Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 103
2.
Microb Drug Resist ; 27(11): 1555-1559, 2021 Nov.
Article En | MEDLINE | ID: mdl-33956523

Apramycin and florfenicol are two antimicrobial agents exclusively used in veterinary medicine. Resistance determinants to these antimicrobial agents have been described in several staphylococci, yet no inhibition zone-based epidemiological cutoff (ECOFF) values are available to detect populations harboring resistance mechanisms. In this study, we propose disk diffusion inhibition zone ECOFF values of Staphylococcus aureus for apramycin and florfenicol. The susceptibility to apramycin and florfenicol was evaluated by disk diffusion of five S. aureus collections, comprising 352 isolates of animal (n = 265) and human (n = 87) origin. The aggregated distributions of inhibition zone diameters were analyzed by the normalized resistance interpretation method to obtain normalized wild-type (WT) population distributions and corresponding ECOFF values. The putative WT populations of S. aureus were characterized by an inhibition zone ≥15 mm (ECOFF = 15 mm) for apramycin and ≥21 mm for florfenicol (ECOFF = 21 mm). Five nonwild-type (NWT) isolates were detected for apramycin, all without inhibition zone and harboring the apmA gene, whereas five NWT isolates were identified for florfenicol, all carrying the fexA gene. The proposed ECOFF values for apramycin and florfenicol may be a valuable tool in future antimicrobial resistance monitoring and surveillance studies to identify S. aureus NWT populations toward these antimicrobial agents.


Anti-Bacterial Agents/pharmacology , Microbial Sensitivity Tests/standards , Nebramycin/analogs & derivatives , Staphylococcus aureus/drug effects , Thiamphenicol/analogs & derivatives , Drug Resistance, Bacterial , Nebramycin/pharmacology , Thiamphenicol/pharmacology , Veterinary Medicine
3.
Front Microbiol ; 10: 1295, 2019.
Article En | MEDLINE | ID: mdl-31244807

The aim of this study was to identify and characterize LA-MRSA in a very recent collection of staphylococci isolated from bovine quarter milk samples. All milk samples (n = 14,924) sent to the MBFG in March 2017 were included into this study. The samples originated from 3,887 cows with 3,367 samples from 2,280 animals being positive for bacteria, prototheca and/or yeast. The second most common infectious agent was Staphylococcus and 659 isolates were investigated. Staphylococcus aureus was confirmed by PCR for the spa gene. A CC398-specific PCR was performed for all S. aureus isolates. Susceptibility to penicillin was tested for all isolates by agar disk diffusion. All oxacillin resistant isolates were analyzed by microarray and tested for their susceptibility to 30 antimicrobial agents. Of the isolates 372 were S. aureus from Germany with 214 isolates being not epidemiologically related. Among the independent isolates nine were identified as oxacillin resistant. In addition five isolates epidemiologically related to these nine were MRSA. One of them showed differences to the other MRSA isolate from the same farm resulting in altogether ten different MRSA isolates. All ten belonged to the clonal complex CC398. These ten LA-MRSA isolates had three to six antimicrobial resistance genes. The gene mecA was in all cases located on a SCCmec V element. Among the remaining S. aureus seven independent isolates belonged to CC398. In conclusion this study showed a high detection rate of staphylococci in bovine quarter milk samples. In contrast MRSA was rarely detected and belonged in all cases to CC398. Only 7/214 MSSA (3.3%) belonged to this CC.

4.
Article En | MEDLINE | ID: mdl-30834392

The draft genome sequences of three Streptococcus suis isolates, IMT40343, IMT40201, and IMT40738, are presented here. These isolates were obtained from bronchoalveolar lavage fluid of healthy and diseased weaners from different German piglet-producing farms and differed in their susceptibility to penicillin.

5.
Vet Microbiol ; 230: 235-240, 2019 Mar.
Article En | MEDLINE | ID: mdl-30827394

Pasteurella multocida is an important respiratory tract pathogen in intensive livestock farming, especially in pigs. Antimicrobial agents are frequently used to combat infections caused by this pathogen. In a study on antimicrobial resistance among respiratory tract pathogens of pigs from 30 German pig-producing farms, P. multocida isolates (n = 9) with high minimal inhibitory concentration (MIC) values of 16/304 mg/L (n = 2), 32/608 mg/L (n = 3) or ≥64/1216 mg/L (n = 4) for trimethoprim/sulfamethoxazole (1:19) and of ≥512 mg/L (n = 9) for trimethoprim (TMP) were detected in three of these farms. The genetic relatedness of the isolates was investigated via capsule-specific PCR and macrorestriction analyses with ApaI and SmaI. Pulsed-field gel electrophoresis revealed indistinguishable restriction patterns per farm, with slight differences between the three farms. All isolates represented capsular type A. Four representative isolates, that were subjected to whole genome sequencing, shared the multi-locus sequence type (ST) 3. Their plasmids were transformed into E. coli TOP10 with subsequent selection on TMP-containing agar plates. Antimicrobial susceptibility testing and plasmid analysis of the transformants confirmed that they were resistant to sulfonamides and trimethoprim and carried only a single small plasmid. This plasmid was completely sequenced and revealed a size of 6050 bp. Sequence analyses identified the presence of a resistance gene cluster comprising the genes sul2-ΔstrA-dfrA14-ΔstrA-ΔstrB. Further analysis identified a dfrA14 gene cassette being integrated into the strA reading frame. Neither the gene dfrA14 nor this gene cluster have been detected before in P. multocida.


Genes, Bacterial , Pasteurella multocida/genetics , Swine/microbiology , Animals , Anti-Bacterial Agents/pharmacology , Electrophoresis, Gel, Pulsed-Field , Farms , Germany , Livestock/microbiology , Microbial Sensitivity Tests , Multigene Family , Pasteurella multocida/drug effects , Pasteurella multocida/isolation & purification , Plasmids/genetics , Sulfamethoxazole/pharmacology , Swine Diseases/genetics , Swine Diseases/microbiology , Trimethoprim/pharmacology , Whole Genome Sequencing
6.
J Antimicrob Chemother ; 74(4): 851-853, 2019 04 01.
Article En | MEDLINE | ID: mdl-30561662

OBJECTIVES: To identify and analyse the first ESBL gene from Mannheimia haemolytica. METHODS: Susceptibility testing was performed according to CLSI. Plasmids were extracted via alkaline lysis and transferred by electrotransformation. The sequence was determined by WGS and confirmed by Sanger sequencing. RESULTS: The M. haemolytica strain 48 showed high cephalosporin MICs. A single plasmid, designated pKKM48, with a size of 4323 bp, was isolated. Plasmid pKKM48 harboured a novel blaROB gene, tentatively designated blaROB-2, and was transferred to Pasteurella multocida B130 and to Escherichia coli JM107. PCR assays and susceptibility testing confirmed the presence and activity of the blaROB-2 gene in the P. multocida and in the E. coli recipient carrying plasmid pKKM48. The transformants had high MICs of all ß-lactam antibiotics. An ESBL phenotype was seen in the E. coli transformant when applying the CLSI double-disc confirmatory test for E. coli. The blaROB-2 gene from plasmid pKKM48 differed in three positions from blaROB-1, resulting in two amino acid exchanges and one additional amino acid in the deduced ß-lactamase protein. In addition to blaROB-2, pKKM48 harboured mob genes and showed high similarity to other plasmids from Pasteurellaceae. CONCLUSIONS: This study described the first ESBL gene in Pasteurellaceae, which may limit the therapeutic options for veterinarians. The transferability to Enterobacteriaceae with the functional activity of the gene in the new host underlines the possibility of the spread of this gene across species or genus boundaries.


Chromosome Mapping , Mannheimia haemolytica/enzymology , Mannheimia haemolytica/genetics , Plasmids/analysis , beta-Lactamases/genetics , Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial , Electroporation , Escherichia coli/genetics , Microbial Sensitivity Tests , Pasteurella multocida/genetics , Transformation, Bacterial , Whole Genome Sequencing , beta-Lactams
7.
Front Microbiol ; 9: 2063, 2018.
Article En | MEDLINE | ID: mdl-30283407

Livestock-associated methicillin-resistant Staphylococcus aureus (LA-MRSA) isolates of the clonal complex 398 are often resistant to a number of antimicrobial agents. Studies on the genetic basis of antimicrobial resistance in these bacteria identified SCCmec cassettes, various transposons and plasmids of different sizes that harbor antimicrobial resistance genes. While large plasmids that carry multiple antimicrobial resistance genes - occasionally together with heavy metal resistance genes and/or virulence genes - are frequently seen in LA-MRSA ST398, certain resistance genes are also associated with small plasmids of up to 15 kb in size. These small resistance plasmids usually carry only one, but in rare cases also two or three antimicrobial resistance genes. In the current review, we focus on small plasmids that carry the macrolide-lincosamide-streptogramin B resistance genes erm(C) or erm(T), the lincosamide resistance gene lnu(A), the pleuromutilin-lincosamide-streptogramin A resistance genes vga(A) or vga(C), the spectinomycin resistance gene spd, the apramycin resistance gene apmA, or the trimethoprim resistance gene dfrK. The detailed analysis of the structure of these plasmids allows comparisons with similar plasmids found in other staphylococci and underlines in many cases an exchange of such plasmids between LA-MRSA ST398 and other staphylococci including also coagulase-negative staphylococci.

8.
Microbiol Spectr ; 6(4)2018 07.
Article En | MEDLINE | ID: mdl-30027886

Bordetella bronchiseptica is involved in respiratory tract infections mainly in dogs and pigs but may also cause infections in humans. Valid and representative data on antimicrobial susceptibility of B. bronchiseptica is rare. Approved antimicrobial susceptibility testing methods have been published, but very few clinical breakpoints are available. The MIC values are low for most agents but high for ß-lactam antibiotics and macrolides. Information on the genetic basis of resistance is scarce. For a small number of isolates that are resistant or show elevated MICs, the molecular basis of resistance was identified. Three tetracycline resistance genes, tet(A), tet(C), and tet(31), coding for major facilitator superfamily efflux pumps, were identified. Two other major facilitator superfamily exporter genes confer resistance to chloramphenicol (cmlB1) or to chloramphenicol and florfenicol (floR). Two class B chloramphenicol acetyltransferase genes (catB1 and catB3), which confer resistance to nonfluorinated phenicols by enzymatic inactivation, have been identified in B. bronchiseptica. Like the trimethoprim resistance genes dfrA1 and dfrB1, which code for trimethoprim-insensitive dihydrofolate reductases, the genes catB1 and catB3 were located on gene cassettes and found in class 1 integrons also harboring the sulfonamide resistance gene sul1. In addition, the gene sul2 has also been detected. Both sul1 and sul2 code for sulfonamide-insensitive dihydropteroate synthases. A gene cassette harboring the ß-lactamase gene blaOXA-2 was also identified, whereas ß-lactam resistance in B. bronchiseptica seems to be more likely due to reduced influx in combination with the species-specific ß-lactamase encoded by blaBOR-1. The resistance genes were mostly located on conjugative plasmids.


Anti-Bacterial Agents/pharmacology , Bordetella bronchiseptica/drug effects , Bordetella bronchiseptica/genetics , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Genes, Bacterial/genetics , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Bacterial Proteins/genetics , Bordetella Infections/microbiology , Bordetella Infections/veterinary , Bordetella bronchiseptica/pathogenicity , Drug Resistance, Multiple, Bacterial/genetics , Humans , Macrolides/pharmacology , Microbial Sensitivity Tests/methods , Species Specificity , Zoonoses/drug therapy
9.
Vet Microbiol ; 221: 153-158, 2018 Jul.
Article En | MEDLINE | ID: mdl-29981702

The aim of the study was to investigate methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-resistant Staphylococcus pseudintermedius (MRSP) among employees of a small animal hospital and the hospital environment. In total, 96 swabs from employees and 73 swabs from the clinic environment were investigated. Cation-adjusted-Mueller-Hinton broth (CAMHB) + 6.5% NaCl was used for enrichment before plating on Mueller-Hinton (MH) agar with 2% NaCl and 0.25 mg/L oxacillin. The staphylococcal species was determined using MALDI-TOF MS. The isolates were subjected to mecA-PCR, macrorestriction analysis, and antimicrobial susceptibility testing. MRSA were present in five nasal swabs of the 55 employees tested and in six environmental samples, MRSP in two employees (nasal and hand swabs, each) and in three environmental samples. All isolates harboured mecA. Susceptibility testing revealed that all but one of the isolates were multiresistant. All isolates were resistant to ß-lactams and fluoroquinolones. All but one of the isolates were resistant to macrolides and lincosamides. A single MRSA was resistant to gentamicin. All MRSP were resistant to trimethoprim/sulfamethoxazole and non-susceptible to gentamicin. One isolate was also resistant to tetracycline. Macrorestriction analysis revealed three main SmaI patterns for MRSA and two main SmaI patterns for MRSP. All environmental isolates were found in areas of high people and animal traffic, such as dog ward areas, waiting and triage rooms. The finding of indistinguishable MRSA or MRSP among employees and in the environment of the small animal hospital suggests the possibility of transfer of these bacteria between humans, animals, and the hospital environment.


Anti-Bacterial Agents/pharmacology , Environmental Microbiology , Hospitals, Animal , Methicillin Resistance , Staphylococcus/classification , Staphylococcus/drug effects , Animals , Carrier State , Humans , Nasal Mucosa/microbiology , Staphylococcal Infections , Veterinarians , Zoonoses
10.
Microbiol Spectr ; 6(4)2018 07.
Article En | MEDLINE | ID: mdl-29992898

Antimicrobial resistance among staphylococci of animal origin is based on a wide variety of resistance genes. These genes mediate resistance to many classes of antimicrobial agents approved for use in animals, such as penicillins, cephalosporins, tetracyclines, macrolides, lincosamides, phenicols, aminoglycosides, aminocyclitols, pleuromutilins, and diaminopyrimidines. In addition, numerous mutations have been identified that confer resistance to specific antimicrobial agents, such as ansamycins and fluoroquinolones. The gene products of some of these resistance genes confer resistance to only specific members of a class of antimicrobial agents, whereas others confer resistance to the entire class or even to members of different classes of antimicrobial agents, including agents approved solely for human use. The resistance genes code for all three major resistance mechanisms: enzymatic inactivation, active efflux, and protection/modification/replacement of the cellular target sites of the antimicrobial agents. Mobile genetic elements, in particular plasmids and transposons, play a major role as carriers of antimicrobial resistance genes in animal staphylococci. They facilitate not only the exchange of resistance genes among members of the same and/or different staphylococcal species, but also between staphylococci and other Gram-positive bacteria. The observation that plasmids of staphylococci often harbor more than one resistance gene points toward coselection and persistence of resistance genes even without direct selective pressure by a specific antimicrobial agent. This chapter provides an overview of the resistance genes and resistance-mediating mutations known to occur in staphylococci of animal origin.


Anti-Bacterial Agents/pharmacology , Drug Resistance, Bacterial/drug effects , Drug Resistance, Bacterial/genetics , Genes, Bacterial/genetics , Staphylococcus/drug effects , Staphylococcus/genetics , Animals , Anti-Bacterial Agents/classification , Drug Resistance, Multiple, Bacterial/genetics , Gene Transfer, Horizontal , Gram-Positive Bacteria/genetics , Humans , Plasmids , Staphylococcal Infections
11.
Vet Microbiol ; 219: 165-170, 2018 Jun.
Article En | MEDLINE | ID: mdl-29778191

The collaboration project VASIB aims at reducing the antibiotic consumption in pig production by integrating information from consulting expertise in clinical inspection, hygiene, epidemiology, microbiology and pharmacology. In this VASIB subproject, we investigated the antimicrobial susceptibility and relatedness of porcine respiratory tract pathogens. Bordetella bronchiseptica (n = 47), Pasteurella multocida (n = 18) and Streptococcus suis (n = 58) were obtained from weaner pigs at two farms. Antimicrobial susceptibility testing was performed by broth microdilution according to CLSI standards. Resistance genes were detected via specific PCR assays. Macrorestriction analysis was conducted to determine the relatedness of the isolates and to identify clones. The B. bronchiseptica isolates showed indistinguishable (farm 1) or two closely related XbaI-patterns (farm 2). Different SmaI-PFGE patterns of P. multocida isolates were obtained at three different time points. In contrast, PFGE analysis of S. suis indicated more than one fragment pattern per pig and time point. Isolates exhibiting indistinguishable PFGE patterns were considered to represent the same clone. This study showed that only two closely related B. bronchiseptica clones were present in both farms, which had low MICs to all antimicrobials, except to ß-lactams. Different P. multocida clones were present at the three time points. They showed overall low MIC values, with two clones being resistant and one intermediate to tetracycline. S. suis clones were resistant to tetracycline (n = 19) and/or erythromycin/clindamycin (n = 16). They harboured the tetracycline resistance genes tet(O), tet(M) or tet(L) and/or the macrolide/lincosamide/streptogramin B resistance gene erm(B). Five penicillin-resistant S. suis clones were also detected.


Anti-Infective Agents/pharmacology , Bacteria/drug effects , Bacteria/genetics , Bordetella Infections/veterinary , Pasteurella Infections/veterinary , Respiratory Tract Infections/veterinary , Streptococcal Infections/veterinary , Animals , Bacteria/classification , Bacteria/isolation & purification , Bordetella Infections/microbiology , Bordetella bronchiseptica/genetics , Bordetella bronchiseptica/isolation & purification , Bordetella bronchiseptica/pathogenicity , Drug Resistance, Multiple, Bacterial/genetics , Lincosamides/pharmacology , Macrolides/pharmacology , Microbial Sensitivity Tests , Pasteurella Infections/microbiology , Pasteurella multocida/genetics , Pasteurella multocida/isolation & purification , Pasteurella multocida/pathogenicity , Respiratory Tract Infections/microbiology , Streptococcal Infections/microbiology , Streptococcus suis/genetics , Streptococcus suis/isolation & purification , Streptococcus suis/pathogenicity , Swine , Swine Diseases/microbiology , Tetracycline/pharmacology , Weaning , beta-Lactams/pharmacology
12.
Vet Microbiol ; 219: 63-69, 2018 Jun.
Article En | MEDLINE | ID: mdl-29778206

Extended-spectrum ß-lactamase (ESBL)-producing Escherichia coli isolates have been increasingly reported in different reservoirs. The aims of this study were to investigate the presence of ESBL-producing E. coli in fresh vegetables and to characterise their ESBL gene-carrying plasmids. Among the 245 samples from vegetables investigated during 2011-2013, seven putative ESBL-producing E. coli (salad n = 2, sprouts n = 5) were found. They were subjected to ESBL phenotypic confirmatory tests, detection/sequencing of ESBL genes, antimicrobial susceptibility testing (AST), phylotyping, XbaI-macrorestriction analysis, multilocus sequence typing and transformation. Transformants were characterised by AST, S1-nuclease PFGE, replicon typing, conjugation and investigated for co-located antimicrobial resistance genes. Two ESBL gene-carrying plasmids were sequenced using a HiSeq 2500 system. The seven isolates were confirmed as ESBL producers, displayed unrelated XbaI-patterns and unique sequence types (STs) and belonged to the phylogroups A, B1 or D. The ESBL genes were located on plasmids. Two plasmids carrying blaCTX-M-14 genes (incompatibility group IncK or IncHI2) were seen in isolates from salad (ST973) and sprout (ST527). Two blaCTX-M-15- (IncFIB; non-typeable) and the IncN blaCTX-M-65- and IncHI2 blaCTX-M-125-carrying plasmids were found in isolates from sprouts (ST410, ST847, ST10, ST542). All plasmids were conjugative, except for the IncFIA-FIB blaCTX-M-2-carrying plasmid. Sequence analysis of two plasmids identified the ESBL genes in close location to other resistance genes: sulfonamide resistance gene sul2, streptomycin resistance genes strA and strB, the plasmid-mediated quinolone resistance gene qnrS1 and blaTEM-1 (sul2-strA-strB-IS66-blaTEM-1-tnpR-ΔtnpA-ISEcp1-blaCTX-M-15-Δorf477-ΔtnpA-qnrS1) or the fosfomycin resistance gene fosA3 (ΔISEcp1-blaCTX-M-125-ΔIS903B-fosA3). These observations underline the importance of vegetables as reservoirs for multidrug resistant ESBL-producing E. coli.


Anti-Bacterial Agents/pharmacology , Escherichia coli/isolation & purification , Plasmids/isolation & purification , Vegetables/microbiology , beta-Lactamases/genetics , Drug Resistance, Multiple, Bacterial/genetics , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/genetics , Fosfomycin/pharmacology , Gene Transfer, Horizontal , Germany , Microbial Sensitivity Tests , Multilocus Sequence Typing , Plasmids/genetics , Replicon , beta-Lactamases/biosynthesis
13.
Vet Microbiol ; 217: 36-46, 2018 Apr.
Article En | MEDLINE | ID: mdl-29615254

The most common livestock-associated lineage of methicillin-resistant Staphylococcus aureus (MRSA) in Western Europe is currently clonal complex (CC) 398. CC398-MRSA spread extensively across livestock populations in several Western European countries, and livestock-derived CC398-MRSA strains can also be detected in humans. Based on their SCCmec elements, different CC398 strains can be distinguished. SCCmec elements of 100 veterinary and human CC398-MRSA isolates from Germany and Austria were examined using DNA microarray-based assays. In addition, 589 published SCC and/or genome sequences of CC398-MRSA (including both, fully finished and partially assembled sequences) were analysed by mapping them to the probe sequences of the microarrays. Several isolates and sequences showed an insertion of a large fragment of CC9 genomic DNA into the CC398 chromosome. Fifteen subtypes of SCCmec elements were detected among the 100 CC398 isolates and 41 subtypes could be discerned among the published CC398 sequences. Eleven of these were also experimentally detected within our strain collection, while four subtypes identified in the isolates where not found among the sequences. A high prevalence of heavy metal resistance genes, especially of czrC, was observed among CC398-MRSA. A possible co-selection of resistances to antibiotics and zinc/copper supplements in animal feed as well as a spill-over of SCCmec elements that have evolved in CC398-MRSA to other, possibly more virulent and/or medically relevant S. aureus lineages might pose public health problems in future.


Genetic Variation , Livestock/microbiology , Methicillin-Resistant Staphylococcus aureus/genetics , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Staphylococcal Infections/veterinary , Animal Feed , Animals , Anti-Bacterial Agents/pharmacology , Austria/epidemiology , Copper/administration & dosage , Copper/pharmacology , Dietary Supplements , Drug Resistance, Multiple, Bacterial , Genome, Bacterial , Germany/epidemiology , Humans , Methicillin-Resistant Staphylococcus aureus/classification , Methicillin-Resistant Staphylococcus aureus/drug effects , Mutagenesis, Insertional/genetics , Oligonucleotide Array Sequence Analysis , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Swine/microbiology , Zinc/administration & dosage , Zinc/pharmacology
14.
J Antimicrob Chemother ; 72(6): 1589-1596, 2017 06 01.
Article En | MEDLINE | ID: mdl-28333184

Objectives: This study aimed at characterizing 23 Escherichia coli isolates from various sources and their respective bla SHV-12 -carrying plasmids and sequencing one of these plasmids completely. Methods: Isolates were typed by XbaI-PFGE, MLST and PCR-based phylotyping. Transformed bla SHV-12 -carrying plasmids were examined by replicon typing, S1-nuclease, conjugation, EcoRI-HindIII-BamHI digests and plasmid MLST. Co-located resistance genes and integrons as well as the bla SHV-12 genetic environment were analysed by PCR and sequencing. One IncI1 plasmid was sequenced completely using HiSeq 2500 and gap closure by PCRs and Sanger sequencing. Results: Among the 23 SHV-12-positive E. coli , some isolates from different sources showed the same characteristics: ST23/phylogroup A (human, dog, livestock), ST57/D (wild bird, chicken meat) and ST117/D (chicken meat, chicken). All bla SHV-12 genes were horizontally transferable via 30-120 kb plasmids of incompatibility groups IncI1 ( n = 17), IncK ( n = 3), IncF ( n = 1), IncX3 ( n = 1) and a non-typeable plasmid. IncK plasmids, indistinguishable in size and restriction patterns, were found in isolates from different sources (ST57/D, meat; ST131/B2, meat; ST57/B1, dog). The IncI1- bla SHV-12 -carrying plasmids were mostly assigned to plasmid ST (pST) 26 and pST3. Three plasmids showed novel pSTs (pST214, pST215). The majority of the IncI1 transformants exhibited resistance to ß-lactams, chloramphenicol and streptomycin (in relation with a class 1 integron containing an estX - psp - aadA2 - cmlA1 - aadA1 - qacI gene cassette array), and to tetracycline. A novel bla SHV-12 environment was detected and whole plasmid sequencing revealed a Tn 21 -derived- bla SHV12 -ΔTn 1721 resistance complex. Conclusions: Results from this study suggest that the dissemination of bla SHV-12 genes occurs by vertical (clonal) and horizontal transfer, the latter mainly mediated through IncI1 multidrug-resistance plasmids.


Escherichia coli/genetics , Food Microbiology , Plasmids , beta-Lactamases/genetics , Animals , Anti-Bacterial Agents/pharmacology , Chickens/microbiology , Chloramphenicol/pharmacology , Dogs/microbiology , Escherichia coli/drug effects , Escherichia coli/enzymology , Escherichia coli/isolation & purification , Gene Transfer, Horizontal , Genes, MDR , Humans , Integrons , Meat/microbiology , Microbial Sensitivity Tests , Multilocus Sequence Typing , Plasmids/isolation & purification , Replicon , Sequence Analysis, DNA
15.
Vet Microbiol ; 200: 95-100, 2017 Feb.
Article En | MEDLINE | ID: mdl-27476981

The multiresistance plasmid pAFS11, obtained from a bovine methicillin-resistant Staphylococcus aureus (MRSA) isolate, was completely sequenced and analysed for its structure and organisation. Moreover, the susceptibility to the heavy metals cadmium and copper was determined by broth macrodilution. The 49,189-bp plasmid harboured the apramycin resistance gene apmA, two copies of the macrolide/lincosamide/streptogramin B resistance gene erm(B) (both located on remnants of a truncated transposon Tn917), the kanamycin/neomycin resistance gene aadD, the tetracycline resistance gene tet(L) and the trimethoprim resistance gene dfrK. The latter three genes were part of a 7,284-bp segment which was bracketed by two copies of IS431. In addition, the cadmium resistance operon cadDX as well as the copper resistance genes copA and mco were located on the plasmid and mediated a reduced susceptibility to cadmium and copper. Moreover, a complete novel ica-like gene cluster of so far unknown genetic origin was detected on this plasmid. The ica-like gene cluster comprised four different genes whose products showed 64.4-76.9% homology to the Ica proteins known to be involved in biofilm formation of the S. aureus strains Mu50, Mu3 and N315. However, 96.2-99.4% homology was seen to proteins from S. sciuri NS1 indicating an S. sciuri origin. The finding of five different antibiotic resistance genes co-located on a plasmid with heavy metal resistance genes and an ica-like gene cluster is alarming. With the acquisition of this plasmid, antimicrobial multiresistance, heavy metal resistances and potential virulence properties may be co-selected and spread via a single horizontal gene transfer event.


Biofilms/growth & development , Cattle Diseases/microbiology , Drug Resistance, Multiple, Bacterial/genetics , Methicillin-Resistant Staphylococcus aureus/genetics , Plasmids/genetics , Staphylococcal Infections/veterinary , Animals , Anti-Infective Agents/pharmacology , Bacterial Proteins/genetics , Cattle , Cattle Diseases/epidemiology , Metals, Heavy/toxicity , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/isolation & purification , Methicillin-Resistant Staphylococcus aureus/pathogenicity , Microbial Sensitivity Tests/veterinary , Multigene Family , Sequence Analysis, DNA/veterinary , Staphylococcal Infections/epidemiology , Staphylococcal Infections/microbiology , Virulence/genetics
16.
Vet Microbiol ; 200: 52-58, 2017 Feb.
Article En | MEDLINE | ID: mdl-27062479

Antimicrobial peptides (AMPs) represent an important part of the innate host immune system. Although they are active against a broad range of pathogens, bacteria have evolved different resistance mechanisms to avoid killing by AMPs. Since not much is known about the impact of efflux pumps on the susceptibility of Gram-positive bacteria to AMPs, especially to the cathelicidins, the aim of this study was to analyze whether Staphylococcus aureus can use efflux pumps to resist the antimicrobial effects of cathelicidins derived from different animal species (human, mouse, rabbit or cattle). For this purpose the minimal inhibitory concentrations (MICs) of S. aureus field isolates for the cathelicidins LL-37, mCRAMP, CAP18, BMAP-27 and BMAP-28 in the presence and absence of different efflux pump inhibitors were determined. Furthermore, the MICs of mutants lacking SecDF, a member of the RND efflux pump family, were determined and compared to the MICs of their respective wildtype and complemented strains. The data demonstrated that after blocking RND-type efflux pumps with 1-(1-naphthylmethyl)-piperazine, the MICs for CAP18, but not those for the other cathelicidins tested, were significantly decreased. In good correlation with these data, significantly decreased MICs for CAP18 and also BMAP-27 have been observed for SecDF knockout mutants as compared to their isogenic wildtype strains. In addition, the MIC values increased again after re-introducing a cloned secDF via plasmid complementation. These results indicated an involvement of SecDF in a reduced efficacy of species-specific cathelicidins against S. aureus.


Anti-Infective Agents/pharmacology , Bacterial Proteins/metabolism , Cathelicidins/pharmacology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Staphylococcal Infections/veterinary , Animals , Antimicrobial Cationic Peptides/pharmacology , Cattle , Disease Susceptibility , Enzyme Inhibitors , Humans , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/genetics , Mice , Microbial Sensitivity Tests , Piperazines/pharmacology , Proteins/pharmacology , Rabbits , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology
17.
Vet Dermatol ; 28(1): 82-e19, 2017 Feb.
Article En | MEDLINE | ID: mdl-27581211

BACKGROUND: Antimicrobial resistance has become a major challenge in veterinary medicine, particularly in the context of bacterial pathogens that play a role in both humans and animals. OBJECTIVES: This review serves as an update on acquired resistance mechanisms in bacterial pathogens of human and animal origin, including examples of transfer of resistant pathogens between hosts and of resistance genes between bacteria. RESULTS: Acquired resistance is based on resistance-mediating mutations or on mobile resistance genes. Although mutations are transferred vertically, mobile resistance genes are also transferred horizontally (by transformation, transduction or conjugation/mobilization), contributing to the dissemination of resistance. Mobile genes specifying any of the three major resistance mechanisms - enzymatic inactivation, reduced intracellular accumulation or modification of the cellular target sites - have been found in a variety of bacteria that may be isolated from animals. Such resistance genes are associated with plasmids, transposons, gene cassettes, integrative and conjugative elements or other mobile elements. Bacteria, including zoonotic pathogens, can be exchanged between animals and humans mainly via direct contact, but also via dust, aerosols or foods. Proof of the direction of transfer of resistant bacteria can be difficult and depends on the location of resistance genes or mutations in the chromosomal DNA or on a mobile element. CONCLUSION: The wide variety in resistance and resistance transfer mechanisms will continue to ensure the success of bacterial pathogens in the future. Our strategies to counteract resistance and preserve the efficacy of antimicrobial agents need to be equally diverse and resourceful.


Anti-Bacterial Agents/therapeutic use , Bacterial Infections/drug therapy , Drug Resistance, Bacterial , Animals , Bacterial Infections/veterinary , Humans , Zoonoses/drug therapy , Zoonoses/microbiology
18.
Vet Microbiol ; 200: 151-156, 2017 Feb.
Article En | MEDLINE | ID: mdl-27566885

Extended-spectrum ß-lactamase (ESBL)-producing isolates have been increasingly reported during recent years. The aims of this study were to characterize ESBL-producing Escherichia coli from bovine mastitis as well as their ESBL gene-carrying plasmids. A culture collection of E. coli isolated from bovine quarter milk samples (2009-2013), was screened for ESBL production using ESBL selective agar plates. Putative ESBL producers (n=16) were investigated by phenotypic confirmatory tests and were characterized by the detection/sequencing of ESBL genes, XbaI macrorestriction analysis, multilocus sequence typing (MLST), phylotyping and antimicrobial susceptibility testing. ESBL gene-carrying plasmids were investigated by transfer experiments, PCRs for the detection of co-located antimicrobial resistance genes, PCR-based replicon typing and S1-nuclease pulsed-field gel electrophoresis. Twelve ESBL-producing isolates were found. They showed eleven different XbaI patterns and were distributed among eight MLST types [ST10 (n=3), ST117 (n=2), ST361 (n=1), ST362 (n=1), ST540 (n=1), ST1431 (n=2), ST1508 (n=1), and the novel ST5447 (n=1)] and the phylogenetic groups A (n=6), B1 (n=2), B2 (n=1) and D (n=3). ESBL genes blaCTX-M-1 (n=5), blaCTX-M-2 (n=2), blaCTX-M-14 and blaCTX-M-15 (n=4) were found on conjugative plasmids (35-225kb) of diverse incompatibility groups (e.g. IncF, IncI1 or HI2+P). Co-located resistance to sulfonamides, tetracycline, trimethoprim, and chloramphenicol/florfenicol was detected on five ESBL gene-carrying plasmids, but seven plasmids conferred solely resistance to ß-lactam antibiotics. The presence of additional resistance genes on the ESBL gene-carrying plasmids suggests that co-selection of ESBL genes may occur even in the absence of ß-lactam antibiotics.


Escherichia coli Infections/veterinary , Escherichia coli/genetics , Mastitis, Bovine/microbiology , Plasmids/genetics , beta-Lactamases/genetics , Animals , Anti-Bacterial Agents/pharmacology , Bacterial Typing Techniques/veterinary , Cattle , Escherichia coli/enzymology , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Female , Germany/epidemiology , Integrons/genetics , Mastitis, Bovine/epidemiology , Microbial Sensitivity Tests/veterinary , Multilocus Sequence Typing/veterinary , Phylogeny , Replicon/genetics , beta-Lactams/pharmacology
19.
Vet Microbiol ; 200: 142-150, 2017 Feb.
Article En | MEDLINE | ID: mdl-27634182

The aim of this study was to identify extended-spectrum ß-lactamase (ESBL)-producing Escherichia coli collected from diseased food-producing animals in Germany. A total of 6849 E. coli isolates, collected from diseased cattle, pigs and poultry in the German national monitoring program GERM-Vet (2008-2014), were characterized by antimicrobial susceptibility testing and screened for the ESBL phenotype. ESBL genes were identified by PCR and sequencing. The isolates were further characterized by PCR-based phylotyping. The 419/6849 (6.1%) ESBL-producers identified included 324/2896 (11.2%) isolates from cattle, 75/1562 (4.8%) from pigs and 20/2391 (0.8%) from poultry. The ESBL genes detected were: blaCTX-M-1 (69.9%), blaCTX-M-15 (13.6%), blaCTX-M-14 (11.7%), blaTEM-52 (1.9%), blaSHV-12 (1.4%), blaCTX-M-3 (1.0%), and blaCTX-M-2 (0.5%). The phylogroup A was the dominant phylogroup (57.0%) followed by phylogroups D (23.4%), B1 (17.9%), and B2 (1.7%). Bovine isolates belonged predominantly to the phylogroups A and D, whereas the porcine and avian isolates mainly belonged to A and B1. The majority of the ESBL-producing isolates found in each phylogroup were from animals suffering from gastrointestinal infections. In 399/419 isolates (95.2%), additional resistance to non-ß-lactam antibiotics was seen. Multidrug-resistance [resistance to aminoglycosides, fluoro(quinolones), sulphonamides, tetracyclines, and trimethoprim] was seen in 369/419 (88.1%) isolates, which may facilitate the co-selection of ESBL genes, when located on the same mobile genetic element as the others resistance genes, and may compromise the therapeutic options.


Cattle Diseases/microbiology , Drug Resistance, Bacterial/genetics , Escherichia coli Infections/veterinary , Escherichia coli/genetics , Poultry Diseases/microbiology , Swine Diseases/microbiology , beta-Lactamases/genetics , Animals , Anti-Bacterial Agents/pharmacology , Cattle , Cattle Diseases/epidemiology , Escherichia coli/enzymology , Escherichia coli/isolation & purification , Escherichia coli Infections/epidemiology , Escherichia coli Infections/microbiology , Germany/epidemiology , Poultry/microbiology , Poultry Diseases/epidemiology , Sequence Analysis, DNA/veterinary , Swine , Swine Diseases/epidemiology
20.
Article En | MEDLINE | ID: mdl-27549310

Lincosamides, streptogramins, phenicols, and pleuromutilins (LSPPs) represent four structurally different classes of antimicrobial agents that inhibit bacterial protein synthesis by binding to particular sites on the 50S ribosomal subunit of the ribosomes. Members of all four classes are used for different purposes in human and veterinary medicine in various countries worldwide. Bacteria have developed ways and means to escape the inhibitory effects of LSPP antimicrobial agents by enzymatic inactivation, active export, or modification of the target sites of the agents. This review provides a comprehensive overview of the mode of action of LSPP antimicrobial agents as well as of the mutations and resistance genes known to confer resistance to these agents in various bacteria of human and animal origin.


Anti-Bacterial Agents/pharmacology , Chloramphenicol/pharmacology , Lincosamides/pharmacology , Streptogramins/pharmacology , Animals , Bacteria/drug effects , Bacterial Proteins/genetics , Diterpenes/pharmacology , Humans , Microbial Sensitivity Tests , Models, Biological , Polycyclic Compounds , Ribosomes/metabolism , Pleuromutilins
...