Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
2.
Euro Surveill ; 29(16)2024 Apr.
Article in English | MEDLINE | ID: mdl-38639094

ABSTRACT

In 2023, an increase of OXA-48-producing Klebsiella pneumoniae was noticed by the Lithuanian National Public Health Surveillance Laboratory. Whole genome sequencing (WGS) of 106 OXA-48-producing K. pneumoniae isolates revealed three distinct clusters of carbapenemase-producing K. pneumoniae high-risk clones, including sequence type (ST) 45 (n = 35 isolates), ST392 (n = 32) and ST395 (n = 28), involving six, six and nine hospitals in different regions, respectively. These results enabled targeted investigation and control, and underscore the value of national WGS-based surveillance for antimicrobial resistance.


Subject(s)
Klebsiella Infections , Klebsiella pneumoniae , Humans , Klebsiella pneumoniae/genetics , Lithuania/epidemiology , Multilocus Sequence Typing , Klebsiella Infections/epidemiology , Klebsiella Infections/drug therapy , beta-Lactamases/genetics , Bacterial Proteins/genetics , Hospitals , Disease Outbreaks , Microbial Sensitivity Tests , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
3.
EFSA J ; 22(4): e8735, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38576537

ABSTRACT

Avian influenza viruses (AIV) remain prevalent among wild bird populations in the European Union and European Economic Area (EU/EEA), leading to significant illness in and death of birds. Transmission between bird and mammal species has been observed, particularly in fur animal farms, where outbreaks have been reported. While transmission from infected birds to humans is rare, there have been instances of exposure to these viruses since 2020 without any symptomatic infections reported in the EU/EEA. However, these viruses continue to evolve globally, and with the migration of wild birds, new strains carrying potential mutations for mammalian adaptation could be selected. If avian A(H5N1) influenza viruses acquire the ability to spread efficiently among humans, large-scale transmission could occur due to the lack of immune defences against H5 viruses in humans. The emergence of AIV capable of infecting mammals, including humans, can be facilitated by various drivers. Some intrinsic drivers are related to virus characteristics or host susceptibility. Other drivers are extrinsic and may increase exposure of mammals and humans to AIV thereby stimulating mutation and adaptation to mammals. Extrinsic drivers include the ecology of host species, such as including wildlife, human activities like farming practices and the use of natural resources, climatic and environmental factors. One Health measures to mitigate the risk of AIV adapting to mammals and humans focus on limiting exposure and preventing spread. Key options for actions include enhancing surveillance targeting humans and animals, ensuring access to rapid diagnostics, promoting collaboration between animal and human sectors, and implementing preventive measures such as vaccination. Effective communication to different involved target audiences should be emphasised, as well as strengthening veterinary infrastructure, enforcing biosecurity measures at farms, and reducing wildlife contact with domestic animals. Careful planning of poultry and fur animal farming, especially in areas with high waterfowl density, is highlighted for effective risk reduction.

4.
Euro Surveill ; 28(47)2023 11.
Article in English | MEDLINE | ID: mdl-37997662

ABSTRACT

BackgroundPreliminary unpublished results of the survey of carbapenem- and/or colistin-resistant Enterobacterales (CCRE survey) showed the expansion of carbapenemase-producing Klebsiella pneumoniae (CPKP) sequence type (ST) 39 in 12 of 15 participating Greek hospitals in 2019.AimWe conducted a rapid survey to determine the extent of spread of CPKP high-risk clones in Greek hospitals in 2022 and compare the distribution of circulating CPKP clones in these hospitals since 2013.MethodsWe analysed whole genome sequences and epidemiological data of 310 K. pneumoniae isolates that were carbapenem-resistant or 'susceptible, increased exposure' from Greek hospitals that participated in the European survey of carbapenemase-producing Enterobacteriaceae (EuSCAPE, 2013-2014), in the CCRE survey (2019) and in a national follow-up survey (2022) including, for the latter, an estimation of transmission events.ResultsFive K. pneumoniae STs including ST258/512 (n = 101 isolates), ST11 (n = 93), ST39 (n = 56), ST147 (n = 21) and ST323 (n = 13) accounted for more than 90% of CPKP isolates in the dataset. While ST11, ST147 and ST258/512 have been detected in participating hospitals since 2013 and 2014, KPC-2-producing ST39 and ST323 emerged in 2019 and 2022, respectively. Based on the defined genetic relatedness cut-off, 44 within-hospital transmission events were identified in the 2022 survey dataset, with 12 of 15 participating hospitals having at least one within-hospital transmission event.ConclusionThe recent emergence and rapid spread of new high-risk K. pneumoniae clones in the Greek healthcare system related to within-hospital transmission is of concern and highlights the need for molecular surveillance and enhanced infection prevention and control measures.


Subject(s)
Carbapenem-Resistant Enterobacteriaceae , Klebsiella Infections , Humans , Klebsiella pneumoniae/genetics , Carbapenem-Resistant Enterobacteriaceae/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Greece/epidemiology , Klebsiella Infections/epidemiology , Klebsiella Infections/drug therapy , Multilocus Sequence Typing , beta-Lactamases/genetics , Bacterial Proteins/genetics , Carbapenems/pharmacology , Hospitals , Clone Cells , Microbial Sensitivity Tests
5.
mBio ; : e0222423, 2023 Nov 10.
Article in English | MEDLINE | ID: mdl-37947415

ABSTRACT

Four molluscum contagiosum virus (MOCV) genotypes (MOCV1-4) and four subtype variants (MOCV1p, MOCV1va, MOCV1vb, and MOCV1vc) were partially characterized using restriction enzyme profiling in the early 1980s/1990s. However, complete genome sequences of only MOCV1 and MOCV2 are available. The evolutionary pathways of MOCV genotypes and subtype variants with unavailable sequences remain unclear, and also whether all MOCV genotypes/subtype variants can be reliably detected and appropriately categorized using available PCR-based protocols. We de novo fully characterized and functionally annotated 47 complete MOCV genomes, including two putative non-MOCV1/2 isolates, expanding the number of fully characterized MOCV genomes to 66. To ascertain the placement of any putative novel MOCV sequence into the restriction profiling typing scheme, we developed an original framework for extracting complete MOCV genome sequence-based restriction profiles and matching them with reference restriction profiles. We confirmed that two putative non-MOCV1/2 isolates represent the first complete genomes of MOCV3. Comprehensive phylogenomic, recombination, and restriction enzyme recognition site analysis of all 66 currently available MOCV genomes showed that they can be agglomerated into six phylogenetic subgroups (PG1-6), corresponding to the subtype variants from the pioneering studies. PG5 was a novel subtype variant of MOCV2, but no PGs corresponded to the subtype variants MOCV1vb or MOCV4. We showed that the phylogenetic subgroups may have diverged from the prototype MOCV genotype lineages following large-scale recombination events and hinted at partial sequence content of MOCV4 and direction of recombinant transfer in the events that spawned PG5 and the yet undetected subtype variant MOCV1vb.IMPORTANCEFour molluscum contagiosum virus (MOCV) genotypes (MOCV1-4) and four subtype variants were partially characterized using restriction enzyme profiling in the 1980s/1990s, but complete genome sequences of only MOCV1 and MOCV2 are available. The evolutionary pathways whereby genotypes/subtype variants with unavailable sequences emerged and whether all MOCVs can be detected using current diagnostic approaches remain unclear. We fully characterized 47 novel complete MOCV genomes, including the first complete MOCV3 genome, expanding the number of fully characterized genomes to 66. For reliably classifying the novel non-MOCV1/2 genomes, we developed and validated a framework for matching sequence-derived restriction maps with those defining MOCV subtypes in pioneering studies. Six phylogenetic subgroups (PG1-6) were identified, PG5 representing a novel MOCV2 subtype. The phylogenetic subgroups diverged from the prototype lineages following large-scale recombination events and hinted at partial sequence content of MOCV4 and direction of recombinant transfer in the events spawning PG5 and yet undetected MOCV1vb variant.

6.
Euro Surveill ; 28(19)2023 05.
Article in English | MEDLINE | ID: mdl-37166762

ABSTRACT

Whole genome sequencing data of 874 Escherichia coli isolates carrying bla NDM-5 from 13 European Union/European Economic Area countries between 2012 and June 2022 showed the predominance of sequence types ST167, ST405, ST410, ST361 and ST648, and an increasing frequency of detection. Nearly a third (30.6%) of these isolates were associated with infections and more than half (58.2%) were predicted to be multidrug-resistant. Further spread of E. coli carrying bla NDM-5 would leave limited treatment options for serious E. coli infections.


Subject(s)
Escherichia coli Infections , Escherichia coli , Humans , beta-Lactamases/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/drug therapy , Escherichia coli Infections/epidemiology , European Union , Microbial Sensitivity Tests , Europe/epidemiology
8.
Euro Surveill ; 27(15)2022 04.
Article in English | MEDLINE | ID: mdl-35426364

ABSTRACT

In the WHO European Region, COVID-19 non-pharmaceutical interventions continued slowing influenza circulation in the 2021/22 season, with reduced characterisation data. A(H3) predominated and, in some countries, co-circulated with A(H1)pdm09 and B/Victoria viruses. No B/Yamagata virus detections were confirmed. Substantial proportions of characterised circulating virus subtypes or lineages differed antigenically from their respective northern hemisphere vaccine components. Appropriate levels of influenza virus characterisations should be maintained until the season end and in future seasons, when surveillance is adapted to integrate SARS-CoV-2.


Subject(s)
COVID-19 , Influenza Vaccines , Influenza, Human , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza B virus/genetics , Influenza, Human/epidemiology , Influenza, Human/prevention & control , SARS-CoV-2 , Seasons , World Health Organization
9.
Euro Surveill ; 27(17)2022 04.
Article in English | MEDLINE | ID: mdl-35485272

ABSTRACT

Many countries, including some within the EU/EEA, are in the process of transitioning from the acute pandemic phase. During this transition, it is crucial that countries' strategies and activities remain guided by clear COVID-19 control objectives, which increasingly will focus on preventing and managing severe outcomes. Therefore, attention must be given to the groups that are particularly vulnerable to severe outcomes of SARS-CoV-2 infection, including individuals in congregate and healthcare settings. In this phase of pandemic management, a strong focus must remain on transitioning testing approaches and systems for targeted surveillance of COVID-19, capitalising on and strengthening existing systems for respiratory virus surveillance. Furthermore, it will be crucial to focus on lessons learned from the pandemic to enhance preparedness and to enact robust systems for the preparedness, detection, rapid investigation and assessment of new and emerging SARS-CoV-2 variants. Filling existing knowledge gaps, including behavioural insights, can help guide the response to future resurgences of SARS-CoV-2 and/or the emergence of other pandemics. Finally, 'vaccine agility' will be needed to respond to changes in people's behaviours, changes in the virus, and changes in population immunity, all the while addressing issues of global health equity.


Subject(s)
COVID-19 , Humans , Pandemics/prevention & control , Public Health , SARS-CoV-2
10.
PLoS Negl Trop Dis ; 15(12): e0010110, 2021 12.
Article in English | MEDLINE | ID: mdl-34968388

ABSTRACT

BACKGROUND: The kinetoplastid protozoan Leishmania tropica mainly causes cutaneous leishmaniasis in humans in the Middle East, and relapse or treatment failure after treatment are common in this area. L. tropica's digenic life cycle includes distinct stages in the vector sandfly and the mammalian host. Sexual reproduction and genetic exchange appear to occur more frequently than in other Leishmania species. Understanding these processes is complicated by chromosome instability during cell division that yields aneuploidy, recombination and heterozygosity. This combination of rare recombination and aneuploid permits may reveal signs of hypothetical parasexual mating, where diploid cells fuse to form a transient tetraploid that undergoes chromosomal recombination and gradual chromosomal loss. METHODOLOGY/PRINCIPAL FINDINGS: The genome-wide SNP diversity from 22 L. tropica isolates showed chromosome-specific runs of patchy heterozygosity and extensive chromosome copy number variation. All these isolates were collected during 2007-2017 in Sweden from patients infected in the Middle East and included isolates from a patient possessing two genetically distinct leishmaniasis infections three years apart with no evidence of re-infection. We found differing ancestries on the same chromosome (chr36) across multiple samples: matching the reference genome with few derived alleles, followed by blocks of heterozygous SNPs, and then by clusters of homozygous SNPs with specific recombination breakpoints at an inferred origin of replication. Other chromosomes had similar marked changes in heterozygosity at strand-switch regions separating polycistronic transcriptional units. CONCLUSION/SIGNIFICANCE: These large-scale intra- and inter-chromosomal changes in diversity driven by recombination and aneuploidy suggest multiple mechanisms of cell reproduction and diversification in L. tropica, including mitotic, meiotic and parasexual processes. It underpins the need for more genomic surveillance of Leishmania, to detect emerging hybrids that could spread more widely and to better understand the association between genetic variation and treatment outcome. Furthering our understanding of Leishmania genome evolution and ancestry will aid better diagnostics and treatment for cutaneous leishmaniasis caused by L.tropica in the Middle East.


Subject(s)
Genome, Protozoan , Leishmania tropica/genetics , Leishmaniasis, Cutaneous/parasitology , Afghanistan , Chromosomes/genetics , DNA Copy Number Variations , DNA, Protozoan/genetics , Genetic Variation , Humans , Iran , Leishmania tropica/classification , Leishmania tropica/isolation & purification , Phylogeny , Polymorphism, Single Nucleotide , Recombination, Genetic , Syria
12.
EFSA J ; 19(3): e06459, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33717355

ABSTRACT

American mink and ferret are highly susceptible to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but no information is available for other mustelid species. SARS-CoV-2 spreads very efficiently within mink farms once introduced, by direct and indirect contact, high within-farm animal density increases the chance for transmission. Between-farm spread is likely to occur once SARS-CoV-2 is introduced, short distance between SARS-CoV-2 positive farms is a risk factor. As of 29 January 2021, SARS-CoV-2 virus has been reported in 400 mink farms in eight countries in the European Union. In most cases, the likely introduction of SARS-CoV-2 infection into farms was infected humans. Human health can be at risk by mink-related variant viruses, which can establish circulation in the community, but so far these have not shown to be more transmissible or causing more severe impact compared with other circulating SARS-CoV-2. Concerning animal health risk posed by SARS-CoV-2 infection the animal species that may be included in monitoring plans are American mink, ferrets, cats, raccoon dogs, white-tailed deer and Rhinolophidae bats. All mink farms should be considered at risk of infection; therefore, the monitoring objective should be early detection. This includes passive monitoring (in place in the whole territory of all countries where animals susceptible to SARS-CoV-2 are bred) but also active monitoring by regular testing. First, frequent testing of farm personnel and all people in contact with the animals is recommended. Furthermore randomly selected animals (dead or sick animals should be included) should be tested using reverse transcriptase-polymerase chain reaction (RT-PCR), ideally at weekly intervals (i.e. design prevalence approximately 5% in each epidemiological unit, to be assessed case by case). Suspected animals (dead or with clinical signs and a minimum five animals) should be tested for confirmation of SARS-CoV-2 infection. Positive samples from each farm should be sequenced to monitor virus evolution and results publicly shared.

13.
F1000Res ; 10: 80, 2021.
Article in English | MEDLINE | ID: mdl-35847383

ABSTRACT

Next Generation Sequencing technologies significantly impact the field of Antimicrobial Resistance (AMR) detection and monitoring, with immediate uses in diagnosis and risk assessment. For this application and in general, considerable challenges remain in demonstrating sufficient trust to act upon the meaningful information produced from raw data, partly because of the reliance on bioinformatics pipelines, which can produce different results and therefore lead to different interpretations. With the constant evolution of the field, it is difficult to identify, harmonise and recommend specific methods for large-scale implementations over time. In this article, we propose to address this challenge through establishing a transparent, performance-based, evaluation approach to provide flexibility in the bioinformatics tools of choice, while demonstrating proficiency in meeting common performance standards. The approach is two-fold: first, a community-driven effort to establish and maintain "live" (dynamic) benchmarking platforms to provide relevant performance metrics, based on different use-cases, that would evolve together with the AMR field; second, agreed and defined datasets to allow the pipelines' implementation, validation, and quality-control over time. Following previous discussions on the main challenges linked to this approach, we provide concrete recommendations and future steps, related to different aspects of the design of benchmarks, such as the selection and the characteristics of the datasets (quality, choice of pathogens and resistances, etc.), the evaluation criteria of the pipelines, and the way these resources should be deployed in the community.


Subject(s)
Benchmarking , High-Throughput Nucleotide Sequencing , Anti-Bacterial Agents/pharmacology , Computational Biology/methods , Drug Resistance, Bacterial/genetics , High-Throughput Nucleotide Sequencing/methods
14.
J Virol Methods ; 288: 114010, 2021 02.
Article in English | MEDLINE | ID: mdl-33152410

ABSTRACT

Hepatitis A virus (HAV) is mainly transmitted via contaminated food or water or through person-to-person contact. Here, we describe development and evaluation of a reverse transcription droplet digital PCR (RT-ddPCR) and reverse transcription real-time PCR (RT-qPCR) assay for detection of HAV in food and clinical specimens. The assay was evaluated by assessing limit of detection, precision, matrix effects, sensitivity and quantitative agreement. The 95 % limit of detection (LOD95 %) was 10 % higher for RT-ddPCR than for RT-qPCR. A Bayesian model was used to estimate precision on different target concentrations. From this, we found that RT-ddPCR had somewhat greater precision than RT-qPCR within runs and markedly greater precision between runs. By analysing serum from naturally infected persons and a naturally contaminated food sample, we found that the two methods agreed well in quantification and had comparable sensitivities. Tests with artificially contaminated food samples revealed that neither RT-ddPCR nor RT-qPCR was severely inhibited by presence of oysters, raspberries, blueberries or leafy-green vegetables. For this assay, we conclude that RT-qPCR should be considered if rapid, qualitative detection is the main interest and that RT-ddPCR should be considered if precise quantification is the main interest. The high precision of RT-ddPCR allows for detection of small changes in viral concentration over time, which has direct implications for both food control and clinical studies.


Subject(s)
Hepatitis A virus , Bayes Theorem , Hepatitis A virus/genetics , Humans , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcription
15.
Euro Surveill ; 25(46)2020 11.
Article in English | MEDLINE | ID: mdl-33213683

ABSTRACT

The COVID-19 pandemic negatively impacted the 2019/20 WHO European Region influenza surveillance. Compared with previous 4-year averages, antigenic and genetic characterisations decreased by 17% (3,140 vs 2,601) and 24% (4,474 vs 3,403). Of subtyped influenza A viruses, 56% (26,477/47,357) were A(H1)pdm09, 44% (20,880/47,357) A(H3). Of characterised B viruses, 98% (4,585/4,679) were B/Victoria. Considerable numbers of viruses antigenically differed from northern hemisphere vaccine components. In 2020/21, maintaining influenza virological surveillance, while supporting SARS-CoV-2 surveillance is crucial.


Subject(s)
Coronavirus Infections/epidemiology , Disease Notification/statistics & numerical data , Epidemiological Monitoring , Influenza A virus/isolation & purification , Influenza B virus/isolation & purification , Influenza, Human/epidemiology , Influenza, Human/virology , Antigens, Viral/genetics , Betacoronavirus , COVID-19 , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza A virus/genetics , Influenza B virus/genetics , Pandemics , Pneumonia, Viral , Population Surveillance , RNA, Viral/genetics , SARS-CoV-2 , Sequence Analysis, DNA
16.
Emerg Microbes Infect ; 9(1): 2124-2135, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32896234

ABSTRACT

Global dissemination of ciprofloxacin-resistant Salmonella Kentucky has been observed over the past decades. In recent years, there have been reports of extended-spectrum ß-lactamase (ESBL) producing S. Kentucky. Routine surveillance at the European Centre for Disease Prevention and Control (ECDC) detected cases with a ciprofloxacin-resistant S. Kentucky with the ESBL-gene bla CTX-M-14b. Ensuing research identified 78 cases in 2013-2018 in eight European countries. Compared to other S. Kentucky and non-typhoidal Salmonella infections, reported to the European Surveillance System, these cases were more likely to be elderly and to present urinary-tract infections. Bayesian time-scaled phylogeny on whole genome sequences of isolates from these cases and supplementary isolates from public sequence databases was used to infer the origin and spread of this clone. We dated the origin of the bla CTX-M-14b clone to approximately 2005 in Northern Africa, most likely in Egypt. The geographic origin predicted by the phylogenetic analysis is consistent with the patients' travel history. Next to multiple introductions of the clone to Europe from Egypt, our analysis suggests that in some parts of Europe the clone might have formed a stable population, from which further spread has occurred. Comparative genomics indicated that the bla CTX-M-14b gene is present on the bacterial chromosome, within the type VI secretion system region. The bla CTX-M-14b gene is integrated downstream of the hcp1 gene, on a 2854 bp plasmid fragment containing also ISEcp1. This is the first report of a chromosomally integrated CTX-M gene in Salmonella spp. in Europe, previous studies having identified similar genes only on plasmids.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Salmonella Infections/epidemiology , Salmonella enterica/genetics , beta-Lactamases/genetics , Adolescent , Adult , Africa, Northern/epidemiology , Aged , Aged, 80 and over , Bayes Theorem , Child , Child, Preschool , Chromosomes, Bacterial , Ciprofloxacin/pharmacology , Egypt/epidemiology , Europe/epidemiology , Female , Genomics , Humans , Infant , Infant, Newborn , Male , Microbial Sensitivity Tests , Middle Aged , Molecular Epidemiology , Phylogeny , Plasmids , Salmonella Infections/microbiology , Salmonella enterica/drug effects , Salmonella enterica/isolation & purification , Whole Genome Sequencing , Young Adult
17.
Euro Surveill ; 25(32)2020 Aug.
Article in English | MEDLINE | ID: mdl-32794443

ABSTRACT

We show the distribution of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) genetic clades over time and between countries and outline potential genomic surveillance objectives. We applied three genomic nomenclature systems to all sequence data from the World Health Organization European Region available until 10 July 2020. We highlight the importance of real-time sequencing and data dissemination in a pandemic situation, compare the nomenclatures and lay a foundation for future European genomic surveillance of SARS-CoV-2.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/epidemiology , Coronavirus/genetics , Genome, Viral/genetics , Pandemics , Pneumonia, Viral/epidemiology , RNA, Viral/analysis , RNA-Dependent RNA Polymerase/genetics , Base Sequence , Betacoronavirus/pathogenicity , COVID-19 , Coronavirus/isolation & purification , Coronavirus Infections/virology , Europe/epidemiology , Humans , Phylogeography , Pneumonia, Viral/virology , RNA, Viral/genetics , SARS-CoV-2 , Severe Acute Respiratory Syndrome , Spatio-Temporal Analysis , World Health Organization
18.
Euro Surveill ; 25(20)2020 05.
Article in English | MEDLINE | ID: mdl-32458791

ABSTRACT

Analysis of sequencing data for 143 blaNDM-1- and blaOXA-48-positive Klebsiella pneumoniae isolates from 13 European national collections and the public domain resulted in the identification of 15 previously undetected multi-country transmission clusters. For 10 clusters, cases had prior travel/hospitalisation history in countries outside of the European Union including Egypt, Iran, Morocco, Russia, Serbia, Tunisia and Turkey. These findings highlight the benefit of European whole genome sequencing-based surveillance and data sharing for control of antimicrobial resistance.


Subject(s)
Bacterial Proteins/genetics , Carbapenem-Resistant Enterobacteriaceae/genetics , Carbapenem-Resistant Enterobacteriaceae/isolation & purification , Disease Outbreaks , Klebsiella Infections/epidemiology , Klebsiella pneumoniae/genetics , Whole Genome Sequencing/methods , beta-Lactamases/genetics , Anti-Bacterial Agents/therapeutic use , Carbapenem-Resistant Enterobacteriaceae/drug effects , Carbapenems/therapeutic use , Emigration and Immigration , Humans , Klebsiella Infections/drug therapy , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/isolation & purification , Microbial Sensitivity Tests/methods
19.
Article in English | MEDLINE | ID: mdl-32122893

ABSTRACT

In this retrospective study, whole-genome sequencing (WGS) data generated on an Ion Torrent platform was used to predict phenotypic drug resistance profiles for first- and second-line drugs among Swedish clinical Mycobacterium tuberculosis isolates from 2016 to 2018. The accuracy was ∼99% for all first-line drugs and 100% for four second-line drugs. Our analysis supports the introduction of WGS into routine diagnostics, which might, at least in Sweden, replace phenotypic drug susceptibility testing in the future.


Subject(s)
Drug Resistance, Multiple, Bacterial/drug effects , Drug Resistance, Multiple, Bacterial/genetics , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Tuberculosis/microbiology , Whole Genome Sequencing , Humans , Sweden , Tuberculosis, Multidrug-Resistant/drug therapy
20.
J Glob Antimicrob Resist ; 17: 233-235, 2019 06.
Article in English | MEDLINE | ID: mdl-30611929

ABSTRACT

OBJECTIVES: Historically, vancomycin-resistant enterococci (VRE) have been very common among farm animals in Europe, and they can still be readily isolated using media supplemented with vancomycin. An increase in the occurrence of VRE among broilers in Sweden was reported during 2000-2005. This was due to the spread of one clone of VRE in the apparent absence of selective pressure. The aims of this study were to estimate the current occurrence of VRE among Swedish broilers and to investigate if there had been any changes with regards to the dominating clone and antimicrobial resistance pattern. METHODS: Caecal samples (n=100) collected at slaughter from healthy broilers were cultured on Slanetz-Bartley agar supplemented with vancomycin (16 mg/L). Presumptive VRE were identified to species using MALDI-TOF MS, susceptibility tested using broth micro-dilution, and whole genome sequenced to investigate the genetic relationship with previous isolates. RESULTS: Eleven (11%) of the samples were positive for VRE. This was a statistically significant decrease in the proportion of positive samples from 2010. The same clone as before still prevailed. CONCLUSIONS: The occurrence of VRE among broilers in Sweden has decreased. However, just as the occurrence of VRE among Swedish broilers increased in the apparent absence of selective pressure, the reasons for the decrease is unknown.


Subject(s)
Chickens/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Enterococcus faecium/drug effects , Enterococcus faecium/genetics , Vancomycin-Resistant Enterococci/genetics , Vancomycin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Feces/microbiology , Genome, Bacterial , Microbial Sensitivity Tests , Multilocus Sequence Typing , Sweden , Whole Genome Sequencing
SELECTION OF CITATIONS
SEARCH DETAIL
...