Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Antimicrob Agents Chemother ; 68(2): e0092523, 2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38169291

ABSTRACT

We describe the inter-regional spread of a novel ESBL-producing Escherichia coli subclone (ST131H89) in long-term care facility residents, general population, and environmental water sources in Western Switzerland between 2017 and 2020. The study highlights the importance of molecular surveillance for tracking emerging antibiotic-resistant pathogens in healthcare and community settings.


Subject(s)
Escherichia coli Infections , Escherichia coli Proteins , Humans , Escherichia coli Infections/epidemiology , Switzerland , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Anti-Bacterial Agents , beta-Lactamases , Molecular Epidemiology
2.
BMC Infect Dis ; 22(1): 33, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991516

ABSTRACT

BACKGROUND: Data on antimicrobial resistance mechanisms are scanty for Cedecea spp., with very variable antibiotic resistance patterns documented. Here we report the first in vivo resistance evolution of a C. davisae clinical isolate in a patient with a complex hand trauma and provide insight in the resistance mechanism, leading to therapeutic implications for this pathogen. CASE PRESENTATION: Cedecea davisae was isolated from a patient with hand trauma during a first surgical debridement. Six days after primary surgical treatment and under antimicrobial treatment with amoxicillin-clavulanic acid and later cefepime, follow up cultures yielded C. davisae which demonstrated a resistance development. The susceptible parental isolate and its resistant derivative were characterized by whole genome sequencing, ampC, ompC and ompF by RT- PCR. The resistant derivative demonstrated an A224G SNP in ampD, the transcriptional regulator of ampC, leading to a His75Arg change in the corresponding AmpD protein. AmpC transcription of the resistant derivative was 362-times higher than the susceptible isolate. Transcription levels of ompF and ompC were 8.5-fold and 1.3-fold lower, respectively, in the resistant derivative. Downregulation of OmpF putatively resulted from a mutation in the presumed promoter region upstream of the dusB-Fis operon, a proposed regulator for ompF. CONCLUSIONS: This case demonstrates the in vivo resistance development of C. davisae within 7 days similar to that of the members of the Enterobacter cloacae complex. Our findings add valuable information for future therapeutic management of these opportunistic pathogens as they warrant the same empirical treatment as AmpC producers.


Subject(s)
Bacterial Proteins , beta-Lactamases , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Enterobacteriaceae , Humans , Microbial Sensitivity Tests , beta-Lactamases/genetics
3.
Antimicrob Agents Chemother ; 58(10): 6320-3, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25092703

ABSTRACT

We evaluated the pet food contained in 30 packages as a potential origin of extended-spectrum cephalosporin-resistant Gram-negative organisms and ß-lactamase genes (bla). Live bacteria were not detected by selective culture. However, PCR investigations on food DNA extracts indicated that samples harbored the blaCTX-M-15 (53.3%), blaCMY-4 (20%), and blaVEB-4-like (6.7%) genes. Particularly worrisome was the presence of blaOXA-48-like carbapenemases (13.3%). The original pet food ingredients and/or the production processes were highly contaminated with bacteria carrying clinically relevant acquired bla genes.


Subject(s)
Bacterial Proteins/genetics , Plasmids/genetics , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/genetics , Microbial Sensitivity Tests , Polymerase Chain Reaction
4.
Antimicrob Agents Chemother ; 58(4): 2446-9, 2014.
Article in English | MEDLINE | ID: mdl-24468781

ABSTRACT

Here, we report a case of OXA-48-producing Salmonella enterica serovar Kentucky of sequence type 198 (ST198) from perianal screening cultures of a patient transferred from Libya to Switzerland. The blaOXA-48 gene was carried by Tn1999.2 and located on an ∼60-kb IncL/M plasmid. This Salmonella strain also possessed the blaVEB-8, aac(6)-Ib, tet(A), sul1, and mphA resistance genes and substitutions in GyrA (Ser83Phe and Asp87Asn) and ParC (Ser80Ile). This finding emphasizes that prompt screening strategies are essential to prevent the dissemination of carbapenemase producers imported from countries where they are endemic.


Subject(s)
Bacterial Proteins/metabolism , Salmonella enterica/enzymology , beta-Lactamases/metabolism , Drug Resistance, Multiple, Bacterial/genetics , Humans , Libya , Salmonella enterica/genetics , Switzerland
5.
Drug Resist Updat ; 16(1-2): 22-45, 2013.
Article in English | MEDLINE | ID: mdl-23395305

ABSTRACT

Escherichia coli, Salmonella spp. and Acinetobacter spp. are important human pathogens. Serious infections due to these organisms are usually treated with extended-spectrum cephalosporins (ESCs). However, in the past two decades we have faced a rapid increasing of infections and colonization caused by ESC-resistant (ESC-R) isolates due to production of extended-spectrum-ß-lactamases (ESBLs), plasmid-mediated AmpCs (pAmpCs) and/or carbapenemase enzymes. This situation limits drastically our therapeutic armamentarium and puts under peril the human health. Animals are considered as potential reservoirs of multidrug-resistant (MDR) Gram-negative organisms. The massive and indiscriminate use of antibiotics in veterinary medicine has contributed to the selection of ESC-R E. coli, ESC-R Salmonella spp. and, to less extent, MDR Acinetobacter spp. among animals, food, and environment. This complex scenario is responsible for the expansion of these MDR organisms which may have life-threatening clinical significance. Nowadays, the prevalence of food-producing animals carrying ESC-R E. coli and ESC-R Salmonella (especially those producing CTX-M-type ESBLs and the CMY-2 pAmpC) has reached worryingly high values. More recently, the appearance of carbapenem-resistant isolates (i.e., VIM-1-producing Enterobacteriaceae and NDM-1 or OXA-23-producing Acinetobacter spp.) in livestock has even drawn greater concerns. In this review, we describe the aspects related to the spread of the above MDR organisms among pigs, cattle, and poultry, focusing on epidemiology, molecular mechanisms of resistance, impact of antibiotic use, and strategies to contain the overall problem. The link and the impact of ESC-R organisms of livestock origin for the human scenario are also discussed.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Disease Reservoirs/veterinary , Gram-Negative Bacterial Infections/veterinary , beta-Lactam Resistance/genetics , beta-Lactamases/genetics , Animals , Cattle , Disease Reservoirs/microbiology , Europe/epidemiology , Gene Transfer, Horizontal , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/enzymology , Gram-Negative Bacteria/genetics , Gram-Negative Bacterial Infections/drug therapy , Gram-Negative Bacterial Infections/epidemiology , Gram-Negative Bacterial Infections/transmission , Humans , Poultry/microbiology , Prevalence , Swine/microbiology , beta-Lactam Resistance/drug effects , beta-Lactamases/metabolism
6.
Antimicrob Agents Chemother ; 57(6): 2867-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545526

ABSTRACT

Two homosexual men were colonized in the urethra with Haemophilus parainfluenzae nonsusceptible to ampicillin (MIC, 8 µg/ml), amoxicillin-clavulanate (MIC, 4 µg/ml), cefotaxime (MIC, 1.5 µg/ml), cefepime (MIC, 3 µg/ml), meropenem (MIC, 0.5 µg/ml), cefuroxime, azithromycin, ciprofloxacin, tetracycline, and chloramphenicol (all MICs, ≥ 32 µg/ml). Repetitive extragenic palindromic PCR (rep-PCR) showed that the strains were indistinguishable. The isolates had amino acid substitutions in PBP3, L4, GyrA, and ParC and possessed Mef(A), Tet(M), and CatS resistance mechanisms. This is the first report of extensively drug-resistant (XDR) H. parainfluenzae.


Subject(s)
Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial/genetics , Haemophilus Infections/microbiology , Haemophilus parainfluenzae/drug effects , Urethritis/microbiology , Amino Acid Substitution , Anti-Bacterial Agents/classification , Bacterial Proteins/genetics , Haemophilus Infections/drug therapy , Haemophilus parainfluenzae/classification , Haemophilus parainfluenzae/genetics , Haemophilus parainfluenzae/isolation & purification , Homosexuality, Male , Humans , Male , Microbial Sensitivity Tests , Sequence Analysis, DNA , Switzerland , Urethra/microbiology , Urethritis/drug therapy
7.
J Antimicrob Chemother ; 68(10): 2249-54, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23759671

ABSTRACT

OBJECTIVES: Resistance to extended-spectrum cephalosporins (ESCs) in Escherichia coli can be due to the production of ESBLs, plasmid-mediated AmpCs (pAmpCs) or chromosomal AmpCs (cAmpCs). Information regarding type and prevalence of ß-lactamases, clonal relations and plasmids associated with the bla genes for ESC-R E. coli (ESC-R-Ec) detected in Switzerland is lacking. Moreover, data focusing on patients referred to the specialized outpatient clinics (SOCs) are needed. METHODS: We analysed 611 unique E. coli isolated during September-December 2011. ESC-R-Ec were studied with microarrays, PCR/DNA sequencing for blaESBLs, blapAmpCs, promoter region of blacAmpC, IS elements, plasmid incompatibility group, and also implementing transformation, aIEF, rep-PCR and MLST. RESULTS: The highest resistance rates were observed in the SOCs, whereas those in the hospital and community were lower (e.g. quinolone resistance of 22.6%, 17.2% and 9.0%, respectively; P = 0.003 for SOCs versus community). The prevalence of ESC-R-Ec in the three settings was 5.3% (n = 11), 7.8% (n = 22) and 5.7% (n = 7), respectively. Thirty isolates produced CTX-M ESBLs (14 were CTX-M-15), 5 produced CMY-2 pAmpC and 5 hyper-expressed cAmpCs due to promoter mutations. Fourteen isolates were of sequence type 131 (ST131; 10 with CTX-M-15). blaCTX-M and blaCMY-2 were associated with an intact or truncated ISEcp1 and were mainly carried by IncF, IncFII and IncI1plasmids. CONCLUSIONS: ST131 producing CTX-M-15 is the predominant clone. The prevalence of ESC-R-Ec (overall 6.5%) is low, but an unusual relatively high frequency of AmpC producers (25%) was noted. The presence of ESC-R-Ec in the SOCs and their potential ability to be exchanged between hospital and community should be taken into serious consideration.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cephalosporins/pharmacology , Community-Acquired Infections/microbiology , Cross Infection/microbiology , Escherichia coli Infections/microbiology , Escherichia coli/drug effects , beta-Lactam Resistance , Ambulatory Care Facilities , Chromosomes , Cluster Analysis , Community-Acquired Infections/epidemiology , Cross Infection/epidemiology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Escherichia coli/classification , Escherichia coli/genetics , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Genotype , Hospitals , Humans , Microarray Analysis , Plasmids , Polymerase Chain Reaction , Sequence Analysis, DNA , Switzerland/epidemiology , beta-Lactamases/genetics , beta-Lactamases/metabolism
8.
Front Cell Infect Microbiol ; 13: 1098944, 2023.
Article in English | MEDLINE | ID: mdl-37180441

ABSTRACT

Background: Increasing reports of multidrug resistance (MDR) in clinical Pseudomonas aeruginosa have led to a necessity for new antimicrobials. Ceftazidime-avibactam (CZA) is indicated for use against MDR P. aeruginosa across a broad range of infection types and particularly those that are carbapenem resistant. This study sought to determine the molecular mechanisms of CZA and imipenem (IPM)-resistance in clinical P. aeruginosa isolates obtained from Swiss hospitals. Methods: Clinical P. aeruginosa isolates were obtained from inpatients in three hospitals in Switzerland. Susceptibility was determined by either antibiotic disc testing or broth microdilution according to EUCAST methodology. AmpC activity was determined using cloxacillin and efflux activity was determined using phenylalanine-arginine ß-naphthylamide, in agar plates. Whole Genome Sequencing was performed on 18 clinical isolates. Sequence types (STs) and resistance genes were ascertained using the Centre for Genomic Epidemiology platform. Genes of interest were extracted from sequenced isolates and compared to reference strain P. aeruginosa PAO1. Results: Sixteen different STs were identified amongst the 18 isolates in this study indicating a high degree of genomic diversity. No carbapenemases were detected but one isolate did harbor the ESBL bla PER-1. Eight isolates were CZA-resistant with MICs ranging from 16-64 mg/L, and the remaining ten isolates had either low/wildtype MICs (n=6; 1-2 mg/L) or elevated, but still susceptible, MICs (n=4; 4-8 mg/L). Ten isolates were IPM-resistant, seven of which had mutations resulting in truncations of OprD, and the remaining nine IPM-susceptible isolates had intact oprD genes. Within CZA-R isolates, and those with reduced susceptibility, mutations resulting in ampC derepression, OprD loss, mexAB overexpression and ESBL (bla PER-1) carriage were observed in various combinations and one harbored a truncation of the PBP4 dacB gene. Within the six isolates with wildtype-resistance levels, five had no mutations that would affect any antimicrobial resistance (AMR) genes of interest when compared to PAO1. Conclusion: This preliminary study highlights that CZA-resistance in P. aeruginosa is multifactorial and could be caused by the interplay between different resistance mechanisms including ESBL carriage, increased efflux, loss of permeability and derepression of its intrinsic ampC.


Subject(s)
Pseudomonas Infections , Pseudomonas aeruginosa , Humans , Pseudomonas aeruginosa/genetics , Anti-Bacterial Agents/pharmacology , Switzerland , Drug Combinations , beta-Lactamases/genetics , Microbial Sensitivity Tests
9.
Sci Rep ; 12(1): 19826, 2022 11 18.
Article in English | MEDLINE | ID: mdl-36400917

ABSTRACT

At our tertiary children's hospital, infections with newly detected methicillin-resistant Staphylococcus aureus (MRSA) among children attending primary (age 6-12 years) and secondary school (age 13-16 years) nearly doubled in 2018 compared to previous years. This observation initiated an epidemiological outbreak investigation including phenotypic (susceptibility testing) and genotypic (whole genome sequencing) characterization of the isolates. In addition, a cross-sectional study was conducted to determine source of the outbreak, colonization frequency and to identify risk factors for transmission using a questionnaire. As a result, 49 individuals were detected with 57 corresponding isolates. Based on the case definition combined with whole genome sequencing, a core cluster was identified that shared common genetic features and a similar antimicrobial susceptibility pattern (efflux-mediated macrolide resistance, tetracycline susceptibility along with presence of Panton-Valentine leukocidin). Epidemiologic evaluation identified a distinct school as a common risk factor. However, the source of the clustered infections within that school could not be further specified. No further cases could be detected after decolonization of infected and colonized children.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Humans , Child , Adolescent , Methicillin-Resistant Staphylococcus aureus/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Cross-Sectional Studies , Switzerland/epidemiology , Drug Resistance, Bacterial , Macrolides , Disease Outbreaks , Schools
10.
Antimicrob Resist Infect Control ; 11(1): 12, 2022 01 21.
Article in English | MEDLINE | ID: mdl-35063032

ABSTRACT

BACKGROUND: Despite the adoption of strict infection prevention and control measures, many hospitals have reported outbreaks of multidrug-resistant organisms (MDRO) during the Coronavirus 2019 (COVID-19) pandemic. Following an outbreak of carbapenem-resistant Acinetobacter baumannii (CRAB) in our institution, we sought to systematically analyse characteristics of MDRO outbreaks in times of COVID-19, focussing on contributing factors and specific challenges in controlling these outbreaks. METHODS: We describe results of our own CRAB outbreak investigation and performed a systematic literature review for MDRO (including Candida auris) outbreaks which occurred during the COVID-19 pandemic (between December 2019 and March 2021). Search terms were related to pathogens/resistance mechanisms AND COVID-19. We summarized outbreak characteristics in a narrative synthesis and contrasted contributing factors with implemented control measures. RESULTS: The CRAB outbreak occurred in our intensive care units between September and December 2020 and comprised 10 patients (thereof seven with COVID-19) within two distinct genetic clusters (both ST2 carrying OXA-23). Both clusters presumably originated from COVID-19 patients transferred from the Balkans. Including our outbreak, we identified 17 reports, mostly caused by Candida auris (n = 6) or CRAB (n = 5), with an overall patient mortality of 35% (68/193). All outbreaks involved intensive care settings. Non-adherence to personal protective equipment (PPE) or hand hygiene (n = 11), PPE shortage (n = 8) and high antibiotic use (n = 8) were most commonly reported as contributing factors, followed by environmental contamination (n = 7), prolonged critical illness (n = 7) and lack of trained HCW (n = 7). Implemented measures mainly focussed on PPE/hand hygiene audits (n = 9), environmental cleaning/disinfection (n = 9) and enhanced patient screening (n = 8). Comparing potentially modifiable risk factors and control measures, we found the largest discrepancies in the areas of PPE shortage (risk factor in 8 studies, addressed in 2 studies) and patient overcrowding (risk factor in 5 studies, addressed in 0 studies). CONCLUSIONS: Reported MDRO outbreaks during the COVID-19 pandemic were most often caused by CRAB (including our outbreak) and C. auris. Inadequate PPE/hand hygiene adherence, PPE shortage, and high antibiotic use were the most commonly reported potentially modifiable factors contributing to the outbreaks. These findings should be considered for the prevention of MDRO outbreaks during future COVID-19 waves.


Subject(s)
Acinetobacter Infections/prevention & control , Acinetobacter baumannii , COVID-19/complications , COVID-19/epidemiology , Candida auris , Candidiasis/prevention & control , Pandemics , SARS-CoV-2 , Acinetobacter Infections/complications , Acinetobacter baumannii/drug effects , Aged , Candidiasis/complications , Carbapenems/pharmacology , Cross Infection/prevention & control , Disease Outbreaks/prevention & control , Drug Resistance, Multiple, Bacterial , Female , Humans , Infection Control/methods , Male , Middle Aged , Retrospective Studies , Switzerland/epidemiology
11.
J Am Med Dir Assoc ; 23(3): 475-481.e5, 2022 03.
Article in English | MEDLINE | ID: mdl-34297981

ABSTRACT

OBJECTIVES: We aimed to assess the burden of extended-spectrum ß-lactamase (ESBL)-producing Enterobacterales in Swiss long-term care facilities (LTCFs) to describe the molecular epidemiology, describe the intrainstitutional and regional clusters of resistant pathogens, and identify independent institution- and resident-level factors associated with colonization. DESIGN: Cross-sectional study. SETTING AND PARTICIPANTS: From August to October 2019, we performed a point prevalence study among residents from 16 LTCFs in Western and Eastern Switzerland (8 per region). METHODS: Residents underwent screening for ESBL-producing Enterobacterales (ESBL-E); whole-genome sequencing (WGS) was performed. We gathered institution-level (eg, number of beds, staff-resident ratio, alcoholic hand rub consumption) and resident-level [eg, anthropometric data, time in facility, dependency, health care exposure, antibiotic treatment, proton-pump inhibitor (PPI) use] characteristics. Factors associated with colonization were identified using a generalized linear model. RESULTS: Among 1185 eligible residents, 606 (51%) consented to the study. ESBL-E prevalence was 11.6% (70/606), ranging from 1.9% to 33.3% between institutions, with a median of 12.5% in the West and 6.9% in the East (P = .03). Among 59 Escherichia coli (from 58 residents), multilocus sequence type (ST) 131 was most common (n = 43/59, 73%), predominantly its subclone H30R1 (n = 37/43, 86%). WGS data identified multiple intrainstitutional and regional clusters. Independent risk factors for ESBL carriage were previous ESBL colonization [adjusted odds ratio (aOR) 23.5, 95% confidence interval (CI) 6.6-83.8, P < .001), male gender (aOR 2.6, 95% CI 1.5-4.6, P = .002), and use of PPIs (aOR 2.2, 95% CI 1.2-3.8, P = .01). CONCLUSIONS AND IMPLICATIONS: Overall ESBL-E prevalence in Swiss LTCF residents is low. Yet, we identified several clusters of residents with identical pathogens within the same institution. This implies that particularly affected institutions might benefit from targeted infection control interventions. PPI use was the only modifiable factor associated with carriage of ESBL producers. This study adds to the growing list of adverse outcomes associated with PPIs, calling for action to restrict their use in the long-term care setting.


Subject(s)
Enterobacteriaceae Infections/epidemiology , Long-Term Care , Molecular Epidemiology , Cross-Sectional Studies , Enterobacteriaceae , Humans , Prevalence , Risk Factors , beta-Lactamases
13.
Front Microbiol ; 8: 407, 2017.
Article in English | MEDLINE | ID: mdl-28360894

ABSTRACT

The blaCMY -2/4-carrying IncB/O/K-like plasmids of seven Escherichia coli strains from poultry, poultry meat and human urine samples were examined using comparative analysis of whole plasmid sequences. The incompatibility group was determined by analysis of the incRNAI region and conjugation assays with strains containing the IncK and IncB/O reference plasmids. Strains were additionally characterized using MLST and MIC determination. The complete DNA sequences of all plasmids showed an average nucleotide identity of 91.3%. Plasmids were detected in E. coli sequence type (ST) 131, ST38, ST420, ST1431, ST1564 and belonged to a new plasmid variant (IncK2) within the IncK and IncB/O groups. Notably, one E. coli from poultry meat and one from human contained the same plasmid. The presence of a common recently recognized IncK2 plasmid in diverse E. coli from human urine isolates and poultry meat production suggests that the IncK2 plasmids originated from a common progenitor and have the capability to spread to genetically diverse E. coli in different reservoirs. This discovery is alarming and stresses the need of rapidly introducing strict hygiene measures throughout the food chain, limiting the spread of such plasmids in the human settings.

15.
PLoS One ; 10(5): e0123063, 2015.
Article in English | MEDLINE | ID: mdl-25933288

ABSTRACT

INTRODUCTION: blaOXA-48, blaNDM-1 and blaCTX-M-3 are clinically relevant resistance genes, frequently associated with the broad-host range plasmids of the IncL/M group. The L and M plasmids belong to two compatible groups, which were incorrectly classified together by molecular methods. In order to understand their evolution, we fully sequenced four IncL/M plasmids, including the reference plasmids R471 and R69, the recently described blaOXA-48-carrying plasmid pKPN-El.Nr7 from a Klebsiella pneumoniae isolated in Bern (Switzerland), and the blaSHV-5 carrying plasmid p202c from a Salmonella enterica from Tirana (Albania). METHODS: Sequencing was performed using 454 Junior Genome Sequencer (Roche). Annotation was performed using Sequin and Artemis software. Plasmid sequences were compared with 13 fully sequenced plasmids belonging to the IncL/M group available in GenBank. RESULTS: Comparative analysis of plasmid genomes revealed two distinct genetic lineages, each containing one of the R471 (IncL) and R69 (IncM) reference plasmids. Conjugation experiments demonstrated that plasmids representative of the IncL and IncM groups were compatible with each other. The IncL group is constituted by the blaOXA-48-carrying plasmids and R471. The IncM group contains two sub-types of plasmids named IncM1 and IncM2 that are each incompatible. CONCLUSION: This work re-defines the structure of the IncL and IncM families and ascribes a definitive designation to the fully sequenced IncL/M plasmids available in GenBank.


Subject(s)
Anti-Infective Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Plasmids/genetics , Base Sequence , Genome, Bacterial/genetics , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Restriction Mapping , Sequence Analysis, DNA
16.
J Food Prot ; 77(11): 1976-81, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25364933

ABSTRACT

The spread of antibiotic-resistant bacteria through food has become a major public health concern because some important human pathogens may be transferred via the food chain. Acinetobacter baumannii is one of the most life-threatening gram-negative pathogens; multidrug-resistant (MDR) clones of A. baumannii are spreading worldwide, causing outbreaks in hospitals. However, the role of raw meat as a reservoir of A. baumannii remains unexplored. In this study, we describe for the first time the antibiotic susceptibility and fingerprint (repetitive extragenic palindromic PCR [rep-PCR] profile and sequence types [STs]) of A. baumannii strains found in raw meat retailed in Switzerland. Our results indicate that A. baumannii was present in 62 (25.0%) of 248 (CI 95%: 19.7 to 30.9%) meat samples analyzed between November 2012 and May 2013, with those derived from poultry being the most contaminated (48.0% [CI 95%: 37.8 to 58.3%]). Thirty-nine strains were further tested for antibiotic susceptibility and clonality. Strains were frequently not susceptible (intermediate and/or resistant) to third- and fourth-generation cephalosporins for human use (i.e., ceftriaxone [65%], cefotaxime [32%], ceftazidime [5%], and cefepime [2.5%]). Resistance to piperacillin-tazobactam, ciprofloxacin, colistin, and tetracycline was sporadically observed (2.5, 2.5, 5, and 5%, respectively), whereas resistance to carbapenems was not found. The strains were genetically very diverse from each other and belonged to 29 different STs, forming 12 singletons and 6 clonal complexes (CCs), of which 3 were new (CC277, CC360, and CC347). RepPCR analysis further distinguished some strains of the same ST. Moreover, some A. baumannii strains from meat belonged to the clonal complexes CC32 and CC79, similar to the MDR isolates responsible for human infections. In conclusion, our findings suggest that raw meat represents a reservoir of MDR A. baumannii and may serve as a vector for the spread of these pathogens into both community and hospital settings.


Subject(s)
Acinetobacter baumannii/isolation & purification , Anti-Bacterial Agents/pharmacology , Drug Resistance, Multiple, Bacterial , Meat/microbiology , Phylogeny , Acinetobacter baumannii/classification , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/genetics , Animals , Cattle , Food Contamination/analysis , Food Contamination/statistics & numerical data , Microbial Sensitivity Tests , Molecular Sequence Data , Poultry , Sheep , Swine , Switzerland
17.
Int J Antimicrob Agents ; 44(3): 260-2, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25123809

ABSTRACT

Extensively drug-resistant (XDR) Klebsiella pneumoniae isolates usually carry a single carbapenemase (e.g. KPC, NDM, OXA-48-like). Here we describe an XDR K. pneumoniae of sequence type 101 that was detected in the screening rectal swab of a patient transferred from the intensive care unit of a hospital located in Belgrade (Serbia) to Bern University Hospital (Switzerland). The isolate was resistant to all antibiotics with the exception of colistin [minimum inhibitory concentration] (MIC ≤ 0.125 µg/mL), tigecycline (MIC = 0.5 µg/mL) and fosfomycin (MIC = 2 µg/mL). The isolate co-possessed class B (NDM-1) and class D (OXA-48) carbapenemases, class A extended-spectrum ß-lactamase (CTX-M-15), class C cephalosporinase (CMY-16), ArmA 16S rRNA methyltransferase, substitutions in GyrA and ParC, loss of OmpK35 porin, as well as other genes conferring resistance to quinolones (qnrA), tetracyclines [tet(A)], sulfonamides (sul1, sul2), trimethoprim (dfrA12, dfrA14), rifampicin (arr-1), chloramphenicol (cmlA1, floR) and streptomycin (aadA1). The patient was placed under contact isolation precautions preventing the spread of this nearly untreatable pathogen.


Subject(s)
Bacterial Proteins/genetics , Carrier State/microbiology , Escherichia coli Proteins/genetics , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Klebsiella pneumoniae/genetics , Methyltransferases/genetics , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/biosynthesis , Cluster Analysis , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Drug Resistance, Multiple, Bacterial , Escherichia coli Proteins/biosynthesis , Feces/microbiology , Humans , Intensive Care Units , Klebsiella pneumoniae/isolation & purification , Male , Methyltransferases/biosynthesis , Microbial Sensitivity Tests , Middle Aged , Switzerland , beta-Lactamases/biosynthesis
18.
mBio ; 5(1): e00960-13, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24425732

ABSTRACT

UNLABELLED: The Mycobacterium tuberculosis genome includes the large family of pe_pgrs genes, whose functions are unknown. Because of precedents in other pathogens in which gene families showing high sequence variation are involved in antigenic variation, a similar role has been proposed for the pe_pgrs genes. However, the impact of immune selection on pe_pgrs genes has not been examined. Here, we sequenced 27 pe_pgrs genes in 94 clinical strains from five phylogenetic lineages of the M. tuberculosis complex (MTBC). We found that pe_pgrs genes were overall more diverse than the remainder of the MTBC genome, but individual members of the family varied widely in their nucleotide diversity and insertion/deletion (indel) content: some were more, and others were much less, diverse than the genome average. Individual pe_pgrs genes also differed in the ratio of nonsynonymous to synonymous mutations, suggesting that different selection pressures act on individual pe_pgrs genes. Using bioinformatic methods, we tested whether sequence diversity in pe_pgrs genes might be selected by human T cell recognition, the major mechanism of adaptive immunity to MTBC. We found that the large majority of predicted human T cell epitopes were confined to the conserved PE domain and experimentally confirmed the antigenicity of this domain in tuberculosis patients. In contrast, despite being genetically diverse, the PGRS domains harbored few predicted T cell epitopes. These results indicate that human T cell recognition is not a significant force driving sequence diversity in pe_pgrs genes, which is consistent with the previously reported conservation of human T cell epitopes in the MTBC. IMPORTANCE: Recognition of Mycobacterium tuberculosis antigens by T lymphocytes is known to be important for immune protection against tuberculosis, but it is unclear whether human T cell recognition drives antigenic variation in M. tuberculosis. We previously discovered that the known human T cell epitopes in the M. tuberculosis complex are highly conserved, but we hypothesized that undiscovered epitopes with naturally occurring sequence variants might exist. To test this hypothesis, we examined the pe_pgrs genes, a large family of genes that has been proposed to function in immune evasion by M. tuberculosis. We found that the pe_pgrs genes exhibit considerable sequence variation, but the regions containing T cell epitopes and the regions of variation are distinct. These findings confirm that the majority of human T cell epitopes of M. tuberculosis are highly conserved and indicate that selection forces other than T cell recognition drive sequence variation in the pe_pgrs genes.


Subject(s)
Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Genetic Variation , Membrane Proteins/genetics , Membrane Proteins/immunology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , T-Lymphocytes/immunology , Computational Biology , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Genotype , Humans , INDEL Mutation , Molecular Sequence Data , Mycobacterium tuberculosis/isolation & purification , Selection, Genetic , Sequence Analysis, DNA
19.
Int J Antimicrob Agents ; 41(3): 236-49, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23313399

ABSTRACT

Despite many years of clinical experience with cefepime, data regarding the outcome of patients suffering from bloodstream infections (BSIs) due to Enterobacter cloacae (Ecl) are scarce. To address the gap in our knowledge, 57 Ecl responsible for 51 BSIs were analysed implementing phenotypic and molecular methods (microarrays, PCRs for bla and other genes, rep-PCR to analyse clonality). Only two E. cloacae (3.5%) were ESBL-producers, whereas 34 (59.6%) and 18 (31.6%) possessed inducible (Ind-Ecl) or derepressed (Der-Ecl) AmpC enzymes, respectively. All isolates were susceptible to imipenem, meropenem, gentamicin and ciprofloxacin. Der-Ecl were highly resistant to ceftazidime and piperacillin/tazobactam (both MIC90≥256 µg/mL), whereas cefepime retained its activity (MIC90 of 3 µg/mL). rep-PCR indicated that the isolates were sporadic, but Ecl collected from the same patients were indistinguishable. In particular, three BSIs initially due to Ind-Ecl evolved (under ceftriaxone or piperacillin/tazobactam treatment) into Der-Ecl because of mutations or a deletion in ampD or insertion of IS4321 in the promoter. These last two mechanisms have never been described in Ecl. Mortality was higher for BSIs due to Der-Ecl than Ind-Ecl (3.8% vs. 29.4%; P=0.028) and was associated with the Charlson co-morbidity index (P=0.046). Using the following directed treatments, patients with BSI showed a favourable treatment outcome: cefepime (16/18; 88.9%); carbapenems (12/13; 92.3%); ceftriaxone (4/7; 57.1%); piperacillin/tazobactam (5/7; 71.4%); and ciprofloxacin (6/6; 100%). Cefepime represents a safe therapeutic option and an alternative to carbapenems to treat BSIs due to Ecl when the prevalence of ESBL-producers is low.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Bacteremia/microbiology , Cephalosporins/therapeutic use , Enterobacter cloacae/drug effects , Enterobacteriaceae Infections/microbiology , Infections/microbiology , Adult , Aged , Anti-Bacterial Agents/pharmacology , Bacteremia/drug therapy , Bacteremia/mortality , Cefepime , Cephalosporins/pharmacology , Drug Therapy, Combination/methods , Enterobacter cloacae/classification , Enterobacter cloacae/genetics , Enterobacter cloacae/isolation & purification , Enterobacteriaceae Infections/drug therapy , Enterobacteriaceae Infections/mortality , Female , Genotype , Hospitals, University , Humans , Infections/drug therapy , Male , Microbial Sensitivity Tests , Middle Aged , Molecular Typing , Survival Analysis , Switzerland , Tertiary Care Centers , Treatment Outcome , beta-Lactamases/genetics , beta-Lactamases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL