Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Restoration of susceptibility to amikacin by 8-hydroxyquinoline analogs complexed to zinc

  • Jesus Magallon,

    Roles Formal analysis, Investigation, Writing – review & editing

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

  • Kevin Chiem,

    Roles Formal analysis, Investigation, Writing – review & editing

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

  • Tung Tran,

    Roles Formal analysis, Investigation, Writing – review & editing

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

  • Maria S. Ramirez,

    Roles Formal analysis, Writing – review & editing

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

  • Veronica Jimenez,

    Roles Formal analysis, Investigation, Writing – review & editing

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

  • Marcelo E. Tolmasky

    Roles Conceptualization, Formal analysis, Investigation, Writing – original draft, Writing – review & editing

    mtolmasky@fullerton.edu

    Affiliation Center for Applied Biotechnology Studies, Department of Biological Science, College of Natural Sciences and Mathematics, California State University Fullerton, Fullerton, CA, United States of America

Abstract

Gram-negative pathogens resistant to amikacin and other aminoglycosides of clinical relevance usually harbor the 6’-N-acetyltransferase type Ib [AAC(6')-Ib], an enzyme that catalyzes inactivation of the antibiotic by acetylation using acetyl-CoA as donor substrate. Inhibition of the acetylating reaction could be a way to induce phenotypic conversion to susceptibility in these bacteria. We have previously observed that Zn2+ acts as an inhibitor of the enzymatic acetylation of aminoglycosides by AAC(6')-Ib, and in complex with ionophores it effectively reduced the levels of resistance in cellulo. We compared the activity of 8-hydroxyquinoline, three halogenated derivatives, and 5-[N-Methyl-N-Propargylaminomethyl]-8-Hydroxyquinoline in complex with Zn2+ to inhibit growth of amikacin-resistant Acinetobacter baumannii in the presence of the antibiotic. Two of the compounds, clioquinol (5-chloro-7-iodo-8-hydroxyquinoline) and 5,7-diiodo-8-hydroxyquinoline, showed robust inhibition of growth of the two A. baumannii clinical isolates that produce AAC(6')-Ib. However, none of the combinations had any activity on another amikacin-resistant A. baumannii strain that possesses a different, still unknown mechanism of resistance. Time-kill assays showed that the combination of clioquinol or 5,7-diiodo-8-hydroxyquinoline with Zn2+ and amikacin was bactericidal. Addition of 8-hydroxyquinoline, clioquinol, or 5,7-diiodo-8-hydroxyquinoline, alone or in combination with Zn2+, and amikacin to HEK293 cells did not result in significant toxicity. These results indicate that ionophores in complex with Zn2+ could be developed into potent adjuvants to be used in combination with aminoglycosides to treat Gram-negative pathogens in which resistance is mediated by AAC(6')-Ib and most probably other related aminoglycoside modifying enzymes.

Introduction

Among many mechanisms bacteria have evolved to resist antibiotics, enzymatic modification is one of the most efficient [1]. In the case of aminoglycosides, bactericidal antibiotics used to treat a wide range of bacterial infections, the most relevant mechanisms of resistance in the clinics are enzymatic inactivation by acetylation, nucleotidylation, or phosphorylation [13]. Although more than hundred aminoglycoside modifying enzymes have been identified in bacterial pathogens, the acetyltransferase AAC(6')-Ib, which mediates resistance to amikacin and other aminoglycosides, is the most widespread among Gram-negative clinical isolates [46]. The progressive acquisition of this gene is eroding the usefulness of amikacin as well as other aminoglycosides. One way to overcome this problem is the design of new antimicrobials such as the recent introduction of plazomicin [7]. However, since this is a slow and expensive process and resistance will inevitably develop against the new antibiotics, these efforts must be complemented by other strategies to prolong the useful life of existing drugs [1, 2, 811]. In the case of aminoglycosides, in addition to the design of new molecules [7, 1214], there is active research to find inhibitors of expression of aminoglycoside modifying enzymes [1518] and to design enzymatic inhibitors [1, 2, 9, 10, 1922]. A recent breakthrough in the search for inhibitors of enzymatic inactivation of aminoglycoside was the finding that Zn2+ and other metal ions inhibit the acetylation of aminoglycosides mediated by AAC(6')-Ib in vitro [23]. Although concentrations beyond toxic levels were needed to interfere with resistance in growing bacteria, further research showed that the action of the metal was enhanced when complexed to ionophores, in which case low concentrations were sufficient to overcome resistance in several aminoglycoside-resistant bacteria [2326]. We recently showed that two classes of ionophores, clioquinol (5-chloro-7-iodo-8-hydroxyquinoline)(CI8HQ) and pyrithione (N-hydroxypyridine-2-thione), when complexed to Zn2+ or Cu2+, significantly reduce the levels of resistance to amikacin in Escherichia coli, Klebsiella pneumoniae, and Acinetobacter baumannii strains harboring the aac(6')-Ib gene [2426]. CI8HQ and other substituted 8-hydroxyquinolines are being tested as treatment for cancer, neurodegenerative conditions such as Alzheimer’s, Parkinson’s, and Huntington’s diseases, and lead poisoning [2730]. The ongoing studies and uses of these compounds indicate that human toxicity is not a serious impediment in their development as drugs for diverse diseases [29, 31]. These facts make CI8HQ and other substituted 8-hydroxyquinolines excellent candidates to be used in combination with aminoglycosides in the treatment of resistant infections. In this work, we compared the effect of commercially available substituted 8-hydroxyquinolines complexed to Zn2+ on growth of amikacin-resistant A. baumannii clinical isolates.

Materials and methods

Bacterial strains and reagents

The A. baumannii A155 [32], A144 [33], and Ab33405 [34] clinical isolates were used in growth and time-killing experiments to test the ability of the ionophores complexed to zinc to reduce resistance to amikacin. A. baumannii A118 [35], A42 [36], and ATCC 17978 [37] were used to determine minimal inhibitory concentrations (MIC) of susceptible strains. All three strains, A155, A144, and Ab33405, are resistant to amikacin but only A144 and A155 naturally carry aac(6′)-Ib [32, 33]. Ionophores and amikacin sulfate were purchased from MilliporeSigma. [Acetyl-1-14C]-Acetyl Coenzyme A was purchased from Perkin-Elmer. Etest strips were purchased from bioMérieux.

Enzymatic acetylation assays

Acetylation activity was assessed using the phosphocellulose paper binding assay as described previously [38]. Amikacin and [Acetyl-1-14C]-Acetyl Coenzyme A were used as substrates in reactions carried out in the presence of the soluble content of cells that were disrupted by sonication as described previously [39]. The reactions were carried out in a final volume of 25 μl containing 200 mM Tris-HCl, pH 7.6, 200 μM amikacin, 0.5 μCi [Acetyl-1-14C]-Acetyl Coenzyme A (specific activity, 60 mCi/mmol), and the enzymatic extract (120 μg protein). The reaction mixtures were incubated at 37°C for 1 h and then 20 μl were spotted on phosphocellulose paper strips. The unreacted radioactive donor substrate was eliminated from the phosphocellulose paper by submersion in 1 l hot water (80°C) followed by several washes with water at room temperature. The phosphocellulose paper strips were allowed to dry before determining the radioactivity.

Growth inhibition, time-kill, and MIC assays

The inhibition of growth of A. baumannii strains by amikacin and ionophore-zinc complexes was tested inoculating 100-μl Mueller-Hinton broth in microtiter plates with the specified additions using the BioTek Synergy 5 microplate reader [23]. The cultures were carried out at 37°C with shaking and contained dimethyl sulfoxide (DMSO) at a final concentration of 0.5%. The optical density at 600 nm (OD600) of the cultures was determined every 20 minutes for 20 h. Time-kill assays were carried out as described before [40]. Briefly, cells were cultured to 106 cfu/ml in Mueller-Hinton broth. At this point the indicated concentrations of amikacin, ionophore, and zinc were added, and the cultures were continued at 37°C with shaking. Samples were removed at 0, 4, 8, 20, and 32 h, serially diluted, plated on Mueller-Hinton agar, and incubated at 37°C for 20 hours to determine the number of cfu/ml. MIC values were determined by the gradient diffusion method (Etest) with commercial strips (bioMérieux) following the procedures recommended by the supplier.

Cytotoxicity assays

Levels of cytotoxicity were determined using the LIVE/DEAD Viability/Cytotoxicity Kit for mammalian cells (Molecular Probes) as described [41]. HEK293 cells plated at a density of 103 cells/well were cultured overnight under standard conditions in flat bottom, 96-well, black microtiter plates. The compounds being tested, dissolved in dimethyl sulfoxide (DMSO), were then added to the cells at increasing concentrations as indicated, and incubation was continued. As control DMSO was added to duplicate wells at same final concentration reached when adding the compounds being tested. After 24 h, the cells were washed with sterile D-PBS and incubated with the LIVE/DEAD reagent (2 μM ethidium homodimer 1 and 1 μM calcein-AM) for 30 min at 37°C, and the fluorescence level at 645 nm (dead cells) and 530 nm (live cells) was measured. The percentage of dead cells was calculated relative to the cells treated with DMSO. The maximum toxicity control was determined using cells incubated in the presence of 0.1% Triton X-100 for 10 min. Experiments were conducted in triplicate. The results were expressed as mean ± SD of three independent experiments.

Results

Combination therapies consisting of an antibiotic and an inhibitor of resistance can be an invaluable tool in the search for solutions to the multidrug resistance problem [10]. While this strategy has already been reduced to practice in the case of pathogens resistant to β-lactams [42], efforts to develop inhibitors of resistance to aminoglycosides are still in experimental stages. We have recently found that ionophores complexed to Zn2+ or Cu2+ could be potentiators that decrease the levels of resistance to amikacin in K. pneumoniae and A. baumannii clinical isolates [2325]. Since one of the ionophores that in complex with Zn2+ demonstrated activity as an inhibitor of the resistance to amikacin was CI8HQ, a substituted 8-hydroxyquinoline (8HQ), we expanded our studies to other compounds with these characteristics. Fig 1 shows the compounds tested in this work. The tests were carried out using as models three A. baumannii clinical isolates, two of them, A155 and A144, harboring the aac(6')-Ib gene [34, 43]. The third strain, Ab33405 does not carry this gene and exhibits resistance to amikacin by a different mechanism. Although this mechanism remains to be elucidated, it most probably consists of phosphorylation mediated by the aphA6 gene found in its genome [34, 44].

thumbnail
Fig 1. Chemical structures of 8-hydroxyquinoline and derivative compounds.

Structures, names, and abbreviations of the compounds used in this study.

https://doi.org/10.1371/journal.pone.0217602.g001

Growth curves in the presence of incremental concentrations of amikacin showed that the strains harboring aac(6')-Ib, A144 and A155, can grow in up to 16 μg/ml of the antibiotic (S1A and S1B Fig). Conversely, strain Ab33405 had a different behavior, while the lag phase became longer as the amikacin concentration was increased, healthy growth was observed at all tested concentrations (S1C Fig). These results are in agreement with the finding that the latter strain resists amikacin using a mechanism different from that in strains A144 and A155. The MIC values of control amikacin susceptible A. baumannii strains A118, A42, and ATCC 17978 were 2, 1.5, and 1.5 μg/ml, respectively.

To confirm that A. baumannii Ab33405 is not able to mediate enzymatic acetylation of amikacin, the total soluble protein extracts of all three strains were used in in vitro acetylation assays using amikacin and AcetylCoA as substrates. Table 1 shows that while extracts from strains A144 and A155 mediated incorporation of radioactive acetyl groups to the acceptor substrate, the extract obtained from strain Ab33405 lacked acetylation activity.

The growth of all three A. baumannii strains was unaffected by the presence of 25 or 50 μM ZnCl2 or up to 10 μM 8HQ, CI8HQ, 5-[N-Methyl-N-propargylaminomethyl]-8-hydroxyquinoline (MP8HQ), or 5,7-diiodo-8-hydroxyquinoline (II8HQ) (S1A–S1C Fig). Conversely, 10 μM 7-Bromo-8-hydroxyquinoline (B8HQ) was toxic to all three strains, and while strains A155 and Ab33405 could grow in the presence of up to 5 μM, strain A144 growth was inhibited at 1 μM B8HQ (S1A–S1C Fig).

Once concentrations of the ionophores and ZnCl2 that were not toxic to growing bacteria were identified, their activity as potentiators of amikacin was determined. These assays showed that CI8HQ and II8HQ were the only 8HQ derivatives that mediated phenotypic conversion to susceptibility to amikacin in strains A144 and A155 (Fig 2). Inspection of these results also showed that after 16 h, strain A155 started to grow when the ionophore tested was II8HQ. We do not yet have a satisfactory explanation for this observation. The ionophores 8HQ and MP8HQ were unable to induce any modification in the growth of strains A144 and A155 in the presence of amikacin and ZnCl2 (Fig 2). The tests where the ionophore used was B8HQ showed a reduction in growth in the presence of combinations that included B8HQ but either amikacin or ZnCl2 were omitted. These results suggested that the toxic effect of B8HQ is playing a role in growth inhibition rather than interference with acetylation of amikacin (Fig 2). Strain Ab33405 showed healthy growth in the presence of either of the ionophores plus amikacin and ZnCl2 confirming that the inhibition by Zn2+ is specific for resistance mediated by the modifying enzyme. Only one condition showed modest inhibition of growth (see Fig 2, strain Ab33405, CI8HQ) but some reduction in growth is also observed in the absence of ZnCl2, which may indicate unspecific inhibition. These results taken together with previous studies, especially those by Li et al. [26], where the authors show than Zn2+ inhibits several modifying enzymes, indicate that ionophores complexed to metal ions can be an excellent strategy to interfere with resistance to aminoglycosides. However, this option might be effective only in cases of resistance mediated by selected aminoglycoside modifying enzymes. Interestingly, a recent report described that the metal homeostasis-disrupting action of ionophore-zinc complexes potentiates several antibiotics to restore susceptibility in resistant Gram-positive bacteria [45].

thumbnail
Fig 2. Effect of ionophore-zinc complexes on resistance to amikacin in A. baumannii strains.

A. baumannii A155 (panels to the left), A144 (center panels) or Ab33405 (panels to the right) were cultured in 100 μl Mueller-Hinton broth in microtiter plates at 37°C, with the additions indicated in the figure and the OD600 was periodically determined. The concentrations used were 8 μg/ml amikacin, 25 μM ZnCl2, 5 μM ionophore. A, amikacin; Z, ZnCl2.

https://doi.org/10.1371/journal.pone.0217602.g002

The results described above showed that CI8HQ and II8HQ were the most efficient ionophores that in complex with Zn2+ were able to mediate a conversion to susceptibility to amikacin in those A. baumannii strains in which resistance is mediated by AAC(6')-Ib. The bactericidal effect of the combination ionophore-zinc and amikacin was confirmed using time-kill assays. Amikacin at a concentration as low as 8 μg/ml showed a robust bactericidal activity on A. baumannii A144 and A155 strains in the presence of the complexes (Fig 3). As expected, these strains did not lose viability when incubated with the antibiotic or any other combination of components that did not include all three of them (Fig 3). Also expected was the absence of bactericidal effect when the combinations ionophore-zinc plus amikacin were added to cultures of A. baumannii Ab33405 or the ionophore utilized was 8HQ (Fig 3). These results confirmed that amikacin can regain its bactericidal power in the presence of Zn2+ ions when resistance is due to AAC(6')-Ib-mediated acetylation.

thumbnail
Fig 3. Time-kill assay curves for amikacin in the presence of ionophore-zinc complexes.

A. baumannii A155 (panels to the left), A144 (center panels) or Ab33405 (panels to the right) were cultured in 100 μl Mueller-Hinton broth in microtiter plates at 37°C, with the additions indicated in the figure and the OD600 was periodically determined. A, amikacin; Z, ZnCl2; I, ionophore.

https://doi.org/10.1371/journal.pone.0217602.g003

The ionophores tested in this work were subjected to a standard cytotoxicity assay using HEK293 cells as described in the Materials and Methods section. Addition of 8HQ, CI8HQ, or II8HQ, alone (S2 Fig) or in combination with amikacin and Zn2Cl to the cells did not result in significant toxicity (Fig 4).

thumbnail
Fig 4. Cytotoxicity tests.

Cytotoxicity on HEK293 cells treated with the indicated concentrations of the different compounds for 24 h was assayed using a LIVE/DEAD kit. The percentage of dead cells was calculated relative to the cells treated with DMSO. Cells incubated with 0.1% Triton X-100 for 10 min were used as a control for maximum toxicity. Experiments were conducted in triplicate and the values are mean ± SD. Black bars show survival in the presence of 5 μM ionophore. Stippled bars show survival in the presence of 5 μM ionophore, 25 μM ZnCl2, and 8 μg/ml amikacin. The same concentrations were used to determine survival in amikacin (white bar) and ZnCl2 (hatched bar). The concentration of DMSO used in the control was μM (gray bar).

https://doi.org/10.1371/journal.pone.0217602.g004

Discussion

Numerous approaches are being pursued to combat the current crisis of antibiotic resistance [10, 12]. In addition to the efforts to find or design new classes of antibiotics, researchers are looking for new scaffolds or attempting to modify existing antimicrobial families or designing compounds that act as adjuvant of these antibiotics by interfering with resistance [12, 4650]. We have previously found that Zn2+, when complexed to ionophores such as pyrithione or CI8HQ, significantly reduces the levels of resistance to amikacin mediated by the AAC(6')-Ib enzyme [2325]. Since this enzyme is the most prevalent in amikacin resistant infections in the clinics [5], this finding represented a significant advance in the search for compounds that in combination with the antibiotic can help extend its useful life. The obvious potential these compounds have to be part of formulations composed of amikacin and the inhibitor warrant further research to find the best ionophores. Since CI8HQ is a derivative of 8HQ, in this work we tested combinations of Zn2+ with 8HQ and other commercially available derivatives. While CI8HQ and II8HQ show similar capacity to reverse resistance to amikacin, 8HQ and MP8HQ did not show any of the desired inhibitory activity, and B8HQ exhibited antimicrobial activity in the absence of the antibiotic. The disparity of effects found among these chemically related compounds shows the importance of assessing the activity of ionophores with similar structures. Since one of the most crucial problems exhibited by numerous compounds that are otherwise good drug or adjuvant candidates is their toxicity, it was interesting that the ionophores tested in this work did not show cytotoxicity in our assays. Furthermore, as substituted 8HQ derivatives are being researched as treatments of other human conditions, their low toxicity has also been established by other laboratories [29, 31]. Taken together, the results described in this work indicate that Zn2+ or other cations complexed to ionophores are firm candidates to be developed as potentiators to aminoglycosides to overcome resistance. In particular, CI8HQ and II8HQ are excellent candidates as adjuvants to overcome AAC(6')-Ib -mediated resistance to amikacin.

Supporting information

S1 Fig. Effect of addition of different reagents on growth of A. baumannii strains.

https://doi.org/10.1371/journal.pone.0217602.s001

(PDF)

References

  1. 1. Ramirez MS, Tolmasky ME. Aminoglycoside modifying enzymes. Drug Resist Updat. 2010;13: 151–71. pmid:20833577
  2. 2. Labby KJ, Garneau-Tsodikova S. Strategies to overcome the action of aminoglycoside-modifying enzymes for treating resistant bacterial infections. Future Med Chem. 2013;5: 1285–309. pmid:23859208
  3. 3. Garneau-Tsodikova S, Labby KJ. Mechanisms of resistance to aminoglycoside antibiotics: overview and perspectives. MedChemComm. 2016;7: 11–27. pmid:26877861
  4. 4. Ramirez MS, Nikolaidis N, Tolmasky ME. Rise and dissemination of aminoglycoside resistance: the aac(6')-Ib paradigm. Front Microbiol. 2013;4: 121. pmid:23730301
  5. 5. Vakulenko SB, Mobashery S. Versatility of aminoglycosides and prospects for their future. Clin Microbiol Rev. 2003;16: 430–450. pmid:12857776
  6. 6. Ramirez MS, Tolmasky ME. Amikacin: uses, resistance, and prospects for inhibition. Molecules. 2017;22: 2267. pmid:29257114
  7. 7. Castanheira M, Deshpande LM, Woosley LN, Serio AW, Krause KM, Flamm RK. Activity of plazomicin compared with other aminoglycosides against isolates from European and adjacent countries, including Enterobacteriaceae molecularly characterized for aminoglycoside-modifying enzymes and other resistance mechanisms. J Antimicrob Chemother. 2018;73: 3346–3354. pmid:30219857
  8. 8. Ju LC, Cheng Z, Fast W, Bonomo RA, Crowder MW. The continuing challenge of metallo-beta-lactamase inhibition: mechanism matters. Trends Pharmacol Sci. 2018;39: 635–647. pmid:29680579
  9. 9. Vong K, Auclair K. Understanding and overcoming aminoglycoside resistance caused by N-6'-acetyltransferase. MedChemComm. 2012;3: 397–407. pmid:28018574
  10. 10. Tolmasky ME. Strategies to prolong the useful life of existing antibiotics and help overcoming the antibiotic resistance crisis. In: Rahman A-u, editor. Frontiers in Clinical Drug Research—Anti-Infectives. 4: Bentham Science Publishers; 2017. p. 3–29.
  11. 11. Chandrika NT, Garneau-Tsodikova S. Comprehensive review of chemical strategies for the preparation of new aminoglycosides and their biological activities. Chem Soc Rev. 2018;47: 1189–1249. pmid:29296992
  12. 12. Chandrika N, Garneau-Tsodikova S. A review of patents (2011–2015) towards combating resistance to and toxicity of aminoglycosides. MedChemComm. 2015;7: 50–68. pmid:27019689
  13. 13. Chandrika N, Green KD, Houghton JL, Garneau-Tsodikova S. Synthesis and Biological Activity of Mono- and Di-N-acylated Aminoglycosides. ACS Med Chem Lett. 2015;6: 1134–1139. pmid:26617967
  14. 14. Matsushita T, Sati GC, Kondasinghe N, Pirrone MG, Kato T, Waduge P, et al. Design, multigram synthesis, and in vitro and in vivo evaluation of propylamycin: a semisynthetic 4,5-deoxystreptamine class aminoglycoside for the treatment of drug-resistant Enterobacteriaceae and other Gram-negative pathogens. J Am Chem Soc. 2019;141: 5051–5061. pmid:30793894
  15. 15. Jackson A, Jani S, Sala CD, Soler-Bistue AJ, Zorreguieta A, Tolmasky ME. Assessment of configurations and chemistries of bridged nucleic acids-containing oligomers as external guide sequences: a methodology for inhibition of expression of antibiotic resistance genes. Biol Methods Protoc. 2016;1: bpw001. pmid:27857983
  16. 16. Lopez C, Arivett BA, Actis LA, Tolmasky ME. Inhibition of AAC(6')-Ib-mediated resistance to amikacin in Acinetobacter baumannii by an antisense peptide-conjugated 2',4'-bridged nucleic acid-NC-DNA hybrid oligomer. Antimicrob Agents Chemother. 2015;59: 5798–5803. pmid:26169414
  17. 17. Sarno R, Ha H, Weinsetel N, Tolmasky ME. Inhibition of aminoglycoside 6'-N-acetyltransferase type Ib-mediated amikacin resistance by antisense oligodeoxynucleotides. Antimicrob Agents Chemother. 2003;47: 3296–3304. pmid:14506044
  18. 18. Soler Bistue AJ, Martin FA, Vozza N, Ha H, Joaquin JC, Zorreguieta A, et al. Inhibition of aac(6')-Ib-mediated amikacin resistance by nuclease-resistant external guide sequences in bacteria. Proc Natl Acad Sci USA. 2009;106: 13230–13235. pmid:19666539
  19. 19. Guan J, Vong K, Wee K, Fakhoury J, Dullaghan E, Auclair K. Cellular studies of an aminoglycoside potentiator reveal a new inhibitor of aminoglycoside resistance. Chembiochem. 2018;19: 2107–2113. pmid:30059603
  20. 20. Wright GD. Mechanisms of resistance to antibiotics. Curr Opin Chem Biol. 2003;7: 563–569. pmid:14580559
  21. 21. Zarate SG, De la Cruz Claure ML, Benito-Arenas R, Revuelta J, Santana AG, Bastida A. Overcoming aminoglycoside enzymatic resistance: design of novel antibiotics and inhibitors. Molecules. 2018;23: 284. Epub 2018/02/02. pmid:29385736
  22. 22. Chiem K, Jani S, Fuentes B, Lin DL, Rasche M, Tolmasky ME. Identification of an inhibitor of the aminoglycoside 6'-N-acetyltransferase type Ib [AAC(6')-Ib] by glide molecular docking. MedChemComm. 2016;7: 184–189. pmid:26973774
  23. 23. Lin DL, Tran T, Alam JY, Herron SR, Ramirez MS, Tolmasky ME. Inhibition of aminoglycoside 6'-N-acetyltransferase type Ib by zinc: reversal of amikacin resistance in Acinetobacter baumannii and Escherichia coli by a zinc ionophore. Antimicrob Agents Chemother. 2014;58: 4238–4241. pmid:24820083
  24. 24. Chiem K, Fuentes BA, Lin DL, Tran T, Jackson A, Ramirez MS, et al. Inhibition of aminoglycoside 6'-N-acetyltransferase type Ib-mediated amikacin resistance in Klebsiella pneumoniae by zinc and copper pyrithione. Antimicrob Agents Chemother. 2015;59: 5851–5853. pmid:26169410
  25. 25. Chiem K, Hue F, Magallon J, Tolmasky ME. Inhibition of aminoglycoside 6'-N-acetyltransferase type Ib-mediated amikacin resistance by zinc complexed to clioquinol, an ionophore active against tumors and neurodegenerative diseases. Int J Antimicrob Agents. 2017;51: 271–273. pmid:28782708
  26. 26. Li Y, Green KD, Johnson BR, Garneau-Tsodikova S. Inhibition of aminoglycoside acetyltransferase resistance enzymes by metal salts. Antimicrob Agents Chemother. 2015;59: 4148–4156. pmid:25941215
  27. 27. Lind SE, Park JS, Drexler JW. Pyrithione and 8-hydroxyquinolines transport lead across erythrocyte membranes. Transl Res. 2009;154: 153–159. pmid:19665691
  28. 28. Chan SH, Chui CH, Chan SW, Kok SH, Chan D, Tsoi MY, et al. Synthesis of 8-hydroxyquinoline derivatives as novel antitumor agents. ACS Med Chem Lett. 2013;4: 170–174. pmid:24900641
  29. 29. Bareggi SR, Cornelli U. Clioquinol: review of its mechanisms of action and clinical uses in neurodegenerative disorders. CNS Neurosci Ther. 2012;18: 41–46. pmid:21199452
  30. 30. Mao X, Li X, Sprangers R, Wang X, Venugopal A, Wood T, et al. Clioquinol inhibits the proteasome and displays preclinical activity in leukemia and myeloma. Leukemia. 2009;23: 585–590. pmid:18754030
  31. 31. Mao X, Schimmer AD. The toxicology of clioquinol. Toxicol Lett. 2008;182: 1–6. pmid:18812216
  32. 32. Arivett BA, Fiester SE, Ream DC, Centron D, Ramirez MS, Tolmasky ME, et al. Draft genome of the multidrug-resistant Acinetobacter baumannii strain A155 clinical isolate. Genome Announc. 2015;3. pmid:25814610
  33. 33. Vilacoba E, Deraspe M, Traglia GM, Roy PH, Ramirez MS, Centron D. Draft genome sequence of an international clonal lineage 1 Acinetobacter baumannii strain from Argentina. Genome Announc. 2014;2. pmid:25428965
  34. 34. Traglia G, Chiem K, Quinn B, Fernandez JS, Montana S, Almuzara M, et al. Genome sequence analysis of an extensively drug-resistant Acinetobacter baumannii indigo-pigmented strain depicts evidence of increase genome plasticity. Sci Rep. 2018;8: 16961. pmid:30446709
  35. 35. Traglia GM, Chua K, Centron D, Tolmasky ME, Ramirez MS. Whole-genome sequence analysis of the naturally competent Acinetobacter baumannii clinical isolate A118. Genome Biol Evol. 2014;6: 2235–2239. pmid:25164683
  36. 36. Vilacoba E, Almuzara M, Gulone L, Traglia GM, Figueroa SA, Sly G, et al. Emergence and spread of plasmid-borne tet(B)::ISCR2 in minocycline-resistant Acinetobacter baumannii isolates. Antimicrob Agents Chemother. 2013;57: 651–654. pmid:23147737
  37. 37. Smith MG, Gianoulis TA, Pukatzki S, Mekalanos JJ, Ornston LN, Gerstein M, et al. New insights into Acinetobacter baumannii pathogenesis revealed by high-density pyrosequencing and transposon mutagenesis. Genes Dev. 2007;21: 601–614. pmid:17344419
  38. 38. Haas MJ, Dowding JE. Aminoglycoside-modifying enzymes. Methods Enzymol. 1975;43: 611–628. pmid:166284
  39. 39. Woloj M, Tolmasky ME, Roberts MC, Crosa JH. Plasmid-encoded amikacin resistance in multiresistant strains of Klebsiella pneumoniae isolated from neonates with meningitis. Antimicrob Agents Chemother. 1986;29: 315–319. pmid:3521478
  40. 40. Petersen PJ, Labthavikul P, Jones CH, Bradford PA. In vitro antibacterial activities of tigecycline in combination with other antimicrobial agents determined by chequerboard and time-kill kinetic analysis. J Antimicrob Chemother. 2006;57: 573–576. pmid:16431863
  41. 41. Tran T, Chiem K, Jani S, Arivett BA, Lin DL, Lad R, et al. Identification of a small molecule inhibitor of the aminoglycoside 6'-N-acetyltransferase type Ib [AAC(6')-Ib] using mixture-based combinatorial libraries. Int J Antimicrob Agents. 2018;51: 752–761. pmid:29410367
  42. 42. Papp-Wallace KM, Bonomo RA. New beta-lactamase inhibitors in the clinic. Infect Dis Clin North Am. 2016;30: 441–464. pmid:27208767
  43. 43. Ramirez MS, Vilacoba E, Stietz MS, Merkier AK, Jeric P, Limansky AS, et al. Spreading of AbaR-type genomic islands in multidrug resistance Acinetobacter baumannii strains belonging to different clonal complexes. Curr Microbiol. 2013;67: 9–14. pmid:23397241
  44. 44. Traglia G, Vilacoba E, Almuzara M, Diana L, Iriarte A, Centron D, et al. Draft genome sequence of an extensively drug-resistant Acinetobacter baumannii indigo-pigmented strain. Genome Announc. 2014;2: e01146–14. pmid:25395633
  45. 45. Bohlmann L, De Oliveira DMP, El-Deeb IM, Brazel EB, Harbison-Price N, Ong CY, et al. Chemical Synergy between Ionophore PBT2 and Zinc Reverses Antibiotic Resistance. MBio. 2018;9: e02391–18. pmid:30538186
  46. 46. Schmidt M, Harmuth S, Barth ER, Wurm E, Fobbe R, Sickmann A, et al. Conjugation of Ciprofloxacin with Poly(2-oxazoline)s and Polyethylene Glycol via End Groups. Bioconjug Chem. 2015;26: 1950–1962. pmid:26284608
  47. 47. Tolmasky ME. Aminoglycoside-modifying enzymes: characteristics, localization, and dissemination. In: Bonomo RA, Tolmasky ME, editors. Enzyme-Mediated Resistance to Antibiotics: Mechanisms, Dissemination, and Prospects for Inhibition. Washington, DC: ASM Press; 2007. p. 35–52.
  48. 48. Wright PM, Seiple IB, Myers AG. The evolving role of chemical synthesis in antibacterial drug discovery. Angew Chem Int Ed Engl. 2014;53: 8840–8869. pmid:24990531
  49. 49. Maianti JP, Hanessian S. Structural hybridization of three aminoglycoside antibiotics yields a potent broad-spectrum bactericide that eludes bacterial resistance enzymes. Medchemcomm. 2015;7: 170–177.
  50. 50. Davies-Sala C, Soler-Bistue A, Bonomo RA, Zorreguieta A, Tolmasky ME. External guide sequence technology: a path to development of novel antimicrobial therapeutics. Ann N Y Acad Sci. 2015;1354: 98–110. pmid:25866265