Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Euro Surveill ; 29(18)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38699902

RESUMEN

BackgroundThe pet industry is expanding worldwide, particularly raw meat-based diets (RMBDs). There are concerns regarding the safety of RMBDs, especially their potential to spread clinically relevant antibiotic-resistant bacteria or zoonotic pathogens.AimWe aimed to investigate whether dog food, including RMBD, commercially available in Portugal can be a source of Salmonella and/or other Enterobacteriaceae strains resistant to last-line antibiotics such as colistin.MethodsFifty-five samples from 25 brands (21 international ones) of various dog food types from 12 suppliers were screened by standard cultural methods between September 2019 and January 2020. Isolates were characterised by phenotypic and genotypic methods, including whole genome sequencing and comparative genomics.ResultsOnly RMBD batches were contaminated, with 10 of 14 containing polyclonal multidrug-resistant (MDR) Escherichia coli and one MDR Salmonella. One turkey-based sample contained MDR Salmonella serotype 1,4,[5],12:i:- ST34/cgST142761 with similarity to human clinical isolates occurring worldwide. This Salmonella exhibited typical antibiotic resistance (bla TEM + strA-strB + sul2 + tet(B)) and metal tolerance profiles (pco + sil + ars) associated with the European epidemic clone. Two samples (turkey/veal) carried globally dispersed MDR E. coli (ST3997-complexST10/cgST95899 and ST297/cgST138377) with colistin resistance (minimum inhibitory concentration: 4 mg/L) and mcr-1 gene on IncX4 plasmids, which were identical to other IncX4 circulating worldwide.ConclusionSome RMBDs from European brands available in Portugal can be a vehicle for clinically relevant MDR Salmonella and pathogenic E. coli clones carrying genes encoding resistance to the last-line antibiotic colistin. Proactive actions within the One Health context, spanning regulatory, pet-food industry and consumer levels, are needed to mitigate these public health risks.


Asunto(s)
Antibacterianos , Escherichia coli , Carne , Salmonella , Animales , Salmonella/aislamiento & purificación , Salmonella/genética , Salmonella/efectos de los fármacos , Humanos , Portugal , Escherichia coli/aislamiento & purificación , Escherichia coli/genética , Escherichia coli/efectos de los fármacos , Perros , Antibacterianos/farmacología , Carne/microbiología , Farmacorresistencia Bacteriana Múltiple/genética , Mascotas/microbiología , Secuenciación Completa del Genoma , Microbiología de Alimentos , Pruebas de Sensibilidad Microbiana , Proteínas de Escherichia coli/genética , Colistina/farmacología , Alimentación Animal/microbiología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/epidemiología
2.
Emerg Infect Dis ; 27(8): 2221-2224, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34287135

RESUMEN

We describe enterococci in raw-frozen dog food commercialized in Europe as a source of genes encoding resistance to the antibiotic drug linezolid and of strains and plasmids enriched in antibiotic-resistance and virulence genes in hospitalized patients. Whole-genome sequencing was fundamental to linking isolates from dog food to human cases across Europe.


Asunto(s)
Enterococcus , Infecciones por Bacterias Grampositivas , Alimentación Animal , Animales , Antibacterianos/farmacología , Perros , Farmacorresistencia Bacteriana , Europa (Continente) , Infecciones por Bacterias Grampositivas/epidemiología , Infecciones por Bacterias Grampositivas/veterinaria , Humanos , Linezolid/farmacología , Pruebas de Sensibilidad Microbiana
3.
J Antimicrob Chemother ; 76(11): 2757-2764, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34450635

RESUMEN

BACKGROUND: Vancomycin resistance is mostly associated with Enterococcus faecium due to Tn1546-vanA located on narrow- and broad-host plasmids of various families. This study's aim was to analyse the effects of acquiring Tn1546-carrying plasmids with proven epidemicity in different bacterial host backgrounds. METHODS: Widespread Tn1546-carrying plasmids of different families RepA_N (n = 5), Inc18 (n = 4) and/or pHTß (n = 1), and prototype plasmids RepA_N (pRUM) and Inc18 (pRE25, pIP501) were analysed. Plasmid transferability and fitness cost were assessed using E. faecium (GE1, 64/3) and Enterococcus faecalis (JH2-2/FA202/UV202) recipient strains. Growth curves (Bioscreen C) and Relative Growth Rates were obtained in the presence/absence of vancomycin. Plasmid stability was analysed (300 generations). WGS (Illumina-MiSeq) of non-evolved and evolved strains (GE1/64/3 transconjugants, n = 49) was performed. SNP calling (Breseq software) of non-evolved strains was used for comparison. RESULTS: All plasmids were successfully transferred to different E. faecium clonal backgrounds. Most Tn1546-carrying plasmids and Inc18 and RepA_N prototypes reduced host fitness (-2% to 18%) while the cost of Tn1546 expression varied according to the Tn1546-variant and the recipient strain (9%-49%). Stability of Tn1546-carrying plasmids was documented in all cases, often with loss of phenotypic resistance and/or partial plasmid deletions. SNPs and/or indels associated with essential bacterial functions were observed on the chromosome of evolved strains, some of them linked to increased fitness. CONCLUSIONS: The stability of E. faecium Tn1546-carrying plasmids in the absence of selective pressure and the high intra-species conjugation rates might explain the persistence of vancomycin resistance in E. faecium populations despite the significant burden they might impose on bacterial host strains.


Asunto(s)
Infección Hospitalaria , Enterococcus faecium , Infecciones por Bacterias Grampositivas , Proteínas Bacterianas/genética , Infección Hospitalaria/epidemiología , Elementos Transponibles de ADN , Brotes de Enfermedades , Enterococcus faecium/genética , Infecciones por Bacterias Grampositivas/epidemiología , Humanos , Plásmidos , Vancomicina/farmacología
4.
Environ Microbiol ; 22(7): 2829-2842, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32281716

RESUMEN

Emergence and expansion of frequent multidrug-resistant (MDR) major Salmonella clones/serotypes has been a significant threat in the last years. Metal compounds, such as copper, commonly used in animal-production settings, have been pointed out as possible contributors for the selection of such strains/clones. However, the scarcity of studies limits our understanding of the impact of other metal environmental contaminants as arsenic (used in insecticides/herbicides/coccidiostats). We analysed arsenic tolerance (AsT) dispersion by phenotypic and genotypic (PCR/sequencing/I-CeuI/S1/XbaI-PFGE/hybridization) assays among Salmonella with diverse epidemiological and genetic backgrounds. Then, to better understand ars operon genetic contexts, the whole genome of five representative strains was sequenced. We found a high dispersion of ars operons conferring AsT, especially among copper-tolerant and relevant serotypes/clones related to pig-production setting. The acr3-type was found dispersed in the chromosome of diverse serotypes, including the emergent S. Rissen. Conversely, arsBII was almost confined to the MDR ST34 European clone of S. Typhimurium/S. 4,[5],12:i:-, always along with copper/silver tolerance sil + pco clusters in an integrative conjugative element. These data suggest that AsT is an essential adaptive feature for the ecological success of these epidemic clones/serotypes and alerts for global strategies to reduce arsenic-based compounds' impact thus preventing environmental/food contamination with frequent MDR foodborne pathogens.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple/genética , Operón/genética , Salmonella/efectos de los fármacos , Salmonella/genética , Animales , Arsénico/toxicidad , Células Clonales , Cobre/toxicidad , Contaminantes Ambientales/toxicidad , Genotipo , Fenotipo , Reacción en Cadena de la Polimerasa , Infecciones por Salmonella/microbiología , Serogrupo , Porcinos
5.
J Antimicrob Chemother ; 75(1): 30-35, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31605129

RESUMEN

OBJECTIVES: Increasing numbers of linezolid-resistant Enterococcus carrying optrA are being reported across different niches worldwide. We aimed to characterize the first optrA-carrying Enterococcus faecalis obtained from food-producing animals and retail meat samples in Tunisia. METHODS: Seven optrA-carrying E. faecalis obtained from chicken faeces (n=3, August 2017) and retail chicken meat (n=4, August 2017) in Tunisia were analysed. Antimicrobial susceptibility was determined by disc diffusion, broth microdilution and Etest against 13 antibiotics, linezolid and tedizolid, respectively (EUCAST/CLSI). optrA stability (∼600 bacterial generations), transfer (filter mating) and location (S1-PFGE/hybridization) were characterized. WGS (Illumina-HiSeq) was done for four representatives that were analysed through in silico and genomic mapping tools. RESULTS: Four MDR clones carrying different virulence genes were identified in chicken faeces (ST476) and retail meat (the same ST476 clone plus ST21 and ST859) samples. MICs of linezolid and tedizolid were stably maintained at 8 and 1-2 mg/L, respectively. optrA was located in the same transferable chromosomal Tn6674-like element in ST476 and ST21 clones, similar to isolates from pigs in Malaysia and humans in China. ST859 carried a non-conjugative plasmid of ∼40 kb with an impB-fexA-optrA segment, similar to plasmids from pigs and humans in China. CONCLUSIONS: The same chromosomal and transferable Tn6674-like element was identified in different E. faecalis clones from humans and animals. The finding of retail meat contaminated with the same linezolid-resistant E. faecalis strain obtained from a food-producing animal highlights the potential role of the food chain in the worrisome dissemination of optrA that can be stably maintained without selective pressure over generations.


Asunto(s)
Animales Domésticos/microbiología , Antibacterianos/farmacología , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecalis/genética , Infecciones por Bacterias Grampositivas/veterinaria , Linezolid/farmacología , Aves de Corral/microbiología , Animales , Pollos , ADN Bacteriano/genética , Farmacorresistencia Bacteriana/genética , Enterococcus faecalis/clasificación , Microbiología de Alimentos , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus , Túnez , Secuenciación Completa del Genoma
6.
J Antimicrob Chemother ; 75(9): 2416-2423, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32607549

RESUMEN

OBJECTIVES: poxtA is the most recently described gene conferring acquired resistance to linezolid, a relevant antibiotic for treating enterococcal infections. We retrospectively screened for poxtA in diverse enterococci and aimed to characterize its genetic/genomic contexts. METHODS: poxtA was screened by PCR in 812 enterococci from 458 samples (hospitals/healthy humans/wastewater/animals/retail food) obtained in Portugal/Angola/Tunisia (1996-2019). Antimicrobial susceptibility testing was performed for 13 antibiotics (EUCAST/CLSI). poxtA stability (∼500 generations), transfer (filter mating), clonality (SmaI-PFGE) and location (S1-PFGE/hybridization) were tested. WGS (Illumina-HiSeq) was performed for clonal representatives. RESULTS: poxtA was detected in Enterococcus faecium from six samples (1.3%): a healthy human (rectal swab) in Porto, Portugal (ST32/2001); four farm cows (milk) in Mateur, Tunisia (ST1058/2015); and a hospitalized patient (faeces) in Matosinhos, Portugal (ST1058/2015). All expressed resistance to linezolid (MIC = 8 mg/L), chloramphenicol, tetracycline and erythromycin, with variable resistance to ciprofloxacin and streptomycin. ST1058-poxtA-carrying isolates from Tunisia and Portugal differed by two SNPs and had similar plasmid content. poxtA, located in an IS1216-flanked Tn6246-like element, co-hybridized with fexB on one or more plasmids per isolate (one to three plasmids of 30-100 kb), was stable after several generations and transferred only from ST1058. ST1058 strains carried resistance/virulence genes (Efmqnr/acm) possibly induced under selective quinolone treatment. CONCLUSIONS: poxtA has been circulating in Portugal since at least 2001, corresponding to the oldest description worldwide to date. We also extend the reservoir of poxtA to bovines. The similar linezolid-resistant poxtA-carrying strains colonizing humans and livestock on different continents, and without a noticeable relationship, suggests a recent transmission event or convergent evolution of E. faecium populations in different hosts and geographic regions.


Asunto(s)
Enterococcus faecium , Infecciones por Bacterias Grampositivas , Angola , Animales , Antibacterianos/farmacología , Bovinos , Farmacorresistencia Bacteriana , Enterococcus faecalis , Enterococcus faecium/genética , Infecciones por Bacterias Grampositivas/epidemiología , Infecciones por Bacterias Grampositivas/veterinaria , Humanos , Linezolid/farmacología , Pruebas de Sensibilidad Microbiana , Portugal/epidemiología , Estudios Retrospectivos , Túnez
7.
Environ Microbiol ; 21(8): 3046-3062, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31162871

RESUMEN

Wild birds have been suggested to be reservoirs of antimicrobial resistant and/or pathogenic Enterococcus faecalis (Efs) strains, but the scarcity of studies and available sequences limit our understanding of the population structure of the species in these hosts. Here, we analysed the clonal and plasmid diversity of 97 Efs isolates from wild migratory birds. We found a high diversity, with most sequence types (STs) being firstly described here, while others were found in other hosts including some predominant in poultry. We found that pheromone-responsive plasmids predominate in wild bird Efs while 35% of the isolates entirely lack plasmids. Then, to better understand the ecology of the species, the whole genome of fivestrains with known STs (ST82, ST170, ST16 and ST55) were sequenced and compared with all the Efs genomes available in public databases. Using several methods to analyse core and accessory genomes (AccNET, PLACNET, hierBAPS and PANINI), we detected differences in the accessory genome of some lineages (e.g. ST82) demonstrating specific associations with birds. Conversely, the genomes of other Efs lineages exhibited divergence in core and accessory genomes, reflecting different adaptive trajectories in various hosts. This pangenome divergence, horizontal gene transfer events and occasional epidemic peaks could explain the population structure of the species.


Asunto(s)
Aves/microbiología , Enterococcus faecalis/genética , Filogenia , Animales , Animales Salvajes , Regulación Bacteriana de la Expresión Génica , Transferencia de Gen Horizontal , Genoma Bacteriano , Especificidad del Huésped
8.
J Antimicrob Chemother ; 74(10): 2865-2869, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31243458

RESUMEN

OBJECTIVES: The epidemiology of Enterococcus resistant to priority antibiotics including linezolid has mainly been investigated in developed countries and especially in hospitals. We aimed to evaluate the contribution of different non-human reservoirs for the burden of MDR enterococci in Tunisia, where scarce data are available. METHODS: Samples (n = 287) were collected from urban wastewater (n = 57), retail meat (n = 29; poultry/bovine/ovine), milk (n = 89; bovine/ovine), farm animal faeces (n = 80; poultry/bovine/ovine) and pets (n = 32; rabbit/dogs/cats/birds) in different Tunisian regions (2014-17). They were plated onto Slanetz-Bartley agar after pre-enrichment without antibiotics. Standard methods were used for bacterial identification and characterization of antibiotic resistance and virulence genes (PCR), antibiotic susceptibility testing (disc diffusion/broth microdilution; EUCAST/CLSI) and clonality (SmaI-PFGE/MLST). RESULTS: All samples carried Enterococcus (n = 377 isolates) resistant to antibiotics considered to be critical or highly important by WHO. Even without antibiotic selection, 38% of Enterococcus faecalis (Efs) and 22% of Enterococcus faecium (Efm) were identified as MDR. Linezolid-resistant isolates (5%; MIC = 8 mg/L) comprised six poxtA-carrying Efm (cow milk), seven optrA-carrying Efs (chicken faeces/meat) and five Efm lacking cfr/optrA/poxtA (poultry/bovine/ovine/wastewater). Clinically relevant Efm clones (clade A1) were identified in animal/meat sources. Ampicillin resistance (1%) was confined to ST18/ST78-like MDR Efm clones from bovine meat/milk samples carrying relevant virulence markers (e.g. ptsD/IS16). CONCLUSIONS: This study provides evidence of the contribution of livestock and foodstuffs to the dispersal of acquired linezolid resistance genes including poxtA and optrA. We report the first poxtA-carrying Efm in Tunisia, and for the first time in bovine samples, stressing the urgent need for alternative measures to counteract the spread of linezolid-resistant enterococci globally.


Asunto(s)
Antibacterianos/farmacología , Farmacorresistencia Bacteriana , Enterococcus/efectos de los fármacos , Microbiología de Alimentos , Infecciones por Bacterias Grampositivas/veterinaria , Linezolid/farmacología , Factores de Virulencia/genética , Animales , Animales Domésticos , Medios de Cultivo , Enterococcus/clasificación , Enterococcus/genética , Enterococcus/aislamiento & purificación , Microbiología Ambiental , Infecciones por Bacterias Grampositivas/microbiología , Pruebas de Sensibilidad Microbiana , Mascotas , Reacción en Cadena de la Polimerasa , Túnez
9.
Appl Environ Microbiol ; 85(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562170

RESUMEN

Chlorhexidine (CHX) is a broad-spectrum antiseptic widely used in community and clinical contexts for many years that has recently acquired higher relevance in nosocomial infection control worldwide. Despite this, CHX tolerance among Enterococcus faecium bacteria, representing one of the leading agents causing nosocomial infections, has been poorly understood. This study provides new phenotypic and molecular data for better identification of CHX-tolerant E. faecium subpopulations in community and clinical contexts. The chlorhexidine MIC (MICCHX) distribution of 106 E. faecium isolates suggested the occurrence of tolerant subpopulations in diverse sources (human, animal, food, environment) and phylogenomic backgrounds (clades A1/A2/B), with predominance in clade A1. They carried a specific variant of the 2CS-CHXT operon, identified here. It encodes glucose and amino acid-polyamine-organocation family transporters, besides the DNA-binding response regulator ChtR, with a P102H mutation previously described only in CHX-tolerant clade A1 E. faecium, and the ChtS sensor. 2CS-CHXT seems to be associated with three regulons modulating diverse bacterial biological functions. Combined data from normal MIC distribution and 2CS-CHXT operon characterization support a tentative epidemiological cutoff (ECOFF) of 8 mg/liter to CHX, which is useful to detect tolerant E. faecium populations in future surveillance studies. The spread of tolerant E. faecium in diverse epidemiological backgrounds calls for the prudent use of CHX in multiple contexts.IMPORTANCE Chlorhexidine is one of the substances included in the World Health Organization's list of essential medicines, which comprises the safest and most effective medicines needed in global health systems. Although it has been widely applied as a disinfectant and antiseptic in health care (skin, hands, mouthwashes, eye drops) since the 1950s, its use in hospitals to prevent nosocomial infections has increased worldwide in recent years. Here, we provide a comprehensive study on chlorhexidine tolerance among strains of Enterococcus faecium, one of the leading nosocomial agents worldwide, and identify a novel 2CS-CHXT operon as a signature of tolerant strains occurring in diverse phylogenomic groups. Our data allowed for the proposal of a tentative epidemiological cutoff of 8 mg/liter, which is useful to detect tolerant E. faecium populations in surveillance studies in community and clinical contexts. The prediction of 2CS-CHXT regulons will also facilitate the design of future experimental studies to better uncover chlorhexidine tolerance among E. faecium bacteria.


Asunto(s)
Antiinfecciosos Locales/farmacología , Clorhexidina/farmacología , Farmacorresistencia Bacteriana/genética , Enterococcus faecium/efectos de los fármacos , Operón/fisiología , Enterococcus faecium/genética , Enterococcus faecium/fisiología , Filogenia
10.
Drug Resist Updat ; 40: 25-39, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30447411

RESUMEN

Vancomycin-resistant enterococci (VRE) are important nosocomial pathogens. Invasive VRE infections are difficult to treat since common therapeutic options including ampicillin and glycopeptides often fail. In vitro, most VRE remain susceptible to last-resort antibiotics such as linezolid, tigecycline and daptomycin. However, neither tigecycline nor linezolid act in a bactericidal manner, and daptomycin has proven activity only at high dosages licensed for treating enterococcal endocarditis. Despite these pharmacological and therapeutic limitations, reports on resistance to these last-resort drugs in VRE, and enterococci in general, have increased in recent years. In this review, we briefly recapitulate the current knowledge on the mode of action as well as the known and novel mechanisms of resistance and describe surveillance data on resistance to linezolid, tigecycline and daptomycin in enterococci. In addition, we also suggest a common nomenclature for designating enterococci and VRE with resistances to these important last-resort antibiotics.


Asunto(s)
Antibacterianos/farmacología , Daptomicina/farmacología , Linezolid/farmacología , Tigeciclina/farmacología , Resistencia a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Ligasas de Carbono-Oxígeno/genética , Daptomicina/uso terapéutico , Genotipo , Infecciones por Bacterias Grampositivas/tratamiento farmacológico , Humanos , Linezolid/uso terapéutico , Pruebas de Sensibilidad Microbiana , Mutación , Tigeciclina/uso terapéutico , Resistencia a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/genética
11.
J Antimicrob Chemother ; 73(2): 306-319, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29149293

RESUMEN

Objectives: The criteria for identification of Enterococcus faecium (Efm) with the ability to cause human infections are currently being debated by the European Food Safety Authority (EFSA). Strains that have an MIC of ampicillin of ≤ 2 mg/L and lack IS16/esp/hyl genes should be regarded as safe for use as feed additives in animal nutrition, despite the lack of knowledge about putative virulence marker (PVM) distribution in community Efm. We analysed the distribution of major PVM and ampicillin phenotypes in large Efm collections to investigate further the safety of strains from a public health perspective. Methods: Thirty-three PVM were assessed by PCR/sequencing among clonally disparate Efm (n = 328; 1986-2015) from different origins. We analysed ampicillin susceptibility (Etest/broth microdilution) according to EUCAST guidelines, clonal relationship (MLST) and genomic location of PVM (S1-PFGE/hybridization). Results: Infection-derived Efm were more enriched in PVM and the increase in ampicillin MIC was positively correlated with an enrichment in different PVM. PVM coding for surface (esp/sgrA/ecbA/complete acm) and pili proteins, or others enhancing colonization (hyl/ptsD/orf1481) or plasticity (IS16), were strongly associated with clinical Efm (mostly clade A1), but also observed in clades A2/B at different rates. ptsD was a good marker of ampicillin-resistant Efm. ptsD, IS16, orf1481, sgrA and hospital variants of complete pili gene clusters are proposed as markers to assess the safety of Efm strains. Conclusions: Our study expands on the distribution of PVM in diverse Efm lineages and demonstrates the enrichment in infection-derived strains of PVM not previously included in EFSA's list of Efm safety criteria. The evidence of relevant Efm infection markers can impact the risk assessment of Efm strains in different public health contexts.


Asunto(s)
Resistencia a la Ampicilina , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/patogenicidad , Infecciones por Bacterias Grampositivas/microbiología , Infecciones por Bacterias Grampositivas/veterinaria , Factores de Virulencia/genética , Animales , Electroforesis en Gel de Campo Pulsado , Enterococcus faecium/genética , Humanos , Pruebas de Sensibilidad Microbiana , Tipificación de Secuencias Multilocus , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
12.
J Antimicrob Chemother ; 72(12): 3245-3251, 2017 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-29029072

RESUMEN

OBJECTIVES: Oxazolidinone resistance is a serious limitation in the treatment of MDR Enterococcus infections. Plasmid-mediated oxazolidinone resistance has been strongly linked to animals where the use of phenicols might co-select resistance to both antibiotic families. Our goal was to assess the diversity of genes conferring phenicol/oxazolidinone resistance among diverse enterococci and to characterize the optrA genetic environment. METHODS: Chloramphenicol-resistant isolates (>16 mg/L, n = 245) from different sources (hospitals/healthy humans/wastewaters/animals) in Portugal, Angola and Tunisia (1996-2016) were selected. Phenicol (eight cat variants, fexA, fexB) or phenicol + oxazolidinone [cfr, cfr(B), optrA] resistance genes were searched for by PCR. Susceptibility (disc diffusion/microdilution), filter mating, stability of antibiotic resistance (500 bacterial generations), plasmid typing (S1-PFGE/hybridization), MLST and WGS (Illumina-HiSeq) were performed for optrA-positive isolates. RESULTS: Resistance to phenicols (n = 181, 74%) and phenicols + oxazolidinones (n = 2, 1%) was associated with the presence of cat(A-8) (40%, predominant in hospitals and swine), cat(A-7) (29%, predominant in poultry and healthy humans), cat(A-9) (2%), fexB (2%) and fexA + optrA (1%). fexA and optrA genes were co-located in a transferable plasmid (pAF379, 72 918 bp) of two ST86 MDR Tunisian Enterococcus faecalis (wastewaters) carrying several putative virulence genes. MICs of chloramphenicol, linezolid and tedizolid were stably maintained at 64, 4 and 1 mg/L, respectively. The chimeric pAF379 comprised relics of genetic elements from different Gram-positive bacteria and origins (human/porcine). CONCLUSIONS: To the best of our knowledge, we report the first detection of optrA in an African country (Tunisia) within a transferable mosaic plasmid of different origins. Its identification in isolates from environmental sources is worrisome and alerts for the need of a concerted global surveillance on the occurrence and spread of optrA.


Asunto(s)
Farmacorresistencia Bacteriana , Enterococcus faecalis/genética , Enterococcus faecalis/aislamiento & purificación , Orden Génico , Genes Bacterianos , Plásmidos/aislamiento & purificación , Aguas Residuales/microbiología , Antibacterianos/farmacología , Cloranfenicol/farmacología , Ciudades , ADN Bacteriano/genética , Pruebas Antimicrobianas de Difusión por Disco , Enterococcus faecalis/efectos de los fármacos , Tipificación de Secuencias Multilocus , Oxazolidinonas/farmacología , Reacción en Cadena de la Polimerasa , Túnez , Secuenciación Completa del Genoma
13.
Antimicrob Agents Chemother ; 60(3): 1736-46, 2016 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-26729505

RESUMEN

This work describes the diversity and evolution of Tn5801 among enterococci, staphylococci, and streptococci based on analysis of the 5,073 genomes of these bacterial groups available in gene databases. We also examined 610 isolates of Enterococcus (from 10 countries, 1987 to 2010) for the presence of this and other known CTn-tet(M) elements due to the scarcity of data about Tn5801 among enterococci. Genome location (by ICeu-I-pulsed-field gel electrophoresis [PFGE] hybridization/integration site identification), conjugation and fitness (by standard methods), Tn5801 characterization (by long-PCR mapping/sequencing), and clonality (by PFGE/multilocus sequence typing [MLST]) were studied. Twenty-three Tn5801 variants (17 unpublished) clustered in two groups, designated "A" (25 kb; n = 14; predominant in Staphylococcus aureus) and "B" (20 kb; n = 9; predominant in Streptococcus agalactiae). The percent GC content of the common backbone suggests a streptococcal origin of Tn5801 group B, with further acquisition of a 5-kb fragment that resulted in group A. Deep sequence analysis allowed identification of variants associated with clonal lineages of S. aureus (clonal complex 8 [CC8], sequence type 239 [ST239]), S. agalactiae (CC17), Enterococcus faecium (ST17/ST18), or Enterococcus faecalis (ST8), local variants, or variants located in different species and geographical areas. All Tn5801 elements were chromosomally located upstream of the guaA gene, which serves as an integration hot spot. Transferability was demonstrated only for Tn5801 type B among E. faecalis clonal backgrounds, which eventually harbored another Tn5801 copy. The study documents early acquisition of Tn5801 by Enterococcus, Staphylococcus, and Streptococcus. Clonal waves of these pathogens seem to have contributed to the geographical spread and local evolution of the transposon. Horizontal transfer, also demonstrated, could explain the variability observed, with the isolates often containing sequences of different origins.


Asunto(s)
Elementos Transponibles de ADN/genética , Enterococcus faecium/genética , Variación Genética/genética , Staphylococcus aureus/genética , Streptococcus agalactiae/genética , Secuencia de Bases , ADN Bacteriano/genética , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/aislamiento & purificación , Humanos , Pruebas de Sensibilidad Microbiana , Análisis de Secuencia de ADN , Staphylococcus aureus/efectos de los fármacos , Streptococcus agalactiae/efectos de los fármacos
14.
J Antimicrob Chemother ; 71(8): 2147-57, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27118781

RESUMEN

OBJECTIVES: Factors driving the expansion of particular MDR Salmonella serotypes/clones are not completely understood. We assessed if emergent MDR Salmonella serotypes/clones were more enriched in metal tolerance genes (e.g. to Cu/Ag) than other less frequent ones, as an additional feature to survive in environments contaminated with metals. METHODS: Metal (Cu pco/Ag,Cu sil/Hg mer/As ars/Te ter) tolerance genes screening (PCR/sequencing), MICs of CuSO4/AgNO3 (aerobiosis/anaerobiosis), genetic element characterization (S1/I-CeuI PFGE) and conjugation assays were performed in a well-characterized Salmonella collection (n = 275 isolates; 2000-14; 49 serotypes/clones). RESULTS: The sil ±â€Špco genes were detected in 37% of isolates from diverse serotypes, mainly in emergent Rissen/ST469 and Typhimurium/ST34 European clone (100%), which are mostly associated with pig settings where Cu is highly used. These genes were frequently co-located with merA ±â€ŠterF and/or antibiotic resistance genes in plasmids (100-270 kb; IncHI2/IncHI1/IncN/IncFIIA; mostly transferable by conjugation) or in the chromosome. Most sil ±â€Špco(+) isolates (77%) were MDR contrasting with sil ±â€Špco(-) ones (48%). The sil ±â€Špco(+) isolates presented significantly higher MICCuSO4 under anaerobiosis (MIC50/MIC90 = 28/32 mM) and MICAgNO3 after previous Ag contact (MIC50/MIC90 > 3 mM) than sil(-) ones (MIC50/MIC90 = 2/8 mM to CuSO4; MIC50/MIC90 = 0.125/0.16 mM to AgNO3). Use of these modified methodological approaches allowed the establishment of CuSO4/AgNO3 tolerance cut-offs to differentiate sil(+) and sil(-) isolates, here firstly proposed. CONCLUSIONS: This study demonstrates that acquisition of Cu/Ag tolerance genes (sil/pco genes) might contribute to the emergence of particular clinically relevant MDR Salmonella serotypes/clones by facilitating their survival in diverse metal-contaminated settings, particularly in pig production. Assessment of control measures for the use and/or accumulation of metals in diverse environments are needed to prevent a wider expansion of such strains or the emergence of new ones.


Asunto(s)
Cobre/toxicidad , Farmacorresistencia Bacteriana Múltiple , Tolerancia a Medicamentos , Metales Pesados/toxicidad , Salmonella/efectos de los fármacos , Anaerobiosis , Crianza de Animales Domésticos/métodos , Animales , Conjugación Genética , Genes Bacterianos , Humanos , Pruebas de Sensibilidad Microbiana , Reacción en Cadena de la Polimerasa , Serogrupo
15.
J Antimicrob Chemother ; 71(12): 3351-3366, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27530756

RESUMEN

OBJECTIVES: Vancomycin-resistant Enterococcus faecium (VREfm) have been increasingly reported since the 1980s. Despite the high number of published studies about VRE epidemiology, the dynamics and evolvability of these microorganisms are still not fully understood. A multilevel population genetic analysis of VREfm outbreak strains since 1986, representing the first comprehensive characterization of plasmid content in E. faecium, was performed to provide a detailed view of potential transmissible units. METHODS: From a comprehensive MeSH search, we identified VREfm strains causing hospital outbreaks (1986-2012). In total, 53 VanA and 18 VanB isolates (27 countries, 5 continents) were analysed and 82 vancomycin-susceptible E. faecium (VSEfm) were included for comparison. Clonal relatedness was established by PFGE and MLST (goeBURST/Bayesian Analysis of Population Structure, BAPS). Characterization of van transposons (PCR mapping, RFLP, sequencing), plasmids (transfer, ClaI-RFLP, PCR typing of relaxases, replication-initiation proteins and toxin-antitoxin systems, hybridization, sequencing), bacteriocins and virulence determinants (PCR, hybridization, sequencing) was performed. RESULTS: VREfm were mainly associated with major human lineages ST17, ST18 and ST78. VREfm and VSEfm harboured plasmids of different families [RCR, small theta plasmids, RepA_N (pRUM/pLG1) and Inc18] able to yield mosaic elements. Tn1546-vanA was mainly located on pRUM/Axe-Txe (USA) and Inc18-pIP186 (Europe) plasmids. The VanB2 type (Tn5382/Tn1549) was predominant among VanB strains (chromosome and plasmids). CONCLUSIONS: Both strains and plasmids contributed to the spread and persistence of vancomycin resistance among E. faecium. Horizontal gene transfer events among genetic elements from different clonal lineages (same or different species) result in chimeras with different stability and host range, complicating the surveillance of epidemic plasmids.


Asunto(s)
Proteínas Bacterianas/genética , Ligasas de Carbono-Oxígeno/genética , Brotes de Enfermedades , Enterococcus faecium/clasificación , Variación Genética , Infecciones por Bacterias Grampositivas/epidemiología , Enterococos Resistentes a la Vancomicina/clasificación , Bacteriocinas/análisis , Infección Hospitalaria/epidemiología , Infección Hospitalaria/microbiología , Elementos Transponibles de ADN , Electroforesis en Gel de Campo Pulsado , Enterococcus faecium/genética , Enterococcus faecium/aislamiento & purificación , Transferencia de Gen Horizontal , Genética de Población , Genotipo , Salud Global , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Tipificación de Secuencias Multilocus , Plásmidos/análisis , Enterococos Resistentes a la Vancomicina/genética , Enterococos Resistentes a la Vancomicina/aislamiento & purificación , Factores de Virulencia/genética
17.
J Antimicrob Chemother ; 69(4): 899-906, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24343895

RESUMEN

OBJECTIVES: We studied the occurrence of diverse copper (Cu) tolerance genes from Gram-positive bacteria and their co-transfer with antibiotic resistance genes among Enterococcus from diverse sources. METHODS: Enterococcus (n = 922) of several species and from human, animal, environment and food samples were included. Antimicrobial and CuSO4 susceptibility and conjugation assays were performed by standard procedures, bacterial screening of Cu and antibiotic resistance genes by PCR, and clonality by PFGE/multilocus sequence typing. RESULTS: tcrB and cueO genes occurred in 15% (n = 137/922) and 14% (n = 128/922) of isolates, respectively, with the highest occurrence in piggeries (P < 0.05). They were more frequent among Enterococcus faecium (tcrB: 23% versus 8% in Enterococcus faecalis and 12% in other species; cueO: 25% versus 5% and 9%, respectively; P < 0.05). A correlation between phenotypic and genotypic assays was observed for most E. faecium (CuSO4 MIC50 = 24 mM in tcrB/cueO(+) versus CuSO4 MIC50 = 12 mM in tcrB/cueO(-)), but not for other species. Co-transfer of Cu tolerance (associated with tcrB, cueO or an unknown mechanism) with erythromycin, tetracycline, vancomycin, aminoglycosides or ampicillin resistance was demonstrated. A variety of PFGE types was detected among isolates carrying Cu tolerance mechanisms, some identified in sequence types (STs) often linked to human infections (E. faecium from ST18 and ST78 clonal lineages and E. faecalis clonal complex 2). CONCLUSIONS: Cu tolerance might contribute to the selection/maintenance of multidrug-resistant Enterococcus (including resistance to first-line antibiotics used to treat enterococcal infections) due to the use of Cu compounds (e.g. antiseptics/animal feed supplements). The distribution of the multicopper oxidase cueO and the co-transfer of ampicillin resistance along with Cu tolerance genes are described for the first time.


Asunto(s)
Antibacterianos/farmacología , Cobre/farmacología , Farmacorresistencia Bacteriana , Enterococcus/efectos de los fármacos , Microbiología Ambiental , Microbiología de Alimentos , Infecciones por Bacterias Grampositivas/microbiología , Animales , Conjugación Genética , ADN Bacteriano/genética , Electroforesis en Gel de Campo Pulsado , Enterococcus/clasificación , Enterococcus/genética , Enterococcus/aislamiento & purificación , Transferencia de Gen Horizontal , Genes Bacterianos , Infecciones por Bacterias Grampositivas/veterinaria , Humanos , Tipificación de Secuencias Multilocus , Reacción en Cadena de la Polimerasa
18.
Front Microbiol ; 15: 1365011, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38746750

RESUMEN

The rise of antibiotic resistance in the food chain is influenced by the use of antimicrobial agents, such as antibiotics, metals, and biocides, throughout the entire farm-to-fork continuum. Besides, non-clinical reservoirs potentially contribute to the transmission of critical pathogens such as multidrug-resistant (MDR) Klebsiella pneumoniae. However, limited knowledge exists about the population structure and genomic diversity of K. pneumoniae circulating in conventional poultry production. We conducted a comprehensive characterization of K. pneumoniae across the whole chicken production chain (7 farms; 14 flocks + environment + meat, 56 samples; 2019-2022), exploring factors beyond antibiotics, like copper and quaternary ammonium compounds (QACs). Clonal diversity and adaptive features of K. pneumoniae were characterized through cultural, molecular (FT-IR), and whole-genome-sequencing (WGS) approaches. All except one flock were positive for K. pneumoniae with a significant increase (p < 0.05) from early (n = 1/14) to pre-slaughter (n = 11/14) stages, most (n = 6/7) persisting in chicken meat batches. Colistin-resistant K. pneumoniae rates were low (4%-n = 1/24 positive samples), while most samples carried MDR strains (67%-n = 16/24) and copper-tolerant isolates (63%-n = 15/24, with sil and pco gene clusters; MICCuSO4 ≥ 16 mM), particularly at pre-slaughter. Benzalkonium chloride consistently exhibited activity against K. pneumoniae (MIC/MBC range = 4-64 mg/L) from representative strains independently of the presence or absence of genes linked to QACs tolerance. A polyclonal K. pneumoniae population, discriminated by FT-IR and WGS, included various lineages dispersed throughout the chicken's lifecycle at the farm (ST29-KL124, ST11-KL106, ST15-KL19, ST1228-KL38), until the meat (ST1-KL19, ST11-KL111, ST6405-KL109, and ST6406-CG147-KL111), or over years (ST631-49 KL109, ST6651-KL107, ST6406-CG147-KL111). Notably, some lineages were identical to those from human clinical isolates. WGS also revealed F-type multireplicon plasmids carrying sil + pco (copper) co-located with qacEΔ1 ± qacF (QACs) and antibiotic resistance genes like those disseminated in humans. In conclusion, chicken farms and their derived meat are significant reservoirs for diverse K. pneumoniae clones enriched in antibiotic resistance and metal tolerance genes, some exhibiting genetic similarities with human clinical strains. Further research is imperative to unravel the factors influencing K. pneumoniae persistence and dissemination within poultry production, contributing to improved food safety risk management. This study underscores the significance of understanding the interplay between antimicrobial control strategies and non-clinical sources to effectively address the spread of antimicrobial resistance.

19.
Microbiol Spectr ; 12(3): e0372423, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38329344

RESUMEN

Enterococcus faecium (Efm) is a leading cause of hospital-associated (HA) infections, often enriched in putative virulence markers (PVMs). Recently, the Efm clade B was assigned as Enterococcus lactis (Elts), which usually lack HA-Efm infection markers. Available databases for extracting PVM are incomplete and/or present an intermix of genes from Efm and Enterococcus faecalis, with distinct virulence profiles. In this study, we constructed a new database containing 27 PVMs [acm, scm, sgrA, ecbA, fnm, sagA, hylEfm, ptsD, orf1481, fms15, fms21-fms20 (pili gene cluster 1, PGC-1), fms14-fms17-fms13 (PGC-2), empA-empB-empC (PGC-3), fms11-fms19-fms16 (PGC-4), ccpA, bepA, gls20-glsB1, and gls33-glsB] from nine reference genomes (seven Efm + two Elts). The database was validated against these reference genomes and further evaluated using a collection of well-characterized Efm (n = 43) and Elts (n = 7) control strains, by assessing PVM presence/absence and its variants together with a genomic phylogeny constructed as single-nucleotide polymorphisms. We found a high concordance between the phylogeny and in silico findings of the PVM, with Elts clustering separately and mostly carrying Elts-specific PVM gene variants. Based on our validation results, we recommend using the database with raw reads instead of assemblies to avoid missing gene variants. This newly constructed database of 27 PVMs will enable a more comprehensive characterization of Efm and Elts based on WGS data. The developed database exhibits scalability and boasts a range of applications in public health, including diagnostics, outbreak investigations, and epidemiological studies. It can be further used in risk assessment for distinguishing between safe and unsafe enterococci.IMPORTANCEThe newly constructed database, consisting of 27 putative virulence markers, is highly scalable and serves as a valuable resource for the comprehensive characterization of these closely related species using WGS data. It holds significant potential for various public health applications, including hospital outbreak investigations, surveillance, and risk assessment for probiotics and feed additives.


Asunto(s)
Enterococcus faecium , Infecciones por Bacterias Grampositivas , Humanos , Enterococcus faecium/genética , Virulencia/genética , Enterococcus/genética , Enterococcus faecalis/genética , Antibacterianos , Infecciones por Bacterias Grampositivas/epidemiología
20.
J Antimicrob Chemother ; 68(12): 2746-54, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23861310

RESUMEN

OBJECTIVES: The aim of this study was to discover the potential role of the pig farm environment in the spread of multidrug-resistant (MDR) Enterococcus strains, including high-risk clones, to animals and humans. METHODS: Enterococcus isolates were recovered from a variety of samples (n = 82; swine, feed/medicines/antiseptics and pig farm facilities) from six Portuguese farms, most using antibiotics. Antimicrobial susceptibility/conjugation assays were performed by standard procedures, bacterial identification/screening of antibiotic resistance genes were performed by PCR and clonality was determined using PFGE/multilocus sequence typing. RESULTS: Enterococcus isolates resistant to antibiotics (n= 473) were recovered from samples of different origin (swine, feed/antiseptics, animal residues and pig farm facilities), but only the clinically relevant species Enterococcus faecium (n = 171) and Enterococcus faecalis (n = 78) were included for further comprehensive molecular analysis. Isolates resistant to vancomycin, ampicillin, tetracyclines, erythromycin and aminoglycosides were better recovered in Slanetz-Bartley medium with these antibiotics present than in media not supplemented with antibiotics (P < 0.05). E. faecium was more frequently resistant to ampicillin, ciprofloxacin or nitrofurantoin and E. faecalis to tetracyclines, chloramphenicol or aminoglycosides (P < 0.05). Glycopeptide and erythromycin resistance rates were similar in both species. The transfer of resistance to several antibiotics, including vancomycin and ampicillin, was demonstrated. Clones associated with human infections were detected in different samples from the same farm [E. faecium from sequence type (ST) 78 lineage and E. faecalis ST16; manure, waste lagoons, faeces and drinking water] and in geographically distant farms [E. faecium clonal complex (CC) 5; E. faecalis CC21 and ST16]. CONCLUSIONS: The pig farm environment has an underestimated potential role in the transmission of MDR Enterococcus to animals and, possibly, to humans. The continuous contact of swine with MDR Enterococcus by different routes (e.g. feed, dust, air and rooms) might decrease the impact of restrictive antibiotic use policies and reinforces the need for different and preliminary interventions at the husbandry management level.


Asunto(s)
Farmacorresistencia Bacteriana Múltiple , Enterococcus faecalis/efectos de los fármacos , Enterococcus faecium/efectos de los fármacos , Microbiología Ambiental , Infecciones por Bacterias Grampositivas/transmisión , Infecciones por Bacterias Grampositivas/veterinaria , Animales , Animales Domésticos , Antibacterianos/farmacología , Análisis por Conglomerados , Conjugación Genética , ADN Bacteriano/genética , Electroforesis en Gel de Campo Pulsado , Enterococcus faecalis/clasificación , Enterococcus faecalis/genética , Enterococcus faecalis/aislamiento & purificación , Enterococcus faecium/clasificación , Enterococcus faecium/genética , Enterococcus faecium/aislamiento & purificación , Genotipo , Infecciones por Bacterias Grampositivas/microbiología , Humanos , Pruebas de Sensibilidad Microbiana , Tipificación Molecular , Fenotipo , Portugal , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA