inactivates diverse TIV-REs. Overall, our findings advance our understanding of phage defense in drug-resistant E. faecalis and provide mechanistic insight into how phages evolve to overcome antiphage defense systems.


Sujet(s)
Bactériophages , Enterococcus faecalis , Protéines virales , Enterococcus faecalis/virologie , Enterococcus faecalis/génétique , Bactériophages/génétique , Bactériophages/physiologie , Protéines virales/métabolisme , Protéines virales/génétique , DNA restriction enzymes/métabolisme , DNA restriction enzymes/génétique , Multirésistance bactérienne aux médicaments/génétique , Plasmides/génétique , Entérocoques résistants à la vancomycine/génétique , Antibactériens/pharmacologie , Protéines bactériennes/métabolisme , Protéines bactériennes/génétique
11.
World J Microbiol Biotechnol ; 40(10): 289, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39102038

RÉSUMÉ

This study presents the empirical findings of an in-depth genomic analysis of Enterococcus faecalis and Enterococcus lactis isolates from South Africa. It offers valuable insights into their genetic characteristics and their significant implications for public health. The study uncovers nuanced variations in the gene content of these isolates, despite their similar GC contents, providing a comprehensive view of the evolutionary diversity within the species. Genomic islands are identified, particularly in E. faecalis, emphasizing its propensity for horizontal gene transfer and genetic diversity, especially in terms of antibiotic resistance genes. Pangenome analysis reveals the existence of a core genome, accounting for a modest proportion of the total genes, with 2157 core genes, 1164 shell genes, and 4638 cloud genes out of 7959 genes in 52 South African E. faecalis genomes (2 from this study, 49 south Africa genomes downloaded from NCBI, and E. faecalis reference genome). Detecting large-scale genomic rearrangements, including chromosomal inversions, underscores the dynamic nature of bacterial genomes and their role in generating genetic diversity. The study uncovers an array of antibiotic resistance genes, with trimethoprim, tetracycline, glycopeptide, and multidrug resistance genes prevalent, raising concerns about the effectiveness of antibiotic treatment. Virulence gene profiling unveils a diverse repertoire of factors contributing to pathogenicity, encompassing adhesion, biofilm formation, stress resistance, and tissue damage. These empirical findings provide indispensable insights into these bacteria's genomic dynamics, antibiotic resistance mechanisms, and virulence potential, underlining the pressing need to address antibiotic resistance and implement robust control measures.


Sujet(s)
Antibactériens , Enterococcus faecalis , Variation génétique , Génome bactérien , Facteurs de virulence , République d'Afrique du Sud , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/pathogénicité , Enterococcus faecalis/isolement et purification , Virulence/génétique , Antibactériens/pharmacologie , Facteurs de virulence/génétique , Humains , Résistance bactérienne aux médicaments/génétique , Ilots génomiques/génétique , Infections bactériennes à Gram positif/microbiologie , Enterococcus/génétique , Enterococcus/effets des médicaments et des substances chimiques , Enterococcus/pathogénicité , Enterococcus/isolement et purification , Enterococcus/classification , Phylogenèse , Transfert horizontal de gène , Génomique , Tests de sensibilité microbienne
12.
Virus Res ; 347: 199426, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38960003

RÉSUMÉ

Enterococci are robust Gram-positive bacteria that pose a significant threat in healthcare settings due to antibiotic resistance, with vancomycin-resistant enterococci (VRE) most prominent. To tackle this issue, bacteriophages (bacterial viruses) can be exploited as they specifically and efficiently target bacteria. Here, we successfully isolated and characterised a set of novel phages: SHEF10, SHEF11, SHEF13, SHEF14, and SHEF16 which target E. faecalis (SHEF10,11,13), or E. faecium (SHEF13, SHEF14 & SHEF16) strains including a range of clinical and VRE isolates. Genomic analysis shows that all phages are strictly lytic and diverse in terms of genome size and content, quickly and effectively lysing strains at different multiplicity of infections. Detailed analysis of the broad host-range SHEF13 phage revealed the crucial role of the enterococcal polysaccharide antigen (EPA) variable region in its infection of E. faecalis V583. In parallel, the discovery of a carbohydrate-targeting domain (CBM22) found conserved within the three phage genomes indicates a role in cell surface interactions that may be important in phage-bacterial interactons. These findings advance our comprehension of phage-host interactions and pave the way for targeted therapeutic strategies against antibiotic-resistant enterococcal infections.


Sujet(s)
Bactériophages , Enterococcus faecalis , Génome viral , Spécificité d'hôte , Bactériophages/génétique , Bactériophages/physiologie , Bactériophages/classification , Bactériophages/isolement et purification , Enterococcus faecalis/virologie , Enterococcus faecalis/génétique , Enterococcus faecium/virologie , Enterococcus faecium/génétique , Enterococcus/virologie , Enterococcus/génétique , Entérocoques résistants à la vancomycine/virologie , Entérocoques résistants à la vancomycine/génétique , Infections bactériennes à Gram positif/microbiologie , Humains
13.
BMC Infect Dis ; 24(1): 671, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965470

RÉSUMÉ

BACKGROUND: Vancomycin-resistant enterococci (VRE) are important pathogens categorized as high-priority bacteria in the Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics published by the World Health Organization. The aim of this study was to determine the risk factors, resistance, virulence, mobilomes associated with multidrug-resistant and clonal lineages of Enterococcus faecium and faecalis circulating among hospitalized patients following the health system in South Africa, using whole genome sequencing (WGS). METHODS: A cross-sectional study was conducted during a two-month periods among hospitalized patients in 2017. Rectal swabs were collected from patients admitted to medical and surgical wards in an urban tertiary hospital, and a rural district hospital in uMgungundlovu district, South Africa. Enterococci were screened for vancomycin resistance on bile esculin azide agar supplemented with 6 mg/L of vancomycin and confirmation of VRE was done using ROSCO kits. Conventional and real-time PCR methods were used to ascertain the presence of VanA, VanB, VanC-2/3 and VanC-1 genes. All six multidrug-resistant Enterococcus faecalis and faecium selected were identified using multiplexed paired-end libraries (2 × 300 bp) with the Nextera XT DNA sample preparation kit (Illumina, San Diego, CA, USA) and genome sequencing was done using Illumina MiSeq instrument with 100× coverage at the National Institute of Communicable Diseases Sequencing Core Facility, South Africa. Antibiotic resistance genes, virulence factors, plasmids, integrons and CRISPR were characterized using RAST, ResFinder, VirulenceFinder, PlasmidFinder, PHAST and ISFinder respectively. RESULTS: Sequencing analysis revealed that these strains harbouring numerous resistance genes to glycopeptides (vanC[100%], vex3[100%], vex2[83,33%] and vanG[16,66%]), macrolides, lincosamides, sterptogramine B (ermB[33,32%], Isa[16,66%], emeA[16,66%]) and tetracyclines (tetM[33,32%]) in both district and tertiary hospitals. Multidrug efflux pumps including MATE, MFS and pmrA conferring resistance to several classes of antibiotics were also identified. The main transposable elements observed were in the Tn3 family, specifically Tn1546. Four single sequence types (STs) were identified among E. faecium in the district hospital, namely ST822, ST636, ST97 along with a novel ST assigned ST1386, while one lineage, ST29 was detected in the tertiary hospital. CONCLUSION: The study reveals the genetic diversity and high pathogenicity of multidrug-resistant Enterococcus faecalis and faecium circulating among hospitalized patients. It underlines the necessity to implement routine screening of admitted patients coupled with infection control procedures, antimicrobial stewardship and awareness should be strengthened to prevent and/or contain the carriage and spread of multidrug resistant E. faecium and E. faecalis in hospitals and communities in South Africa.


Sujet(s)
Multirésistance bactérienne aux médicaments , Enterococcus faecalis , Enterococcus faecium , Infections bactériennes à Gram positif , Séquençage du génome entier , Humains , République d'Afrique du Sud/épidémiologie , Enterococcus faecium/génétique , Enterococcus faecium/effets des médicaments et des substances chimiques , Enterococcus faecium/isolement et purification , Études transversales , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/isolement et purification , Mâle , Infections bactériennes à Gram positif/microbiologie , Infections bactériennes à Gram positif/épidémiologie , Multirésistance bactérienne aux médicaments/génétique , Femelle , Adulte , Adulte d'âge moyen , Antibactériens/pharmacologie , Jeune adulte , Entérocoques résistants à la vancomycine/génétique , Entérocoques résistants à la vancomycine/isolement et purification , Entérocoques résistants à la vancomycine/effets des médicaments et des substances chimiques , Sujet âgé , Tests de sensibilité microbienne , Adolescent , Génome bactérien , Facteurs de virulence/génétique , Hospitalisation , Virulence/génétique
14.
BMC Med Genomics ; 17(1): 174, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38951840

RÉSUMÉ

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.


Sujet(s)
Enterococcus faecium , Infections bactériennes à Gram positif , Linézolide , Tests de sensibilité microbienne , Linézolide/pharmacologie , Humains , Chine/épidémiologie , Enterococcus faecium/génétique , Enterococcus faecium/effets des médicaments et des substances chimiques , Enterococcus faecium/isolement et purification , Infections bactériennes à Gram positif/microbiologie , Infections bactériennes à Gram positif/épidémiologie , Infections bactériennes à Gram positif/traitement médicamenteux , Mâle , Adulte d'âge moyen , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/isolement et purification , Femelle , Vancomycine/pharmacologie , Antibactériens/pharmacologie , Épidémiologie moléculaire , Adulte , Résistance à la vancomycine/génétique , Sujet âgé , Études rétrospectives , Entérocoques résistants à la vancomycine/génétique , Entérocoques résistants à la vancomycine/effets des médicaments et des substances chimiques , Entérocoques résistants à la vancomycine/isolement et purification , Jeune adulte , Enterococcus/génétique , Enterococcus/effets des médicaments et des substances chimiques , Enterococcus/isolement et purification
15.
Nature ; 632(8023): 174-181, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38987594

RÉSUMÉ

Changes in the gut microbiome have pivotal roles in the pathogenesis of acute graft-versus-host disease (aGVHD) after allogenic haematopoietic cell transplantation (allo-HCT)1-6. However, effective methods for safely resolving gut dysbiosis have not yet been established. An expansion of the pathogen Enterococcus faecalis in the intestine, associated with dysbiosis, has been shown to be a risk factor for aGVHD7-10. Here we analyse the intestinal microbiome of patients with allo-HCT, and find that E. faecalis escapes elimination and proliferates in the intestine by forming biofilms, rather than by acquiring drug-resistance genes. We isolated cytolysin-positive highly pathogenic E. faecalis from faecal samples and identified an anti-E. faecalis enzyme derived from E. faecalis-specific bacteriophages by analysing bacterial whole-genome sequencing data. The antibacterial enzyme had lytic activity against the biofilm of E. faecalis in vitro and in vivo. Furthermore, in aGVHD-induced gnotobiotic mice that were colonized with E. faecalis or with patient faecal samples characterized by the domination of Enterococcus, levels of intestinal cytolysin-positive E. faecalis were decreased and survival was significantly increased in the group that was treated with the E. faecalis-specific enzyme, compared with controls. Thus, administration of a phage-derived antibacterial enzyme that is specific to biofilm-forming pathogenic E. faecalis-which is difficult to eliminate with existing antibiotics-might provide an approach to protect against aGVHD.


Sujet(s)
Bactériophages , Enterococcus faecalis , Microbiome gastro-intestinal , Maladie du greffon contre l'hôte , Adulte , Sujet âgé , Animaux , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Jeune adulte , Bactériophages/enzymologie , Bactériophages/génétique , Biofilms/effets des médicaments et des substances chimiques , Biofilms/croissance et développement , Dysbiose/complications , Dysbiose/microbiologie , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/génétique , Enterococcus faecalis/croissance et développement , Enterococcus faecalis/métabolisme , Enterococcus faecalis/virologie , Fèces/microbiologie , Axénie , Maladie du greffon contre l'hôte/complications , Maladie du greffon contre l'hôte/microbiologie , Maladie du greffon contre l'hôte/prévention et contrôle , Maladie du greffon contre l'hôte/thérapie , Transplantation de cellules souches hématopoïétiques/effets indésirables , Techniques in vitro , Intestins/effets des médicaments et des substances chimiques , Intestins/microbiologie , Perforine/métabolisme , Facteurs de risque , Transplantation homologue/effets indésirables , Séquençage du génome entier , Résistance bactérienne aux médicaments/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie
16.
Mol Microbiol ; 122(2): 230-242, 2024 08.
Article de Anglais | MEDLINE | ID: mdl-38994873

RÉSUMÉ

Enterococcus faecalis is an opportunistic pathogen frequently causing nosocomial infections. The virulence of this organism is underpinned by its capacity to evade phagocytosis, allowing dissemination in the host. Immune evasion requires a surface polysaccharide produced by all enterococci, known as the enterococcal polysaccharide antigen (EPA). EPA consists of a cell wall-anchored rhamnose backbone substituted by strain-specific polysaccharides called 'decorations', essential for the biological activity of this polymer. However, the structural determinants required for innate immune evasion remain unknown, partly due to a lack of suitable validated assays. Here, we describe a quantitative, in vitro assay to investigate how EPA decorations alter phagocytosis. Using the E. faecalis model strain OG1RF, we demonstrate that a mutant with a deletion of the locus encoding EPA decorations can be used as a platform strain to express heterologous decorations, thereby providing an experimental system to investigate the inhibition of phagocytosis by strain-specific decorations. We show that the aggregation of cells lacking decorations is increasing phagocytosis and that this process does not involve the recognition of lipoproteins by macrophages. Collectively, our work provides novel insights into innate immune evasion by enterococci and paves the way for further studies to explore the structure/function relationship of EPA decorations.


Sujet(s)
Enterococcus faecalis , Échappement immunitaire , Lipoprotéines , Macrophages , Phagocytose , Enterococcus faecalis/immunologie , Enterococcus faecalis/métabolisme , Enterococcus faecalis/génétique , Lipoprotéines/métabolisme , Lipoprotéines/génétique , Macrophages/microbiologie , Macrophages/immunologie , Macrophages/métabolisme , Polyosides bactériens/métabolisme , Polyosides bactériens/immunologie , Humains , Antigènes bactériens/métabolisme , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Immunité innée , Virulence , Animaux , Souris
17.
Nat Microbiol ; 9(9): 2448-2461, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38965331

RÉSUMÉ

Interactions between microbiota and enteric pathogens can promote colonization resistance or enhance pathogenesis. The pathobiont Enterococcus faecalis increases enterohaemorrhagic E. coli (EHEC) virulence by upregulating Type 3 Secretion System (T3SS) expression, effector translocation, and attaching and effacing (AE) lesion formation on enterocytes, but the mechanisms underlying this remain unknown. Using co-infection of organoids, metabolomics, supplementation experiments and bacterial genetics, here we show that co-culture of EHEC with E. faecalis increases the xanthine-hypoxanthine pathway activity and adenine biosynthesis. Adenine or E. faecalis promoted T3SS gene expression, while transcriptomics showed upregulation of adeP expression, which encodes an adenine importer. Mechanistically, adenine relieved High hemolysin activity (Hha)-dependent repression of T3SS gene expression in EHEC and promoted AE lesion formation in an AdeP-dependent manner. Microbiota-derived purines, such as adenine, support multiple beneficial host responses; however, our data show that this metabolite also increases EHEC virulence, highlighting the complexity of pathogen-microbiota-host interactions in the gut.


Sujet(s)
Adénine , Enterococcus faecalis , Escherichia coli entérohémorrhagique , Régulation de l'expression des gènes bactériens , Systèmes de sécrétion de type III , Escherichia coli entérohémorrhagique/génétique , Escherichia coli entérohémorrhagique/pathogénicité , Escherichia coli entérohémorrhagique/métabolisme , Virulence , Systèmes de sécrétion de type III/métabolisme , Systèmes de sécrétion de type III/génétique , Enterococcus faecalis/génétique , Enterococcus faecalis/métabolisme , Enterococcus faecalis/pathogénicité , Adénine/métabolisme , Adénine/pharmacologie , Animaux , Protéines Escherichia coli/métabolisme , Protéines Escherichia coli/génétique , Souris , Infections à Escherichia coli/microbiologie , Humains , Hémolysines/métabolisme , Hémolysines/génétique , Interactions hôte-pathogène , Techniques de coculture , Entérocytes/microbiologie , Entérocytes/métabolisme , Xanthine/métabolisme , Hypoxanthine/métabolisme , Facteurs de virulence/métabolisme , Facteurs de virulence/génétique , Microbiome gastro-intestinal
18.
Can J Microbiol ; 70(10): 446-460, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39079170

RÉSUMÉ

With antimicrobial resistance (AMR) rapidly evolving in pathogens, quick and accurate identification of genetic determinants of phenotypic resistance is essential for improving surveillance, stewardship, and clinical mitigation. Machine learning (ML) models show promise for AMR prediction in diagnostics but require a deep understanding of internal processes to use effectively. Our study utilised AMR gene, pangenomic, and predicted plasmid features from 647 Enterococcus faecium and Enterococcus faecalis genomes across the One Health continuum, along with corresponding resistance phenotypes, to develop interpretive ML classifiers. Vancomycin resistance could be predicted with 99% accuracy with AMR gene features, 98% with pangenome features, and 96% with plasmid clusters. Top pangenome features overlapped with the resistance genes of the vanA operon, which are often laterally transmitted via plasmids. Doxycycline resistance prediction achieved approximately 92% accuracy with pangenome features, with the top feature being elements of Tn916 conjugative transposon, a tet(M) carrier. Erythromycin resistance prediction models achieved about 90% accuracy, but top features were negatively correlated with resistance due to the confounding effect of population structure. This work demonstrates the importance of reviewing ML models' features to discern biological relevance even when achieving high-performance metrics. Our workflow offers the potential to propose hypotheses for experimental testing, enhancing the understanding of AMR mechanisms, which are crucial for combating the AMR crisis.


Sujet(s)
Antibactériens , Résistance bactérienne aux médicaments , Enterococcus faecalis , Enterococcus faecium , Génome bactérien , Apprentissage machine , Plasmides , Enterococcus faecalis/génétique , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecium/génétique , Enterococcus faecium/effets des médicaments et des substances chimiques , Antibactériens/pharmacologie , Résistance bactérienne aux médicaments/génétique , Plasmides/génétique , Humains , Tests de sensibilité microbienne , Infections bactériennes à Gram positif/microbiologie , Protéines bactériennes/génétique
19.
Nat Commun ; 15(1): 6332, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068147

RÉSUMÉ

Bacteriocins are antimicrobial peptides that are naturally produced by many bacteria. They hold great potential in the fight against antibiotic resistant bacteria, including ESKAPE pathogens. Engineered live biotherapeutic products (eLBPs) that secrete bacteriocins can be created to deliver targeted bacteriocin production. Here we develop a modular bacteriocin secretion platform that can be used to express and secrete multiple bacteriocins from non-pathogenic Escherichia coli host strains. As a proof of concept we create Enterocin A (EntA) and Enterocin B (EntB) secreting strains that show strong antimicrobial activity against Enterococcus faecalis and Enterococcus faecium in vitro, and characterise this activity in both solid culture and liquid co-culture. We then develop a Lotka-Volterra model that can be used to capture the interactions of these competitor strains. We show that simultaneous exposure to EntA and EntB can delay Enterococcus growth. Our system has the potential to be used as an eLBP to secrete additional bacteriocins for the targeted killing of pathogenic bacteria.


Sujet(s)
Antibactériens , Bactériocines , Enterococcus faecalis , Enterococcus faecium , Escherichia coli , Bactériocines/pharmacologie , Bactériocines/métabolisme , Bactériocines/biosynthèse , Enterococcus faecalis/métabolisme , Enterococcus faecalis/effets des médicaments et des substances chimiques , Enterococcus faecalis/génétique , Enterococcus faecium/métabolisme , Enterococcus faecium/génétique , Enterococcus faecium/effets des médicaments et des substances chimiques , Escherichia coli/métabolisme , Escherichia coli/effets des médicaments et des substances chimiques , Escherichia coli/génétique , Antibactériens/pharmacologie , Tests de sensibilité microbienne , Techniques de coculture
20.
BMC Microbiol ; 24(1): 280, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-39068414

RÉSUMÉ

BACKGROUND: Enterococcus faecium and Staphylococcus aureus are the Gram-positive pathogens of the ESKAPE group, known to represent a great threat to human health due to their high virulence and multiple resistances to antibiotics. Combined, enterococci and S. aureus account for 26% of healthcare-associated infections and are the most common organisms responsible for blood stream infections. We previously showed that the peptidyl-prolyl cis/trans isomerase (PPIase) PpiC of E. faecium elicits the production of specific, opsonic, and protective antibodies that are effective against several strains of E. faecium and E. faecalis. Due to the ubiquitous characteristics of PPIases and their essential function within Gram-positive cells, we hypothesized a potential cross-reactive effect of anti-PpiC antibodies. RESULTS: Opsonophagocytic assays combined with bioinformatics led to the identification of the foldase protein PrsA as a new potential vaccine antigen in S. aureus. We show that PrsA is a stable dimeric protein able to elicit opsonic antibodies against the S. aureus strain MW2, as well as cross-binding and cross-opsonic in several S. aureus, E. faecium and E. faecalis strains. CONCLUSIONS: Given the multiple antibiotic resistances S. aureus and enterococci present, finding preventive strategies is essential to fight those two nosocomial pathogens. The study shows the potential of PrsA as an antigen to use in vaccine formulation against the two dangerous Gram-positive ESKAPE bacteria. Our findings support the idea that PPIases should be further investigated as vaccine targets in the frame of pan-vaccinomics strategy.


Sujet(s)
Protéines bactériennes , Enterococcus faecalis , Enterococcus faecium , Peptidylpropyl isomerase , Staphylococcus aureus , Staphylococcus aureus/immunologie , Staphylococcus aureus/génétique , Enterococcus faecium/immunologie , Enterococcus faecium/génétique , Protéines bactériennes/immunologie , Protéines bactériennes/génétique , Peptidylpropyl isomerase/immunologie , Peptidylpropyl isomerase/génétique , Enterococcus faecalis/immunologie , Enterococcus faecalis/génétique , Humains , Infections bactériennes à Gram positif/prévention et contrôle , Infections bactériennes à Gram positif/immunologie , Infections bactériennes à Gram positif/microbiologie , Vaccins antibactériens/immunologie , Opsonines/immunologie , Anticorps antibactériens/immunologie , Anticorps antibactériens/sang , Animaux , Réactions croisées , Souris , Antigènes bactériens/immunologie , Antigènes bactériens/génétique , Phagocytose , Infections à staphylocoques/prévention et contrôle , Infections à staphylocoques/immunologie , Infections à staphylocoques/microbiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE