<<

Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Topoisomerase Inhibitors: Fluoroquinolone Mechanisms of Action and Resistance

David C. Hooper1 and George A. Jacoby2

1Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts 02114 2Lahey Hospital and Medical Center, Burlington, Massachusetts 01805 Correspondence: [email protected]

Quinolone are widely used in clinical medicine and are the only current class of agents that directly inhibit bacterial DNA synthesis. Quinolones dually target DNA gyrase and IV binding to specific domains and conformations so as to block DNA strand passage catalysis and stabilize DNA–enzyme complexes that block the DNA repli- cation apparatus and generate double breaks in DNA that underlie their bactericidal activity. Resistance has emerged with clinical use of these agents and is common in some bacterial . Mechanisms of resistance include mutational alterations in target affinity and efflux pump expression and acquisition of resistance-conferring genes. Resistance mu- tations in one or both of the two drug target enzymes are commonly in a localized domain of the GyrA and ParC subunits of gyrase and topoisomerase IV, respectively, and reduce drug binding to the enzyme–DNA complex. Other resistance occur in regulatory genes that control the expression of native efflux pumps localized in the bacterial membrane(s). These pumps have broad substrate profiles that include other antimicrobials as well as quin- olones. Mutations of both types can accumulate with selection pressure and produce highly resistant strains. Resistance genes acquired on plasmids confer low-level resistance that promotes the selection of mutational high-level resistance. Plasmid-encoded resistance is because of Qnr proteins that protect the target enzymes from quinolone action, a mutant aminoglycoside-modifying enzyme that also modifies certain quinolones, and mobile efflux pumps. Plasmids with these mechanisms often encode additional resistances and can transfer multidrug resistance that includes quinolones. www.perspectivesinmedicine.org uinolones have been used extensively for a (Lesher et al. 1962). Medicinal chemists from Qwide range of clinical applications (Owens a number of companies, however, subsequently and Ambrose 2000; Kim and Hooper 2014). modified the core quinolone and related chem- , a related naphthyridone struc- ical scaffolds, generating compounds with ture and the first member of the class used clin- greater potency, broader spectra of activity, im- ically, was discovered by George Lesher as a proved pharmacokinetics, and lower frequency byproduct of synthesis in 1962. of development of resistance (Domagala and Its use was limited to treatment of urinary tract Hagen 2003). An important addition of a fluo- and resistance emerged quickly rine substituent at position 6 added to po-

Editors: Lynn L. Silver and Karen Bush Additional Perspectives on and Resistance available at www.perspectivesinmedicine.org Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a025320 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320

1 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

tency and became the common feature of the 2013; Bax et al. 2010; Wohlkonig et al. 2010). fluoroquinolone class with the introductions In GyrA, a noncatalytic Mg2þ ion coordinated of norfloxacin in 1986 and ciprofloxacin in with four water molecules appears as a bridge 1987 that showed substantially greater potency for hydrogen bonding between quinolone and against Gram-negative bacteria. Subsequently, Ser83 and Asp87 (Wohlkonig et al. 2010), the other fluoroquinolones, such as levofloxacin, two most commonly mutated amino acids in gatifloxacin, moxifloxacin, and gemifloxacin quinolone-resistant mutants. Although quino- were developed with increased activity against lones can bind to mycobacterial gyrase in the Gram-positive bacteria. Because of their po- absence of DNA (Kumar et al. 2014), in E. coli, tency, spectrum of activity, oral bioavailability, the gyrase–DNA complex shows increases in and generally good safety profile, fluoroquino- the amount and specificity of quinolone bind- lones were used widely for a range of clinical ing relative to gyrase alone (Shen et al. 1989; indications worldwide. Although still clinically Willmott and Maxwell 1993). valuable, fluoroquinolone use has been com- Quinolones inhibit enzyme function by promised by the emergence of bacterial resis- blocking the resealing of the DNA double- tance because of and acquisition of strand break, but, in addition, this process stabi- plasmid-encoded genes. In the sections that fol- lizes a catalytic intermediate covalent complex low, we review the mechanisms of quinolone of enzyme and DNA that serves as a barrier to action and resistance, which are summarized movement of the DNA replication fork (Went- in Table 1. zell and Maxwell 2000) or transcription com- plexes (Willmott et al. 1994) and can be con- verted to permanent double-strand DNA breaks QUINOLONE MECHANISM OF ACTION (Drlica et al. 2008), thereby functioning as Quinolones target two essential bacterial type II topoisomerase poisons (Kreuzer and Cozzarelli topoisomerase enzymes, DNA gyrase and DNA 1979). Quinolone interactions with DNA gyrase topoisomerase IV (Hooper 1997). Both en- appear to result in more rapid inhibition of zymes are heterotetramers with two subunits, DNA replication than quinolone interactions gyrase being constituted as GyrA2GyrB2 and with topoisomerase IV (Khodursky et al. 1995; topoisomerase IV as ParC2ParE2. GyrA is ho- Fournier et al. 2000), possibly relating to the mologous to ParC, and GyrB homologous to overall proximity to the DNA replication com- ParE (Wang 1996). In Staphylococcus aureus, plex of enzyme-binding sites on chromosomal topoisomerase IV subunits historically have DNA, with gyrase positioned ahead of the com- also been referred to as GrlA and GrlB. Both plex and topoisomerase IV behind it (Khodur- www.perspectivesinmedicine.org enzymes act by catalyzing a DNA double-strand sky and Cozzarelli 1998). Quinolones can differ break, passing another DNA strand through the in their potency for the two enzymes, with a break, and resealing the break (Aldred et al. general pattern among quinolones in clinical 2014). DNA strand passing occurs via domains use that there is greater activity against DNA that are localized in GyrA and ParC, whereas gyrase in Gram-negative bacteria and greater ATPase activity, which is required for enzyme activity against topoisomerase IV in Gram-pos- catalysis, occurs via domains localized in GyrB itive bacteria; but exceptions occur, and some and ParE. quinolones have similar potency against both Quinolones bind reversibly to the complex- enzymes (Blanche et al. 1996; Pan and Fisher es of DNA with gyrase and topoisomerase IV at 1997; Strahilevitz and Hooper 2005). the interface between protein and DNA near the DNA strand breaks trigger bacterial SOS active site tyrosine (Tyr122 for GyrA, Tyr120 for DNA repair responses (Piddock et al. 1990; ParC in numbering), which is Drlica et al. 2008) and, to the extent that repair transiently covalently linked to DNA during is incomplete, generate quinolone bactericidal DNA strand passage, with intercalation into activity (Hiasa et al. 1996; Drlica and Zhao the cleaved DNA (Laponogov et al. 2009, 1997; Drlica et al. 2008, 2009). Inhibition of

2 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from

www.perspectivesinmedicine.org http://perspectivesinmedicine.cshlp.org/ ieti ril as article this Cite Table 1. Mechanisms of quinolone resistance Mechanism Gene alteration Effector genes Species Other factors Altered target with Mutation in chromosomal gyrA All Primary and secondary targets vary by reduced drug structural genes gyrB drug and species with gyrA and/or odSrn abPrpc Med Perspect Harb Spring Cold binding parC (grlA) Most except mycobacteria, parC most often mutated; mutations parE (grlB) , Treponema in both targets with additive resistance Increased expression Mutation in chromosomal MFS family: norA, norB, Staphylococcus aureus Regulators include mgrA, norG, mepR, of efflux pumps genes of transcriptional norC, mdeA, lmrS, sdrM, arlRS regulators of expression qacB(III) of chromosomally MATE family: mepA

encoded pumps onOctober1,2021-PublishedbyColdSpringHarborLaboratoryPress MFS family: bmr, bmr3, blt Bacillus subtilis MFS family: lmrP Lactococcus lactis ABC family: lmrA

2016;6:a025320 MFS family: pmrA pneumoniae

ABC family: patAB Resistance and Action of Mechanisms Fluoroquinolone ABC family: satAB Streptococcus suis MFS family: lde monocytogenes Regulator fepR MATE family: fepA RND family: acrAB-tolC Escherichia coli Regulators include marR, soxRS, rob, MFS family: emrAB-tolC, emrR mdfA RND family: acrAB-tolC spp. RND family: oqxAB-tolC Regulators include oqxR, rarA RND family: acrAB-tolC aerogenes RND family: cmeABC Campylobacter jejuni MATE family: norM Vibrio parahaemolyticus RND family: mexAB-oprM, aeruginosa Regulators include mexR, nfxB, nfxC, mexCD-oprJ, mexEF- mvaT, mexZ oprN, mexXY-oprM RND family: adeIJK, baumannii Regulators include adeRS, adeL adeABC, adeFGH RND family: smeDEF Stenotrophomonas maltophilia MATE family: norA Bacteroides fragilis 3 Continued Downloaded from

www.perspectivesinmedicine.org http://perspectivesinmedicine.cshlp.org/ ..Hoe n ..Jacoby G.A. and Hooper D.C. 4

Table 1. Continued Mechanism Gene alteration Effector genes Species Other factors

MATE family: bexA Bacteroides thetaiotaomicron onOctober1,2021-PublishedbyColdSpringHarborLaboratoryPress Plasmid-acquired efflux RND family: oqxAB, qepA Multiples species of pump gene Enterobacteriaceae

ieti ril as article this Cite Target protection Chromosomal genes mfpA Mycobacterium tuberculosis and Mycobacterium smegmatis (among others) ahqnr Aeromonas hydrophila Protection of gyrase and topoisomerase efsqnr Enterococcus faecalis IV from quinolone action qnrVC Plasmid-acquired genes qnrA, qnrB, qnrS, qnrC, Multiple species of odSrn abPrpc Med Perspect Harb Spring Cold qnrD, qnrVC Enterobacteriaceae, also Acinetobacter, Aeromonas, Pseudomonas, and Vibrio spp. Drug modification Plasmid-acquired gene aac(60)-Ib-cr Multiples species of Acetylation of the secondary amine of Enterobacteriaceae the piperazinyl group of ciprofloxacin and norfloxacin MFS, Major facilitator superfamily; MATE, multiple antibiotic and toxin extrusion; ABC, ATP-binding cassette; RND, resistance–nodulation–division. 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

protein synthesis does not interfere with quin- ase-targeting class of compounds (Aldred et olone-mediated inhibition of DNA replication al. 2012). Mutations in the Ser and nearby acid- but can reduce bacterial killing, with variation ic residues differ in their effects on catalytic ef- in the magnitude of reduction seen with differ- ficiency of gyrase and topoisomerase IV, with ent quinolones (Lewin et al. 1991; Howard et al. mutation of the Ser residue generally having 1993). These phenomena suggest that events in little effect and mutation in the acidic residue addition to DNA replication inhibition that resulting in a five- to 10-fold decrease in cata- may affect DNA or other cellular damage may lytic efficiency (Hiasa 2002; Aldred et al. 2014). also contribute to quinolone bactericidal activ- Resistance mutations in GyrB and ParE are ity, but the molecular mechanisms are not yet substantially less common than those in GyrA understood. and ParC in resistant clinical isolates. In E. coli, mutations at Asp426 and Lys447 of GyrB and Leu445 of ParE, as well as mutations at similar QUINOLONE RESISTANCE BECAUSE positions in other species, can cause resistance OF MUTATIONAL ALTERATION (Yoshida et al. 1991; Ito et al. 1994; Gensberg IN TARGET ENZYMES et al. 1995; Breines et al. 1997; Weigel et al. Single amino acid changes in either gyrase or 2001). Binding of to enzyme–DNA topoisomerase IV that confer quinolone resis- complexes constituted with resistant mutant tance have been most commonly localized to GyrB is reduced (Yoshida et al. 1993). The crys- the amino-terminal domains of GyrA (residues tal structures of some conformations of yeast 67 to 106 for E. coli numbering) or ParC (resi- topoisomerase II show proximity of the regions dues 63 to 102). These domains are near the homologous to the QRDRs of GyrA and GyrB active site tyrosines (Tyr122 for GyrA, Tyr120 (Fass et al. 1999). In E. coli crystal structures, for ParC) of both enzymes (Morais Cabral et al. the basic substituents at position C7 of cipro- 1997; Laponogov et al. 2009, 2013; Wohlkonig floxacin and moxifloxacin were shown to be et al. 2010). These domains have been termed facing the GyrB subunit, and they could be the “quinolone resistance-determining region” cross-linked to GyrB Cys466 (Mustaev et al. (QRDR) of GyrA and ParC (Yoshida et al. 2014). In addition, in another crystal structure 1990). The most common site of mutation in Arg418 of Acinetobacter baumannii topoisom- GyrA of E. coli is at Ser83 followed by Asp87, erase IV is in proximity to the moxifloxacin C7 which as noted above are key residues in the basic substituent (Wohlkonig et al. 2010). No- binding of quinolones to GyrA or ParC. There tably, resistance mutations in acidic residues in is conservation of a Ser and another acidic res- this domain of GyrB in E. coli (Asp426Asn) as www.perspectivesinmedicine.org idue separated by four amino acids in both well as in ParC, suggest that drug–enzyme con- GyrA and ParC in other species, and mutation tacts could be mediated by charge interactions in these residues can be frequently selected with (Wohlkonig et al. 2010). Thus, resistance muta- quinolones in laboratory mutants and are fre- tions in the QRDRs of both GyrA/ParC and quently found in resistant clinical isolates GyrB/ParE appear to act by reducing the affin- (Hooper 2003; Drlica et al. 2009; Aldred et al. ity of quinolones for the enzyme–DNA com- 2014). Ser83Trp and Ser83Leu mutations of plex. Although direct drug binding data are not E. coli GyrA have been associated with reduced yet available for mutant topoisomerase IV– binding of the quinolone norfloxacin and enox- DNA complexes, the similarity of structures acin to gyrase–DNA complexes (Willmott and between gyrase and topoisomerase IV and the Maxwell 1993; Yoshida et al. 1993; Willmott conservation of key residues predict that resis- et al. 1994). A Ser81Phe resistance mutation in tance is similarly mediated by reduced affinity ParC of appears also to show for both enzyme–DNA complexes. decreased quinolone binding to the enzyme– The magnitude of resistance caused by a DNA complex based on competition experi- single-target mutation in one of the subunits ments with quinazolinediones, another gyr- of gyrase or topoisomerase IV varies by quino-

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 5 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

lone and bacterial species (Pan and Fisher 1997; both enzymes and is the only quinolone target Fournier et al. 2000). Because quinolone in- (Hooper 2003). Thus, selection of mutations teraction with either target enzyme–DNA com- with substantial resistance phenotypes is pre- plex is sufficient to block cell growth and trigger dicted to occur readily in these pathogens, cell death (Drlica and Zhao 1997), the level of a prediction consistent with the frequent occur- susceptibility of a wild-type bacterium is deter- rence of resistance with clinical use of quino- mined by the more sensitive of the two tar- lones without use of other active agents to treat get enzymes, as noted above, often gyrase in infections with M. tuberculosis and H. pylori Gram-negative bacteria and topoisomerase IV (Tsukamura et al. 1985; Me´graud 1998). in Gram-positive bacteria (Blanche et al. 1996; Pan and Fisher 1997). Thus, under quinolone selection pressure, resistance mutation in the QUINOLONE RESISTANCE BECAUSE OF DECREASED DRUG ACCESS TO more sensitive or primary target enzyme will TARGET ENZYMES generally occur first because mutation in the secondary less sensitive target enzyme alone Quinolones must cross the bacterial envelope does not have a resistance phenotype because to interact with their cytoplasmic gyrase and of the dominance of the quinolone–primary topoisomerase IV targets. Active quinolone target interaction (Trucksis et al. 1991; Ng et efflux, reductions in influx, or both can de- al. 1996; Breines et al. 1997). The magnitude crease cytoplasmic quinolone concentrations of the increase in resistance from such a first- and confer resistance. In Gram-positive bacte- step mutation in the primary target is then de- ria, reduced diffusion across the cytoplasmic termined by either the magnitude of the effect membrane has not been found to cause resis- of the mutation on enzyme sensitivity or the tance, but active efflux transporters that include intrinsic level of sensitivity of the secondary tar- quinolones in their substrate profiles have been get enzyme. Thus, the level of sensitivity of the shown to cause low-level resistance. In contrast, secondary target enzyme can set a cap on the in Gram-negative bacteria, reduced diffusion magnitude of resistance conferred by mutation through outer membrane porin diffusion chan- in the primary target enzyme. The dominance nels can contribute to resistance. Reduced in- of sensitivity over resistance in the two target flux often acts in concert with basal or increased enzymes also implies that quinolones with sim- expression of efflux transporters with both con- ilar potency against both gyrase and topoisom- tributing additively to resistance (Lomovskaya erase IV in an organism may require mutations et al. 1999; Li and Nikaido 2009). Quinolones in both enzymes before the mutant bacterium themselves generally do not induce expression www.perspectivesinmedicine.org shows a substantial resistance phenotype (Pan of efflux pumps. With the exception of plasmid- and Fisher 1998, 1999; Strahilevitz and Hooper mediated quinolone resistance discussed later, 2005). Fluoroquinolones currently in clinical acquired quinolone resistance by altered drug use generally have differences in potency be- permeation occurs largely by mutations in tween the two target enzymes, and single target genes encoding regulatory proteins that control mutations produce eight- to 16-fold increases the transcription of efflux pump or porin genes in resistance. (Grkovic et al. 2002). Uncommonly, mutations Accumulating mutations in both target en- in efflux pump structural genes have caused zymes have been shown to cause increasing changes in pump substrate profiles that add quinolone resistance. In many species, high-lev- quinolones (Blairet al. 2015). The levels of quin- el quinolone resistance is generally associated olone resistance because of regulatory mutation with mutations in both gyrase and topoisomer- and pump overexpression are often limited to ase IV (Schmitz et al. 1998). In several species, about four- to eightfold increases in inhibitory Mycobacterium tuberculosis, Helicobacter pylori, concentrations, likely because of counterbal- and Treponema pallidum, there is no topoisom- ancing regulatory factors and cellular toxicities erase IV, and gyrase provides the functions of of high levels of pump overexpression.

6 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

Altered Permeation in Gram-Positive Bacteria tive levels of expression of NorA, NorB, and NorC are modified in response to a variety of In Gram-positive bacteria, the major facilitator environmental conditions. Notably, norB ex- superfamily (MFS) of transporters contains pression is selectively increased in an abscess the largest number of efflux transporters that environment in response to low-free iron con- include quinolones in their substrate profiles. ditions and contributes to fitness and bacterial These efflux pumps are transporters energized survival in abscesses (Ding et al. 2008), a com- by the proton gradient acrossthe bacterial mem- mon form of S. aureus . The natural brane and are generally antiporters with ex- substrate of NorB, transport of which may con- change of substrate and protons in opposite di- tribute to improving fitness in an abscess envi- rections. The Nor MFS pumps of S. aureus have ronment, is not known. In addition, physiologic been most extensively studied (Li and Nikaido increased expression of NorB at the site of in- 2009; Schindler et al. 2015). NorA (Ubukata et al. fection would suggest that susceptibility testing 1989; Yuet al. 2002), NorB (Truong-Bolduc et al. under clinical laboratory conditions may not 2005), and NorC (Truong-Bolduc et al. 2006) fully reflect susceptibility at the site of infection. efflux pumps cause four- to eight-fold increases NorG, a member of the GntR-like transcrip- in resistance to quinolones when overexpressed. tional regulators, can also modulate pump ex- NorA confers resistance to hydrophilic quino- pression and levels of quinolone resistance; lones, such as norfloxacin and ciprofloxacin, it is a direct activator of norA and norB expres- whereas NorB and NorC each confer resistance sion but a direct repressor of norC expression to both hydrophilic quinolones and hydropho- (Truong-Bolduc and Hooper 2007; Truong- bic quinolones, such as sparfloxacin and moxi- Bolduc et al. 2011b). ArlRS, a two-component floxacin (Yu et al. 2002; Truong-Bolduc et al. regulatory system, has been shown to affect ex- 2005, 2006); these pumps also have structurally pression of norA as well (Fournier and Hooper unrelated substrates in addition to quinolones, 2000; Fournier et al. 2001). There are often hi- in keeping with broad substrate profiles of many erarchies in regulatory networks, and other reg- MFS transporters. Although there are natural ulators can affect expression of MgrA and quinolone-like compounds (Heeb et al. 2011), NorG. Such complex regulatory networks af- it is unlikely that synthetic antibacterial quino- fecting pump expression imply the importance lones are themselves the natural pump sub- of modulation of pump functions in cellular strates, which are as yet unknown for Gram-pos- physiology and may contribute to different bac- itive quinolone resistance pumps. terial responses to quinolones in different envi- Regulation of expression of these transport- ronments that affect pump expression. www.perspectivesinmedicine.org ers is complex and involves several transcrip- Other MFS efflux transporters that can con- tional regulators. MgrA, the most studied, acts tribute to quinolone resistance in S. aureus in- as a positive regulator of norA expression and clude MdeA (norfloxacin and ciprofloxacin) a negative regulator of norB and norC expres- (Huang et al. 2004), SdrM (norfloxacin) (Ya- sion (Ingavale et al. 2005; Truong-Bolduc et al. mada et al. 2006), QacB(III) (norfloxacin and 2005). Posttranslational phosphorylation of ciprofloxacin) (Nakaminami et al. 2010), and MgrA by the PknB kinase results in the loss of LmrS (gatifloxacin) (Floyd et al. 2010). MFS the ability of MgrA dimers to bind the norA transporters in other Gram-positive bacteria promoter and an increase in their binding have also been shown to include quinolones to the norB promoter (Truong-Bolduc et al. in their substrate profiles. These transporters 2008; Truong-Bolduc and Hooper 2010). Acidic include Bmr, Bmr3, and Blt of B. subtilis conditions alter the proportions of phosphory- (Klyachko et al. 1997; Ohki and Murata 1997); lated and unphosphorylated MgrA, and oxida- PmrA of (Gill et al. tive and aeration conditions also affect dimeri- 1999); LmrP of Lactococcus lactis (Bolhuis zation and promoter binding (Chen et al. 2006; et al. 1995), and Lde of Listeria monocytogenes Truong-Bolduc et al. 2011a, 2012). Thus, rela- (Godreuil et al. 2003).

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 7 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

In addition to the MFS transporters, less- export of substrates across both inner and outer er numbers of efflux pumps of the multiple membranes (Li and Nikaido 2009). The best- antibiotic and toxin extrusion (MATE) and studied systems have been in E. coli and Pseudo- ATP-binding cassette (ABC) families have monas aeruginosa. been shown to confer quinolone resistance in In E. coli, the AcrAB-TolC pump complex Gram-positive bacteria. MATE family pumps, has been extensively studied. Crystal structures like those of the MFS, are secondary transport- of the complex have revealed a trimer of AcrB ers energized by the membrane electrochemical pump monomers that rotate around a central gradient. MepA confers resistance to norfloxa- axis perpendicular to the membrane, with each cin, ciprofloxacin, moxifloxacin, and sparflox- monomer as its rotation position changes as- acin, as well as other antimicrobials and dyes suming a different conformation mediating dif- (Kaatz et al. 2006). MepA is negatively regulated ferent steps in substrate binding and extrusion by MepR, and pentamidine, a MepA substrate, through the channel (Nikaido and Takatsuka reduces MepR binding to the mepA promoter, 2009). Substrates enter the vestibule of AcrB thereby increasing mepA expression (Kumara- from the periplasmic space between the inner swami et al. 2009; Schindler et al. 2013). In and outer membranes or the outer leaflet of the L. monocytogenes, the FepA MATE family inner membrane. Binding sites for ciprofloxacin pump is overexpressed in quinolone-resistant and other substrates of diverse chemical types strains and is regulated by the FepR transcrip- have been identified in the central cavity of tional regulator, a member of the TetR family. the periplasmic domain of AcrB (Yu et al. Mutation in FepR causes FepA overexpression 2003, 2005; Li and Nikaido 2004). Fluoroquin- and resistance to norfloxacin and ciprofloxacin olones, which are zwitterionic, are presumed to (Guerin et al. 2014). cross the outer membrane through OmpF and Members of the ABC family of transporters OmpC porin diffusion channels, down-regula- are, in contrast to the other pump families dis- tion or mutation of which may amplify resis- cussed, energized by ATP hydrolysis. PatAB of tance. Mutations in the MarR regulator can re- S. pneumoniae (norfloxacin and ciprofloxacin) sult in both an increase in acrB expression as (Boncoeur et al. 2012), SatAB of S. suis (nor- well as a decrease in ompF expression, dually floxacin and ciprofloxacin) (Escudero et al. contributing to quinolone resistance (Alekshun 2011), and LmrA of L. lactis (ciprofloxacin and Levy 1999). Mutations in the E. coli SoxRS and ofloxacin) (Poelarends et al. 2000; Putman (Miller et al. 1994; Chou et al. 1998) and Rob et al. 2000) all have been shown to confer resis- (Jair et al. 1996) regulons can also effect resis- tance to some quinolones. tance to fluoroquinolones in part related to www.perspectivesinmedicine.org reductions in OmpF and in a manner that is dependent on AcrAB-TolC. Expression of ALTERED PERMEATION IN GRAM-NEGATIVE AcrAB-TolC also confers resistance to bile salts BACTERIA and is induced by bile salts, likely one of its In Gram-negative bacteria, the majority of ef- natural substrates (Rosenberg et al. 2003). flux pumps that can effect quinolone resistance Thus, AcrAB supports the ability of E. coli to are members of the resistance–nodulation– survive in its natural habitat, the lower gastro- division (RND) superfamily (Li et al. 2015). intestinal tract, and perhaps only incidentally The RND pumps are secondary antiporters affects quinolone susceptibility. composed of a pump protein localized in the In P. aeruginosa, the OprF porin channel cytoplasmic membrane, an outer membrane has permeability a 100-fold lower than that of channel protein, and a membrane fusion pro- OmpF in E. coli (Nikaido et al. 1991), contrib- tein that links the pump and the outer mem- uting to its intrinsic resistance to quinolones brane protein (Du et al. 2014). Some outer and other antimicrobial agents relative to membrane components may link to more E. coli and other enteric bacteria. The MexAB- than one pump–fusion protein pair, enabling OprM efflux pump, a RND pump similar in

8 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

structure to AcrAB-TolC and expressed in wild- charge at position 7 (e.g., nalidixic acid) often type strains, acts in concert with the low per- select mexR and nfxC-type mutants, differences meability of OprF to augment the intrinsic level presumably reflecting differences in the resis- of resistance of P. aeruginosa to fluoroquino- tance profiles of the regulated pumps. lones (Li et al. 2000b). Mutations in mexA Additional RND pumps that cause quino- and oprM cause increased uptake of norfloxacin lone resistance have been found in a broad range and increased susceptibility to fluoroquino- of Gram-negative bacteria. Salmonella spp. lones (Poole et al. 1996b). Increased expression (Baucheron et al. 2002) and Enterobacter aero- of MexAB-OprM because of mutations in the genes (Pradel and Page`s 2002) have AcrAB MexR negative regulator causes increased re- homologs, and their increased expression has sistance to ciprofloxacin and nalidixic acid, been associated with quinolone resistance. The and mexR mutants can be selected with expo- CmeABC RND pump of Campylobacter jejuni sure to fluoroquinolones (Poole et al. 1993). contributes to the resistance of mutants selected P. aeruginosa also has three other efflux pump with enrofloxacin, a veterinary quinolone sim- systems that include quinolones in their sub- ilar to ciprofloxacin (Lin et al. 2002; Luo et al. strate profiles, MexCD-OprJ, MexEF-OprN, 2003). In Klebsiella pneumoniae, the OqxAB- and MexXY-OprM (Masuda et al. 2000). These TolC pump is encoded on the chromosome pumps vary in expression levels in wild-type (Kim et al. 2009b) and was originally identified strains (Li et al. 2000a), but resistant mutants in E. coli isolates from pigs as a cause of plasmid- overexpressing these pumps can be selected mediated resistance to olaquindox, a growth with fluoroquinolones and other antimicrobial promotant used in swine production; it also substrates (Ko¨hler et al. 1997b). Mutation in the confers resistance to quinolones. NfxB repressor, which is encoded upstream of Among nonenteric bacteria, in A. bauman- the mexCD-oprJ operon, causes increased ex- nii, the AdeIJK RND pump (Fernando et al. pression of MexCD-OprJ and resistance to 2014) is constitutively expressed and its broad fluoroquinolones (Poole et al. 1996a). Mutation resistance profile includes fluoroquinolones. In in nfxC results in overexpression of MexEF-OprN, addition, overexpression of the AdeABC and but the exact regulatory mechanism is not yet AdeFGH RND pumps because of mutation in known (Ko¨hler et al. 1997a). Mutations in the their respective regulators, AdeRS, a two-com- global regulator MvaT, which affects quorum ponent sensor-regulator system, and AdeL, a sensing and virulence, also causes increased ex- LysR family transcriptional regulator, also con- pression of mexEF-oprM and resistance to nor- fer a similarly broad resistance profile contain- floxacin (Westfall et al. 2006). Expression of ing fluoroquinolones (Yoon et al. 2013, 2015). www.perspectivesinmedicine.org both MexEF-OprN and MexCD-OprJ vary in- In Stenotrophomonas maltophilia, the SmeDEF versely with the level of expression of MexAB- pump (Alonso et al. 2000; Zhang et al. 2001) OprM, but the mechanisms underlying this has been shown to contribute to quinolone property have not yet been elucidated (Li et al. resistance based on pump knockout mutants 2000a). Mutations in the MexZ repressor cause with increased susceptibility, resistant isolates increased expression of MexXY-OprM and re- with increased pump gene expression, and its sistance to fluoroquinolones, aminoglycosides, ability to confer resistance when overexpressed and other pump substrates (Matsuo et al. 2004; in E. coli. Hay et al. 2013). Specific quinolones differ in Non-RND efflux pumps are much less com- the mutations they most commonly select mon in Gram-negative bacteria. A few cases (Ko¨hler et al. 1997b). Most quinolones in clin- MFS and MATE pumps associated with quino- ical use have a fluorine at position 6 and a pos- lone resistance have been identified. Among itively charged substituent at position 7 (e.g., MFS pumps, in E. coli, EmrAB-TolC, an MFS norfloxacin, ciprofloxacin, levofloxacin, and pump that functions in tripartite structure like moxifloxacin) and tend to select nfxB-type mu- the RND pumps, is negatively regulated by tants. In contrast, quinolones lacking a positive EmrR and confers resistance to nalidixic acid

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 9 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

but not fluoroquinolones (Lomovskaya et al. by chromosomal mutations (Courvalin 1990). 1995). MdfA, originally termed CmlA, confers The first PMQR was discovered in a multiresis- resistance to both and fluoro- tant urinary isolate of K. pneumoniae from Ala- quinolones (Yang et al. 2003). MATE pumps bama that could transfer low-level ciprofloxacin include the NorM pump, which can confer resistance to a variety of Gram-negative bacte- quinolone resistance in Vibrio parahaemolyticus ria. When the responsible gene, named qnr and (Morita et al. 2000), and in anaerobic Gram- later qnrA, was cloned and sequenced facilitat- negative bacteria the NorA pump of Bacteroides ing its identification by PCR (Tran and Jacoby fragilis (Miyamae et al. 1998) and the BexA 2002), qnr was soon found at low-frequency on pump of B. thetaiotaomicron (Miyamae et al. plasmids in Gram-negative isolates around the 2001), which have been shown to efflux fluoro- world. One qnrA plasmid from Shanghai con- quinolones. ferred an unusually high-level of resistance and There are additional examples in both further study disclosed that it carried an addi- Gram-positive and Gram-negative bacteria in tional mechanism for PMQR, namely, modifi- which there is evidence of efflux in quinolone- cation of certain quinolones by a variant of the resistant isolates determined by either reduction common aminoglycoside-modifying acetyl- in resistance with addition of a broad efflux transferase AAC(60)-Ib (Robicsek et al. 2006). pump inhibitor or reduced quinolone accumu- A third mechanism for PMQR was the discov- lation in resistant cells, but the contributing ery of two plasmid-encoded quinolone efflux pump or its regulator have not been identified pumps: OqxAB and QepA (Sorensen et al. (Li and Nikaido 2009; Nikaido and Takatsuka 2003, Pe´richon et al. 2007; Yamane et al. 2009). Information on efflux mechanisms and 2007). In the last decade, PMQR genes have resistance in .50 bacterial species has recent- been found in bacterial isolates worldwide. ly been extensively reviewed (Li and Nikaido They reduce bacterial susceptibility to quino- 2009; Li et al. 2015) and is beyond the scope lones, usually not to the level of clinical non- of this review. Thus, efflux-mediated resistance susceptibility, but facilitate the selection of mu- to quinolones and many other antimicrobials tants with higher level quinolone resistance and is widespread. The broad substrate profiles of promote treatment failure. these pumps link quinolone resistance to mul- tidrug resistance and constitute mechanisms Qnr Structure and Function by which use of non-quinolone antimicrobials can also increase quinolone resistance. A simi- Cloning and sequencing qnrA revealed that it lar linkage to multidrug resistance occurs with encoded a 218-residue protein with a tandemly www.perspectivesinmedicine.org plasmid-mediated quinolone resistance, which repeating unit of five amino acids that indicated is discussed in the next section. membership in the many thousand-member pentapeptide repeat family of proteins (Tran and Jacoby 2002). Further searches led to the PLASMID-MEDIATED QUINOLONE discovery of related genes for plasmid-mediated RESISTANCE pentapeptide repeat proteins qnrS (Hata et al. Plasmid-mediatedquinoloneresistance(PMQR) 2005), qnrB (Jacoby et al. 2006), qnrC (Wang was reported in 1998, 31 years after nalidixic etal.2009),qnrD(Cavacoetal.2009),andqnrVC acid began to be used clinically and 12 years (Fonseca and Vicente 2013), as well as chromo- after modern fluoroquinolones were approved somal qnr genes in bacteria from avarietyof clin- for use (Martı´nez-Martı´nez et al. 1998). Trans- ical and environmental sources (Rodrı´guez-Mar- ferable nalidixic acid resistance had been sought tı´nez et al. 2008a; Sa´nchez et al. 2008). These new unsuccessfully in the 1970s (Burman 1977), and qnr genes generally differed by 35% or more in plasmid-mediated resistance was thought un- sequence from qnrA and each other. Allelic vari- likely to exist because quinolones are synthetic eties that differ by 10% or less have been de- compounds, and adequate resistance can arise scribed in almost all families: currently seven

10 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

for QnrA, 78 for QnrB, one for QnrC, two for et al. 2011; Xiong et al. 2011). Removal of even a QnrD, nine for QnrS, and six for QnrVC (see single amino acid in the larger loop compro- lahey.org/qnrstudies) (Jacoby et al. 2008). mises protection. Other essential residues in The first pentapeptide-repeat protein to QnrB are found in pentapeptide repeat posi- have its structure determined by X-ray crystal- tions i and i-2, in which alanine substitution lography was MfpA, which is encoded on the for the native amino acid eliminates protection chromosome of M. smegmatus and other my- as does deletion of .10 amino acids at the ami- cobacteria (Hegde et al. 2005) and implicated in no terminus or as few as three amino acids from quinolone resistance (Montero et al. 2001). the dimerization module at the carboxyl termi- MfpA is a dimer, linked carboxyl terminus to nus (Jacoby et al. 2013). EfsQnr lack loops, but carboxyl terminus, and folded into a right- EfsQnr differs from MfpA in having a 25-ami- handed quadrilateral b helix with size, shape, no-acid flexible extension required for full pro- and charge mimicking the B-form of DNA and tective activity (Hegde et al. 2011). just the size to fit into the cationic G segment In vitro, more Qnr is required to protect DNA-binding saddle of DNA gyrase and topo- DNA gyrase as the inhibiting concentration of isomerase IV. In vitro, MfpA inhibits DNA su- quinolone is increased (Tran and Jacoby 2002). percoiling by gyrase and, although it fails to In a gel-displacement assay (Tran et al. 2005) or block gyrase inhibition by quinolone, it can still bacterial two-hybrid system (Kim et al. 2015), confer quinolone resistance to whole cells by Qnr binds to both gyrase holoenzyme and its competing with DNA to reduce the number of A and B subunits. Binding to GyrA is reduced lethal double-strand breaks produced by quin- by the same amino- and carboxy-terminal and olone (Hegde et al. 2005). loop B deletions in QnrB that destroy its pro- As shown first with purified QnrA1 (Tran tective activity, whereas subinhibitory concen- and Jacoby 2002), and subsequently with plas- trations of ciprofloxacin reduce binding to mid-encoded QnrB1 (Jacoby et al. 2006) and GyrA but not to GyrB, suggesting that Qnr pro- QnrS1 (Tavio et al. 2014), and with chromo- tects gyrase by blocking access of quinolone to somally encoded AhQnr from Aeromonas hy- GyrA sites essential for its lethal action. drophila (Xiong et al. 2011) and EfsQnr from Many naturally occurring antibiotics and E. faecalis (Hegde et al. 2011), Qnr proteins do synthetic agents also target DNA gyrase. Qnr protect DNA gyrase from quinolone inhibition protects against compounds with a somewhat and only inhibit the enzyme at high concentra- quinolone-like structure (Jacobyet al. 2015), for tion. Like MfpA, they form rod-like dimers but example, 2-pyridone (Flamm et al. 1995), quin- have additional structural features. The struc- azoline-2,4-dione (Huband et al. 2007), or spi- www.perspectivesinmedicine.org ture of QnrB1 is shown in Figure 1. The quad- ropyrimidinetrione (Kern et al. 2011), so it is rilateral b-helix is stabilized by interactions not strictly quinolone-specific. Qnr, however, between the middle, usually hydrophobic, ami- does not block agents acting on the GyrB sub- no acid (i) of the pentapeptide repeat and the unit, and it also does not block simocyclinone first polar or hydrophobic residue (i-2), which D8, which, like quinolones, binds to the amino point inward, whereas the remaining amino terminus of GyrA and blocks DNA binding acids (i-1, iþ1, iþ2) are oriented outward, (Hearnshaw et al. 2014). forming a generally anionic surface. Hydrogen bonding between backbone atoms of neighbor- Qnr ORIGIN ing coils stabilizes the helix. The monomers of QnrB1 and AhQnr have Qnr homologs can be found encoded on the projecting loops of eight and 12 amino acids chromosome of many Gram-positive as well as that are important for their activity. Deletion Gram-negative bacteria, including species of of the small A loop reduces quinolone protec- Bacillus, Enterococcus, Listeria, and Mycobacteri- tion, whereas deletion of the larger B loop or um, and anaerobes such as Clostridium difficile both loops destroys protective activity (Vetting and Clostridium perfringens (Rodrı´guez-Martı´-

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 11 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

Loop B ′ Loop A′ C-term Subunit B 102–113 46–53 N-term α1′

N-term′

α1 Loop A Loop B′ 46–53 C-term Subunit A 102–113

Face1 Face2 Face3 Face4 N-term i–2i–1iii+1i+2 i–2i–1i i+1i+2 –2i–1iii+1i+2 –2i–1i i+1i+2 Coil0 MA LALVG EKIDR NRFTG 17 Coil1 EKIEN STFFN CDFSG ADLSG 37 Coil2 TEFIG CQF * CNFSR AMLKD 63 Coil3 AIFKS CDLSM ADFRN SSALG 83 Coil4 IEIRH CRAQG ADFRG ASF** 101 Coil5 AYITN TNLSY ANFSK VVLEK 133 Coil6 CELWE NRWIG AQVLG ATFSG 153 Coil7 SDLSG GEFST FDWRA ANFTH 173 Coil8 CDLTN SELGD LDIRG VDLQG 193 Coil9 VKLDN YQASL LMERL GIAVIG 214

Loop A (*) 46 Y D R E S Q K G 53 Loop B (**) 102 M N M I T T R T W F C S 113

Figure 1. The rod-like structure of the QnrB1 dimer is shown (above) with the sequence of the monomer (below). The sequence is divided into four columns representing the four faces of the right-handed quadrilateral b-helix. Face names and color are shown at the top along with the naming convention for the five residues of the pentapeptide repeats. Loops A and B are indicated by one and two asterisks, respectively, with their sequences indicated below and the loops shown as black traces on the diagram. The carboxy-terminal a-helix is colored salmon. The molecular twofold symmetry is indicated with a black diamond. Type II turn containing faces are shown as spheres and type IV-containing faces as strands. N-term, Amino terminal; C-term, carboxy terminal. (From Jacoby et al. 2014; reproduced, with permission, from the authors.) www.perspectivesinmedicine.org nez et al. 2008b; Sa´nchez et al. 2008; Boulund complex (Jacoby et al. 2011; Ribeiro et al. et al. 2012; Jacoby and Hooper 2013). Aquatic 2015). The small, nonconjugative plasmids bacteria are especially well represented, includ- that carry qnrD are especially likely to be found ing species of Aeromonas, Photobacterium, She- in Proteeae, such as Proteus mirabilis, P.vulgaris, wanella, and Vibrio (Poirel et al. 2005a,b). and Providencia rettgeri and may have originat- QnrA1 is 98% identical to the chromosomally ed there (Guillard et al. 2012, 2014; Zhang et al. determined Qnrof Shewanella algae (Poirel et al. 2013). 2005a). QnrS1 is 97% identical to Qnr from Vib- The worldwide distribution of qnr sug- rio parahemolyticus S022 (GenBank accession gests an origin well before quinolones were number WP_029823919) or Vibrio mytili (Gen- discovered. Indeed, qnrB genes and pseudo- Bank WP_041155100), and QnrC is 97% genes have been discovered on the chromo- identical to Qnr in V. parahemolyticus S145 some of C. freundii strains collected in the (GenBank WP_025518018). QnrB homologs, 1930s (Saga et al. 2013). What the native func- on the other hand, are encoded on the chromo- tion of Qnr may have been is an as-yet unan- some of members of the Citrobacter freundii swered question.

12 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

AAC(60)-Ib-cr farmworkers in China, where olaquindox is AAC(60)-Ib-cr is a bifunctional variant of a used, 40% of E. coli isolated from animals and common acetyltransferase, providing resistance 30% from humans carried plasmid-mediated to such aminoglycosides as , kanamy- oqxAB, making it much more common than cin, and tobramycin, but also able to acetylate other types of PMQR in this setting (Zhao those fluoroquinolones with an amino nitrogen et al. 2010). on the piperazinyl ring such as ciprofloxacin The QepA efflux pump was reported in and norfloxacin (Robicsek et al. 2006). Com- 2007 by investigators in Japan and France on pared with other AAC(60)-Ib enzymes, the -cr plasmids in clinical isolates of E. coli often as- variant has two unique amino acid substitu- sociated with aminoglycoside resistance be- tions—Trp102Arg and Asp179Tyr—both of cause of ribosomal methylase rmtB (Pe´richon which are required for quinolone acetylating et al. 2007; Yamane et al. 2007). It has subse- activity. Models of enzyme action suggest that quently been found worldwide (Habeeb et al. the Asp179Tyr replacement is particularly im- 2014; Zhao et al. 2015). portant in permitting p-stacking interactions with the quinolone ring to facilitate quinolone QUINOLONE RESISTANCE PLASMIDS binding. The role of Trp102Arg is to position the Tyr face for optimal interaction (Vetting Genes for quinolone resistance have been found et al. 2008) or to hydrogen bond to keto or on plasmids varying in size and incompatibility carboxyl groups of the quinolone to fix it in specificity, indicating that the spread of multi- place (Maurice et al. 2008). The aac(60)-Ib-cr ple plasmids has been responsible for the dis- gene is usually found in a cassette as part of semination of this resistance around the world an integron in a multiresistance plasmid, which and that plasmid acquisition of qnr and other may contain other PMQR genes. Several alleles quinolone resistance determinants has occurred have been described (Quiroga et al. 2015). The independently multiple times. A mobile or gene has been found worldwide in a variety of transposable element is almost invariably asso- Enterobacteriaceae and even in P. aeruginosa ciated with qnr genes, especially ISCR1 and (Ogbolu et al. 2011). Association with extended IS26. qnrD and qnrS2 are located within mobile spectrum b-lactamase (ESBL) CTX-M-15 is insertion cassettes, elements with bracketing in- particularly common (Oteo et al. 2009; Sabtch- verted repeats but lacking a transposase (Pica˜o eva et al. 2009). et al. 2008; Guillard et al. 2014), whereas qnrVC is so far the only qnr gene located in a cassette with a linked attC site (Fonseca et al. 2008; Be- www.perspectivesinmedicine.org OqxAB AND QepA lotti et al. 2015). OqxAB was first identified as a plasmid-medi- qnr genes are usually found in multiresis- ated efflux pump conferring resistance to the tance plasmids linked to other resistance deter- olaquindox, a food additive enhancing growth minants. b-lactamase genes, including genes in pigs (Sorensen et al. 2003) and later shown to for ESBLs, AmpC enzymes, and carbapene- confer resistance to ciprofloxacin and norflox- mases, have been conspicuously common (Ja- acin as well as other antimicrobials, including coby et al. 2014). chloramphenicol, , and trimeth- PMQR genes have been found in a variety of oprim (Hansen et al. 2007; Ho et al. 2016). Enterobacteriaceae, especially E. coli and species Genes for oqxAB are commonly found on the of Enterobacter, Klebsiella, and Salmonella (Ja- chromosome of K. pneumoniae and Entero- coby et al. 2014). They have rarely been found in bacter spp. but are expressed at a much higher nonfermenters but have occasionally been re- level when captured on plasmids, often in asso- ported in P. aeruginosa and A. baumannii. qnr ciation with the IS26 mobilizing element (Rod- genes are also found in a variety of Gram-pos- rı´guez-Martı´nez et al. 2013; Wong et al. 2015). itive organisms but are chromosomal and not In a study of fecal samples from animals and plasmid-mediated. The earliest known qnr out-

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 13 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

side of Citrobacter spp. dates from 1988 (Jacoby concentration (MIC) commonly yields mutants et al. 2009). Studies in the last decade suggest with alterations in the QRDR of gyrA. Surpris- that the prevalence of PMQR is increasing (Kim ingly, gyrase mutants are rarely selected from et al. 2009a). In a recent study of .500 isolates the same strain carrying qnr (Cesaro et al. of E. coli from 30 county hospitals in China, 2008). Rather, mutants have increased expres- 37.3% carried at least one PMQR gene, includ- sion of acrAB, mdtEF,orydhE pumps, which ing 19.7% with aac(60)-Ib-cr, 14.4% with qepA, efflux quinolones, or alterations in genes of li- 3.8% with oqxAB, and 3.7% with qnr (Zhao popolysaccharide core biosynthesis, which may et al. 2015). reduce quinolone entry via reduction in porin expression (Vinue´ et al. 2015). Despite the modest effect of PMQR genes RESISTANCE BECAUSE OF PMQR on susceptibility, their presence makes in- DETERMINANTS fections in animal models harder to treat (Rod- Table 2 shows the effect on susceptibility of a rı´guez-Martı´nez et al. 2008a; Jakobsen et al. common E. coli host of various PMQR genes. 2012), and there is some evidence for poorer qnr alleles decrease ciprofloxacin or levofloxacin outcomes from human infections with qnr con- susceptibility 30-fold to a level similar to that of taining pathogens (Chong et al. 2010; Liao et al. the common Ser83Leu GyrA mutation but have 2013). much less effect on nalidixic acid resistance. The other PMQR genes reduce susceptibility even SUMMARY less but with more specificity. Levofloxacin and nalidixic acid are unaffected by AAC(60)- Resistance to quinolones has increased to sub- Ib-cr because they lack the C7 amino target stantial levels, despite these agents being syn- for acetylation, and QepA affects ciprofloxacin thetic and having two essential bacterial targets. susceptibility more than that of the other quin- The microbial resistance has been affected by an olones (Pe´richon et al. 2007). By themselves impressive diversity of mechanisms that have none reaches the CLSI breakpoint for loss of linked quinolone resistance to multidrug resis- susceptibility. In combination, however, they tance, compounding the current public health may do so, and all facilitate selection of high- and medical challenges of broadly resistant bac- er-level quinolone resistance (Martı´nez-Martı´- teria. Dual topoisomerase targets with differing nez et al. 1998; Robicsek et al. 2006; Rodrı´guez- sensitivities to many quinolones in clinical use Martı´nez et al. 2007). create a pathway for additive mutational resis- From a PMQR-free E. coli strain selection on tance to high levels. This core target-based www.perspectivesinmedicine.org ciprofloxacin at 3 times minimum inhibitory resistance has been further supplemented and

Table 2. Minimum inhibitory concentrations (MICs) produced in E. coli MIC (mg/ml)

E. coli strain Ciprofloxacin Levofloxacin Nalidixic acid J53 0.008 0.015 4 J53 gyrA S83L 0.25 0.5 512 J53 pMG252 (qnrA1) 0.25 0.5 16 J53 pMG299 (qnrB1) 0.25 0.5 16 J53 pMG306 (qnrS1) 0.25 0.38 16 J53 pMG320 (aac(60)-Ib-cr) 0.06 0.015 4 J53 pAT851 (qepA) 0.064 0.032 4 CLSI susceptibility breakpoint 1.0 2.0 16 From Jacoby et al. 2014. MIC, Minimum inhibitory concentration.

14 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

facilitated by more insidious low-level resis- 1996. Differential behaviors of Staphylococcus aureus tance. This low-level resistance has come from and Escherichia coli type II DNA . Anti- microb Agents Chemother 40: 2714–2720. both overexpression of native multidrug efflux Bolhuis H, Poelarends G, Van Veen HW, Poolman B, Dries- pumps as well as the unanticipated emergence sen AJM, Konings WN. 1995. The lactococcal lmrP gene of multiple mechanisms of plasmid-mediated encodes a proton motive force-dependent drug trans- quinolone resistance, which may not be readily porter. J Biol Chem 270: 26092–26098. Boncoeur E, Durmort C, Bernay B, Ebel C, Di Guilmi AM, detected in clinical laboratories but can facili- Croize J, Vernet T, Jault JM. 2012. PatA and PatB form a tate selection of higher-level resistance and adds functional heterodimeric ABC multidrug efflux trans- a plasmid linkage to multidrug resistance. porter responsible for the resistance of Streptococcus pneumoniae to fluoroquinolones. Biochemistry 51: 7755–7765. Boulund F, Johnning A, Pereira MB, Larsson DG, Kristians- ACKNOWLEDGMENTS son E. 2012. A novel method to discover fluoroquinolone This work is supported by Grants R01 AI057576 antibiotic resistance (qnr) genes in fragmented nucleo- tide sequences. BMC Genomics 13: 695. (to D.C.H and G.A.J.), R37 AI023988 (to Breines DM, Ouabdesselam S, Ng EY, Tankovic J, Shah S, D.C.H.), and P01 AI083214 (to D.C.H.) from Soussy CJ, Hooper DC. 1997. Quinolone resistance locus the U.S. Public Health Service, National Insti- nfxD of Escherichia coli is a mutant allele of parE gene encoding a subunit of topoisomerase IV. Antimicrob tutes of Health. Agents Chemother 41: 175–179. Burman LG. 1977. Apparent absence of transferable resis- tance to nalidixic acid in pathogenic Gram-negative bac- REFERENCES teria. J Antimicrob Chemother 3: 509–516. Cavaco LM, Hasman H, Xia S, Aarestrup FM. 2009. qnrD,a Aldred KJ, McPherson SA, Wang P, Kerns RJ, Graves DE, novel gene conferring transferable quinolone resistance Turnbough CL Jr, Osheroff N. 2012. Drug interactions in Salmonella enterica serovar Kentucky and Bovismor- with Bacillus anthracis topoisomerase IV: Biochemical bificans strains of human origin. Antimicrob Agents Che- basis for quinolone action and resistance. Biochemistry mother 53: 603–608. 51: 370–381. Cesaro A, Bettoni RR, Lascols C, Merens A, Soussy CJ, Cam- Aldred KJ, Kerns RJ, Osheroff N. 2014. Mechanism of quin- bau E. 2008. Low selection of topoisomerase mutants olone action and resistance. Biochemistry 53: 1565–1574. from strains of Escherichia coli harbouring plasmid- Alekshun MN, Levy SB. 1999. The mar regulon: Multiple borne qnr genes. J Antimicrob Chemother 61: 1007–1015. resistance to antibiotics and other toxic chemicals. Trends Chen PR, Bae T,Williams WA,Duguid EM, Rice PA, Schnee- Microbiol 7: 410–413. wind O, He C. 2006. An oxidation-sensing mechanism is Alonso A, Martinez JL. 2000. Cloning and characterization used by the global regulator MgrA in Staphylococcus au- of SmeDEF, a novel multidrug efflux pump from Steno- reus. Nat Chem Biol 2: 591–595. trophomonas maltophilia. Antimicrob Agents Chemother 44: 3079–3086. Chong YP,Choi SH, Kim ES, Song EH, Lee EJ, Park KH, Cho OH, Kim SH, Lee SO, Kim MN, et al. 2010. Bloodstream Baucheron S, Imberechts H, Chaslus-Dancla E, Cloeckaert infections caused by qnr-positive Enterobacteriaceae: A. 2002. The AcrB multidrug transporter plays a major

www.perspectivesinmedicine.org Clinical and microbiologic characteristics and outcomes. role in high-level fluoroquinolone resistance in Salmo- Diagn Microbiol Infect Dis 67: 70–77. nella enterica serovar typhimurium phage type DT204. Microb Drug Resist 8: 281–289. Chou JH, Greenberg JT,Demple B. 1998. Postranscriptional repression of Escherichia coli OmpF protein in response Bax BD, Chan PF, Eggleston DS, Fosberry A, Gentry DR, to redox stress: Positive control of the micF antisense Gorrec F, Giordano I, Hann MM, Hennessy A, Hibbs M, RNA by the soxRS locus. J Bacteriol 175: 1026–1031. et al. 2010. Type IIA topoisomerase inhibition by a new class of antibacterial agents. Nature 466: 935–940. Courvalin P. 1990. Plasmid-mediated 4-quinolone resis- Belotti PT, Thabet L, Laffargue A, Andre C, Coulange- tance: A real or apparent absence? Antimicrob Agents Che- Mayonnove L, Arpin C, Messadi A, M’Zali F, Quentin mother 34: 681–684. C, Dubois V. 2015. Description of an original integron Ding Y, Onodera Y, Lee JC, Hooper DC. 2008. NorB, an encompassing blaVIM-2, qnrVC1 and genes encoding bac- efflux pump in Staphylococcus aureus MW2, contributes terial group II intron proteins in Pseudomonas aerugi- to bacterial fitness in abscesses. J Bacteriol 190: 7123– nosa. J Antimicrob Chemother 70: 2237–2240. 7129. Blair JM, Bavro VN, Ricci V, Modi N, Cacciotto P, Kleine- Domagala JM, Hagen SE. 2003. Structure-activity relation- kathfer U, Ruggerone P, Vargiu AV, Baylay AJ, Smith HE, ships of the quinolone antibacterials in the new millen- et al. 2015. AcrB drug-binding pocket substitution con- nium: Some things change and some do not. In Quino- fers clinically relevant resistance and altered substrate lone antimicrobial agents, 3rd ed. (ed. Hooper DC, specificity. Proc Natl Acad Sci 112: 3511–3516. Rubinstein E), pp. 3–18. ASM, Washington, DC. Blanche F, Cameron B, Bernard FX, Maton L, Manse B, Drlica K, Zhao XL. 1997. DNA gyrase, topoisomerase IV, Ferrero L, Ratet N, Lecoq C, Goniot A, Bisch D, et al. and the 4-quinolones. Microbiol Rev 61: 377–392.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 15 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

Drlica K, Malik M, Kerns RJ, Zhao X. 2008. Quinolone- olone resistance in Listeria monocytogenes. Antimicrob mediated bacterial death. Antimicrob Agents Chemother Agents Chemother 47: 704–708. 52: 385–392. Grkovic S, Brown MH, Skurray RA. 2002. Regulation of Drlica K, Hiasa H, Kerns R, Malik M, Mustaev A, Zhao X. bacterial drug export systems. Microbiol Mol Biol Rev 2009. Quinolones: Action and resistance updated. Curr 66: 671–701. Top Med Chem 9: 981–998. Guerin F, Galimand M, Tuambilangana F, Courvalin P,Cat- Du D, Wang Z, James NR, Voss JE, Klimont E, Ohene-Agyei toir V. 2014. Overexpression of the novel MATE fluoro- T, Venter H, Chiu W, Luisi BF. 2014. Structure of the quinolone efflux pump FepA in Listeria monocytogenes is AcrAB-TolC multidrug efflux pump. Nature 509: 512– driven by inactivation of its local repressor FepR. PLoS 515. ONE 9: e106340. Escudero JA, San MA, Gutierrez B, Hidalgo L, La Ragione Guillard T, Cambau E, Neuwirth C, Nenninger T, Mbadi A, RM, AbuOun M, Galimand M, Ferrandiz MJ, Domin- Brasme L, Vernet-Garnier V, Bajolet O, de Champs C. guez L, de la Campa AG, et al. 2011. Fluoroquinolone 2012. Description of a 2,683-base-pair plasmid contain- efflux in Streptococcus suis is mediated by SatAB and not ing qnrD in two Providencia rettgeri isolates. Antimicrob by SmrA. Antimicrob Agents Chemother 55: 5850–5860. Agents Chemother 56: 565–568. Fass D, Bogden CE, Berger JM. 1999. Quaternary changes in Guillard T, Grillon A, de Champs C, Cartier C, Madoux J, topoisomerase II may direct orthogonal movement of Bercot B, Lebreil AL, Lozniewski A, Riahi J, Vernet-Gar- two DNA strands. Nature Struct Biol 6: 322–326. nier V, et al. 2014. Mobile insertion cassette elements found in small non-transmissible plasmids in Proteeae Fernando DM, Xu W, Loewen PC, Zhanel GG, Kumar A. may explain qnrD mobilization. PLoS ONE 9: e87801. 2014. Triclosan can select for an AdeIJK-overexpressing mutant of Acinetobacter baumannii ATCC 17978 that Habeeb MA, Haque A, Iversen A, Giske CG. 2014. Occur- displays reduced susceptibility to multiple antibiotics. rence of virulence genes, 16S rRNA methylases, and Antimicrob Agents Chemother 58: 6424–6431. plasmid-mediated quinolone resistance genes in CTX- M-producing Escherichia coli from Pakistan. Eur J Clin Flamm RK, Vojtko C, Chu DT, Li Q, Beyer J, Hensey D, Microbiol Infect Dis 33: 399–409. Ramer N, Clement JJ, Tanaka SK. 1995. In vitro evalua- tion of ABT-719, a novel DNA gyrase inhibitor. Antimi- Hansen LH, Jensen LB, Sorensen HI, Sorensen SJ. 2007. crob Agents Chemother 39: 964–970. Substrate specificity of the OqxAB multidrug resistance pump in Escherichia coli and selected enteric bacteria. Floyd JL, Smith KP, Kumar SH, Floyd JT, Varela MF. 2010. J Antimicrob Chemother 60: 145–147. LmrS is a multidrug efflux pump of the major facilitator superfamily from Staphylococcus aureus. Antimicrob Hata M, Suzuki M, Matsumoto M, TakahashiM, Sato K, Ibe Agents Chemother 54: 5406–5412. S, Sakae K. 2005. Cloning of a novel gene for quinolone resistance from a transferable plasmid in Shigella flexneri Fonseca EL, Vicente AC. 2013. Epidemiology of qnrVC al- 2b. Antimicrob Agents Chemother 49: 801–803. leles and emergence out of the Vibrionaceae family. JMed Hay T, Fraud S, Lau CH, Gilmour C, Poole K. 2013. Anti- Microbiol 62: 1628–1630. biotic inducibility of the mexXY multidrug efflux operon Fonseca EL, Dos Santos Freitas F, Vieira VV, Vicente AC. of : Involvement of the MexZ 2008. New qnr gene cassettes associated with superinte- anti-repressor ArmZ. PLoS ONE 8: e56858. gron repeats in Vibrio cholerae O1. Emerg Infect Dis 14: Hearnshaw SJ, Edwards MJ, Stevenson CE, Lawson DM, 1129–1131. Maxwell A. 2014. A new crystal structure of the bifunc- Fournier B, Hooper DC. 2000. A new two-component reg- tional antibiotic simocyclinone D8 bound to DNA gyrase ulatory system involved in adhesion autolysis, and extra- gives fresh insight into the mechanism of inhibition. cellular proteolytic activity of Staphylococcus aureus. J www.perspectivesinmedicine.org J Mol Biol 426: 2023–2033. Bacteriol 182: 3955–3964. Heeb S, Fletcher MP, Chhabra SR, Diggle SP, Williams P, Fournier B, Zhao X, Lu T, Drlica K, Hooper DC. 2000. Camara M. 2011. Quinolones: From antibiotics to auto- Selective targeting of topoisomerase IV and DNA gyrase inducers. FEMS Microbiol Rev 35: 247–274. in Staphylococcus aureus: Different patterns of quino- Hegde SS, Vetting MW, Roderick SL, Mitchenall LA, Max- lone-induced inhibition of DNA synthesis. Antimicrob well A, Takiff HE, Blanchard JS. 2005. A fluoroquinolone Agents Chemother 44: 2160–2165. resistance protein from Mycobacterium tuberculosis that Fournier B, Klier A, Rapoport G. 2001. The two-component mimics DNA. Science 308: 1480–1483. system ArlS-ArlR is a regulator of virulence gene expres- Hegde SS, Vetting MW, Mitchenall LA, Maxwell A, Blan- sion in Staphylococcus aureus. Mol Microbiol 41: 247– chard JS. 2011. Structural and biochemical analysis of the 261. pentapeptide repeat protein EfsQnr, a potent DNA gyrase Gensberg K, Jin YF,Piddock LJ. 1995. A novel gyrB mutation inhibitor. Antimicrob Agents Chemother 55: 110–117. in a fluoroquinolone-resistant clinical isolate of Salmo- Hiasa H. 2002. The Glu-84 of the ParC subunit plays criti- nella typhimurium. FEMS Microbiol Lett 132: 57–60. cal roles in both topoisomerase IV-quinolone and topo- Gill MJ, Brenwald NP, Wise R. 1999. Identification of an isomerase IV-DNA interactions. Biochemistry 41: 11779– efflux pump gene, pmrA, associated with fluoroquino- 11785. lone resistance in Streptococcus pneumoniae. Antimicrob Hiasa H, YousefDO, Marians KJ. 1996. DNA strand cleavage Agents Chemother 43: 187–189. is required for replication fork arrest by a frozen topo- Godreuil S, Galimand M, Gerbaud G, Jacquet C, Courvalin isomerase-quinolone-DNA ternary complex. J Biol Chem P. 2003. Efflux pump Lde is associated with fluoroquin- 271: 26424–26429.

16 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

Ho PL, Ng KY, Lo WU, Law PY, Lai EL, Wang Y, Chow KH. DNA gyrase. Antimicrob Agent Chemother 59: 6689– 2016. Plasmid-mediated OqxAB is an important mech- 6695. anism for nitrofurantoin resistance in Escherichia coli. Jair KW, Yu X, Skarstad K, Tho¨ny B, Fujita N, Ishihama A, Antimicrob Agents Chemother 60: 537–543. WolfRE Jr, 1996. Transcriptional activation of promoters Hooper DC. 1997. Bacterial topoisomerases, anti-topo- of the superoxide and multiple antibiotic resistance reg- isomerases, and anti-topoisomerase resistance. Clin In- ulons by Rob, a binding protein of the Escherichia coli fect Dis 27: S54–S63. origin of chromosomal replication. J Bacteriol 178: Hooper DC. 2003. Mechanisms of quinolone resistance. In 2507–2513. Quinolone antimicrobial agents, 3rd ed. (ed. Hooper DC, Jakobsen L, Cattoir V, Jensen KS, Hammerum AM, Nord- Rubinstein E), pp. 41–67. ASM, Washington, DC. mann P, Frimodt-Moller N. 2012. Impact of low-level fluoroquinolone resistance genes qnrA1, qnrB19 and Howard BM, Pinney RJ, Smith JT. 1993. 4-Quinolone bac- qnrS1 on ciprofloxacin treatment of isogenic Escherichia tericidal mechanisms. Arzneimittel-Forschung 43: 1125– coli strains in a murine model. 1129. J Antimicrob Chemother 67: 2438–2444. Huang J, O’Toole PW, Shen W, Amrine-Madsen H, Jiang X, Kaatz GW,DeMarco CE, Seo SM. 2006. MepR, a repressor of Lobo N, Palmer LM, Voelker L, Fan F, Gwynn MN, et al. the Staphylococcus aureus MATE family multidrug efflux 2004. Novel chromosomally encoded multidrug efflux pump MepA, is a substrate-responsive regulatory pro- transporter MdeA in Staphylococcus aureus. Antimicrob tein. Antimicrob Agents Chemother 50: 1276–1281. Agents Chemother 48: 909–917. Kern G, Basarab GS, Andrews B, Schuck V, Stone G, Huband MD, Cohen MA, Zurack M, Hanna DL, Skerlos LA, Kutschke A, Beaudoin M-E, San Martin M, Brassil P, Sulavik MC, Gibson GW,Gage JW,Ellsworth E, Stier MA, Fan JH, et al. 2011. A DNA gyrase inhibitor with a novel et al. 2007. In vitro and in vivo activities of PD 0305970 mode of inhibition and in vivo efficacy. 51st Interscience and PD 0326448, new bacterial gyrase/topoisomerase Conference on Antimicrobial Agents and inhibitors with potent antibacterial activities versus mul- (ICAAC), Abstract F1-1840. Chicago, September 17–20. tidrug-resistant gram-positive and fastidious organism Khodursky AB, Cozzarelli NR. 1998. The mechanism of groups. Antimicrob Agents Chemother 51: 1191–1201. inhibition of topoisomerase IV by quinolone antibacte- Ingavale S, Van Wamel W, Luong TT, Lee CY, Cheung AL. rials. J Biol Chem 273: 27668–27677. 2005. Rat/MgrA, a regulator of autolysis, is a regulator of Khodursky AB, Zechiedrich EL, Cozzarelli NR. 1995. Topo- virulence genes in Staphylococcus aureus. Infect Immun isomerase IV is a target of quinolones in Escherichia coli. 73: 1423–1431. Proc Natl Acad Sci 92: 11801–11805. Ito H, Yoshida H, Bogaki-Shonai M, Niga T, Hattori H, Kim ES, Hooper DC. 2014. Clinical importance and epide- Nakamura S. 1994. Quinolone resistance mutations in miology of quinolone resistance. Infect Chemother 46: the DNA gyrase gyrA and gyrB genes of Staphylococcus 226–238. aureus. Antimicrob Agents Chemother 38: 2014–2023. Kim HB, Park CH, Kim CJ, Kim EC, Jacoby GA, Hooper Jacoby GA, Hooper DC. 2013. Phylogenetic analysis of chro- DC. 2009a. Prevalence of plasmid-mediated quinolone mosomally determined Qnr and related proteins. Anti- resistance determinants over a 9-year period. Antimicrob microb Agents Chemother 57: 1930–1934. Agents Chemother 53: 639–645. Jacoby GA, WalshKE, Mills DM, Walker VJ, Oh H, Robicsek Kim HB, Wang M, Park CH, Kim EC, Jacoby GA, Hooper A, Hooper DC. 2006. qnrB, another plasmid-mediated DC. 2009b. oqxAB encoding a multidrug efflux pump in gene for quinolone resistance. Antimicrob Agents Chemo- human clinical isolates of Enterobacteriaceae. Antimicrob ther 50: 1178–1182. Agents Chemother 53: 3582–3584. Jacoby G, Cattoir V,Hooper D, Martı´nez-Martı´nez L, Nord- Kim ES, Chen C, Braun M, Kim HY, Okumura R, Wang Y, www.perspectivesinmedicine.org mann P, Pascual A, Poirel L, Wang M. 2008. qnr gene Jacoby GA, Hooper DC. 2015. Interactions between nomenclature. Antimicrob Agents Chemother 52: 2297– QnrB, QnrB mutants, and DNA gyrase. Antimicrob 2299. Agents Chemother 59: 5413–5419. Jacoby GA, Gacharna N, Black TA, Miller GH, Hooper DC. Klyachko KA, Schuldiner S, Neyfakh AA. 1997. Mutations 2009. Temporal appearance of plasmid-mediated quino- affecting substrate specificity of the Bacillus subtilis mul- lone resistance genes. Antimicrob Agents Chemother 53: tidrug transporter Bmr. J Bacteriol 179: 2189–2193. 1665–1666. Ko¨hler T, Michea-Hamzehpour M, Henze U, Gotoh N, Curty L, Peche`re JC. 1997a. Characterization of MexE– Jacoby GA, Griffin CM, Hooper DC. 2011. Citrobacter spp. MexF–OprN, a positively regulated multidrug efflux sys- as a source of qnrB alleles. Antimicrob Agents Chemother tem of Pseudomonas aeruginosa. Mol Microbiol 23: 345– 55: 4979–4984. 354. Jacoby GA, Corcoran MA, Mills DM, Griffin CM, Hooper Ko¨hler T, Michea-Hamzehpour M, Plesiat P, Kahr AL, Pe- DC. 2013. Mutational analysis of quinolone resistance che`re JC. 1997b. Differential selection of multidrug efflux protein QnrB1. Antimicrob Agents Chemother 57: systems by quinolones in Pseudomonas aeruginosa. Anti- 5733–5736. microb Agents Chemother 41: 2540–2543. Jacoby GA, Strahilevitz J, Hooper DC. 2014. Plasmid-me- Kreuzer KN, Cozzarelli NR. 1979. Escherichia coli mutants diated quinolone resistance. Microbiol Spectr doi: 10. thermosensitive for deoxyribonucleic acid gyrase subunit 1128/microbiolspec.PLAS-0006-2013. A: Effects on deoxyribonucleic acid replication, tran- Jacoby GA, Corcoran MA, Hooper DC. 2015. The protective scription, and bacteriophage growth. J Bacteriol 140: effect of Qnr on agents other than quinolones that target 424–435.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 17 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

Kumar R, Shankar MB, Nagaraja V. 2014. Molecular basis tions and the function of the CmeABC efflux pump. for the differential quinolone susceptibility of mycobac- Antimicrob Agents Chemother 47: 390–394. terial DNA gyrase. Antimicrob Agents Chemother. Martı´nez-Martı´nez L, Pascual A, Jacoby GA. 1998. Quino- Kumaraswami M, Schuman JT,Seo SM, Kaatz GW,Brennan lone resistance from a transferable plasmid. Lancet 351: RG. 2009. Structural and biochemical characterization of 797–799. MepR, a multidrug binding transcription regulator of Masuda N, Sakagawa E, Ohya S, Gotoh N, Tsujimoto H, the Staphylococcus aureus multidrug efflux pump MepA. Nishino T. 2000. Substrate specificities of MexAB– Nucleic Acids Res 37: 1211–1224. OprM, MexCD–OprJ, and MexXY–OprM efflux pumps Laponogov I, Sohi MK, Veselkov DA, Pan XS, Sawhney R, in Pseudomonas aeruginosa. Antimicrob Agents Chemo- Thompson AW,McAuley KE, Fisher LM, Sanderson MR. ther 44: 3322–3327. 2009. Structural insight into the quinolone-DNA cleav- Matsuo Y, Eda S, Gotoh N, Yoshihara E, Nakae T. 2004. age complex of type IIA topoisomerases. Nat Struct Mol MexZ-mediated regulation of mexXY multidrug efflux Biol 16: 667–669. pump expression in Pseudomonas aeruginosa by binding Laponogov I, Veselkov DA, Crevel IM, Pan XS, Fisher LM, on the mexZ-mexX intergenic DNA. FEMS Microbiol Lett Sanderson MR. 2013. Structure of an “open” clamp type 238: 23–28. II topoisomerase-DNA complex provides a mechanism Maurice F, Broutin I, Podglajen I, Benas P,Collatz E, Dardel for DNA capture and transport. Nucleic Acids Res 41: F. 2008. Enzyme structural plasticity and the emergence 9911–9923. of broad-spectrum antibiotic resistance. EMBO Rep 9: Lesher GY,Forelich ED, Gruet MD, Bailey JH, Brundage RP. 344–349. 1962. 1,8-Naphthyridine derivatives. A new class of che- Me´graud F. 1998. Epidemiology and mechanism of anti- motherapeutic agents. J Med Pharm Chem 5: 1063–1068. biotic resistance in Helicobacter pylori. Gastroenterology Lewin CS, Howard BM, Smith JT. 1991. Protein- and RNA- 115: 1278–1282. synthesis independent bactericidal activity of ciprofloxa- Miller PF, Gambino L, Sulavik MC, Gracheck SJ. 1994. Ge- cin that involves the A subunit of DNA gyrase. JMed netic relationship between soxRS and mar loci in promot- Microbiol 34: 19–22. ing multiple antibiotic resistance in Escherichia coli. Anti- Li XZ, Nikaido H. 2004. Efflux-mediated in microb Agents Chemother 38: 1773–1779. bacteria. 64: 159–204. Miyamae S, Nikaido H, Tanaka Y,Yoshimura F.1998. Active efflux of norfloxacin by Bacteroides fragilis. Antimicrob Li XZ, Nikaido H. 2009. Efflux-mediated drug resistance in Agents Chemother 42: 2119–2121. bacteria: An update. Drugs 69: 1555–1623. Miyamae S, Ueda O, Yoshimura F, Hwang J, Tanaka Y, Ni- Li XZ, Barre´ N, Poole K. 2000a. Influence of the MexA– kaido H. 2001. A MATE family multidrug efflux trans- MexB–OprM multidrug efflux system on expression of porter pumps out fluoroquinolones in Bacteroides the- the MexC–MexD–OprJ and MexE–MexF–OprN mul- taiotaomicron. Antimicrob Agents Chemother 45: 3341– tidrug efflux systems in Pseudomonas aeruginosa. J Anti- 3346. microb Chemother 46: 885–893. Montero C, Mateu G, Rodriguez R, Takiff H. 2001. Intrinsic Li XZ, Zhang L, Poole K. 2000b. Interplay between the resistance of Mycobacterium smegmatis to fluoroquino- MexA–MexB–OprM multidrug efflux system and the lones may be influenced by new pentapeptide protein outer membrane barrier in the multiple antibiotic resis- MfpA. Antimicrob Agents Chemother 45: 3387–3392. tance of Pseudomonas aeruginosa. J Antimicrob Chemo- ther 45: 433–436. Morais Cabral JH, Jackson AP,Smith CV, Shikotra N, Max- well A, Liddington RC. 1997. Crystal structure of the Li XZ, Plesiat P, Nikaido H. 2015. The challenge of efflux- breakage-reunion domain of DNA gyrase. Nature 388: mediated antibiotic resistance in Gram-negative bacteria. www.perspectivesinmedicine.org 903–906. Clin Microbiol Rev 28: 337–418. Morita Y, Kataoka A, Shiota S, Mizushima T, Tsuchiya T. Liao CH, Hsueh PR, Jacoby GA, Hooper DC. 2013. Risk 2000. NorM of Vibrio parahaemolyticus is an Naþ-driven factors and clinical characteristics of patients with qnr- multidrug efflux pump. J Bacteriol 182: 6694–6697. positive Klebsiella pneumoniae bacteraemia. J Antimicrob Mustaev A, Malik M, Zhao X, Kurepina N, Luan G, Oppe- Chemother 68: 2907–2914. gard LM, Hiasa H, Marks KR, Kerns RJ, Berger JM, et Lin J, Michel LO, Zhang QJ. 2002. CmeABC functions as a al. 2014. Fluoroquinolone-gyrase-DNA complexes: Two multidrug efflux system in Campylobacter jejuni. Antimi- modes of drug binding. J Biol Chem 289: 12300–12312. crob Agents Chemother 46: 2124–2131. Nakaminami H, Noguchi N, Sasatsu M. 2010. Fluoroquin- Lomovskaya O, Lewis K, Matin A. 1995. EmrR is a negative olone efflux by the plasmid-mediated multidrug efflux regulator of the Escherichia coli multidrug resistance pump QacB variant QacBIII in Staphylococcus aureus. pump EmrAB. J Bacteriol 177: 2328–2334. Antimicrob Agents Chemother 54: 4107–4111. Lomovskaya O, Lee A, Hoshino K, Ishida H, Mistry A, War- Ng EY,Trucksis M, Hooper DC. 1996. Quinolone resistance ren MS, Boyer E, Chamberland S, Lee VJ. 1999. Use of a mutations in topoisomerase IV: Relationship of the flqA genetic approach to evaluate the consequences of inhibi- locus and genetic evidence that topoisomerase IV is the tion of efflux pumps in Pseudomonas aeruginosa. Anti- primary target and DNA gyrase the secondary target of microb Agents Chemother 43: 1340–1346. fluoroquinolones in Staphylococcus aureus. Antimicrob Luo N, Sahin O, Lin J, Michel LO, Zhang QJ. 2003. In vivo Agents Chemother 40: 1881–1888. selection of Campylobacter isolates with high levels of Nikaido H, Takatsuka Y. 2009. Mechanisms of RND multi- fluoroquinolone resistance associated with gyrA muta- drug efflux pumps. Biochim Biophys Acta 1794: 769–781.

18 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

Nikaido H, Nikaido K, Harayama S. 1991. Identification Poole K, Gotoh N, Tsujimoto H, Zhao QX, Wada A, Yama- and characterization of porins in Pseudomonas aerugi- saki T, Neshat S, Yamagishi JI, Li XZ, Nishino T. 1996a. nosa. J Biol Chem 266: 770–779. Overexpression of the mexC–mexD–oprJ efflux operon Ogbolu DO, Daini OA, Ogunledun A, Alli AO, WebberMA. in nfxB- type multidrug-resistant strains of Pseudomonas 2011. High levels of multidrug resistance in clinical iso- aeruginosa. Mol Microbiol 21: 713–724. lates of Gram-negative pathogens from Nigeria. Int J Poole K, Tetro K, Zhao QX, Neshat S, Heinrichs DE, Bianco Antimicrob Agents 37: 62–66. N. 1996b. Expression of the multidrug resistance operon Ohki R, Murata M. 1997. bmr3, a third multidrug transport- mexA–mexB–oprM in Pseudomonas aeruginosa: mexR er gene of Bacillus subtilis. J Bacteriol 179: 1423–1427. encodes a regulator of operon expression. Antimicrob Agents Chemother 40: 2021–2028. Oteo J, Cuevas O, Lo´pez-Rodrı´guez I, Banderas-Florido A, Pradel E, Page`s JM. 2002. The AcrAB-TolC efflux pump Vindel A, Pe´rez-Va´zquez M, Bautista V, Arroyo M, Gar- contributes to multidrug resistance in the nosocomial cı´a-Caballero J, Marin-Casanova P, et al. 2009. Emer- Enterobacter aerogenes. Antimicrob Agents Che- gence of CTX-M-15-producing Klebsiella pneumoniae mother 46: 2640–2643. of multilocus sequence types 1, 11, 14, 17, 20, 35 and 36 as pathogens and colonizers in newborns and adults. Putman M, Van Veen HW, Konings WN. 2000. Molecular J Antimicrob Chemother 64: 524–528. properties of bacterial multidrug transporters. Microbiol Mol Biol Rev 64: 672–693. Owens RC Jr, Ambrose PG. 2000. Clinical use of the fluo- Quiroga MP,Orman B, Errecalde L, Kaufman S, Centron D. roquinolones. Med Clin N Am 84: 1447–1469. 2015. Characterization of Tn6238 with a new allele of Pan XS, Fisher LM. 1997. Targeting of DNA gyrase in Strep- aac(60)-Ib-cr. Antimicrob Agents Chemother 59: 2893– tococcus pneumoniae by sparfloxacin: Selective targeting 2897. of gyrase or topoisomerase IV by quinolones. Antimicrob Ribeiro TG, Novais A, Branquinho R, Machado E, Peixe L. Agents Chemother 41: 471–474. 2015. Phylogeny and comparative genomics unveil inde- Pan XS, Fisher LM. 1998. DNA gyrase and topoisomerase IV pendent diversification trajectories of qnrB and genetic are dual targets of clinafloxacin action in Streptococcus platforms within particular Citrobacter species. Antimi- pneumoniae. Antimicrob Agents Chemother 42: 2810– crob Agents Chemother 59: 5951–5958. 2816. Robicsek A, Strahilevitz J, Jacoby GA, Macielag M, Abbanat Pan XS, Fisher LM. 1999. Streptococcus pneumoniae DNA D, Park CH, Bush K, Hooper DC. 2006. Fluoroquino- gyrase and topoisomerase IV: Overexpression, purifica- lone-modifying enzyme: A new adaptation of a common tion, and differential inhibition by fluoroquinolones. aminoglycoside acetyltransferase. Nat Med 12: 83–88. Antimicrob Agents Chemother 43: 1129–1136. Rodrı´guez-Martı´nez JM, Velasco C, Garcı´a I, Cano ME, Pe´richon B, Courvalin P, Galimand M. 2007. Transferable Martı´nez-Martı´nez L, Pascual A. 2007. Mutant preven- resistance to aminoglycosides by methylation of G1405 in tion concentrations of fluoroquinolones for Entero- 16S rRNA and to hydrophilic fluoroquinolones by QepA- bacteriaceae expressing the plasmid-carried quinolone mediated efflux in Escherichia coli. Antimicrob Agents resistance determinant qnrA1. Antimicrob Agents Chemo- Chemother 51: 2464–2469. ther 51: 2236–2239. Pica˜o RC, Poirel L, Demarta A, Silva CS, Corvaglia AR, Rodrı´guez-Martı´nez JM, Pichardo C, Garcı´a I, Pacho´n-Iba- Petrini O, Nordmann P. 2008. Plasmid-mediated quino- n˜ez ME, Docobo-Pe´rez F, Pascual A, Pacho´n J, Martı´nez- lone resistance in Aeromonas allosaccharophila recovered Martı´nez L. 2008a. Activity of ciprofloxacin and levoflox- from a Swiss lake. J Antimicrob Chemother 62: 948–950. acin in experimental caused by Klebsiella pneumoniae deficient in porins, expressing active efflux Piddock LJ, Walters RN, Diver JM. 1990. Correlation of and producing QnrA1. Clin Microbiol Infect 14: 691–697.

www.perspectivesinmedicine.org quinolone MIC and inhibition of DNA, RNA, and pro- tein synthesis and induction of the SOS response in Es- Rodrı´guez-Martı´nez JM, Velasco C, Briales A, Garcı´aI,Co- cherichia coli. Antimicrob Agents Chemother 34: 2331– nejo MC, Pascual A. 2008b. Qnr-like pentapeptide repeat 2336. proteins in Gram-positive bacteria. J Antimicrob Chemo- ther 61: 1240–1243. Poelarends GJ, Mazurkiewicz P, Putman M, Cool RH, Veen ´ ´ ´ HW, Konings WN. 2000. An ABC-type multidrug trans- Rodrıguez-Martınez JM, Dıaz de Alba P,Briales A, Machuca ´ ´ ˜ porter of Lactococcus lactis possesses an exceptionally J, Lossa M, Fernandez-Cuenca F, Rodrıguez Bano J, Mar- tı´nez-Martı´nez L, Pascual A. 2013. Contribution of broad substrate specificity. Drug Resist Updat 3: 330–334. OqxAB efflux pumps to quinolone resistance in extend- Poirel L, Liard A, Rodriguez-Martinez JM, Nordmann P. ed-spectrum-b-lactamase-producing Klebsiella pneumo- 2005a. Vibrionaceae as a possible source of Qnr-like niae. J Antimicrob Chemother 68: 68–73. quinolone resistance determinants. J Antimicrob Chemo- Rosenberg EY, Bertenthal D, Nilles ML, Bertrand KP, Ni- ther 56: 1118–1121. kaido H. 2003. Bile salts and fatty acids induce the ex- Poirel L, Rodriguez-Martinez JM, Mammeri H, Liard A, pression of Escherichia coli AcrAB multidrug efflux pump Nordmann P. 2005b. Origin of plasmid-mediated quin- through their interaction with Rob regulatory protein. olone resistance determinant QnrA. Antimicrob Agents Mol Microbiol 48: 1609–1619. Chemother 49: 3523–3525. Sabtcheva S, Kaku M, Saga T, Ishii Y, Kantardjiev T. 2009. Poole K, Krebes K, McNally C, Neshat S. 1993. Multiple High prevalence of the aac(60)-Ib-cr gene and its dissem- antibiotic resistance in Pseudomonas aeruginosa:Evi- ination among Enterobacteriaceae isolates by CTX-M-15 dence for involvement of an efflux operon. J Bacteriol plasmids in Bulgaria. Antimicrob Agents Chemother 53: 175: 7363–7372. 335–336.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 19 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

D.C. Hooper and G.A. Jacoby

Saga T, Sabtcheva S, Mitsutake K, Ishii Y, Tateda K, Yama- Truong-Bolduc QC, Strahilevitz J, Hooper DC. 2006. NorC, guchi K, Kaku M. 2013. Characterization of qnrB-like a new efflux pump regulated by MgrA of Staphylococcus genes in Citrobacter species of the American type culture aureus. Antimicrob Agents Chemother 50: 1104–1107. collection. Antimicrob Agents Chemother 57: 2863–2866. Truong-Bolduc QC, Ding Y,Hooper DC. 2008. Posttransla- Sa´nchez MB, Herna´ndez A, Rodrı´guez-Martı´nez JM, Mar- tional modification influences the effects of MgrA on tı´nez-Martı´nez L, Martı´nez JL. 2008. Predictive analysis norA expression in Staphylococcus aureus. J Bacteriol of transmissible quinolone resistance indicates Stenotro- 190: 7375–7381. phomonas maltophilia as a potential source of a novel Truong-Bolduc QC, Bolduc GR, Okumura R, Celino B, family of Qnr determinants. BMC Microbiol 8: 148–161. Bevis J, Liao CH, Hooper DC. 2011a. Implication of Schindler BD, Patel D, Seo SM, Kaatz GW. 2013. Mutagen- the NorB efflux pump in the adaptation of Staphylococcus esis and modeling to predict structural and functional aureus to growth at acid pH and in resistance to moxi- characteristics of the Staphylococcus aureus MepA multi- floxacin. Antimicrob Agents Chemother 55: 3214–3219. drug efflux pump. J Bacteriol 195: 523–533. Truong-Bolduc QC, Dunman PM, Eidem T, Hooper DC. Schindler BD, Frempong-Manso E, DeMarco CE, Kosmidis 2011b. Transcriptional profiling analysis of the global C, Matta V, Seo SM, Kaatz GW. 2015. Analyses of multi- regulator NorG, a GntR-like protein of Staphylococcus drug efflux pump-like proteins encoded on the Staph- aureus. J Bacteriol 193: 6207–6214. ylococcus aureus chromosome. Antimicrob Agents Chemo- Truong-Bolduc QC, Liao C-H, Villet R, Bolduc GR, Esta- ther 59: 747–748. brooks Z, Taguezem GF,Hooper DC. 2012. Reduced aer- Schmitz FJ, Jones ME, Hofmann B, Hansen B, Scheuring S, ation affects the expression of the NorB efflux pump of Lu¨ckefahr MFA, Verhoef J, Hadding U, Heinz HP,Ko¨hrer Staphylococcus aureus by posttranslational modification K. 1998. Characterization of grlA, grlB, gyrA, and gyrB of MgrA. J Bacteriol 194: 1823–1834. mutations in 116 unrelated isolates of Staphylococcus au- Tsukamura M, Nakamura E, Yoshii S, Amano H. 1985. reus and effects of mutations on ciprofloxacin MIC. Anti- Therapeutic effect of a new antibacterial substance oflox- microb Agents Chemother 42: 1249–1252. acin (DL8280) on pulmonary tuberculosis. Am Rev Resp Shen LL, Kohlbrenner WE, Weigl D, Baranowski J. 1989. Dis 131: 352–356. Mechanism of quinolone inhibition of DNA gyrase. Ap- Ubukata K, Itoh-YamashitaN, Konno M. 1989. Cloning and pearance of unique norfloxacin binding sites in enzyme- expression of the norA gene for fluoroquinolone resis- DNA complexes. J Biol Chem 264: 2973–2978. tance in Staphylococcus aureus . Antimicrob Agents Che- mother 33: 1535–1539. Sorensen AH, Hansen LH, Johannesen E, Sorensen SJ. 2003. Conjugative plasmid conferring resistance to olaquindox. Vetting MW, Park CH, Hegde SS, Jacoby GA, Hooper DC, Antimicrob Agents Chemother 47: 798–799. Blanchard JS. 2008. Mechanistic and structural analysis of aminoglycoside N-acetyltransferase AAC(60)-Ib and Strahilevitz J, Hooper DC. 2005. Dual targeting of topo- its bifunctional fluoroquinolone-active AAC(60)-Ib-cr isomerase IV and gyrase to reduce mutant selection: Di- variant. Biochemistry 47: 9825–9835. rect testing of the paradigm by using WCK-1734, a new fluoroquinolone, and ciprofloxacin. Antimicrob Agents Vetting MW, Hegde SS, Wang M, Jacoby GA, Hooper DC, Chemother 49: 1949–1956. Blanchard JS. 2011. Structure of QnrB1, a plasmid-me- diated fluoroquinolone resistance factor. J Biol Chem 286: Tavio MM, Jacoby GA, Hooper DC. 2014. QnrS1 structure– 25265–25273. activity relationships. J Antimicrob Chemother 69: 2102– ´ 2109. Vinue L, Corcoran MA, Hooper DC, Jacoby GA. 2015. Mu- tations that enhance the ciprofloxacin resistance of Es- Tran JH, Jacoby GA. 2002. Mechanism of plasmid-mediated cherichia coli with qnrA1. Antimicrob Agents Chemother quinolone resistance. Proc Natl Acad Sci 99: 5638–5642. www.perspectivesinmedicine.org 60: 1537–1545. Tran JH, Jacoby GA, Hooper DC. 2005. Interaction of the Wang JC. 1996. DNA topoisomerases. Annu Rev Biochem plasmid-encoded quinolone resistance protein Qnr with 65: 635–692. Escherichia coli DNA gyrase. Antimicrob Agents Chemo- Wang M, Guo Q, Xu X, Wang X, Ye X, Wu S, Hooper DC, ther 49: 118–125. Wang M. 2009. New plasmid-mediated quinolone resis- Trucksis M, Wolfson JS, Hooper DC. 1991. A novel locus tance gene, qnrC, found in a clinical isolate of Proteus conferring fluoroquinolone resistance in Staphylococcus mirabilis. Antimicrob Agents Chemother 53: 1892–1897. aureus. J Bacteriol 173: 5854–5860. Weigel LM, Anderson GJ, Facklam RR, Tenover FC. 2001. Truong-Bolduc QC, Hooper DC. 2007. Transcriptional reg- Genetic analyses of mutations contributing to fluoro- ulators NorG and MgrA modulate resistance to both quinolone resistance in clinical isolates of Streptococcus quinolones and b-lactams in Staphylococcus aureus. J pneumoniae. Antimicrob Agents Chemother 45: 3517– Bacteriol 189: 2996–3005. 3523. Truong-Bolduc QC, Hooper DC. 2010. Phosphorylation of WentzellLM, Maxwell A. 2000. The complex of DNA gyrase MgrA and its effect on expression of the NorA and NorB and quinolone drugs on DNA forms a barrier to the T7 efflux pumps of Staphylococcus aureus. J Bacteriol 192: DNA polymerase replication complex. J Mol Biol 304: 2525–2534. 779–791. Truong-Bolduc QC, Dunman PM, Strahilevitz J, Projan SJ, Westfall LW,Carty NL, Layland N, Kuan P,Colmer-Hamood Hooper DC. 2005. MgrA is a multiple regulator of two JA, Hamood AN. 2006. mvaT mutation modifies the ex- new efflux pumps in Staphylococcus aureus. J Bacteriol pression of the Pseudomonas aeruginosa multidrug efflux 187: 2395–2405. operon mexEF-oprN. FEMS Microbiol Lett 255: 247–254.

20 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Fluoroquinolone Mechanisms of Action and Resistance

Willmott CJ, Maxwell A. 1993. A single point mutation in ic resistance and biofilm formation in Acinetobacter bau- the DNA gyrase A protein greatly reduces binding of mannii. MBio 6: e00309–e00315. fluoroquinolones to the gyrase–DNA complex. Antimi- Yoshida H, Bogaki M, Nakamura M, Nakamura S. 1990. crob Agents Chemother 37: 126–127. Quinolone resistance-determining region in the DNA Willmott CJ, Critchlow SE, Eperon IC, Maxwell A. 1994. gyrase gyrA gene of Escherichia coli. Antimicrob Agents The complex of DNA gyrase and quinolone drugs with Chemother 34: 1271–1272. DNA forms a barrier to transcription by RNA polymer- Yoshida H, Bogaki M, Nakamura M, Yamanaka LM, Naka- ase. J Mol Biol 242: 351–363. mura S. 1991. Quinolone resistance-determining region Wohlkonig A, Chan PF, Fosberry AP, Homes P, Huang J, in the DNA gyrase gyrB gene of Escherichia coli. Antimi- Kranz M, Leydon VR, Miles TJ, Pearson ND, Perera RL, crob Agents Chemother 35: 1647–1650. et al. 2010. Structural basis of quinolone inhibition of Yoshida H, Nakamura M, Bogaki M, Ito H, Kojima T, Hat- type IIA topoisomerases and target-mediated resistance. tori H, Nakamura S. 1993. Mechanism of action of quin- Nat Struct Mol Biol 17: 1152–1153. olones against Escherichia coli DNA gyrase. Antimicrob Wong MH, Chan EW, Chen S. 2015. Evolution and dis- Agents Chemother 37: 839–845. semination of OqxAB-like efflux pumps, an emerging Yu JL, Grinius L, Hooper DC. 2002. NorA functions as a quinolone resistance determinant among members of multidrug efflux protein in both cytoplasmic membrane Enterobacteriaceae. Antimicrob Agents Chemother 59: vesicles and reconstituted proteoliposomes. J Bacteriol 3290–3297. 184: 1370–1377. Xiong X, Bromley EH, Oelschlaeger P,Woolfson DN, Spen- Yu EW, Aires JR, Nikaido H. 2003. AcrB multidrug efflux cer J. 2011. Structural insights into quinolone antibiotic pump of Escherichia coli: Composite substrate-binding resistance mediated by pentapeptide repeat proteins: cavity of exceptional flexibility generates its extremely Conserved surface loops direct the activity of a Qnr pro- wide substrate specificity. J Bacteriol 185: 5657–5664. tein from a Gram-negative bacterium. Nucleic Acids Res Yu EW, Aires JR, McDermott G, Nikaido H. 2005. A peri- 39: 3917–3927. plasmic drug-binding site of the AcrB multidrug efflux Yamada Y, Hideka K, Shiota S, Kuroda T, Tsuchiya T. 2006. pump: A crystallographic and site-directed mutagenesis Gene cloning and characterization of SdrM, a chromo- study. J Bacteriol 187: 6804–6815. somally encoded multidrug efflux pump, from Staphylo- Zhang L, Li XZ, Poole K. 2001. SmeDEF multidrug efflux coccus aureus. Biol Pharm Bull 29: 554–556. pump contributes to intrinsic multidrug resistance in Yamane K, Wachino J, Suzuki S, Kimura K, Shibata N, Kato Stenotrophomonas maltophilia. Antimicrob Agents Che- H, Shibayama K, Konda T, Arakawa Y. 2007. New plas- mother 45: 3497–3503. mid-mediated fluoroquinolone efflux pump, QepA, Zhang S, Sun J, Liao XP, Hu QJ, Liu BT, Fang LX, Deng H, found in an Escherichia coli clinical isolate. Antimicrob Ma J, Xiao X, Zhu HQ, et al. 2013. Prevalence and Agents Chemother 51: 3354–3360. plasmid characterization of the qnrD determinant in En- Yang S, Clayton SR, Zechiedrich EL. 2003. Relative contri- terobacteriaceae isolated from animals, retail meat prod- butions of the AcrAB, MdfA and NorE efflux pumps to ucts, and humans. Microb Drug Resist 19: 331–335. quinolone resistance in Escherichia coli. J Antimicrob Zhao J, Chen Z, Chen S, Deng Y,Liu Y,Tian W,Huang X, Wu Chemother 51: 545–556. C, Sun Y, Zeng Z, et al. 2010. Prevalence and dissemina- Yoon EJ, Courvalin P,Grillot-Courvalin C. 2013. RND-type tion of oqxAB in Escherichia coli Isolates from animals, efflux pumps in multidrug-resistant clinical isolates of farmworkers, and the environment. Antimicrob Agents Acinetobacter baumannii: Major role for AdeABC over- Chemother 54: 4219–4224. expression and AdeRS mutations. Antimicrob Agents Zhao L, Zhang J, Zheng B, Wei Z, Shen P,Li S, Li L, Xiao Y.

www.perspectivesinmedicine.org Chemother 57: 2989–2995. 2015. Molecular epidemiology and genetic diversity of Yoon EJ, Chabane YN, Goussard S, Snesrud E, Courvalin P, fluoroquinolone-resistant Escherichia coli isolates from De E, Grillot-Courvalin C. 2015. Contribution of resis- patients with community-onset infections in 30 Chinese tance-nodulation-cell division efflux systems to antibiot- county hospitals. J Clin Microbiol 53: 766–770.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025320 21 Downloaded from http://perspectivesinmedicine.cshlp.org/ on October 1, 2021 - Published by Cold Spring Harbor Laboratory Press

Topoisomerase Inhibitors: Fluoroquinolone Mechanisms of Action and Resistance

David C. Hooper and George A. Jacoby

Cold Spring Harb Perspect Med 2016; doi: 10.1101/cshperspect.a025320 originally published online July 22, 2016

Subject Collection Antibiotics and Antibiotic Resistance

Fosfomycin: Mechanism and Resistance The Whys and Wherefores of Antibiotic Lynn L. Silver Resistance Cameron R. Strachan and Julian Davies Pleuromutilins: Potent Drugs for Resistant Bugs β-Lactamases: A Focus on Current Challenges −−Mode of Action and Resistance Robert A. Bonomo Susanne Paukner and Rosemarie Riedl Appropriate Targets for Antibacterial Drugs Approved Glycopeptide Antibacterial Drugs: Lynn L. Silver Mechanism of Action and Resistance Daina Zeng, Dmitri Debabov, Theresa L. Hartsell, et al. Lincosamides, Streptogramins, Phenicols, and Mechanism of Action and Resistance to Pleuromutilins: Mode of Action and Mechanisms Daptomycin in Staphylococcus aureus and of Resistance Enterococci Stefan Schwarz, Jianzhong Shen, Kristina Kadlec, William R. Miller, Arnold S. Bayer and Cesar A. et al. Arias Resistance to Antibiotics in Public Polymyxin: Alternative Mechanisms of Action and Health Pathogens Resistance Corey Fyfe, Trudy H. Grossman, Kathy Kerstein, et Michael J. Trimble, Patrik Mlynárcik, Milan Kolár, et al. al. Bacterial Protein Synthesis as a Target for Topoisomerase Inhibitors: Fluoroquinolone Antibiotic Inhibition Mechanisms of Action and Resistance Stefan Arenz and Daniel N. Wilson David C. Hooper and George A. Jacoby Antibacterial : From Development β-Lactams and β-Lactamase Inhibitors: An through Resistance to the Next Generation Overview Alexavier Estrada, Dennis L. Wright and Amy C. Karen Bush and Patricia A. Bradford Anderson Antibacterial Drug Discovery Targeting the , Alone and in Combination Lipopolysaccharide Biosynthetic Enzyme LpxC David M. Rothstein Alice L. Erwin

For additional articles in this collection, see http://perspectivesinmedicine.cshlp.org/cgi/collection/

Copyright © 2016 Cold Spring Harbor Laboratory Press; all rights reserved