Next Article in Journal
Clostridioides difficile Infection Treatment and Outcome Disparities in a National Sample of United States Hospitals
Next Article in Special Issue
First Belgian Report of Ertapenem Resistance in an ST11 Klebsiella Pneumoniae Strain Isolated from a Dog Carrying blaSCO-1 and blaDHA-1 Combined with Permeability Defects
Previous Article in Journal
The Impact of Antibiotics Administration on Mortality for Time in Sepsis and Septic Shock Patients including Possible Reasons for Delayed Administration in Malaysia
Previous Article in Special Issue
Prevalence and Molecular Characterization of Extended-Spectrum β-Lactamase Producing Enterobacterales in Healthy Community Dogs in Israel
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Investigation of In Vitro Susceptibility and Resistance Mechanisms in Skin Pathogens: Perspectives for Fluoroquinolone Therapy in Canine Pyoderma

1
Department of Comparative Biomedical Sciences, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK
2
Department of Clinical Sciences and Services, Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield AL9 7TA, UK
3
Institute of Infection, Veterinary and Ecological Sciences, Department of Veterinary Anatomy, Physiology and Pathology, Veterinary Microbiology Diagnostic, University of Liverpool Leahurst Campus, Neston CH64 7TE, UK
4
Research Support Office, Royal Veterinary College, University of London, London NW1 0TU, UK
*
Author to whom correspondence should be addressed.
Antibiotics 2022, 11(9), 1204; https://doi.org/10.3390/antibiotics11091204
Submission received: 15 August 2022 / Revised: 2 September 2022 / Accepted: 3 September 2022 / Published: 6 September 2022

Abstract

:
Fluoroquinolones (FQ) are commonly used in dogs with bacterial skin infections. Their use as first choice, along with the increased incidence of FQ-resistance, represents a risk to animal and public health. Our study determined minimum inhibitory (MIC) and bactericidal (MBC) concentrations of five FQs in Staphylococcus aureus, Staphylococcus pseudintermedius, and Escherichia coli, together with FQ-resistance mechanisms. MICs, efflux pump (EP) overexpression and MBCs were measured in 249 skin infection isolates following CLSI guidelines (CLSI VET01-A4, CLSI M26-A). Chromosomal and plasmid-mediated resistance genes were investigated after DNA extraction and sequencing. FQ-resistance was detected in 10% of methicillin-susceptible (MS), 90% of methicillin-resistant (MR) staphylococci and in 36% of E. coli. Bactericidal effect was observed except in 50% of MRSA/P for ciprofloxacin and in 20% of MRSPs for enrofloxacin. Highest MICs were associated with double mutation in gyrA (Ser83Leu + Asp87Asn), efflux pumps and three PMQR genes in E. coli, and grlA (Ser80Phe + Glu84Lys) in S. aureus. EP overexpression was high among E. coli (96%), low in S. aureus (1%) and absent in S. pseudintermedius. Pradofloxacin and moxifloxacin showed low MICs with bactericidal effect. Since in vitro FQ resistance was associated with MR, FQ use should be prudently guided by susceptibility testing.

1. Introduction

Canine pyoderma, a cutaneous pyogenic bacterial infection [1], represents one of the most common skin diseases in dogs [2], and is caused, in almost 90% of cases, by Staphylococcus pseudintermedius [3]. Other less-frequently isolated pathogens include Escherichia coli, Pseudomonas aeruginosa, and Streptococcus spp. [1,4]. S. aureus is rarely found on canine skin [5], and the majority of isolates are human-related [6]. However, the increased incidence of infections caused by multidrug-resistant (MDR) and methicillin-resistant (MR) staphylococci (MRSP and MRSA) in dogs is a public health concern [7,8], along with the increased frequency of infections caused by E. coli [9,10,11].
Fluoroquinolones (FQ) are efficacious and licensed for the systemic treatment of bacterial skin infections in dogs [12,13,14,15,16], and their use is advocated in deep pyoderma [17], or widespread and severe superficial pyoderma when antimicrobial susceptibility testing (AST) results are compatible.
FQs have a broad spectrum of activity and are listed as second choice antimicrobials [18], or also classified as category B “prudent” by the European Medicine Agency (EMA), following WHO guidelines on critically important antimicrobials (CIA) in human medicine [19,20]. Their use should be prudently limited to clinical cases where first-line antibiotics have been ineffective [13,21,22], to reduce the risk of antimicrobial resistance both in commensal and pathogenic bacteria [23]. FQ-resistance usually arises in a stepwise manner, when bacteria are sequentially exposed to the drug, and is mediated by mutations of genes encoding for DNA gyrase (gyrA and gyrB), and topoisomerase IV (parC and parE in Gram-negative, or grlA and grlB in Gram-positive). As a result, FQ binding affinity to the topoisomerases is reduced due to alteration of the target protein structure secondary to amino acid substitution [24,25]. Other resistance mechanisms described include: (1) decreased permeability of bacterial cell wall due to down-regulation and under-expression of outer membrane porins, (2) overexpression of efflux pumps [26,27,28,29,30,31,32], (3) resistance conferred by plasmid-mediated quinolone resistance (PMQR) genes, which includes FQ degradation (aac(6′)-Ib-cr) [33], efflux pumps (qepA) [28,29] or disruption of the interaction with FQs by binding to topoisomerases (Qnr family) [34]. Efflux pump upregulation can cause a 4–8-fold increase in MIC. However, major contributions in decreased susceptibility are caused by multiple mutations (up to 128-fold) and, in Gram-negatives by PMQR genes [35].
Previous studies have investigated the susceptibility and prevalence of resistance mechanisms among staphylococci and E. coli of canine origin [6,31,36,37,38,39,40,41,42,43,44,45,46], but limited information exists on the bactericidal activity of isolates collected from canine pyoderma or wound infection cases.
Early veterinary FQs can bind both topoisomerases, conferring bactericidal activity. However, their primary targets are specific for bacterial species: DNA gyrase for Gram-negative and Topoisomerase IV for Gram-positive bacteria, respectively [47,48,49]. In comparison, newer generation pradofloxacin and moxifloxacin may represent an advantage in reducing the likelihood of resistance development as the drugs target both bacterial topoisomerases with increased affinity, conferring low minimum inhibitory concentrations (MIC) and mutant prevention concentrations (MPC) [50,51].
Pradofloxacin, licensed for dogs in Europe in oral formulations [52], has a very similar molecular structure to moxifloxacin, a drug licensed for human use [53]. Silley et al. [54], showed that pradofloxacin exhibits bactericidal activity with regards to minimum bactericidal concentration (MBC), the minimum concentration that kills 99.9% of bacteria. The MBC values were within two MIC doubling dilutions against 90% of selected isolates from unspecified animal species and body sites, but to date no veterinary studies have compared MBC of pradofloxacin with other FQs in isolates specifically from canine skin.
Here we compared MICs and MBCs distributions of selected veterinary and human fluoroquinolones in S. pseudintermedius, S. aureus, and E. coli isolates from canine pyoderma or skin wound infections, with a particular focus on the correlation between methicillin and FQ resistance. It was hypothesized that MICs and MBCs distributions of clinical isolates differed between FQs, and MR in staphylococci was associated with increased MICs and MBCs that predicted FQ-resistance. We also evaluated chromosomal mutations, efflux pump overexpression and the presence of PMQR genes amongst FQ-resistant skin pathogens, with the objective to correlate the resistance mechanisms with susceptibility. We hypothesized that the presence of multiple chromosomal mutations, efflux pumps, and PMQR genes were associated with increased MICs.

2. Results

MICs, MBCs values, MBC/MIC ratios and their respective ranges are shown in Table 1. MICs graphical distributions are shown in Figure 1. Chromosomal mutations, PMQR genes, and efflux pump overexpression detection are represented in Table 2. Supplementary Materials comprise QC growth ranges (Table S1), clinical breakpoints (Table S2), graphical MBCs distributions (Figure S1) and statistical analyses comparing MICs (Table S3) and MBCs (Table S4).

2.1. Minimum Inhibitory Concentration

MICs distributions were bimodal for the three species (Figure 1). In staphylococci, MS and MR resistant isolates showed different modes within each distribution.
S. aureus: 31 out of 34 MSSA (91.2%) and 2 out of 45 MRSA (4.4%) were FQ-susceptible. Among the resistant isolates, two MSSA and two MRSA were resistant to all FQs except for pradofloxacin (MIC 1 µg/mL). Pradofloxacin and moxifloxacin had lower MICs (p < 0.0001) both for MRSA and MSSA (Table S3), compared with enrofloxacin, marbofloxacin and ciprofloxacin. No statistical difference (p > 0.05) was observed between marbofloxacin and ciprofloxacin (p = 0.87) and between pradofloxacin and moxifloxacin (p = 0.34). MICs for MRSA were higher than for MSSA (p < 0.0001) for all FQs tested.
S. pseudintermedius: 50 out of 53 MSSP (94.3%) and 3 out of 52 MRSP (5.8%) were FQ- susceptible. Among the resistant isolates, one MSSP was resistant to all FQs except for pradofloxacin and moxifloxacin (MIC 0.5 µg/mL with both drugs) and two MRSP were susceptible to pradofloxacin (1 µg/mL).
MICs were lower for each FQ tested in MSSP compared with MRSP (p < 0.0001) and lower MICs were observed between both pradofloxacin, moxifloxacin (p < 0.0001) and the other FQs tested.
E. coli: 41 out of 65 isolates (63%) were FQ-susceptible except for moxifloxacin (36 out of 65, 55%). Highly significant differences in MICs and MBCs distribution between fluoroquinolones (p < 0.0001) were observed, and pradofloxacin showed the lowest MICs and MBC (p < 0.0001) when compared with the other FQs tested.

2.2. Minimum Bactericidal Concentration

MBCs distributions were bimodal for the three species (Figure S1). In staphylococci, MS and MR isolates showed different modes within each distribution.
S. aureus: bactericidal effect was observed in all the isolates with an MBC within four two-fold dilutions of the MIC value, except for enrofloxacin (1 out of 34 MSSA, 2.9% and 1 out of 45 MRSA, 2.2%), ciprofloxacin (22 out of 45 MRSA, 48.8%) and moxifloxacin (1 out of 45 MRSA, 2.2%). Pradofloxacin and moxifloxacin had lower MBC values (Table S4), both for MSSA and MRSA, in comparison with the other FQs (p < 0.0001) and higher values were associated with methicillin resistant isolates (p < 0.0001). MBCs for MRSA were higher than MSSA (p < 0.0001) for all FQs tested.
S. pseudintermedius: pradofloxacin and moxifloxacin had lower MBCs (p < 0.0001) compared to the other FQs tested. All the isolates showed MBCs within four doubling dilutions except for enrofloxacin (10 out of 52 MRSP, 19.2%), ciprofloxacin (29 out of 52 MRSP, 55.7%).
E. coli: pradofloxacin showed the lowest MBCs (p < 0.0001) when compared with the other FQs tested. MBC/MIC ratios were within four doubling dilutions only with pradofloxacin and moxifloxacin.

2.3. Resistance Mechanism Detection

S. aureus: in five selected resistant isolates (two MSSA and three MRSA), gyrA showed either single (Ser84Leu, three out of six isolates) or double (Ser84Leu + Gly90Cys, two out of six isolates) chromosomal mutations. A single mutation (ser80Phe) was observed on grlA (Table 2) in four out of six isolates with pradofloxacin MIC ≤ 4 µg/mL whereas two mutations were observed in one out of six isolates with high pradofloxacin MIC (32 µg/mL). No mutations were detected in one isolate showing efflux pump overexpression, which represented 1% (1 out of 79) of the resistant isolates.
S. pseudintermedius: in six selected resistant isolates (3 MSSA and 3 MRSA), single mutations were found on gyrA (Ser84Leu, five out of six, and Glu88Gly, 1 out of six) and on grlA (Ser80Ile, Ser80Arg, Asp84Asn; four, one and one out of six, respectively) (Table 2) both in isolates with intermediate susceptibility (MIC < 2 µg/mL) or low resistance (MIC ≥ 2 µg/mL) to pradofloxacin. None of the isolates showed efflux pump overexpression.
E. coli: all FQ-resistant selected isolates showed double mutations on gyrA (Ser83Leu + Asp87Asn) and single (Ser80Ile) mutation on parC. Double mutations (Ser80Ile + Ala108Val) on parC were observed in three out of six isolates (Table 2).
MICs in the presence of an efflux pump inhibitor (EPI) Phenylalanine-arginine beta-naphthylamide (PAβN) were 4- to 32-fold (median 8-fold) lower (p < 0.0001) than pradofloxacin MICs and 23 out of 24 (96%) of the resistant isolates showed efflux pump overexpression.
Eleven out of twenty-four (46%) resistant E. coli carried PMQR genes: 8 out of 24 (33%) were aac-(6′)-lb-cr, 3 out of 24 (12.5%) qnrS and 2 out of 24 (8.3%) qnrB, whereas the qnrA gene was not detected. Two isolates with high MICs were associated double PMQR genes, the first one with qnrS and aac-(6′)-lb-cr, and the second one with qnrS and qnrB. qepA and oqxAB genes were not detected.

3. Discussion

Our data indicate that pradofloxacin and moxifloxacin overall show low MICs with bactericidal effect both in staphylococci and E. coli, compared to earlier generation veterinary and human FQs. The results are compatible with and expand those previously published on MICs [31,38,40,42,51,55,56,57], in isolates collected from canine skin infections. However, this is the first veterinary study that examined a substantial number of MS and MR staphylococci in canine pyoderma. The percentages of FQ-resistances were high among MR staphylococci (90%) and E. coli (36–40%), in line with previous studies, where resistance was also detected among four non-β-lactam classes [40,58,59,60]. Since the MIC data are obtained from different countries and periods of time, future analysis is needed to investigate the differences in terms of antibiotic pressure and the related resistance development between old and new generation FQs.
This is the first report on MBC of veterinary and human FQs in canine skin isolates. Previous studies showed that the MBC of ciprofloxacin, moxifloxacin, and pradofloxacin in MS and MR S. aureus [61], and pradofloxacin in S. pseudintermedius [54], was within four doubling dilutions of the MIC in all isolates. In E. coli, ciprofloxacin [62], and pradofloxacin [54], showed bactericidal effect in almost all isolates. Our study confirmed the high bactericidal effect in E. coli collected from canine pyoderma and extended to other veterinary and human FQs. However, in staphylococci no bactericidal effect (MBC/MIC ratio > four) was observed for ciprofloxacin in 50% of MRSA and MRSP and in 20% of enrofloxacin in MRSP. A comparison of the MBC90 of ciprofloxacin in MRSA in our study with results from a similar study by Smith and Eng [63], reflects an alarming increase of more than 512-fold over a 30-year period.
Mutations on S. pseudintermedius isolates were found in codons gyrA84 and grlA80 in all selected FQ-resistant isolates as previously reported [45,64], except in one isolate with intermediate/increased susceptibility to pradofloxacin (0.5 µg/mL) with a single mutation on codon 88 (Glu88GLy). The same mutation was previously identified by Descloux et al. [45], in one FQ-resistant isolate with an MIC of enrofloxacin at the breakpoint (4 µg/mL) and one isolate from Japan [65], with intermediate susceptibility to ofloxacin and resistance to enrofloxacin and levofloxacin. In S. aureus, mutations on gyrA (Ser84Leu) and grlA (Ser80Phe) were identified both in isolates with intermediate susceptibility and resistance to pradofloxacin. However, the presence of an additional mutation on gyrA (Gly90Cys), together with grlA (Glu84Lys) as reported by Hiasa et al. [66], conferred high resistance to pradofloxacin (32 µg/mL) and moxifloxacin (16 µg/mL). Further screening is therefore necessary to understand the molecular mechanisms that confer low resistance profiles in pradofloxacin and moxifloxacin when compared to higher resistant MICs in early generation FQs.
All E. coli had two mutations on gyrA (Ser83Leu, Asp87Asn) and one mutation on parC (Ser80Ile). However, additional mutations on parC (Ala108Val, Glu84Val) were screened in isolates with both moderate and, if associated with plasmid genes, high resistance profiles.
Mutations were found to be within the quinolone resistance determining region (QRDR), which are in the proximity of the FQ-binding sites of the primary targets (Tyr122 in E. coli gyrA and Tyr119 in staphylococcal grlA). Mutations are known to reduce the hydrogen bond with FQs [67], with the addition of loss in negative charge with mutations in parC [68]. However, in silico tools such as molecular docking of FQs into the native and mutated protein are necessary to elucidate the primacy of enzyme targeting in veterinary FQs and their associated reduced resistance emergence.
Resistance conferred by efflux pumps was observed to be highly prevalent in E. coli (96%), compared to in S. aureus (1%) or absent in S. pseudintermedius. Pradofloxacin was the only FQ tested against all three bacterial species to detect efflux pump overexpression. Further research is therefore needed to investigate the susceptibility of other fluoroquinolones. It has been suggested that more lipophilic FQs (such as enrofloxacin, and moxifloxacin) are more readily excreted by efflux pumps in comparison to hydrophilic FQs (such as ciprofloxacin, pradofloxacin, and marbofloxacin) [38,69,70,71,72,73]. In S. pseudintermedius, the mechanism was not detected in our study, which is in accordance with previous studies [31,32]. Although efflux pump overexpression was observed in only 1 out of 79 of S. aureus isolates, to our knowledge, this is the first report of FQ resistance due to an efflux pump mechanism in S. aureus from canine pyoderma. Similar results were obtained by Schmitz et al. [74], with moxifloxacin, where the change in MIC after exposure to reserpine were negligible (1–2 dilutions), compared to ciprofloxacin (1–4 dilutions). In contrast to staphylococci, almost all the E. coli isolates showed efflux pump overexpression mechanism, and we identified a higher percentage compared to previous studies on E. coli collected from canine otitis or different body sites [37,38].
With regards to PMQR, nearly 50% of FQ-resistant isolates were associated with at least one PMQR gene. Among the genes screened for, aac-(6′)-lb-cr (33%) and QnrS (12.5%) were the most prevalent, followed by QnrB (8.3%) as also demonstrated in other studies [37,38,75], whereas no QnrA was found. The presence of QnrB together with QnrS was associated with high MICs.
Limitations of the study are represented by the lack of veterinary CBPs for moxifloxacin and ciprofloxacin, that were extrapolated from human guidelines. The higher percentage of resistance of moxifloxacin in E. coli may be associated with discrepancies between EUCAST and CLSI breakpoints guidelines. As other authors have highlighted, this addresses the need for harmonized guidelines across countries [76]. Moreover, CBPs were not available for S. pseudintermedius and were extrapolated from S. aureus as adopted by previous investigators [51].

4. Materials and Methods

4.1. Bacterial Pathogens

A total of 249 isolates from canine skin or wound infections were included in this study. S. aureus (n = 79, 34 methicillin susceptible [MSSA] and 45 methicillin resistant [MRSA]) and S. pseudintermedius (n = 105, 53 MSSP and 52 MRSP), were collected from Germany between 2005–2006 and 2010–2011, respectively. E. coli (n = 65) were collected from the UK (Royal Veterinary College and University of Liverpool) from 2010 and 2017. Species identification was confirmed by the presence of the genes nucA [77,78], in staphylococci and uidA [79], and uspA [80], in E. coli. Methicillin resistance in staphylococci was investigated by the presence of the mecA gene [81].

4.2. Antimicrobial Agents Tested

Enrofloxacin, marbofloxacin, ciprofloxacin, and moxifloxacin were purchased from Merck (Steinheim, Germany). Pradofloxacin was provided by Bayer Animal Health (Monheim, Germany). Stock solutions (1 mg/mL) were dissolved in deionized water, filter sterilized, adjusted for potency according to Clinical and Laboratory Standard Institute guidelines (CLSI VET01-A4, 2013) [82], and stored in darkness at −80 °C for up to one month.

4.3. Minimum Inhibitory and Bactericidal Concentrations

MICs were determined by broth microdilution (CLSI VET01-A4) [82]: isolates were recovered from Brain Heart Infusion (BHI, ThermoFisher, Basingstoke, UK) with 25% glycerol at −80 °C, subcultured onto 5% sheep blood (TCS Bioscience, Botolph Claydon, UK) agar plates (BA, Merck, Steinheim, Germany) and incubated at 37 °C for 18–24 h. After incubation, 3–5 colonies were suspended into glass tubes containing phosphate buffer saline (PBS ThermoFisher, Basingstoke, UK). A spectrophotometer (Densichek®, Biomérieux, Marcy L’Étoile, France) was used to standardize optical density to 0.5 McFarland, equal to 1–2 × 108 colony forming unit/mL (CFU/mL) and the bacterial suspension was diluted to achieve a final concentration of 5 × 105 CFU/mL in a 96-well MIC plate (Sarstedt®, Nümbrecht, Germany) containing two-fold dilution series of the antimicrobial agents. Plates were incubated at 37 °C for 18–24 h and after incubation were manually read against a black paper background; MIC was considered as the lowest concentration where no visible growth was observed for each isolate. MIC50 and MIC90 (50th and 90th percentiles of the distribution) were calculated as the lowest concentrations that inhibited the growth of 50% and 90% of the isolates, respectively. S. aureus ATCC 29213 and E. coli ATCC 25922 were included for quality control purposes. Their respective ranges of MICs are shown in Table S1. Inoculum density and culture purity were confirmed according to EUCAST guidelines (2022) [83].
MICs data were compared with clinical breakpoints published by CLSI guidelines VET01S (2020) [84], for veterinary fluoroquinolones, and CLSI M100 (2022) [85], for human fluoroquinolones. Isolates with “intermediate” susceptibility were considered susceptible with increased exposure as reported by EUCAST. Epidemiological cut-offs (ECOFFs) were also included, if available from datasets, to distinguish between wild- and non-wild-type isolates. Moxifloxacin clinical breakpoints for E. coli were obtained from EUCAST guidelines (2022) [86]. For S. pseudintermedius, moxifloxacin and ciprofloxacin CBPs were adopted from CLSI for S. aureus as previously described in a veterinary study [51]. Clinical breakpoints are listed in Table S2.
After MIC determination, MBC was measured according to CLSI M26-A “Methods for Determining Bactericidal Activity of Antimicrobial Agents; Approved Guidelines” [87].
MBC50 and MBC90 (50th and 90th percentiles of the distribution) were calculated as the lowest concentrations that produced at least a 3log10 reduction (99.9% bacterial killing) of viable bacterial populations (i.e., <5 × 102 CFU/mL) in antibiotic-treated wells where no visible growth was observed in 50% and 90% of the isolates, respectively. Moreover, drugs were considered bactericidal if the MBC/MIC ratio was ≤ 4.

4.4. Efflux Pump Overexpression

Resistant isolates were tested, according to the same MIC method previously described (CLSI VET04), with the addition of EPIs: 20 µg/mL of reserpine and 80 µg/mL of PaβN for E. coli were added to MIC plates containing 2-fold pradofloxacin (used as standard FQ) dilutions. Overexpression of efflux pump was detected if the ratio between MIC in the absence and MIC in the presence of EPI (MIC/MICEPI) was ≥4.

4.5. Chromosomal Mutations and PMQR Genes

Eighteen FQ-resistant isolates (6 S. aureus, 6 S. pseudintermedius, and 6 E. coli, Table 2) were chosen to represent different resistance profiles based on pradofloxacin MICs (low/susceptible with increased exposure, medium, and high resistance).
DNA was sequenced for the presence of chromosomal mutations on the first subunit of the two topoisomerases (DNA gyrase and topoisomerase IV) targeted by fluoroquinolones, namely gyrA and grlA (parC for E. coli).
DNA was extracted from bacterial cells: 5–6 colonies of an overnight culture on sheep blood agar were suspended into Eppendorf tubes containing 1 mL of PBS and centrifuged at 5000 g for 10 min to pellet cells. The supernatant was removed, cell pellet was resuspended in 100 µL of Tris EDTA (TE) buffer and heated at 100 °C for 10 min. For staphylococci, the heated suspension was incubated on ice for 1 min to degrade the cell wall. The final step involved centrifugation at 5000 g for 30 s to pellet cell debris and collect supernatant into sterile Eppendorf tubes.
GyrA and grlA (parC) genes were amplified with polymerase chain reaction (PCR) using previously published protocols [64,88,89]. PCR products were run with electrophoresis on 2% agarose gel together with a positive and a negative control. Results were read with a transilluminator, and results saved on electronic files.
DNA concentration was measured with a fluorometer (Qubit, Invitrogen) and diluted to a standard concentration of 10 ng/mL. Each primer (both forward and reverse) was separately diluted to a standard concentration of 1.3 nmol/µL. Samples were analyzed by Source bioscience (Cambridge) and genetic sequences were aligned and compared with QC isolates with Bioedit 7.2 version.
Moreover, all FQ-resistant E. coli were screened for carriage and prevalence of plasmid-mediated quinolone resistance (PMQR) genes: QnrA, QnrB, QnrS [90], aac(6')-lb-cr, qepA, and oqxAB [91], genes were amplified by PCR according to previously published methods.

4.6. Statistical Analysis

MICs and MBCs data were log2 transformed before statistical analysis and a Shapiro–Wilk test was used to assess normality of the distributions. MS and MR staphylococci were considered as two different bacterial types within the same species.
MICs and MBCs distributions were compared between 5 fluoroquinolones within bacterial type (MS, MR staphylococci and E. coli) using Anderson Darling and Friedman’s tests. Dunn’s post hoc analysis was carried out for pairwise comparisons.
Wilcoxon signed-rank tests were used to compare the median MICs distributions of pradofloxacin in presence and absence of EPI. Analyses were performed using GraphPad Prism version 9.0 statistical software package (San Diego, CA, USA) with p < 0.05 for significance.

5. Conclusions

Lowest MICs and MBCs were measured with pradofloxacin and moxifloxacin. The presence of methicillin resistance can predict FQ-resistance in more than 90% of MR staphylococci and in 36% of E. coli. Bactericidal effect may not be achieved in MR staphylococci with enrofloxacin and ciprofloxacin.
Since FQ-resistance is multifactorial, further molecular screening of resistance mechanisms and its correlation with antimicrobial susceptibility are necessary.
These findings address the need for prudent use of FQs that should be preventively guided by antimicrobial susceptibility testing prior to systemic treatment.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/antibiotics11091204/s1, Table S1: QC growth ranges, Table S2: clinical breakpoints, Figure S1: graphical MBCs distributions, Tables S3 and S4: statistical analysis of MICs and MBCs values.

Author Contributions

Conceptualization, S.A., L.P. and R.B.; methodology, S.A., L.P. and R.B.; software, S.A, L.P. and Y.-M.C.; validation, S.A., L.P. and R.B.; formal analysis, S.A.; investigation, S.A.; resources, A.L., R.B., F.Z. and D.T.; data curation, S.A.; writing—original draft preparation, S.A.; writing—review and editing, S.A., R.B., A.L., F.Z., D.T. and L.P.; visualization, S.A., R.B. and L.P.; supervision, L.P. and R.B.; project administration, L.P.; funding acquisition, L.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Veterinary Medicine Directorate (UK), grant number VM0536.

Acknowledgments

We thank John Readman for kindly providing Klebsiella pneumoniae isolate as a positive control for qepA and oqxAB plasmid genes, and Bayer Animal Health (Monheim, Germany) for kindly providing pradofloxacin powder.

Conflicts of Interest

The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results. LP acted as a consultant for Zoetis, Vetcare, CEVA and Beckton Dickinson, received research grants from DEFRA, WSAVA, PetPlan, Transpharm, UKRI, BBSRC, Horizon 2020 and is an active member of the VetCAST steering committee. FZ, DT, and LP are members of the European Network for Optimization of Veterinary Antimicrobial Treatment (ENOVAT, CA18217).

References

  1. Hillier, A.; Alcorn, J.R.; Cole, L.K.; Kowalski, J.J. Pyoderma caused by Pseudomonas aeruginosa infection in dogs: 20 cases. Vet. Dermatol. 2006, 17, 432–439. [Google Scholar] [CrossRef] [PubMed]
  2. Gortel, K. Recognizing pyoderma: More difficult than it may seem. Vet. Clin. Small Anim. 2013, 43, 1–18. [Google Scholar] [CrossRef] [PubMed]
  3. Petersen, A.D.; Walker, R.D.; Bowman, M.M.; Schott, H.C.; Rosser, E.J., Jr. Frequency of isolation and antimicrobial susceptibility patterns of Staphylococcus intermedius and Pseudomonas aeruginosa isolates from canine skin and ear samples over a 6-year period (1992–1997). J. Am. Anim. Hosp. Assoc. 2002, 38, 407–413. [Google Scholar] [CrossRef] [PubMed]
  4. Summers, J.F.; Brodbelt, D.C.; Forsythe, P.J.; Loeffler, A.; Hendricks, A. The effectiveness of systemic antimicrobial treatment in canine superficial and deep pyoderma: A systematic review. Vet. Dermatol. 2012, 23, 305-e61. [Google Scholar] [CrossRef] [PubMed]
  5. Frank, L.A.; Loeffler, A. Meticillin-resistant Staphylococcus pseudintermedius: Clinical challenge and treatment options. Vet. Dermatol. 2012, 23, 256–283. [Google Scholar] [CrossRef]
  6. Loncaric, I.; Lepuschitz, S.; Ruppitsch, W.; Trstan, A.; Andreadis, T.; Bouchlis, N.; Marbach, H.; Schauer, B.; Szostak, M.P.; Feßler, A.T. Increased genetic diversity of methicillin-resistant Staphylococcus aureus (MRSA) isolated from companion animals. Vet. Microbiol. 2019, 235, 118–126. [Google Scholar] [CrossRef] [PubMed]
  7. Morris, D.O.; Loeffler, A.; Davis, M.F.; Guardabassi, L.; Weese, J.S. Recommendations for approaches to methicillin-resistant staphylococcal infections of small animals: Diagnosis, therapeutic considerations and preventative measures. Clinical Consensus Guidelines of the World Association for Veterinary Dermatology. Vet. Dermatol. 2017, 28, 304-e69. [Google Scholar] [CrossRef]
  8. Lord, J.; Millis, N.; Jones, R.D.; Johnson, B.; Kania, S.A. Patterns of antimicrobial, multidrug and methicillin resistance among Staphylococcus spp. isolated from canine specimens submitted to a diagnostic laboratory in Tennessee, USA: A descriptive study. BMC Vet. Res. 2022, 18, 1–16. [Google Scholar] [CrossRef]
  9. Ishii, S.; Meyer, K.P.; Sadowsky, M.J. Relationship between phylogenetic groups, genotypic clusters, and virulence gene profiles of Escherichia coli strains from diverse human and animal sources. Appl. Environ. Microbiol. 2007, 73, 5703–5710. [Google Scholar] [CrossRef]
  10. Johnson, J.R.; Clabots, C.; Kuskowski, M.A. Multiple-host sharing, long-term persistence, and virulence of Escherichia coli clones from human and animal household members. J. Clin. Microbiol. 2008, 46, 4078–4082. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Platell, J.L.; Cobbold, R.N.; Johnson, J.R.; Heisig, A.; Heisig, P.; Clabots, C.; Kuskowski, M.A.; Trott, D.J. Commonality among fluoroquinolone-resistant sequence type ST131 extraintestinal Escherichia coli isolates from humans and companion animals in Australia. Antimicrob. Agents Chemother. 2011, 55, 3782–3787. [Google Scholar] [CrossRef]
  12. Carlotti, D.N.; Guaguere, E.; Pin, D.; Jasmin, P.; Thomas, E.; Guiral, V. Therapy of difficult cases of canine pyoderma with marbofloxacin: A report of 39 dogs. J. Small Anim. Pract. 1999, 40, 265–270. [Google Scholar] [CrossRef]
  13. Paradis, M.; Abbey, L.; Baker, B.; Coyne, M.; Hannigan, M.; Joffe, D.; Pukay, B.; Trettien, A.; Waisglass, S.; Wellington, J. Evaluation of the clinical efficacy of marbofloxacin (Zeniquin) tablets for the treatment of canine pyoderma: An open clinical trial. Vet. Dermatol. 2001, 12, 163–169. [Google Scholar] [CrossRef]
  14. Horspool, L.J.I.; Van Laar, P.; Van Den Bos, R.; Mawhinney, I. Treatment of canine pyoderma with ibafloxacin and marbofloxacin–fluoroquinolones with different pharmacokinetic profiles. J. Vet. Pharmacol. Ther. 2004, 27, 147–153. [Google Scholar] [CrossRef]
  15. Mueller, R.S.; Stephan, B. Pradofloxacin in the treatment of canine deep pyoderma: A multicentred, blinded, randomized parallel trial. Vet. Dermatol. 2007, 18, 144–151. [Google Scholar] [CrossRef]
  16. Restrepo, C.; Ihrke, P.J.; White, S.D.; Spiegel, I.B.; Affolter, V.K. Evaluation of the clinical efficacy of pradofloxacin tablets for the treatment of canine pyoderma. J. Am. Anim. Hosp. Assoc. 2010, 46, 301–311. [Google Scholar] [CrossRef]
  17. Beco, L.; Guaguere, E.; Méndez, C.L.; Noli, C.; Nuttall, T.; Vroom, M. Suggested guidelines for using systemic antimicrobials in bacterial skin infections: Part 2—Antimicrobial choice, treatment regimens and compliance. Vet. Rec. 2013, 172, 156. [Google Scholar] [CrossRef] [PubMed]
  18. Hillier, A.; Lloyd, D.H.; Weese, J.S.; Blondeau, J.M.; Boothe, D.; Breitschwerdt, E.; Guardabassi, L.; Papich, M.G.; Rankin, S.; Turnidge, J.D. Guidelines for the diagnosis and antimicrobial therapy of canine superficial bacterial folliculitis (A ntimicrobial G uidelines W orking G roup of the I nternational S ociety for C ompanion A nimal I nfectious D iseases). Vet. Dermatol. 2014, 25, 163-e43. [Google Scholar] [CrossRef]
  19. European Medicine Agency. Categorisation of Antibiotics in the European Union. Available online: https://www.ema.europa.eu/en/documents/report/categorisation-antibiotics-european-union-answer-request-european-commission-updating-scientific_en.pdf (accessed on 26 August 2022).
  20. World Health Organization (WHO). Critically Important Antimicrobials for Human Medicine—6th Revision 2018—Ranking of Medically Important Antimicrobials for Risk Management of Antimicrobial Resistance Due to Non-Human Use. Available online: https://www.who.int/publications/i/item/9789241515528 (accessed on 26 August 2022).
  21. Ihrke, P.J.; Papich, M.G.; Demanuelle, T.C. The use of fluoroquinolones in veterinary dermatology. Vet. Dermatol. 1999, 10, 193–204. [Google Scholar] [CrossRef]
  22. Authier, S.; Paquette, D.; Labrecque, O.; Messier, S. Comparison of susceptibility to antimicrobials of bacterial isolates from companion animals in a veterinary diagnostic laboratory in Canada between 2 time points 10 years apart. Can. Vet. J. 2006, 47, 774. [Google Scholar]
  23. Guardabassi, L.; Schwarz, S.; Lloyd, D.H. Pet animals as reservoirs of antimicrobial-resistant bacteria. J. Antimicrob. Chemother. 2004, 54, 321–332. [Google Scholar] [CrossRef] [PubMed]
  24. Piddock, L.J.V. Mechanisms of fluoroquinolone resistance: An update 1994–1998. Drugs 1999, 58, 11–18. [Google Scholar] [CrossRef] [PubMed]
  25. Hooper, D.C. Mechanisms of action and resistance of older and newer fluoroquinolones. Clin. Infect. Dis. 2000, 31, S24–S28. [Google Scholar] [CrossRef] [PubMed]
  26. Ng, E.Y.; Trucksis, M.; Hooper, D.C. Quinolone resistance mediated by norA: Physiologic characterization and relationship to flqB, a quinolone resistance locus on the Staphylococcus aureus chromosome. Antimicrob. Agents Chemother. 1994, 38, 1345–1355. [Google Scholar] [CrossRef]
  27. Everett, M.J.; Jin, Y.F.; Ricci, V.; Piddock, L.J. Contributions of individual mechanisms to fluoroquinolone resistance in 36 Escherichia coli strains isolated from humans and animals. Antimicrob. Agents Chemother. 1996, 40, 2380–2386. [Google Scholar] [CrossRef] [PubMed]
  28. Périchon, B.; Courvalin, P.; Galimand, M. Transferable resistance to aminoglycosides by methylation of G1405 in 16S rRNA and to hydrophilic fluoroquinolones by QepA-mediated efflux in Escherichia coli. Antimicrob. Agents Chemother. 2007, 51, 2464–2469. [Google Scholar] [CrossRef] [PubMed]
  29. Yamane, K.; Wachino, J.; Suzuki, S.; Kimura, K.; Shibata, N.; Kato, H.; Shibayama, K.; Konda, T.; Arakawa, Y. New plasmid-mediated fluoroquinolone efflux pump, QepA, found in an Escherichia coli clinical isolate. Antimicrob. Agents Chemother. 2007, 51, 3354–3360. [Google Scholar] [CrossRef] [PubMed]
  30. Kim, H.B.; Wang, M.; Park, C.H.; Kim, E.C.; Jacoby, G.A.; Hooper, D.C. oqxAB encoding a multidrug efflux pump in human clinical isolates of Enterobacteriaceae. Antimicrob. Agents Chemother. 2009, 53, 3582–3584. [Google Scholar] [CrossRef]
  31. Gebru Awji, E.; Tassew, D.D.; Lee, J.S.; Lee, S.J.; Choi, M.J.; Reza, M.A.; Rhee, M.H.; Kim, T.H.; Park, S.C. Comparative mutant prevention concentration and mechanism of resistance to veterinary fluoroquinolones in Staphylococcus pseudintermedius. Vet. Dermatol. 2012, 23, 376-e69. [Google Scholar] [CrossRef]
  32. Shimizu, T.; Harada, K.; Kataoka, Y. Mutant prevention concentration of orbifloxacin: Comparison between Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus pseudintermedius of canine origin. Acta Vet. Scand. 2013, 55, 37. [Google Scholar] [CrossRef] [PubMed]
  33. Robicsek, A.; Strahilevitz, J.; Jacoby, G.A.; Macielag, M.; Abbanat, D.; Park, C.H.; Bush, K.; Hooper, D.C. Fluoroquinolone-modifying enzyme: A new adaptation of a common aminoglycoside acetyltransferase. Nat. Med. 2006, 12, 83–88. [Google Scholar] [CrossRef] [PubMed]
  34. Xiong, X.; Bromley, E.H.C.; Oelschlaeger, P.; Woolfson, D.N.; Spencer, J. Structural insights into quinolone antibiotic resistance mediated by pentapeptide repeat proteins: Conserved surface loops direct the activity of a Qnr protein from a gram-negative bacterium. Nucleic Acids Res. 2011, 39, 3917–3927. [Google Scholar] [CrossRef] [PubMed]
  35. Redgrave, L.S.; Sutton, S.B.; Webber, M.A.; Piddock, L.J.V. Fluoroquinolone resistance: Mechanisms, impact on bacteria, and role in evolutionary success. Trends Microbiol. 2014, 22, 438–445. [Google Scholar] [CrossRef] [PubMed]
  36. Shaheen, B.W.; Boothe, D.M.; Oyarzabal, O.A.; Wang, C.; Johnson, C.M. Evaluation of the contribution of gyrA mutation and efflux pumps to fluoroquinolone and multidrug resistance in pathogenic Escherichia coli isolates from dogs and cats. Am. J. Vet. Res. 2011, 72, 25–32. [Google Scholar] [CrossRef]
  37. Shaheen, B.W.; Nayak, R.; Foley, S.L.; Boothe, D.M. Chromosomal and plasmid-mediated fluoroquinolone resistance mechanisms among broad-spectrum-cephalosporin-resistant Escherichia coli isolates recovered from companion animals in the USA. J. Antimicrob Chemother 2013, 68, 1019–1024. [Google Scholar] [CrossRef]
  38. Vingopoulou, E.I.; Delis, G.A.; Batzias, G.C.; Kaltsogianni, F.; Koutinas, A.; Kristo, I.; Pournaras, S.; Saridomichelakis, M.N.; Siarkou, V.I. Prevalence and mechanisms of resistance to fluoroquinolones in Pseudomonas aeruginosa and Escherichia coli isolates recovered from dogs suffering from otitis in Greece. Vet. Microbiol. 2018, 213, 102–107. [Google Scholar] [CrossRef]
  39. Ruiz-Ripa, L.; Simón, C.; Ceballos, S.; Ortega, C.; Zarazaga, M.; Torres, C.; Gómez-Sanz, E. S. pseudintermedius and S. aureus lineages with transmission ability circulate as causative agents of infections in pets for years. BMC Vet. Res. 2021, 17, 1–10. [Google Scholar] [CrossRef]
  40. Saputra, S.; Jordan, D.; Worthing, K.A.; Norris, J.M.; Wong, H.S.; Abraham, R.; Trott, D.J.; Abraham, S. Antimicrobial resistance in coagulase-positive staphylococci isolated from companion animals in Australia: A one year study. PLoS ONE 2017, 12, e0176379. [Google Scholar] [CrossRef]
  41. Priyantha, R.; Gaunt, M.C.; Rubin, J.E. Antimicrobial susceptibility of Staphylococcus pseudintermedius colonizing healthy dogs in Saskatoon, Canada. Can. Vet. J. 2016, 57, 65. [Google Scholar]
  42. Schink, A.K.; Kadlec, K.; Hauschild, T.; Brenner Michael, G.; Dorner, J.C.; Ludwig, C.; Werckenthin, C.; Hehnen, H.R.; Stephan, B.; Schwarz, S. Susceptibility of canine and feline bacterial pathogens to pradofloxacin and comparison with other fluoroquinolones approved for companion animals. Vet. Microbiol. 2013, 162, 119–126. [Google Scholar] [CrossRef]
  43. Meunier, D.d.; Acar, J.F.; Martel, J.L.; Kroemer, S.; Valle, M. A seven-year survey of susceptibility to marbofloxacin of pathogenic strains isolated from pets. Int. J. Antimicrob. Agents 2004, 24, 592–598. [Google Scholar] [CrossRef] [PubMed]
  44. Ganière, J.P.; Médaille, C.; Limet, A.; Ruvoen, N.; André-Fontaine, G. Antimicrobial activity of enrofloxacin against Staphylococcus intermedius strains isolated from canine pyodermas. Vet. Dermatol. 2001, 12, 171–175. [Google Scholar] [CrossRef] [PubMed]
  45. Descloux, S.; Rossano, A.; Perreten, V. Characterization of new staphylococcal cassette chromosome mec (SCC mec) and topoisomerase genes in fluoroquinolone-and methicillin-resistant Staphylococcus pseudintermedius. J. Clin. Microbiol. 2008, 46, 1818–1823. [Google Scholar] [CrossRef] [PubMed]
  46. Ganiere, J.P.; Medaille, C.; Mangion, C. Antimicrobial drug susceptibility of Staphylococcus intermedius clinical isolates from canine pyoderma. J. Vet. Med. Ser. B 2005, 52, 25–31. [Google Scholar] [CrossRef]
  47. Peterson, L.R. Quinolone molecular structure-activity relationships: What we have learned about improving antimicrobial activity. Clin. Infect. Dis. 2001, 33, S180–S186. [Google Scholar] [CrossRef]
  48. Drlica, K.; Malik, M. Fluoroquinolones: Action and resistance. Curr. Top. Med. Chem. 2003, 3, 249–282. [Google Scholar] [CrossRef]
  49. Drlica, K.; Malik, M.; Kerns, R.J.; Zhao, X. Quinolone-mediated bacterial death. Antimicrob. Agents Chemother. 2008, 52, 385–392. [Google Scholar] [CrossRef]
  50. Wetzstein, H.G. Comparative mutant prevention concentrations of pradofloxacin and other veterinary fluoroquinolones indicate differing potentials in preventing selection of resistance. Antimicrob. Agents Chemother. 2005, 49, 4166–4173. [Google Scholar] [CrossRef]
  51. Plowgian, C.; Blondeau, J.M.; Levinson, M.; Rosenkrantz, W. A pilot study on the comparative minimum inhibitory and mutant prevention concentration values for moxifloxacin and pradofloxacin against canine and human isolates of Staphylococcus pseudintermedius and S. schleiferi. Vet. Dermatol. 2019, 30, 481-e142. [Google Scholar] [CrossRef]
  52. Lees, P. Pharmacokinetics, pharmacodynamics and therapeutics of pradofloxacin in the dog and cat. J. Vet. Pharmacol. Ther. 2013, 36, 209–221. [Google Scholar] [CrossRef] [PubMed]
  53. Messias, A.; Gekeler, F.; Wegener, A.; Dietz, K.; Kohler, K.; Zrenner, E. Retinal safety of a new fluoroquinolone, pradofloxacin, in cats: Assessment with electroretinography. Doc. Ophthalmol. 2008, 116, 177–191. [Google Scholar] [CrossRef] [PubMed]
  54. Silley, P.; Stephan, B.; Greife, H.A.; Pridmore, A. Bactericidal properties of pradofloxacin against veterinary pathogens. Vet. Microbiol. 2012, 157, 106–111. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, B.-T.; Wang, X.-M.; Liao, X.-P.; Sun, J.; Zhu, H.-Q.; Chen, X.-Y.; Liu, Y.-H. Plasmid-mediated quinolone resistance determinants oqxAB and aac (6′)-Ib-cr and extended-spectrum β-lactamase gene bla CTX-M-24 co-located on the same plasmid in one Escherichia coli strain from China. J. Antimicrob. Chemother. 2011, 66, 1638–1639. [Google Scholar] [CrossRef] [PubMed]
  56. Liu, X.; Boothe, D.M.; Jin, Y.; Thungrat, K. In vitro potency and efficacy favor later generation fluoroquinolones for treatment of canine and feline Escherichia coli uropathogens in the United States. World J. Microbiol. Biotechnol. 2013, 29, 347–354. [Google Scholar] [CrossRef]
  57. Ludwig, C.; de Jong, A.; Moyaert, H.; El Garch, F.; Janes, R.; Klein, U.; Morrissey, I.; Thiry, J.; Youala, M. Antimicrobial susceptibility monitoring of dermatological bacterial pathogens isolated from diseased dogs and cats across Europe (ComPath results). J. Appl. Microbiol. 2016, 121, 1254–1267. [Google Scholar] [CrossRef]
  58. Dancer, S.J. The effect of antibiotics on methicillin-resistant Staphylococcus aureus. J. Antimicrob. Chemother. 2007, 61, 246–253. [Google Scholar] [CrossRef]
  59. Harbarth, S.; Fankhauser, C.; Schrenzel, J.; Christenson, J.; Gervaz, P.; Bandiera-Clerc, C.; Renzi, G.; Vernaz, N.; Sax, H.; Pittet, D. Universal screening for methicillin-resistant Staphylococcus aureus at hospital admission and nosocomial infection in surgical patients. JAMA 2008, 299, 1149–1157. [Google Scholar] [CrossRef]
  60. Bemis, D.A.; Jones, R.D.; Frank, L.A.; Kania, S.A. Evaluation of susceptibility test breakpoints used to predict mecA-mediated resistance in Staphylococcus pseudintermedius isolated from dogs. J. Vet. Diagn. Investig. 2009, 21, 53–58. [Google Scholar] [CrossRef]
  61. Berrington, A.W.; Perry, J.D.; Gould, F.K. Bactericidal activity of moxifloxacin against Staphylococcus aureus. Clin. Microbiol. Infect. 2001, 7, 161–163. [Google Scholar] [CrossRef]
  62. Hansen, G.T.; Blondeau, J.M. Comparison of the minimum inhibitory, mutant prevention and minimum bactericidal concentrations of ciprofloxacin, levofloxacin and garenoxacin against enteric Gram-negative urinary tract infection pathogens. J. Chemother. 2005, 17, 484–492. [Google Scholar] [CrossRef]
  63. Smith, S.M.; Eng, R.H. Activity of ciprofloxacin against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 1985, 27, 688–691. [Google Scholar] [CrossRef] [PubMed]
  64. Intorre, L.; Vanni, M.; Di Bello, D.; Pretti, C.; Meucci, V.; Tognetti, R.; Soldani, G.; Cardini, G.; Jousson, O. Antimicrobial susceptibility and mechanism of resistance to fluoroquinolones in Staphylococcus intermedius and Staphylococcus schleiferi. J. Vet. Pharmacol. Ther. 2007, 30, 464–469. [Google Scholar] [CrossRef] [PubMed]
  65. Onuma, K.; Tanabe, T.; Sato, H. Antimicrobial resistance of Staphylococcus pseudintermedius isolates from healthy dogs and dogs affected with pyoderma in Japan. Vet. Dermatol. 2012, 23, 17-e15. [Google Scholar] [CrossRef] [PubMed]
  66. Hiasa, H. The Glu-84 of the ParC subunit plays critical roles in both topoisomerase IV—Quinolone and Topoisomerase IV—DNA interactions. Biochemistry 2002, 41, 11779–11785. [Google Scholar] [CrossRef]
  67. Wohlkonig, A.; Chan, P.F.; Fosberry, A.P.; Homes, P.; Huang, J.; Kranz, M.; Leydon, V.R.; Miles, T.J.; Pearson, N.D.; Perera, R.L. Structural basis of quinolone inhibition of type IIA topoisomerases and target-mediated resistance. Nat. Struct. Mol. Biol. 2010, 17, 1152–1153. [Google Scholar] [CrossRef]
  68. Sáenz, Y.; Zarazaga, M.; Briñas, L.; Ruiz-Larrea, F.; Torres, C. Mutations in gyrA and parC genes in nalidixic acid-resistant Escherichia coli strains from food products, humans and animals. J. Antimicrob. Chemother. 2003, 51, 1001–1005. [Google Scholar] [CrossRef]
  69. Neyfakh, A.A.; Borsch, C.M.; Kaatz, G.W. Fluoroquinolone resistance protein NorA of Staphylococcus aureus is a multidrug efflux transporter. Antimicrob. Agents Chemother. 1993, 37, 128–129. [Google Scholar] [CrossRef]
  70. Kaatz, G.W.; Seo, S.M. Inducible NorA-mediated multidrug resistance in Staphylococcus aureus. Antimicrob. Agents Chemother. 1995, 39, 2650–2655. [Google Scholar] [CrossRef]
  71. Markham, P.N.; Neyfakh, A.A. Inhibition of the multidrug transporter NorA prevents emergence of norfloxacin resistance in Staphylococcus aureus. Antimicrob. Agents Chemother. 1996, 40, 2673–2674. [Google Scholar] [CrossRef]
  72. Tejedor, M.a.T.; Martín, J.L.; Navia, M.; Freixes, J.; Vila, J. Mechanisms of fluoroquinolone resistance in Pseudomonas aeruginosa isolates from canine infections. Vet. Microbiol. 2003, 94, 295–301. [Google Scholar] [CrossRef]
  73. Harada, K.; Arima, S.; Niina, A.; Kataoka, Y.; Takahashi, T. Characterization of Pseudomonas aeruginosa isolates from dogs and cats in Japan: Current status of antimicrobial resistance and prevailing resistance mechanisms. Microbiol. Immunol. 2012, 56, 123–127. [Google Scholar] [CrossRef]
  74. Schmitz, F.-J.; Fluit, A.C.; Lückefahr, M.; Engler, B.; Hofmann, B.; Verhoef, J.; Heinz, H.P.; Hadding, U.; Jones, M.E. The effect of reserpine, an inhibitor of multidrug efflux pumps, on the in-vitro activities of ciprofloxacin, sparfloxacin and moxifloxacin against clinical isolates of Staphylococcus aureus. J. Antimicrob. Chemother. 1998, 42, 807–810. [Google Scholar] [CrossRef] [Green Version]
  75. Guillard, T.; de Jong, A.; Limelette, A.; Lebreil, A.L.; Madoux, J.; De Champs, C.; ComPath Study, G. Characterization of quinolone resistance mechanisms in Enterobacteriaceae recovered from diseased companion animals in Europe. Vet. Microbiol. 2016, 194, 23–29. [Google Scholar] [CrossRef]
  76. Cusack, T.P.; Ashley, E.A.; Ling, C.L.; Rattanavong, S.; Roberts, T.; Turner, P.; Wangrangsimakul, T.; Dance, D.A.B. Impact of CLSI and EUCAST breakpoint discrepancies on reporting of antimicrobial susceptibility and AMR surveillance. Clin. Microbiol. Infect. 2019, 25, 910–911. [Google Scholar] [CrossRef]
  77. Brakstad, O.G.; Aasbakk, K.; Maeland, J.A. Detection of Staphylococcus aureus by polymerase chain reaction amplification of the nuc gene. J. Clin. Microbiol. 1992, 30, 1654–1660. [Google Scholar] [CrossRef]
  78. Becker, K.; von Eiff, C.; Keller, B.; Brück, M.; Etienne, J.; Peters, G. Thermonuclease gene as a target for specific identification of Staphylococcus intermedius isolates: Use of a PCR-DNA enzyme immunoassay. Diagn. Microbiol. Infect. Dis. 2005, 51, 237–244. [Google Scholar] [CrossRef]
  79. McDaniels, A.E.; Rice, E.W.; Reyes, A.L.; Johnson, C.H.; Haugland, R.A.; Stelma Jr, G.N. Confirmational identification of Escherichia coli, a comparison of genotypic and phenotypic assays for glutamate decarboxylase and beta-D-glucuronidase. Appl. Environ. Microbiol. 1996, 62, 3350–3354. [Google Scholar] [CrossRef]
  80. Anastasi, E.M.; Matthews, B.; Gundogdu, A.; Vollmerhausen, T.L.; Ramos, N.L.; Stratton, H.; Ahmed, W.; Katouli, M. Prevalence and persistence of Escherichia coli strains with uropathogenic virulence characteristics in sewage treatment plants. Appl. Environ. Microbiol. 2010, 76, 5882–5886. [Google Scholar] [CrossRef]
  81. Brakstad, O.G.; MÆLand, J.A.; Tveten, Y. Multiplex polymerase chain reaction for detection of genes for Staphylococcus aureus thermonuclease and methicillin resistance and correlation with oxacillin resistance. Apmis 1993, 101, 681–688. [Google Scholar] [CrossRef]
  82. Clinical and Laboratory Standard Institue (CLSI). Performance Standards for Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated From Animals; Approved Standard—Fourth Edition—CLSI VET01-A4. 2013. Available online: https://clsi.org/media/1531/vet01a4_sample.pdf (accessed on 1 April 2022).
  83. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Available online: https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Disk_test_documents/2022_manuals/Reading_guide_BMD_v_4.0_2022.pdf (accessed on 1 April 2022).
  84. Clinical and Laboratory Standard Institue (CLSI). Performance Standards for Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated From Animals, 5th Edition—CLSI VET01S. 2020. Available online: https://clsi.org/media/m1gppkx2/vet01sed5_sample.pdf (accessed on 1 April 2022).
  85. Clinical and laboratory standard institue (CLSI). Performance Standards for Antimicrobial Susceptibility Testing, 32nd Edition—CLSI M100. 2022. Available online: https://clsi.org/media/2663/m100ed29_sample.pdf (accessed on 1 April 2022).
  86. European Committee on Antimicrobial Susceptibility Testing (EUCAST). Available online: https://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_12.0_Breakpoint_Tables.pdf (accessed on 1 April 2022).
  87. Clinical and Laboratory Standard institue (CLSI). Methods for Determining Bactericidal Activity of Antimicrobial Agents; Approved Guideline—M26-A. 1999. Available online: https://clsi.org/media/1462/m26a_sample.pdf (accessed on 1 April 2022).
  88. Kim, H.B.; Park, C.H.; Kim, C.J.; Kim, E.-C.; Jacoby, G.A.; Hooper, D.C. Prevalence of plasmid-mediated quinolone resistance determinants over a 9-year period. Antimicrob. Agents Chemother. 2009, 53, 639–645. [Google Scholar] [CrossRef]
  89. Schmitz, F.-J.; Jones, M.E.; Hofmann, B.; Hansen, B.; Scheuring, S.; Lückefahr, M.; Fluit, A.; Verhoef, J.; Hadding, U.; Heinz, H.-P. Characterization of grlA, grlB, gyrA, and gyrB mutations in 116 unrelated isolates of Staphylococcus aureus and effects of mutations on ciprofloxacin MIC. Antimicrob. Agents Chemother. 1998, 42, 1249–1252. [Google Scholar] [CrossRef] [PubMed]
  90. Robicsek, A.; Strahilevitz, J.; Sahm, D.F.; Jacoby, G.A.; Hooper, D.C. qnr prevalence in ceftazidime-resistant Enterobacteriaceae isolates from the United States. Antimicrob. Agents Chemother. 2006, 50, 2872–2874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Ciesielczuk, H.; Hornsey, M.; Choi, V.; Woodford, N.; Wareham, D.W. Development and evaluation of a multiplex PCR for eight plasmid-mediated quinolone-resistance determinants. J. Med. Microbiol. 2013, 62, 1823–1827. [Google Scholar] [CrossRef] [PubMed]
Figure 1. MIC distributions in S. aureus (first row), S. pseudintermedius (second row) and E. coli (third row) of five fluoroquinolones. Blue-dotted lines indicate the clinical breakpoint, the highest MIC value considered susceptible, and the epidemiological cut-off (ECOFF), the MIC value that separates wild-type from non-wild-type bacteria.
Figure 1. MIC distributions in S. aureus (first row), S. pseudintermedius (second row) and E. coli (third row) of five fluoroquinolones. Blue-dotted lines indicate the clinical breakpoint, the highest MIC value considered susceptible, and the epidemiological cut-off (ECOFF), the MIC value that separates wild-type from non-wild-type bacteria.
Antibiotics 11 01204 g001
Table 1. MIC, MBC values and MBC/MIC ratios of five fluoroquinolones among a total of 249 isolates of three bacterial species (S. aureus, S. pseudintermedius, E. coli) and their subtypes (MS and MR staphylococci), isolated from canine pyoderma and/or wound infection cases.
Table 1. MIC, MBC values and MBC/MIC ratios of five fluoroquinolones among a total of 249 isolates of three bacterial species (S. aureus, S. pseudintermedius, E. coli) and their subtypes (MS and MR staphylococci), isolated from canine pyoderma and/or wound infection cases.
Bacterial TypeFluoroquinoloneMIC (μg/mL)MBC (μg/mL)MBC/MIC Ratio *
MIC50MIC90 RangeMBC50 MBC90 Range MBC50/MIC50MBC90/MIC90Range
MSSA
(n = 34)
ENR 0.2520.125–160.2520.125–161 21–8
MAR0.510.25–32120.25–32121–4
PRA0.060.1250.03–20.1250.250.03–4121–2
CIP0.520.25–320.5 80.25–64121–4
MOX0.1250.50.03–4 0.250.50.06–4121–4
MRSA
(n = 45)
ENR16320.25–12832640.5– ≥512241–8
MAR32640.5–12864640.5–128121–2
PRA440.125–32480.25–32121–4
CIP1281280.5–256256≥5120.5 – ≥5124161–32
MOX480.125–16 8160.125–32121–8
MSSP
(n = 53)
ENR0.12520.125–320.2520.125–64 1 21–4
MAR0.2520.25–32 0.5 20.25–641 21–4
PRA0.060.250.016–4 0.06 0.250.03–41 21–8
CIP0.2510.125–160.2520.125– ≥512121–64
MOX0.060.50.03–4 0.060.50.03–8121–2
MRSP
(n = 52)
ENR32640.06–6464≥5120.125– ≥5122321–64
MAR32640.25–6464640.5–64 1 21–2
PRA240.03–8 440.06–16 1 21–2
CIP32320.125–128256 5120.125– ≥5128161–64
MOX480.06–80.0680.125–32 221–8
E. coli
(n = 65)
ENR0.061280.016–512 0.06 2560.016–5121 21–4
MAR0.06640.016–256 0.125640.016–5121 21–8
PRA0.03320.016–1280.03320.016–1281 21–4
CIP0.032560.008– ≥5120.032560.008– ≥5121 21–8
MOX0.125640.016–5120.125640.016–5121 21–4
* Bactericidal effect if MBC/MIC ratio ≤ 4.
Table 2. Molecular analysis of 18 selected FQ-resistant isolates. Comparison between chromosomal mutations on DNA gyrase (gyrA) and topoisomerase IV (grlA/parC), plasmid mediated quinolone resistance (PMQR), ratio between MIC of pradofloxacin and MIC with pradofloxacin + efflux pump inhibitor (MICepi) in selected resistant isolates. The isolates were chosen based on their pradofloxacin susceptibility.
Table 2. Molecular analysis of 18 selected FQ-resistant isolates. Comparison between chromosomal mutations on DNA gyrase (gyrA) and topoisomerase IV (grlA/parC), plasmid mediated quinolone resistance (PMQR), ratio between MIC of pradofloxacin and MIC with pradofloxacin + efflux pump inhibitor (MICepi) in selected resistant isolates. The isolates were chosen based on their pradofloxacin susceptibility.
ISOLATE IDMIC
Enrofloxacin
(µg/mL)
MIC
Marbofloxacin
(µg/mL)
MIC
Pradofloxacin
(µg/mL)
MIC
Ciprofloxacin
(µg/mL)
MIC
Moxifloxacin
(µg/mL)
gyrAgrlA (parC)PMQRMIC/MICepi
MSSA B05316321164Ser84LeuSer80PheNA1
MSSA B0718161164No mutationNo mutationNA16 *
MSSA B0748162324Ser84LeuSer80PheNA1
MRSA A019163211284Ser84LeuSer80PheNA1
MRSA A0696412842561Ser84Leu, Gly90CysSer80PheNA1
MRSA A13216643225616Ser84Leu, Gly90CysSer80Phe, Glu84LysNA1
MSSP 098440.540.5Glu88GlySer80ArgNA1
MSSP 11516162162Ser84LeuAsp84AsnNA1
MSSP 09932324164Ser84LeuSer80IleNA2
MRSP 04532321324Ser84LeuSer80IleNA1
MRSP 03864322322Ser84LeuSer80IleNA1
MRSP 06764644324Ser84LeuSer80IleNA2
E. coli 10L-225383243216Ser83Leu, Asp87AsnSer80Ile, Ala108Val-4 *
E. coli 16L-40636432412816Ser83Leu, Asp87AsnSer80Ile, Glu84Valaac-(6’)-lb-cr2
E. coli 15L-327512864326464Ser83Leu, Asp87AsnSer80IleQnrB4 *
E. coli 28230512864326464Ser83Leu, Asp87AsnSer80IleQnrB8 *
E. coli 13L-486525612864256128Ser83Leu, Asp87AsnSer80Ile, Ala108ValQnrS8 *
E. coli 13L-528312825664128256Ser83Leu, Asp87AsnSer80IleQnrS, aac-(6’)-lb-cr8 *
E. coli 13L-6009512256128256512Ser83Leu, Asp87AsnSer80IleQnrB, QnrS, aac-(6’)-lb-cr8 *
(*): Efflux pump overexpression (MIC/MICepi ≥ 4). Yellow colour Resistant, Green colour Susceptible, increased exposure (according to EUCAST) NA= not assessed.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Azzariti, S.; Bond, R.; Loeffler, A.; Zendri, F.; Timofte, D.; Chang, Y.-M.; Pelligand, L. Investigation of In Vitro Susceptibility and Resistance Mechanisms in Skin Pathogens: Perspectives for Fluoroquinolone Therapy in Canine Pyoderma. Antibiotics 2022, 11, 1204. https://doi.org/10.3390/antibiotics11091204

AMA Style

Azzariti S, Bond R, Loeffler A, Zendri F, Timofte D, Chang Y-M, Pelligand L. Investigation of In Vitro Susceptibility and Resistance Mechanisms in Skin Pathogens: Perspectives for Fluoroquinolone Therapy in Canine Pyoderma. Antibiotics. 2022; 11(9):1204. https://doi.org/10.3390/antibiotics11091204

Chicago/Turabian Style

Azzariti, Stefano, Ross Bond, Anette Loeffler, Flavia Zendri, Dorina Timofte, Yu-Mei Chang, and Ludovic Pelligand. 2022. "Investigation of In Vitro Susceptibility and Resistance Mechanisms in Skin Pathogens: Perspectives for Fluoroquinolone Therapy in Canine Pyoderma" Antibiotics 11, no. 9: 1204. https://doi.org/10.3390/antibiotics11091204

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop