Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Drug Resistance Updates 15 (2012) 162–172

Contents lists available at SciVerse ScienceDirect

Drug Resistance Updates


journal homepage: www.elsevier.com/locate/drup

Evolution of antibiotic resistance at non-lethal drug concentrations


Dan I. Andersson ∗ , Diarmaid Hughes
Department of Medical Biochemistry and Microbiology, Box 582, SE-75123 Uppsala, Sweden

a r t i c l e i n f o a b s t r a c t

Article history: Human use of antimicrobials in the clinic, community and agricultural systems has driven selection for
Received 28 January 2012 resistance in bacteria. Resistance can be selected at antibiotic concentrations that are either lethal or
Received in revised form 22 March 2012 non-lethal, and here we argue that selection and enrichment for antibiotic resistant bacteria is often a
Accepted 26 March 2012
consequence of weak, non-lethal selective pressures – caused by low levels of antibiotics – that operates
We would like to dedicate this paper to on small differences in relative bacterial fitness. Such conditions may occur during antibiotic therapy or
Prof. Fernando Baquero, who has pioneered in anthropogenically drug-polluted natural environments. Non-lethal selection increases rates of mutant
ideas about the importance of low levels of appearance and promotes enrichment of highly fit mutants and stable mutators.
antibiotics for antibiotic resistance © 2012 Elsevier Ltd. All rights reserved.
development.

Keywords:
Antibiotic resistance
Biocide
Minimal inhibitory concentration
Selective window

1. Antibiotic resistance is an ecological problem environment and bacterial species (e.g. in E. coli bacteria present
in urine/feces in antibiotic-treated individuals) but for many other
The extensive human use and misuse of antimicrobials in the types of resistances, in particular for those that are horizontally
clinic, community, animal husbandry and agriculture has resulted transferred, these three levels might be fully separated tempo-
in strong selection pressures for the emergence, enrichment and rally and spatially. Thus, the resistance mechanism could have
spread of various resistance mechanisms in pathogenic bacteria. originated pre-therapeutic use of antibiotics in non-pathogenic
The temporal and spatial dynamics and driving forces of these environmental bacteria, and recent evidence strongly supports the
processes are complex and we are only slowly beginning to under- notion that many types of resistance mechanisms and resistant bac-
stand how resistance genes, resistant bacteria and the selective teria were present long before human production, use and spread of
agents (antibiotics, biocides, environmental pollutants, etc.) move antibiotics expanded rapidly in the second half of the 1900s (Smith,
between different ecosystems and exert their effect. In this review, 1967; Hughes and Datta, 1983; Aminov and Mackie, 2007; Mindlin
resistance is used in the broadest sense to indicate any reduction et al., 2008; Allen et al., 2009; D’Costa et al., 2011). The emergence of
in susceptibility in comparison to the wild type population. resistant pathogens could have followed later when the resistance
With regard to the dynamics of antibiotic resistance develop- was horizontally transferred from an environmental bacterium to
ment it is essential to distinguish between (at least) three levels a human/animal pathogen (Patel et al., 2000; Poirel et al., 2002,
of description, namely (i) where did the resistance mechanism 2005; Hong et al., 2004; Canton, 2009; Wright, 2010), possibly
itself originate, (ii) where did the resistant pathogenic bacteria in an environment where these bacteria only transiently resided
of relevance to human and animal infections emerge and (iii) together. Finally, these resistant human pathogens were enriched
where were these resistant pathogens enriched and transmitted? due to selective pressures present in the clinic, community or agri-
In cases where the resistance is conferred by a mutational mecha- cultural setting. Often it is difficult to distinguish between these
nism these three levels often converge on one particular bacterial levels but from the point of view of trying to restrict the emergence
species and environment. For example, for Escherichia coli resistant and spread of resistance it is important that we attempt to do so
to fluoroquinolones the de novo emergence of the resistance mech- since the design and efficiency of our strategies will vary consider-
anism and the subsequent enrichment might occur in the same ably depending on whether we try to prevent the initial emergence
of resistance, or try to prevent the enrichment of already existing
resistant pathogens that have spread into clinical/community set-
∗ Corresponding author. Tel.: +46 18 4714175. tings. For example, as suggested by increasing amounts of data,
E-mail address: Dan.Andersson@imbim.uu.se (D.I. Andersson). clinically relevant plasmid/ICE mediated resistances may emerge

1368-7646/$ – see front matter © 2012 Elsevier Ltd. All rights reserved.
http://dx.doi.org/10.1016/j.drup.2012.03.005
D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172 163

Fig. 1. Schematic representation of flows of resistant bacteria (blue) and antibiotics (red) between different environments and areas of use.

by HGT from environmental bacteria to pathogens in the out- various aquatic environments and soil and exert a weaker selective
side environment (e.g. in waste water treatment facilities, manure, pressure. Even though antibiotic concentrations in these environ-
sludge, soils or similar environments) (Patel et al., 2000; Poirel et al., ments are several orders lower than where the drugs were initially
2002, 2005; Hong et al., 2004; Canton, 2009; Wright, 2010; Taylor used they could still, as argued below, be of great importance for
et al., 2011). With this knowledge in hand, an important control the evolution of resistance.
measure to reduce the rate of resistance emergence might be active
stewardship in environmental resistance reservoirs (e.g. killing of
2. Which are the significant selectors of resistance?
bacteria and destruction/removal of antibiotic pollutants in waste
water, sludge, manure, etc.) in addition to clinical control programs
With regard to the selective pressures that enrich for resistance,
that are aimed at reducing antibiotic use and selective pressures
it is probable that antibiotics represent the main selector. Thus,
near the patients. Conceivably it might be more efficient to prevent
there exist convincing correlations between amount of antibiotic
the initial emergence of resistant pathogens rather than to later
used at the hospital, community and country level and the fre-
try to control their enrichment and spread in clinical and human
quency of resistance observed (van de Sande-Bruinsma et al., 2008;
community settings by restrictive antibiotic use.
Bergman et al., 2009; Goossens, 2009). However, it is likely that
As outlined schematically in Fig. 1, resistance genes, resistant
other types of selective pressures are involved as well. For example,
bacteria and antibiotics will flow between different compartments
the amount (in metric tons) of human-made biocides that bacteria
and environments, including humans, animals, soil and water,
are exposed to on a global scale is many orders of magnitude higher
etc., creating situations where resistant bacteria may emerge,
than that for antibiotics (SCENIHR, 2009). As the increasing use of
become enriched and spread between hosts and various natural
antibiotics and biocides has been temporally and spatially linked it
environments. Resistance might be selected in any of the above
is difficult to entangle their relative contributions to the resistance
environments but it is difficult to assess the relative contributions
problem but based on the wide use of biocides in numerous types
of various environments and selections to the total problem. For
of environments (for example, in health care, household prod-
example, it is commonly assumed that hospitals represent signifi-
ucts, textiles, food industry and many other environments) and
cant foci for resistance evolution because of the high use (per capita)
fundamental evolutionary principles, it is expected that biocides
of antibiotics that generate a strong local selection pressure, and the
will exacerbate the problem (Maillard, 2007; Tumah, 2009). Bio-
high host population density that facilitates spread of the resistant
cides could contribute to the problem by co-selection mechanisms
bacteria. This notion is most likely true for certain pathogens but
where the biocide resistance conferring genes (e.g. qac, heavy metal
with regard to the total selective pressure asserted globally due
resistance genes, etc.) are present on the same genetic element
to anthropogenic influences, hospital use of antibiotics probably
(e.g. a plasmid or ICE) as the antibiotic resistance genes, thereby
represents only a few percent of the total volume of antibiotic use
and it is conceivable that the weaker selective pressures present
in other environments are equally important breeding grounds for Table 1
the emergence of resistance (Baquero, 2001a,b), and possibly high Fraction of antibiotic dose for different antibiotic classes that is excreted in active
antibiotic-use environments like hospitals mostly act as enrich- form in human urine (Bryskier, 2005). The fractions will vary between different
compounds from the same antibiotic class and these values should therefore be
ment and transmission centers of already existing resistant clones. taken as approximate.
It is also notable that antibiotics exert their effect with widely
different active concentrations during their movement through Antibiotic class Fraction of dose excreted in
human urine in active form
various environments and ecosystems. When antibiotics are used
therapeutically in humans, animals and agriculture they generally Fluoroquinolones 40%
Aminoglycosides 80–90%
are present at high local concentrations and exert strong selection
Tetracycline 40%
pressures in these environments. However, about half of all antibi- Macrolides 20–30%
otics given to humans and animals are excreted in an unchanged Penicillins 50%
active form mainly via urine (Table 1) into waste water, manure and Cephalosporins 70–90%
run-off water, where they will ultimately end up in a diluted form in Trimetoprim 50%
164 D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172

indirectly driving enrichment for antibiotic resistance (Laraki et al., we must determine the lowest concentrations of different antibi-
1999; Bjorland et al., 2001; Sidhu et al., 2001, 2002; Cookson, 2005; otics that selectively enrich for resistant variants. A complication
Noguchi et al., 2005; Sekiguchi et al., 2005, 2007; Dumitrescu et al., is that the interplay between the relative antibiotic susceptibility
2007; Levings et al., 2007). Another mechanism might be by cross- and growth fitness of different bacterial variants will determine
resistance where the biocide and the antibiotic share a common how each is selectively enriched at different drug concentrations.
resistance mechanism, as has been shown for up-regulation of At an antibiotic concentration that we term the minimum selective
efflux pumps (Paulsen et al., 1996; Brown et al., 1999; Putman et al., concentration (MSC) the growth rate of the susceptible strain will
2000; Borges-Walmsley and Walmsley, 2001; Poole, 2001, 2002; be reduced to a level where it equals the growth rate of the resis-
Levy, 2002; McKeegan et al., 2003; Piddock, 2006) alterations of tant strain, and by definition any antibiotic concentration higher
the cell wall (Denyer and Maillard, 2002; Nikaido, 2003; Tkachenko than the MSC will selectively enrich the resistant strain. Thus,
et al., 2007) and modification of target enzymes (McMurry et al., paradoxically, relative to a particular susceptible strain, two dif-
1999; Heath et al., 2000; Parikh et al., 2000). Several studies have ferent resistant mutants with identical MICs but different fitness
shown that cross-resistance to antibiotics can result from selection values would have different MSCs. MSC will be lowest for strains
for biocide resistance in the laboratory, strongly suggesting that with the smallest fitness costs. Experiments have shown that in
biocides in natural settings would have similar effects (Pomposiello general resistant strains have a lower fitness in drug-free envi-
et al., 2001; Braoudaki and Hilton, 2004; Codling et al., 2004; ronments than susceptible strains (Bjorkman et al., 1998; Nagaev
Sanchez et al., 2005; Karatzas et al., 2007; Randall et al., 2007; Fraud et al., 2001), but the range of fitness values can extend up to a level
et al., 2008; Huet et al., 2008). where it can prove very difficult to distinguish it experimentally
from that of an isogenic susceptible strain (Marcusson et al., 2009;
Andersson and Hughes, 2010). In addition, the relative fitness dif-
3. Where are resistant bacteria selected?
ference between isogenic resistant and susceptible strains is not
a constant but instead changes continuously as a function of the
It is commonly assumed that resistant bacteria are selected in
antibiotic concentration to which they are exposed. The question
antibiotic-treated humans or animals in response to the strong
boils down to how small a selective effect can be, and still influence
selective pressures that are generated when antibiotics are used
the outcome of a competition?
therapeutically and the concentrations reached are well above the
We argue here that extremely small differences in relative fit-
minimal inhibitory concentration (MIC) of the susceptible popu-
ness can be effectively selected in nature. An extreme example that
lation of bacterial cells. This notion is certainly true for certain
supports this argument is the widespread evolution of the pref-
types of infections, antibiotic regimens and resistance mechanisms.
erential use of some synonymous codons, so-called codon usage
For example, in Mycobacterium tuberculosis examples exist where
bias, in many free-living microorganisms. The particular set of
mutationally resistant bacilli were selected de novo from an ini-
synonymous codons that are preferentially used differs between
tially susceptible population during inadequate treatment regimes
microorganisms, showing that codon usage bias evolved indepen-
(Burman et al., 2006; Cox et al., 2007; Hu et al., 2011; Mariam
dently in different genetic lineages. Codon usage bias is thought
et al., 2011; Skrahina et al., 2012). Similarly, mutational resistance
to be the result of selection for translationally optimal codons
to fluoroquinolones has been shown to emerge during treatment of,
(Ikemura, 1985; Sharp et al., 2010). Based on this model the growth
for example, E. coli/Salmonella enterica (Cattoir et al., 2006; Corvec
advantage and selection coefficient for an optimal versus a non-
et al., 2008; de Toro et al., 2010; Jeong et al., 2011) as well as fluoro-
optimal codon has been calculated to be in the range of 10−5
quinolone and ␤-lactam resistance in Pseudomonas aeruginosa (Le
(Bulmer, 1991) to 10−8 (Sharp et al., 2010) per codon. The wide
Thomas et al., 2001; Leotard et al., 2001; Nakano et al., 2001). Other
range in these values reflects different assumptions of effective
examples where resistance emerged de novo in treated patients
population size (Ne ) for bacterial species like E. coli (Berg, 1996)
as a result of horizontal gene transfer of resistance genes include
but the major conclusion from the widespread occurrence of bias
the appearance of vancomycin resistant MRSA (Chang et al., 2003;
in synonymous codon usage is that miniscule differences in fitness
Weigel et al., 2003) and carbapenem resistant E. coli (Richter et al.,
(≤10−5 ) can be effectively selected over evolutionary time scales.
2011) where the resistant donor bacteria were either co-infecting
This example provides strong support for the idea that even an
with resistant Enterococcus faecalis or Klebsiella pneumoniae.
extremely weak selective advantage can drive the genome-wide
On the other hand, many types of plasmid/ICE-mediated resis-
evolution of DNA sequences in bacteria.
tances (which represent the largest proportion of all resistance
A relevant clinical example with respect to antibiotic expo-
mechanisms) are unlikely to emerge de novo during treatment.
sure and selection is the situation that arises each time a patient
Thus, the patient/animal is either infected with the susceptible
is treated with an antibiotic. It is expected that, because of the
bacteria (and no resistance development occurs during treatment)
complexity of the human body, the antibiotic will reach differ-
or they are infected with the resistant strain and the antibi-
ent concentrations in different body compartments, and that these
otic treatment mainly causes an enrichment of a pre-existing
concentrations will change over time (Baquero et al., 1997, 1998).
mutant. As described above, it is likely that for many of the
Thus the exposure of different bacteria in the patient to the antibi-
resistance mechanisms found today in human pathogens, their
otic will vary temporally and spatially. If there are within the
origin was in environmental bacteria and anthropogenic changes
patient sub-populations of less susceptible bacteria these could
of the environment via the use of selective antimicrobials and
then be enriched in particular body compartments and may explain
ecological opportunity for gene transfer allowed for transfer of
for example, the apparent incremental step-wise evolution of ␤-
resistance genes into previously susceptible populations of bacte-
lactamases (Baquero et al., 1998). Similar variation in antibiotic
rial pathogens.
selective pressures is also predicted to occur in the highly struc-
tured environments of soil.
4. The power of small selective differences There are examples where small differences in fitness of antibi-
otic resistant strains have been shown experimentally to be
In Section 1 we argued that the wider environment contains lev- selectable in the absence of the drug both in vitro and in vivo
els of antibiotics and other agents with the potential to enrich for (Marcusson et al., 2009). The concept that small differences in
antibiotic-resistant pathogens. To understand quantitatively how fitness could also be selected in a drug-concentration-dependent
environmental contamination influences resistance development window was tested experimentally by competing isogenic strains
D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172 165

carrying variant ␤-lactamases, TEM-1 and TEM-12 that differed an experiment). Below we discuss different methods of measuring
only slightly in MIC for cefotaxime, 0.008 and 0.012 ␮g/mL, respec- and selecting small differences in fitness and resistance.
tively (Negri et al., 2000). Isogenic strains carrying the variant
TEM genes were competed in vitro (laboratory liquid medium)
and in vivo (mouse thigh model) at a range of cefotaxime con- 5. Detection of selection
centrations. The TEM-12 variant was preferentially enriched in
a selection lasting several hours in a narrow drug concentra- To get a quantitative understanding of the problems posed by
tion range, both in vitro and in vivo, supporting the concept of a antibiotics in the environment we need to determine the lowest
drug-concentration selective window (Negri et al., 2000) in which levels of antibiotics that confer a measurable selective advantage
specific resistant mutants would be enriched. Interestingly, when under experimental conditions. Some standard measures of fitness,
the competition between the two TEM variants was continued for a with a discriminatory power that is at best ±3% per generation, such
longer period of several days and with higher drug concentrations, as growth rate measurements in the presence of different concen-
a variant with a secondary mutation in an outer membrane pro- trations of antibiotic, are too crude to be useful. In addition, such
tein, OmpF, was selected. The mutant selection window hypothesis, measurements quantify only the exponential part of a bacterial
that different drug concentrations will selectively enrich specific growth cycle and may completely miss fitness parameters that are
mutant sub-populations, has also been extensively explored and important in the presence of growth inhibiting antibiotics. A bet-
validated in the context of the multi-step evolution of resistance ter approach is to compete isogenic strains differing only in some
to fluoroquinolones (Dong et al., 1999; Marcusson et al., 2005; resistance determinant in mixed cultures where the competition
Drlica and Zhao, 2007). The TEM-1, TEM-12 experiment described involves the complete growth cycle (Andersson and Hughes, 2010).
above (Negri et al., 2000) provides a model framework that poten- A small difference in relative fitness can be amplified by cycling the
tially explains the step-wise evolution of novel ␤-lactamases in competing mixture of strains, with successive rounds of inoculation
response to changing antibiotic selective pressures (Petrosino et al., into fresh media, over several days. The change in the ratio of one
1998; Salverda et al., 2010). However, because specific spontaneous strain relative to the other can be monitored as a function of time
mutations are generally very infrequent (typical eubacterial muta- and the slope of the line generated can be used to calculate a selec-
tion rates are 10−9 to 10−11 per nucleotide per generation) this tion coefficient (Dean et al., 1988). We have used this method to
particular mode of resistance evolution is expected to be depen- measure relative fitness costs in isogenic strains marked with dif-
dent on large bacterial populations to supply the specific mutations ferent antibiotic resistance markers where the two populations are
required for the selected phenotype. One way this limitation can counted on separate agar plates (Bjorkman et al., 1998). However,
be overcome is that several antibiotics increase mutation rates, at drawbacks include the tedium of counting colonies, the statistical
least within a particular concentration range (Phillips et al., 1987; uncertainty introduced by the relatively small numbers counted,
Ren et al., 1999; Perez-Capilla et al., 2005; Kohanski et al., 2010). and the possibility that growth of colonies on different antibiotic-
Another way in which small bacterial populations can bypass the containing media introduces a systematic error into the ratio. A
mutation-supply bottleneck is by high frequency gene amplifica- better method, which avoids using antibiotics to distinguish strains,
tion events, that alter the phenotype of the selected strain, and is to compete isogenic strains where one is marked with a neutral
simultaneously amplify the genetic target for spontaneous muta- mutation (for example araB), which allows the two populations
tions (Andersson and Hughes, 2009). The potential importance of to be distinguished on the same agar plate based on differential
this mechanism for ␤-lactamase evolution was shown experimen- colony colour (Marcusson et al., 2009). This has the advantage
tally when the TEM-1 gene was evolved in vitro to successively that the competing populations are measured as colonies in the
higher levels of resistance to a cephalosporin (cephalothin). Note same environment but a drawback is that the quantitative range
that TEM-1 is associated with a very small increase in MIC to over which the two populations can be measured is limited in
this antibiotic that provides a weak resistance phenotype that practice to about three orders of magnitude. Each of these com-
can be amplified under selection. It was found that the most petition methods discriminates fitness differences in the range of
frequent response to cephalosporin selection in independent lin- ∼
=1% per generation. An elegant chromogenic assay has been devel-
eages was amplification of the copy number of the TEM-1 gene oped that uses bacterial strains expressing either of the proteins
(Sun et al., 2009). The amplification of the TEM-1 gene by itself amilCP and amilGFP, and has the advantage that it permits a quali-
provided a higher level of resistance (amplifying the very weak tative discrimination between competing strains in liquid medium
cephalosporinase activity of TEM-1) but importantly it also allowed (Liu et al., 2011). Although the method is not designed for accu-
the population of bacteria to survive and expand sufficiently to real- rate quantification of small differences in selection coefficients, it
ize additional rare secondary mutations in the genome, including in may prove useful for assaying environmental contamination by
OmpF (Sun et al., 2009). These experiments illustrate the potential antibiotics.
interplay between the diversity of bacterial genotypes present in Our aim was to measure small differences in selection coeffi-
large populations, the possibility for small populations to undergo cients and so we developed an assay that is both sensitive and
high-frequency gene amplification, and the diversity of selective quantitative (Gullberg et al., 2011). The assay is based on growth
pressures that can be produced along the antibiotic concentration competition between isogenic strains genetically tagged with vari-
gradients formed in human body compartments and other natu- ants of the green fluorescent protein gene (yfp and cfp, encoding
ral environments. These antibiotic gradients will potentially result yellow- and cyan-fluorescent proteins, respectively). The compet-
in differential growth rates of resistant bacterial variants (Baquero ing strains, susceptible and resistant, were competed for up to 80
and Negri, 1997; Baquero et al., 1998; Baquero, 2001a,b), regardless generations by serial passage in batch cultures. After each cycle
of the specific mechanisms by which the variants arise and evolve. of growth (10 generations per cycle) large populations of cells
We can thus safely assume that antibiotic selection, especially (approximately 105 cells) were counted by fluorescent activated
if repeated over many generations, can amplify the effects of cell sorting (FACS), thereby significantly reducing any experimental
extremely small differences in fitness, resulting in an increased fre- errors associated with counting small cell populations. This exper-
quency of resistant variants. In an experimental setting however, a imental set-up allows detection of growth rate differences of 0.2%
practical concern is the resolution that can be achieved in measur- per generation (Lind et al., 2010; Gullberg et al., 2011). This level of
ing small selective differences on a short time scale (if we assume discrimination approaches the limit of sensitivity set by the inter-
that we do not have millions of years at our disposal to complete ference caused by periodic selection events (Atwood et al., 1951;
166 D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172

Koch, 1974; Berg, 1995) that are associated with mutational adap- correlation between the magnitude of the reduced fitness of the
tation to the growth media/conditions unrelated to the antibiotic mutant strains in the absence of antibiotic and the magnitude of
selective pressure. We have also begun using pre-adapted strains the measured MSC. Thus, the fitness deficits were smallest for the
for these assays; i.e. strains grown for at least 1000 generations gyrA Ser83Leu mutation and the Tn10 insertion and this correlated
in the growth medium to be used for competition, before the with them having the smallest MSC values. Because each of the
introduction of the fluorescent and resistance markers. The use of above competition experiments had been made with a 1:1 ratio of
pre-adapted strains permits extended competition runs without wild type and mutant we asked whether this experimental set-up
interference from periodic selection, thus increasing the discrimi- could have influenced the outcome of the competition. To test this
natory power of the assay. This highly sensitive fluorescence-based we repeated a series of competition experiments (with wild-type
assay can be adapted for use in natural environments or in in vivo and isogenic Tn10 mutant) but this time used initial strain ratios of
animal models, and we have recently initiated such experiments. 1:1, 10:1, 100:1, 1000:1 and 10,000:1 (wild type:mutant). The data
In conclusion, we have developed a robust assay that can reliably were collected and analyzed as before and for each of the competi-
and quantitatively measure selective differences of ∼ =10−3 per gen- tions, regardless of the initial ratio, the calculated MSC was 1/100.
eration, in a competition experiment that can be completed in 1–2 This confirms that the measured MSC values are robust and are not
weeks. We have used this experimental system to measure the MSC significantly influenced by the initial ratio of wild type to mutant.
of several different antibiotic classes in relation to several clinically The conclusion from these competition experiments is that in every
relevant antibiotic resistance determinants (Gullberg et al., 2011). case tested the MSC is significantly below the MIC of the susceptible
wild type, and in some cases it is very far below MIC (1/100, 1/230).
Results from an independent study agree qualitatively with those
6. Sub-MIC selection from our study and show that antibiotic concentrations that are sig-
nificantly below the wild-type MIC can selectively enrich resistant
We measured, under controlled laboratory conditions, the MSC mutant populations (Liu et al., 2011). The MSC values measured
for three different classes of antibiotic: ciprofloxacin, a fluoro- in our experiments correspond to absolute antibiotic concentra-
quinolone; streptomycin, an aminoglycoside; and tetracycline, tions of 1 ␮g/ml (streptomycin), 15 ng/ml (tetracycline), and from
using the growth competition method described in the previous 2.5 ng/ml down to 100 pg/ml (ciprofloxacin).
section (Gullberg et al., 2011). The aim was to determine the Having shown that very low concentrations of antibiotic could
lowest antibiotic concentration that discriminated between com- selectively enrich resistant mutants in competition experiments
peting isogenic wild type and mutant strains carrying any one we next asked whether sub-MIC concentrations could also select
of six different resistance determinants that are clinically rele- and enrich de novo mutants. For that experiment, 20 indepen-
vant for resistance to these antibiotics. The mutations tested were: dent lineages each of the S. enterica and E. coli wild-types were
two point mutations affecting the fluoroquinolone target DNA grown in batch cultures in the presence of sub-MIC concentra-
gyrase (gyrA Ser83Leu, and Asp87Asn); two deletion mutations of tions of streptomycin (S. typhimurium, (1/4) MIC, 700 generations)
regulators of drug efflux (marR, and acrR); a point mutation or ciprofloxacin (E. coli, 1/10 MIC, 600 generations). Each lineage
in ribosomal protein S12 that causes resistance to streptomycin was monitored periodically throughout the experiment to quan-
(rpsL105 Lys42Arg); and the presence in the chromosome of a tify the frequency of cells with increased levels of resistance. In
transposon causing resistance to tetracycline (Tn10). We made the each case we observed a significant and continuous increase in the
experiments as cyclic growth competitions in laboratory medium frequency of more resistant sub-populations within the lineages.
using two different bacterial species, E. coli and S. enterica serovar For example, for streptomycin, after 400 generations all 20 lineages
Typhimurium. Each strain (wild-type and isogenic mutants) was had sub-populations (at least 1% frequency) that were 8 times the
constructed in two variant forms. One form was tagged with a cfp wild-type MIC, and after 600 generations 14 lineages had subpop-
gene inserted at the galK locus while the other form was tagged ulations that were 16 times MIC. Similarly for E. coli and sub-MIC
with a yfp gene inserted at the same locus. This allowed us to make selection with ciprofloxacin: after 500 generations five of the lin-
parallel competition experiments in which either the wild type eages had sub-populations (≥1% frequency) with MICs from two-
or the mutant strain was tagged with either of the fluorescence- to six-fold higher than the wild-type (Gullberg et al., 2011). Thus,
producing genes, to ensure that the tagging itself did not alter the sub-MIC concentration of antibiotics can select for both low- and
outcome of the competition. Control experiments showed that the high-level resistance mutants de novo from a susceptible popula-
small difference in fitness costs between the cfp and yfp markers tion. We draw two important conclusions from these evolution
had a negligible impact on growth rates. For the competition exper- experiments. First, sub-MIC antibiotic concentrations can select
iments wild-type and mutant cultures were mixed in a 1:1 ratio resistant mutants de novo from a susceptible bacterial population.
and grown in liquid medium containing different antibiotic con- Second, de novo-selected mutants often had MICs that were much
centrations, as well as in the absence of antibiotic, for up to 80 higher than the selective concentration of antibiotic. Thus, both
generations. After each cycle of growth (10 generations) the ratio high- and low-level antibiotic-resistant mutants could potentially
of resistant to susceptible bacteria in the population was scored be selected by exposure to low levels of antibiotics.
by FACS analysis. Over this time scale (40–80 generations) peri- We also developed a mathematical model to calculate how
odic selection was not a significant problem. The data on changing rapidly a de novo resistance mutation could take over (at least 50%
ratios were then plotted to calculate a selection coefficient for each of the population) a susceptible population under selection at sub-
mutant strain as a function of antibiotic concentration. For all six MIC antibiotic concentrations. The calculations took into account
of the competition experiments the data showed that the MSC of the mutation rate (), the population size (N), and the selective
the mutant strain was significantly below the MIC of the isogenic advantage of the mutant (s) as a function of antibiotic concentra-
wild-type strain. The MSC values ranged from (1/4) MIC (strepto- tion based on the data from the competition experiments (Gullberg
mycin; rpsL105), through 1/10 MIC (ciprofloxacin; gyrA Asp87Asn, et al., 2011). There are two terms that dominate the equations: the
marR, and acrR), to 1/100 MIC (tetracycline; Tn10). The lowest stochastic waiting time for the first mutation to appear, and the
MSC was 1/230 MIC measured for gyrA Ser83Leu (ciprofloxacin), subsequent time for the mutant to grow to 50% of the population.
a mutation that is almost always found in clinical isolates of E. For populations where the mutant is already present (N > 1) the
coli that are fluoroquinolone-resistant (Komp Lindgren et al., 2003; time to fixation can be rapid (100–1000 generations) for s values
Christiansen et al., 2011; Nazir et al., 2011). There was also a between 0.1 and 0.01. For populations where N < 1 (for example
D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172 167

Table 2
Differences in outcome for selection at lethal and non-lethal drug concentrations.

Lethal (>MIC) Non-lethal (<MIC)

Selection for rare mutations of big Selection for common


effects mutations of small
effect → Higher rates of
appearance of resistant mutants
Mutations need to pre-exist Mutations can form during
selection growth after selection is
applied → Mutation supply
increased
Selective agent (i.e. antibiotic) Selective agent can modulate
cannot modulate the rate of rates of mutation,
formation of mutants since recombination, and horizontal
bacteria die (or do not grow) gene transfer → Rates of mutant
formation increased
The enrichment for mutators is For step-wise selection of
Fig. 2. Growth rates as a function of antibiotic concentrations. The concentration weak for rare one-step mutations of small effect the
interval where the susceptible strain (blue line) will outcompete the resistant strain mutations of large effect enrichment for mutators is
(red line) is indicated in green. Sub-MIC selective window (orange) and traditional strong → Enrichment for
mutant selective window (red) indicate concentration intervals where the resistant mutators with increased
strain will outcompete the susceptible strain. MICsusc = minimal inhibitory concen- propensity for further resistance
tration of the susceptible strain, MICres = minimal inhibitory concentration of the development
resistant strain and MSC = minimal selective concentration. Fitness of mutant less important Fitness of mutant important
since only resistant mutants
Figure reproduced from PLoS Pathogens (Gullberg et al., 2011).
with a fitness reduction less
than that caused by the
antibiotic in the susceptible
very rare mutations, or small bacterial populations) the stochastic bacteria are selected → Mutants
waiting time for a mutation to appear will dominate. However, it with higher fitness selected

is worth noting here that many antibiotics, including the fluoro-


quinolones, have been shown to increase mutation rates (Kohanski
et al., 2010) which could potentially reduce waiting times and shift
the balance toward N > 1 where takeover is dominated by the beta-lactams can by induction of the SOS response (Ysern et al.,
growth selective advantage of the resistant mutant as a function of 1990; Miller et al., 2004; Perez-Capilla et al., 2005; Cirz et al., 2007;
antibiotic selective pressure. Thus, from the mathematical model Baharoglu and Mazel, 2011; Thi et al., 2011) and translational mis-
we can infer that, especially in large populations where N > 1, reading (Ren et al., 1999; Balashov and Humayun, 2002) as well as
and at antibiotic concentrations where 0.01 < s < 1.0, that resistant by generation of mutagenic oxygen radicals (Kohanski et al., 2010)
mutants will rapidly appear and take over the population in less increase the mutation rate up to 15-fold, an effect that might be
than 1000 generations of growth. relevant under certain selective conditions (Cirz et al., 2005). For
example, by experimentally increasing the mutation rate by as lit-
7. Problems generated by non-lethal (sub-MIC) resistance tle as two-fold, the rate of evolution to fluoroquinolone-resistance
selection in E. coli is significantly increased (Örlén and Hughes, 2006). In addi-
tion, non-lethal concentrations of antibiotics also increase rates of
Clearly resistant mutants can be selected by antibiotic concen- homologous recombination and horizontal gene transfer, includ-
trations both in the traditional selective window and the sub-MIC ing in the biofilms present in many infections (Lopez et al., 2007;
window ranges (Fig. 2) and an important question is whether these Couce and Blazquez, 2009; Canton and Morosini, 2011). Antibiotics
different concentration intervals will generate different endpoints at sub-MIC levels have been shown to induce the SOS response
with regard to which mutants are selected? As outlined below we and activate mobilization of genetic elements and increase rates
would argue that non-lethal selections for resistance, where the of recombination. For example, tetracycline stimulates by up to
antibiotics do not kill the susceptible bacteria but only slow down 1000-fold horizontal transfer rates of both conjugative and inte-
their growth, are more problematic with regard to both the rate grative genetic elements in several significant human pathogens,
by which mutants emerge and the types of resistance mechanisms including Staphylococcus aureus (Barr et al., 1986), E. faecalis and
that are selected (Table 2). The reasons for this are several. Listeria monocytogenes (Doucet-Populaire et al., 1991; Bahl et al.,
First, in a lethal selection (i.e. in the classical selective window), 2004) and various SOS-inducing classes of antibiotics can increase
selection only detects rare pre-existing mutations that provide suf- integron recombination in Vibrio cholerae and E. coli (Guerin et al.,
ficiently high levels of resistance, and if they are not present when 2009).
the selective pressure is applied the susceptible population will Third, another problematic aspect of exposure to sub-lethal
die and go extinct (e.g. the infection is cured). In contrast, in the levels of drug is the increased potential for enrichment of sta-
sub-MIC window selection is not lethal but only detects small dif- ble genetic mutators. It has long been understood that antibiotics
ferences in growth rate between susceptible and resistant bacteria exert a second-order selection for mutator clones, i.e. clones of bac-
(Fig. 2). A consequence is that under conditions when the mutation teria with increased mutation rates, because increased mutation
supply rate (i.e. population size times mutation rate) is limiting rates speed up the rate of adaptation to the selective conditions,
the rate of resistance evolution, sub-MIC concentrations of antibi- especially when stepwise selection of different resistance muta-
otics will allow the population to grow until a (rare) resistance tions occurs (Mao et al., 1997; Taddei et al., 1997; Shaver et al.,
mutation appears. In effect, sub-MIC selections allow an increase 2002). For example, among fluoroquinolone-resistant clinical iso-
in population size and mutation supply. lates of E. coli there is a positive correlation between the number
Second, as shown by recent studies certain antibiotic classes of resistance-associated mutations and an increased mutation rate
can at sub-MIC increase the mutation rate by conferring a muta- (Komp Lindgren et al., 2003). Similarly, natural isolates of E. coli
genic effect. For example, fluoroquinolones, aminoglycosides and with intermediate mutator phenotypes have been found to be
168 D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172

8. Which resistance mechanisms are most likely to be


selected at low antibiotic concentrations?

A priori one might think that selection at low antibiotic con-


centrations preferentially select for mutations that confer low
resistance, but the key factor is that any mechanism that con-
fers a sufficiently high increase in resistance and concomitantly
does not decrease fitness too much can potentially be selected.
However, it has been empirically observed that most studied, high-
level resistances whether mutational or HGT based do confer a
substantial (i.e. >1%) fitness cost and at very low antibiotic concen-
trations (i.e. well below the MIC) these mutants will accordingly
not be selected because the antibiotic selective effect is smaller
than the fitness cost. Thus, many of the presently known and
well-described resistance mechanisms are not expected to enrich
under sub-MIC conditions. One illustrative example of this is pro-
vided by selection for streptomycin resistance in S. typhimurium
Fig. 3. Schematic representation of how selection at high and low antibiotic con- where at high streptomycin concentrations (>100 mg/L) the classi-
centrations influences the characteristics of resistant mutants with regard to their
fitness and level of resistance. The blue area indicates that high levels of antibi-
cal rpsL mutations are almost exclusively isolated. These mutations
otic select high-level resistant mutants, with either high or low fitness costs. The provide a high level of resistance (>1024 mg/L) but they are also
yellow area indicates that low levels of antibiotic select low-fitness cost mutants, generally associated with fitness costs ranging between 3 and
with either high- or low-level resistance. The overlapping box outlined in red indi- 27% (Bjorkman et al., 1998; Maisnier-Patin et al., 2002; Gullberg
cates that mutants with high-level resistance and low fitness costs can potentially
et al., 2011). Instead, if selection occurs at sub-MIC of strepto-
be selected at both high and low levels of antibiotic.
mycin for the susceptible wild type strain, rpsL mutations are not
selected (because they are too costly) and instead a new class of
low cost mutations (in the gidB gene) and with moderate increases
in MIC is found (Okamoto et al., 2007). Note that this does not
more likely to be multidrug resistant (Denamur et al., 2005). exclude the possibility that sub-MIC could select high-level resis-
These data strongly suggest that low levels of antibiotic in the tance. The tendency will be that sub-MIC selects mutants with
environment could have significant effects on resistance evolu- high fitness (regardless of resistance level), whereas greater-than-
tion by secondary selection of increased mutation rates. This is in MIC selects mutants with high resistance (regardless of fitness
contrast to selection at a high level of drug where typically resis- level).
tance is conferred by acquiring in one step a specific mutation Few studies have been aimed at identifying resistance mutations
of large effect, e.g. rpsL and rpoB mutations causing streptomycin at low antibiotic concentration regimes, especially under condi-
and rifampicin resistance, respectively, or HGT of plasmid/ICE that tions of long-term exposure to sub-MIC levels. Several types of
carries a specific resistance gene(s) and the enrichment effect is mutants are known to provide low-level resistance but at present
low. we cannot predict their occurrence, mainly because of limited
Finally, one of the conclusions we can draw from the exper- knowledge of which mutations confer no or very small fitness costs.
iments that measure MSC is that there is a positive correlation Thus, as discussed above the likelihood that low-level resistance
between small fitness costs in the absence of antibiotic, and low will become enriched at sub-lethal concentrations of drug is largely
MSC values (Gullberg et al., 2011). The correlation between small determined by the magnitude of the fitness reduction as compared
fitness costs and low MSC is expected on theoretical grounds to the strength of the selection. Below are outlined some examples
because a fitness cost must first be overcome by the negative of potential mechanisms to be considered:
effect of antibiotics on the susceptible strain before the resis-
tant mutant can be selectively enriched. Thus, only resistance 1. Resistance conferred by activation of efflux systems (either by
mechanisms with a fitness cost smaller than the growth reduc- mutation or regulation) can confer an unspecific and gener-
tion caused by the antibiotic in the susceptible bacteria will be ally low-level type of resistance. For example, activation of the
competitive, and resistance mutations that confer a too high fit- mar system in E. coli results in a several-fold increase in resis-
ness costs will not be enriched at low levels of antibiotic (even tance to a number of different antibiotics (e.g. chloramphenicol,
though they might confer high level of resistance). An important fluoroquinolones, tetracycline) and biocides (e.g. triclosan and
implication from this reasoning is that non-lethal selective con- quaternary ammonium compounds). Although these mutations
ditions will enrich for resistant mutants with higher fitness, and occur at a high frequency (by inactivation of repression), they
these mutants may express either a low level or a high level of often have a considerable fitness cost and they are therefore
resistance (Fig. 3). In contrast, when the selection is lethal (above unlikely to be selected during prolonged exposure at very low
MIC) it is only required that the acquired mechanism (mutation antibiotic concentrations (Gullberg et al., 2011).
or HGT) increases antibiotic resistance above the applied drug 2. Another class of mutations increasingly recognized as being
concentration, and the effect of the resistance mechanism on bac- important for low-level resistance is mutation in porins, proteins
terial fitness is less important because even resistance mechanisms that form water channels in the outer membrane of Gram-
with a very high fitness cost will be selected as long as sus- negative bacteria. Again these mutations typically only cause a
ceptible competitor bacteria are eliminated. In conclusion, from few-fold increase in resistance to for example ␤-lactams, and
the point of view of persistence and potential for reversibility their fitness costs are often quite low. Thus, at least certain types
of resistance mechanisms that are selected at sub-MIC might be of porin mutations are predicted to appear at sub-MIC selections.
especially problematic since the low fitness cost means that such 3. Increased dosage of the antibiotic target molecule or an inef-
bacteria will remain in the population longer when the antibi- ficient enzymatic antibiotic-detoxifying mechanism can also
otic use and selective pressure is reduced (Andersson and Hughes, provide low-level resistance. The latter type of mechanism have
2010). been observed for ␤-lactam resistance where successive gene
D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172 169

amplification of the ampC/bla genes conferred increasing resis- bacteria) whereas levels in the outside environments are typi-
tance from a very low basal level (Normark et al., 1977; Edlund cally substantially lower. Thus, antibiotic concentrations in rivers,
and Normark, 1981; Sun et al., 2009). Interestingly, a change in lakes and soils are usually in the ␮g/L to ng/L range, i.e. they
the opposite direction, by a mutation that reduced drug-target would act as non-lethal selections (Thiele-Bruhn, 2003; Chander
enzyme expression, confers low-level resistance to quinolones et al., 2005; Kummerer, 2009). However, high antibiotic concen-
(Ince and Hooper, 2003). trations can be observed in connection with wastewater outlets
4. Low-level resistance can also occur as a byproduct of a resis- from, for example, pharmaceutical industries (Larsson et al., 2007;
tance mechanism that provides a high-level of resistance to Fick et al., 2009) and hospitals (Kummerer and Henninger, 2003;
a specific antibiotic but a low level of resistance to a related Gomez et al., 2006; Thomas et al., 2007; Duong et al., 2008;
antibiotic of the same structural or chemical class. Examples of Seifrtova et al., 2008; Verlicchi et al., 2010). High antibiotic con-
this includes certain gyrA mutations in E. coli that confer high- centrations are also associated with wastewater from agricultural
level resistance to nalidixic acid but only low level resistance to and aquacultural activities (Pena et al., 2010; Thuy et al., 2011;
ciprofloxacin (Yoshida et al., 1990; Gullberg et al., 2011), CTX- Wei et al., 2011; Zou et al., 2011) and detectable levels are some-
M, TEM and AmpC genes that confer high-level resistance to times found in tap water for human consumption (Fick et al., 2009;
penicillins, but only miniscule increases in resistance to car- Yiruhan et al., 2010). In one extreme case the concentration of
bapenems (Ardanuy et al., 1998; Martinez-Martinez et al., 1999; the fluoroquinolone ciprofloxacin found in river water (31 mg/L)
Mammeri et al., 2008; Girlich et al., 2009; Yang et al., 2009) and downstream of a region with several antibiotic-producing indus-
the aminoglycoside modifying enzyme aminoglycoside (AG) 3 - tries well exceeded the antibiotic concentration measured in the
phosphotransferase II that provide full resistance to kanamycin serum of ciprofloxacin-treated patients (about 2–3 mg/L), indicat-
but only low-level resistance to amikacin (Perlin and Lerner, ing an extremely strong selective pressure (Larsson et al., 2007).
1986).
5. Furthermore, it is likely that there exist a number of resistance
mechanisms that are yet to be identified. It is hard to predict 10. Approaches to reduce the presence of sub-lethal
which these functions might be but as shown by a recent screen selective conditions
for low-level resistance conferred by over-expression of E. coli
genes, increased dosage of 61 ORFs (from a library containing Even though antibiotic levels can be at sub-MIC in treated
over 4000 genes) could confer partial resistance to 86 of 237 humans, animals and plants in clinical, community and agricultural
antibiotic/toxin-containing environments. Of these ORFs, most settings, it is likely that the most common environments where
conferred small but significant increases in MIC against tetracy- low levels are consistently present over long time periods is in
clines, ␤-lactams, antifolates, aminoglycosides, and macrolides animals treated with antibiotics for growth promotion and in vari-
(Soo et al., 2011). This experiment demonstrates that there exists ous aquatic environments (Baquero et al., 2008; Kummerer, 2009;
a considerable reservoir of genes in the so-called intrinsic resis- Martinez, 2009; Hoa et al., 2011) and in soils (Rooklidge, 2004;
tome that could confer low-level resistance when increased in Pico and Andreu, 2007; Sukul and Spiteller, 2007; Martinez, 2008)
dosage. It is also conceivable that amino acid changes in exist- where a big part of the therapeutically used drugs will ultimately
ing ORFs could confer low resistance. A still unanswered key end up if they have not been degraded. It is hard to obtain accurate
question is how such genetic changes would affect fitness but if estimates of global antibiotic use but it is clearly in excess of several
they are not too costly they could potentially be selected during hundred thousands of tons per year and about half of these antibi-
sub-MIC antibiotic exposure. otics will enter natural environments in active form after they have
6. Apart from genetic changes in the bacterium, low-level resis- exerted their therapeutic or growth-promoting action in humans
tance can also be conferred by physiological and regulatory and animals.
responses that activate intrinsic resistance mechanisms or gen- Is there any reasonable way we may deal with these phar-
erate a physiological state where the bacteria are less susceptible maceutical pollutants and prevent them exerting selection for
to the drugs. One example of the former is the activation of a antimicrobial resistance in the environment? Obviously a general
cryptic aminoglycoside resistance gene, aadA, which when acti- reduction of antibiotic use will reduce antibiotic release into the
vated by certain growth conditions provides a moderate level of environment. Thus, a more prudent therapeutic use of antibiotic
resistance (Koskiniemi et al., 2011). Another well-studied exam- for infections will have beneficial effects. Similarly, it is possible
ple is the occurrence of tolerant cells that are refractory to drugs to stop the use of antibiotics for growth promotion without major
(Jayaraman, 2008; Lewis, 2008; Allison et al., 2011) or the for- negative effects on animal production, as shown by bans in Sweden
mation of biofilms in which the bacteria are less susceptible and Denmark introduced in 1986 and 1997, respectively (Aarestrup
(Anderson and O’Toole, 2008; Hoiby et al., 2010). In contrast et al., 2001; Bengtsson and Wierup, 2006; Grave et al., 2006) and the
to genetic mechanisms, these physiological states are rapidly recent ban, applicable within all countries of the European Union,
reversible if environmental conditions change but they could still that was introduced by the European Parliament and the Council of
potentially serve as stepping-stones that provide transient low- Europe in 2006 (2003; Cogliani et al., 2011). As the use of antibiotics
levels of resistance that allow bacteria to subsequently acquire for growth promotion represents a substantial part of total antibi-
stable, high-level resistance mechanisms. otic consumption a global ban would drastically reduce the overall
selective pressure. With regard to release of antibiotics into the
environment one could either prevent antibiotic release into water
9. What are the drug concentrations in different from humans (mainly urine) or inactivate the drugs downstream
environments? from the release site. The latter is clearly achievable and several
methods already exist that are technically feasible and economi-
When examining the actual concentrations of antibiotics that cally viable (Esplugas et al., 2007; MistraPharma, 2011; Wahlberg
are found in different environments there is obviously an enor- et al., 2011). For example, ozone treatment of sewage water is an
mous variation. Generally the concentrations observed in treated effective and relatively cheap method for destruction of pharma-
humans, animals and in farming are relatively high (even though ceuticals, including antibiotics. An additional beneficial effect of
there might exist compartments or time intervals where concen- ozone treatment is that essentially all types of pathogenic infec-
trations are low, i.e. below MICs of the drugs against susceptible tious agents will be efficiently inactivated. Thus, ozone treatment
170 D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172

will not only remove the selective agents but also break transmis- Bjorland, J., Sunde, M., et al., 2001. Plasmid-borne smr gene causes resistance to
sion cycles of both susceptible and resistant microbes. quaternary ammonium compounds in bovine Staphylococcus aureus. Journal of
Clinical Microbiology 39 (11), 3999–4004.
Borges-Walmsley, M.I., Walmsley, A.R., 2001. The structure and function of drug
pumps. Trends in Microbiology 9 (2), 71–79.
Acknowledgements Braoudaki, M., Hilton, A.C., 2004. Adaptive resistance to biocides in Salmonella
enterica and Escherichia coli O157 and cross-resistance to antimicrobial agents.
This work was supported by grants from the Swedish Research Journal of Clinical Microbiology 42 (1), 73–78.
Brown, M.H., Paulsen, I.T., et al., 1999. The multidrug efflux protein NorM is a proto-
Council Medicine, EU project Predicting Antibiotic Resistance (PAR, type of a new family of transporters. Molecular Microbiology 31 (1), 394–395.
grant no. 241476), Swedish Strategic Research Foundation (SSF), Bryskier, A. (Ed.), 2005. Antimicrobial Agents: Antibacterials and Antifungals. ASM
Swedish Innovation Agency (Vinnova) to DIA and DH. We thank Press.
Bulmer, M., 1991. The selection-mutation-drift theory of synonymous codon usage.
Christina Greko and Linus Sandegren for helpful discussion and
Genetics 129 (3), 897–907.
comments on the manuscript. Burman, W., Benator, D., et al., 2006. Acquired rifamycin resistance with twice-
weekly treatment of HIV-related tuberculosis. American Journal of Respiratory
and Critical Care Medicine 173 (3), 350–356.
References Canton, R., 2009. Antibiotic resistance genes from the environment: a perspective
through newly identified antibiotic resistance mechanisms in the clinical set-
Anon., 2003. Regulation (EC) No 1831/2003 of the European Parliament and of the ting. Clinical Microbiology and Infection: The Official Publication of the European
Council of 22 September 2003 on additives for use in animal nutrition. Official Society of Clinical Microbiology and Infectious Diseases 15 (Suppl. 1), 20–25.
Journal of the European Union L268, 29–43. Canton, R., Morosini, M.I., 2011. Emergence and spread of antibiotic resistance fol-
Aarestrup, F.M., Seyfarth, A.M., et al., 2001. Effect of abolishment of the use of antimi- lowing exposure to antibiotics. FEMS Microbiology Review 35 (5), 977–991.
crobial agents for growth promotion on occurrence of antimicrobial resistance Cattoir, V., Lesprit, P., et al., 2006. In vivo selection during ofloxacin therapy of
in fecal enterococci from food animals in Denmark. Antimicrobial Agents and Escherichia coli with combined topoisomerase mutations that confer high resis-
Chemotherapy 45 (7), 2054–2059. tance to ofloxacin but susceptibility to nalidixic acid. Journal of Antimicrobial
Allen, H.K., Moe, L.A., et al., 2009. Functional metagenomics reveals diverse beta- Chemotherapy 58 (5), 1054–1057.
lactamases in a remote Alaskan soil. The ISME Journal 3 (2), 243–251. Chander, Y., Kumar, K., et al., 2005. Antibacterial activity of soil-bound antibiotics.
Allison, K.R., Brynildsen, M.P., et al., 2011. Heterogeneous bacterial persisters and Journal of Environmental Quality 34 (6), 1952–1957.
engineering approaches to eliminate them. Current Opinion in Microbiology 14 Chang, S., Sievert, D.M., et al., 2003. Infection with vancomycin-resistant Staphylo-
(5), 593–598. coccus aureus containing the vanA resistance gene. The New England Journal of
Aminov, R.I., Mackie, R.I., 2007. Evolution and ecology of antibiotic resistance genes. Medicine 348 (14), 1342–1347.
FEMS Microbiology Letters 271 (2), 147–161. Christiansen, N., Nielsen, L., et al., 2011. Fluoroquinolone resistance mechanisms
Anderson, G.G., O’Toole, G.A., 2008. Innate and induced resistance mechanisms of in urinary tract pathogenic Escherichia coli isolated during rapidly increasing
bacterial biofilms. Current Topics in Microbiology and Immunology 322, 85–105. fluoroquinolone consumption in a low-use country. Microbial Drug Resistance
Andersson, D.I., Hughes, D., 2009. Gene amplification and adaptive evolution in 17 (3), 395–406.
bacteria. Annual Review of Genetics 43, 167–195. Cirz, R.T., Chin, J.K., et al., 2005. Inhibition of mutation and combating the evolution
Andersson, D.I., Hughes, D., 2010. Antibiotic resistance and its cost: is it possible to of antibiotic resistance. PLoS Biology 3 (6), e176.
reverse resistance? Nature Reviews Microbiology 8 (4), 260–271. Cirz, R.T., Jones, M.B., et al., 2007. Complete and SOS-mediated response of Staphy-
Ardanuy, C., Linares, J., et al., 1998. Outer membrane profiles of clonally related Kleb- lococcus aureus to the antibiotic ciprofloxacin. Journal of Bacteriology 189 (2),
siella pneumoniae isolates from clinical samples and activities of cephalosporins 531–539.
and carbapenems. Antimicrobial Agents and Chemotherapy 42 (7), 1636–1640. Codling, C.E., Jones, B.V., et al., 2004. Identification of genes involved in the sus-
Atwood, K.C., Schneider, L.K., et al., 1951. Periodic selection in Escherichia coli. Pro- ceptibility of Serratia marcescens to polyquaternium-1. Journal of Antimicrobial
ceedings of the National Academy of Sciences of the United States of America Chemotherapy 54 (2), 370–375.
37 (3), 146–155. Cogliani, C., Goosens, H., et al., 2011. Restricting antimicribial use in food animals:
Baharoglu, Z., Mazel, D., 2011. Vibrio cholerae triggers SOS and mutagenesis in lessons from Europe. Microbe Magazine (American Society for Microbiology) 6
response to a wide range of antibiotics: a route towards multiresistance. Antimi- (6), 274–279.
crobial Agents and Chemotherapy 55 (5), 2438–2441. Cookson, B., 2005. Clinical significance of emergence of bacterial antimicrobial resis-
Bahl, M.I., Sorensen, S.J., et al., 2004. Effect of tetracycline on transfer and establish- tance in the hospital environment. Journal of Applied Microbiology 99 (5),
ment of the tetracycline-inducible conjugative transposon Tn916 in the guts of 989–996.
gnotobiotic rats. Applied and Environmental Microbiology 70 (2), 758–764. Corvec, S., Lepelletier, D., et al., 2008. In vivo selection of an Escherichia coli isolate
Balashov, S., Humayun, M.Z., 2002. Mistranslation induced by streptomycin pro- highly resistant to ciprofloxacin and ceftazidime: role of a 4-bp duplication in
vokes a RecABC/RuvABC-dependent mutator phenotype in Escherichia coli cells. acrR and ampC overexpression. International Journal of Antimicrobial Agents
Journal of Molecular Biology 315 (4), 513–527. 32 (2), 196–198.
Baquero, F., 2001a. Low-level antibacterial resistance: a gateway to clinical resis- Couce, A., Blazquez, J., 2009. Side effects of antibiotics on genetic variability. FEMS
tance. Drug Resistance Updates: Reviews and Commentaries in Antimicrobial Microbiology Review 33 (3), 531–538.
and Anticancer Chemotherapy 4 (2), 93–105. Cox, H.S., Niemann, S., et al., 2007. Risk of acquired drug resistance during short-
Baquero, F., 2001b. Low-level antibiotic resistance. In: Hughes, D., Andersson, D.I. course directly observed treatment of tuberculosis in an area with high levels
(Eds.), Antibiotic Development and Resistance. Taylor and Francis, London and of drug resistance. Clinical Infectious Diseases: An Official Publication of the
New York, pp. 117–136. Infectious Diseases Society of America 44 (11), 1421–1427.
Baquero, F., Martinez, J.L., et al., 2008. Antibiotics and antibiotic resistance in water D’Costa, V.M., King, C.E., et al., 2011. Antibiotic resistance is ancient. Nature 477
environments. Current Opinion in Biotechnology 19 (3), 260–265. (7365), 457–461.
Baquero, F., Negri, M.C., 1997. Selective compartments for resistant microorganisms de Toro, M., Rojo-Bezares, B., et al., 2010. In vivo selection of aac(6 )-Ib-cr and muta-
in antibiotic gradients. BioEssays: News and Reviews in Molecular, Cellular and tions in the gyrA gene in a clinical qnrS1-positive Salmonella enterica serovar
Developmental Biology 19 (8), 731–736. Typhimurium DT104B strain recovered after fluoroquinolone treatment. Journal
Baquero, F., Negri, M.C., et al., 1997. The antibiotic selective process: concentration- of Antimicrobial Chemotherapy 65 (9), 1945–1949.
specific amplification of low-level resistant populations. Ciba Foundation Dean, A.M., Dykhuizen, D.E., et al., 1988. Fitness effects of amino acid replacements
Symposium 207, 93–105 (Discussion 105–111). in the beta-galactosidase of Escherichia coli. Molecular Biology and Evolution 5
Baquero, F., Negri, M.C., et al., 1998. Antibiotic-selective environments. Clinical (5), 469–485.
Infectious Diseases: An Official Publication of the Infectious Diseases Society Denamur, E., Tenaillon, O., et al., 2005. Intermediate mutation frequencies favor
of America 27 (Suppl. 1), S5–S11. evolution of multidrug resistance in Escherichia coli. Genetics 171 (2), 825–827.
Barr, V., Barr, K., et al., 1986. Beta-lactam antibiotics increase the frequency of plas- Denyer, S.P., Maillard, J.Y., 2002. Cellular impermeability and uptake of biocides
mid transfer in Staphylococcus aureus. Journal of Antimicrobial Chemotherapy and antibiotics in Gram-negative bacteria. Journal of Applied Microbiology 92
17 (4), 409–413. (Suppl.), 35S–45S.
Bengtsson, B., Wierup, M., 2006. Antimicrobial resistance in Scandinavia after ban Dong, Y., Zhao, X., et al., 1999. Effect of fluoroquinolone concentration on selec-
of antimicrobial growth promoters. Animal Biotechnology 17 (2), 147–156. tion of resistant mutants of Mycobacterium bovis BCG and Staphylococcus aureus.
Berg, O.G., 1995. Periodic selection and hitchhiking in a bacterial population. Journal Antimicrobial Agents and Chemotherapy 43 (7), 1756–1758.
of Theoretical Biology 173 (3), 307–320. Doucet-Populaire, F., Trieu-Cuot, P., et al., 1991. Inducible transfer of conjugative
Berg, O.G., 1996. Selection intensity for codon bias and the effective population size transposon Tn1545 from Enterococcus faecalis to Listeria monocytogenes in the
of Escherichia coli. Genetics 142 (4), 1379–1382. digestive tracts of gnotobiotic mice. Antimicrobial Agents and Chemotherapy
Bergman, M., Nyberg, S.T., et al., 2009. Association between antimicrobial consump- 35 (1), 185–187.
tion and resistance in Escherichia coli. Antimicrobial Agents and Chemotherapy Drlica, K., Zhao, X., 2007. Mutant selection window hypothesis updated. Clinical
53 (3), 912–917. Infectious Diseases 44 (5), 681–688.
Bjorkman, J., Hughes, D., et al., 1998. Virulence of antibiotic-resistant Salmonella Dumitrescu, O., Boisset, S., et al., 2007. Effect of antibiotics on Staphylococcus aureus
typhimurium. Proceedings of the National Academy of Sciences of the United producing Panton–Valentine leukocidin. Antimicrobial Agents and Chemother-
States of America 95 (7), 3949–3953. apy 51 (4), 1515–1519.
D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172 171

Duong, H.A., Pham, N.H., et al., 2008. Occurrence, fate and antibiotic resistance Larsson, D.G., de Pedro, C., et al., 2007. Effluent from drug manufactures contains
of fluoroquinolone antibacterials in hospital wastewaters in Hanoi, Vietnam. extremely high levels of pharmaceuticals. Journal of Hazardous Materials 148
Chemosphere 72 (6), 968–973. (3), 751–755.
Edlund, T., Normark, S., 1981. Recombination between short DNA homologies causes Le Thomas, I., Couetdic, G., et al., 2001. In vivo selection of a target/efflux double
tandem duplication. Nature 292 (5820), 269–271. mutant of Pseudomonas aeruginosa by ciprofloxacin therapy. Journal of Antimi-
Esplugas, S., Bila, D.M., et al., 2007. Ozonation and advanced oxidation technologies crobial Chemotherapy 48 (4), 553–555.
to remove endocrine disrupting chemicals (EDCs) and pharmaceuticals and per- Leotard, S., Poirel, L., et al., 2001. In vivo selection of oxacillinase-mediated cef-
sonal care products (PPCPs) in water effluents. Journal of Hazardous Materials tazidime resistance in Pseudomonas aeruginosa. Microbial Drug Resistance 7 (3),
149 (3), 631–642. 273–275.
Fick, J., Soderstrom, H., et al., 2009. Contamination of surface, ground, and drinking Levings, R.S., Partridge, S.R., et al., 2007. SGI1-K, a variant of the SGI1 genomic island
water from pharmaceutical production. Environmental Toxicology and Chem- carrying a mercury resistance region, in Salmonella enterica serovar Kentucky.
istry 28 (12), 2522–2527. Antimicrobial Agents and Chemotherapy 51 (1), 317–323.
Fraud, S., Campigotto, A.J., et al., 2008. MexCD-OprJ multidrug efflux system Levy, S.B., 2002. Active efflux, a common mechanism for biocide and antibiotic
of Pseudomonas aeruginosa: involvement in chlorhexidine resistance and resistance. Journal of Applied Microbiology 92 (Suppl.), 65S–71S.
induction by membrane-damaging agents dependent upon the AlgU stress Lewis, K., 2008. Multidrug tolerance of biofilms and persister cells. Current Topics
response sigma factor. Antimicrobial Agents and Chemotherapy 52 (12), in Microbiology and Immunology 322, 107–131.
4478–4482. Lind, P.A., Tobin, C., et al., 2010. Compensatory gene amplification restores fit-
Girlich, D., Poirel, L., et al., 2009. CTX-M expression and selection of ertapenem resis- ness after inter-species gene replacements. Molecular Microbiology 75 (5),
tance in Klebsiella pneumoniae and Escherichia coli. Antimicrobial Agents and 1078–1089.
Chemotherapy 53 (2), 832–834. Liu, A., Fong, A., et al., 2011. Selective advantage of resistant strains at trace levels of
Gomez, M.J., Petrovic, M., et al., 2006. Determination of pharmaceuticals antibiotics: a simple and ultrasensitive color test for detection of antibiotics and
of various therapeutic classes by solid-phase extraction and liquid genotoxic agents. Antimicrobial Agents and Chemotherapy 55 (3), 1204–1210.
chromatography–tandem mass spectrometry analysis in hospital effluent Lopez, E., Elez, M., et al., 2007. Antibiotic-mediated recombination: ciprofloxacin
wastewaters. Journal of Chromatography A 1114 (2), 224–233. stimulates SOS-independent recombination of divergent sequences in
Goossens, H., 2009. Antibiotic consumption and link to resistance. Clinical Microbi- Escherichia coli. Molecular Microbiology 64 (1), 83–93.
ology and Infection: The Official Publication of the European Society of Clinical Maillard, J.Y., 2007. Bacterial resistance to biocides in the healthcare environment:
Microbiology and Infectious Diseases 15 (Suppl. 3), 12–15. should it be of genuine concern? The Journal of Hospital Infection 65 (Suppl. 2),
Grave, K., Jensen, V.F., et al., 2006. Usage of veterinary therapeutic antimicrobials in 60–72.
Denmark, Norway and Sweden following termination of antimicrobial growth Maisnier-Patin, S., Berg, O.G., et al., 2002. Compensatory adaptation to the dele-
promoter use. Preventive Veterinary Medicine 75 (1–2), 123–132. terious effect of antibiotic resistance in Salmonella typhimurium. Molecular
Guerin, E., Cambray, G., et al., 2009. The SOS response controls integron recombina- Microbiology 46 (2), 355–366.
tion. Science 324 (5930), 1034. Mammeri, H., Nordmann, P., et al., 2008. Contribution of extended-spectrum AmpC
Gullberg, E., Cao, S., et al., 2011. Selection of resistant bacteria at very low antibiotic (ESAC) beta-lactamases to carbapenem resistance in Escherichia coli. FEMS
concentrations. PLoS Pathogens 7 (7), e1002158. Microbiology Letters 282 (2), 238–240.
Heath, R.J., Li, J., et al., 2000. Inhibition of the Staphylococcus aureus NADPH- Mao, E.F., Lane, L., et al., 1997. Proliferation of mutators in a cell population. Journal
dependent enoyl-acyl carrier protein reductase by triclosan and hex- of Bacteriology 179 (2), 417–422.
achlorophene. Journal of Biological Chemistry 275 (7), 4654–4659. Marcusson, L.L., Frimodt-Moller, N., et al., 2009. Interplay in the selection of fluoro-
Hoa, P.T., Managaki, S., et al., 2011. Antibiotic contamination and occurrence of quinolone resistance and bacterial fitness. PLoS Pathogens 5 (8), e1000541.
antibiotic-resistant bacteria in aquatic environments of northern Vietnam. The Marcusson, L.L., Olofsson, S.K., et al., 2005. Mutant prevention concentrations of
Science of the Total Environment 409 (15), 2894–2901. ciprofloxacin for urinary tract infection isolates of Escherichia coli. Journal of
Hoiby, N., Bjarnsholt, T., et al., 2010. Antibiotic resistance of bacterial biofilms. Inter- Antimicrobial Chemotherapy 55 (6), 938–943.
national Journal of Antimicrobial Agents 35 (4), 322–332. Mariam, S.H., Werngren, J., et al., 2011. Dynamics of antibiotic resistant Mycobac-
Hong, H.J., Hutchings, M.I., et al., 2004. Characterization of an inducible vancomycin terium tuberculosis during long-term infection and antibiotic treatment. PLoS
resistance system in Streptomyces coelicolor reveals a novel gene (vanK) required One 6 (6), e21147.
for drug resistance. Molecular Microbiology 52 (4), 1107–1121. Martinez, J.L., 2008. Antibiotics and antibiotic resistance genes in natural environ-
Hu, Y., Mathema, B., et al., 2011. Population-based investigation of fluoroquinolones ments. Science 321 (5887), 365–367.
resistant tuberculosis in rural eastern China. Tuberculosis 91 (3), 238–243. Martinez, J.L., 2009. Environmental pollution by antibiotics and by antibiotic resis-
Huet, A.A., Raygada, J.L., et al., 2008. Multidrug efflux pump overexpression in tance determinants. Environmental Pollution 157 (11), 2893–2902.
Staphylococcus aureus after single and multiple in vitro exposures to biocides Martinez-Martinez, L., Pascual, A., et al., 1999. Roles of beta-lactamases and porins
and dyes. Microbiology 154 (Pt 10), 3144–3153. in activities of carbapenems and cephalosporins against Klebsiella pneumoniae.
Hughes, V.M., Datta, N., 1983. Conjugative plasmids in bacteria of the ‘pre-antibiotic’ Antimicrobial Agents and Chemotherapy 43 (7), 1669–1673.
era. Nature 302 (5910), 725–726. McKeegan, K.S., Borges-Walmsley, M.I., et al., 2003. The structure and function of
Ikemura, T., 1985. Codon usage and tRNA content in unicellular and multicellular drug pumps: an update. Trends in Microbiology 11 (1), 21–29.
organisms. Molecular Biology and Evolution 2 (1), 13–34. McMurry, L.M., McDermott, P.F., et al., 1999. Genetic evidence that InhA of
Ince, D., Hooper, D.C., 2003. Quinolone resistance due to reduced target enzyme Mycobacterium smegmatis is a target for triclosan. Antimicrobial Agents and
expression. Journal of Bacteriology 185 (23), 6883–6892. Chemotherapy 43 (3), 711–713.
Jayaraman, R., 2008. Bacterial persistence: some new insights into an old phe- Miller, C., Thomsen, L.E., et al., 2004. SOS response induction by beta-lactams and
nomenon. Journal of Biosciences 33 (5), 795–805. bacterial defense against antibiotic lethality. Science 305 (5690), 1629–1631.
Jeong, H.S., Kim, J.A., et al., 2011. Prevalence of plasmid-mediated quinolone resis- Mindlin, S.Z., Soina, V.S., et al., 2008. Isolation of antibiotic resistance bacterial strains
tance and mutations in the gyrase and topoisomerase IV genes in Salmonella from Eastern Siberia permafrost sediments. Russian Journal of Genetics 44 (1),
isolated from 12 tertiary-care hospitals in Korea. Microbial Drug Resistance 17 27–34.
(4), 551–557. MistraPharma, 2011. Collaboration to reduce the environmental risks of pharma-
Karatzas, K.A., Webber, M.A., et al., 2007. Prolonged treatment of Salmonella enter- ceuticals. Stockholm, MistraPharma (available from info@mistrapharma.se).
ica serovar Typhimurium with commercial disinfectants selects for multiple Nagaev, I., Bjorkman, J., et al., 2001. Biological cost and compensatory evolution
antibiotic resistance, increased efflux and reduced invasiveness. Journal of in fusidic acid-resistant Staphylococcus aureus. Molecular Microbiology 40 (2),
Antimicrobial Chemotherapy 60 (5), 947–955. 433–439.
Koch, A.L., 1974. The pertinence of the periodic selection phenomenon to prokaryote Nakano, M., Yasuda, M., et al., 2001. In vivo selection of Pseudomonas aeruginosa with
evolution. Genetics 77 (1), 127–142. decreased susceptibilities to fluoroquinolones during fluoroquinolone treat-
Kohanski, M.A., DePristo, M.A., et al., 2010. Sublethal antibiotic treatment leads to ment of urinary tract infection. Urology 58 (1), 125–128.
multidrug resistance via radical-induced mutagenesis. Molecular Cell 37 (3), Nazir, H., Cao, S., et al., 2011. Can phylogenetic type predict resistance development?
311–320. Journal of Antimicrobial Chemotherapy 66 (4), 778–787.
Komp Lindgren, P., Karlsson, Å., et al., 2003. Mutation rate and evolution of fluoro- Negri, M.C., Lipsitch, M., et al., 2000. Concentration-dependent selection of small
quinolone resistance in Escherichia coli isolates from patients with urinary tract phenotypic differences in TEM beta-lactamase-mediated antibiotic resistance.
infections. Antimicrobial Agents and Chemotherapy 47 (10), 3222–3232. Antimicrobial Agents and Chemotherapy 44 (9), 2485–2491.
Koskiniemi, S., Pranting, M., et al., 2011. Activation of cryptic aminoglycoside resis- Nikaido, H., 2003. Molecular basis of bacterial outer membrane permeability revis-
tance in Salmonella enterica. Molecular Microbiology 80 (6), 1464–1478. ited. Microbiology and Molecular Biology Reviews: MMBR 67 (4), 593–656.
Kummerer, K., 2009. Antibiotics in the aquatic environment – a review – part I. Noguchi, N., Suwa, J., et al., 2005. Susceptibilities to antiseptic agents and distri-
Chemosphere 75 (4), 417–434. bution of antiseptic-resistance genes qacA/B and smr of methicillin-resistant
Kummerer, K., Henninger, A., 2003. Promoting resistance by the emission of antibi- Staphylococcus aureus isolated in Asia during 1998 and 1999. Journal of Medical
otics from hospitals and households into effluent. Clinical Microbiology and Microbiology 54 (Pt 6), 557–565.
Infection: The Official Publication of the European Society of Clinical Microbiol- Normark, S., Edlund, T., et al., 1977. Escherichia coli K-12 mutants hyperproducing
ogy and Infectious Diseases 9 (12), 1203–1214. chromosomal beta-lactamase by gene repetitions. Journal of Bacteriology 132
Laraki, N., Galleni, M., et al., 1999. Structure of In31, a blaIMP-containing Pseu- (3), 912–922.
domonas aeruginosa integron phyletically related to In5, which carries an Okamoto, S., Tamaru, A., et al., 2007. Loss of a conserved 7-methylguanosine mod-
unusual array of gene cassettes. Antimicrobial Agents and Chemotherapy 43 ification in 16S rRNA confers low-level streptomycin resistance in bacteria.
(4), 890–901. Molecular Microbiology 63 (4), 1096–1106.
172 D.I. Andersson, D. Hughes / Drug Resistance Updates 15 (2012) 162–172

Örlén, H., Hughes, D., 2006. Weak mutators can drive the evolution of fluoro- Sharp, P.M., Emery, L.R., et al., 2010. Forces that influence the evolution of codon bias.
quinolone resistance in Escherichia coli. Antimicrobial Agents and Chemotherapy Philosophical Transactions of the Royal Society of London. Series B, Biological
50 (10), 3454–3456. Sciences 365 (1544), 1203–1212.
Parikh, S.L., Xiao, G., et al., 2000. Inhibition of InhA, the enoyl reductase from Shaver, A.C., Dombrowski, P.G., et al., 2002. Fitness evolution and the rise of mutator
Mycobacterium tuberculosis, by triclosan and isoniazid. Biochemistry 39 (26), alleles in experimental Escherichia coli populations. Genetics 162 (2), 557–566.
7645–7650. Sidhu, M.S., Heir, E., et al., 2002. Frequency of disinfectant resistance genes and
Patel, R., Piper, K., et al., 2000. The biopesticide Paenibacillus popilliae has a genetic linkage with beta-lactamase transposon Tn552 among clinical staphy-
vancomycin resistance gene cluster homologous to the enterococcal VanA van- lococci. Antimicrobial Agents and Chemotherapy 46 (9), 2797–2803.
comycin resistance gene cluster. Antimicrobial Agents and Chemotherapy 44 Sidhu, M.S., Heir, E., et al., 2001. Genetic linkage between resistance to quaternary
(3), 705–709. ammonium compounds and beta-lactam antibiotics in food-related Staphylo-
Paulsen, I.T., Brown, M.H., et al., 1996. Proton-dependent multidrug efflux systems. coccus spp. Microbial Drug Resistance 7 (4), 363–371.
Microbiological Reviews 60 (4), 575–608. Skrahina, A., Hurevich, H., et al., 2012. Alarming levels of drug-resistant tuberculo-
Pena, A., Pina, J., et al., 2010. Fluoroquinolone antibiotics determination in pig- sis in Belarus: results of a survey in Minsk. The European Respiratory Journal:
geries environmental waters. Journal of Environmental Monitoring: JEM 12 (3), Official Journal of the European Society for Clinical Respiratory Physiology, Feb
642–646. 15 (Epub ahead of print).
Perez-Capilla, T., Baquero, M.R., et al., 2005. SOS-independent induction of dinB Smith, D.H., 1967. R factor infection of Escherichia coli lyophilized in 1946. Journal
transcription by beta-lactam-mediated inhibition of cell wall synthesis in of Bacteriology 94 (6), 2071–2072.
Escherichia coli. Journal of Bacteriology 187 (4), 1515–1518. Soo, V.W., Hanson-Manful, P., et al., 2011. Artificial gene amplification reveals an
Perlin, M.H., Lerner, S.A., 1986. High-level amikacin resistance in Escherichia coli due abundance of promiscuous resistance determinants in Escherichia coli. Proceed-
to phosphorylation and impaired aminoglycoside uptake. Antimicrobial Agents ings of the National Academy of Sciences of the United States of America 108
and Chemotherapy 29 (2), 216–224. (4), 1484–1489.
Petrosino, J., Cantu 3rd, C., et al., 1998. beta-Lactamases: protein evolution in real Sukul, P., Spiteller, M., 2007. Fluoroquinolone antibiotics in the environment.
time. Trends in Microbiology 6 (8), 323–327. Reviews of Environmental Contamination and Toxicology 191, 131–162.
Phillips, I., Culebras, E., et al., 1987. Induction of the SOS response by new 4- Sun, S., Berg, O.G., et al., 2009. Contribution of gene amplification to evolution of
quinolones. Journal of Antimicrobial Chemotherapy 20 (5), 631–638. increased antibiotic resistance in Salmonella typhimurium. Genetics 182 (4),
Pico, Y., Andreu, V., 2007. Fluoroquinolones in soil—risks and challenges. Analytical 1183–1195.
and Bioanalytical Chemistry 387 (4), 1287–1299. Taddei, F., Radman, M., et al., 1997. Role of mutator alleles in adaptive evolution.
Piddock, L.J., 2006. Clinically relevant chromosomally encoded multidrug resis- Nature 387 (6634), 700–702.
tance efflux pumps in bacteria. Clinical Microbiology Reviews 19 (2), Taylor, N.G., Verner-Jeffreys, D.W., et al., 2011. Aquatic systems: maintaining, mixing
382–402. and mobilising antimicrobial resistance? Trends in Ecology & Evolution 26 (6),
Poirel, L., Kampfer, P., et al., 2002. Chromosome-encoded Ambler class A beta- 278–284.
lactamase of Kluyvera georgiana, a probable progenitor of a subgroup of CTX-M Thi, T.D., Lopez, E., et al., 2011. Effect of recA inactivation on mutagenesis of
extended-spectrum beta-lactamases. Antimicrobial Agents and Chemotherapy Escherichia coli exposed to sublethal concentrations of antimicrobials. Journal
46 (12), 4038–4040. of Antimicrobial Chemotherapy 66 (3), 531–538.
Poirel, L., Rodriguez-Martinez, J.M., et al., 2005. Origin of plasmid-mediated Thiele-Bruhn, S., 2003. Pharmaceutical antibiotic compounds in soils – a review. The
quinolone resistance determinant QnrA. Antimicrobial Agents and Chemother- Journal of Plant Nutrition and Soil Science 166, 145–167.
apy 49 (8), 3523–3525. Thomas, K.V., Dye, C., et al., 2007. Source to sink tracking of selected human phar-
Pomposiello, P.J., Bennik, M.H., et al., 2001. Genome-wide transcriptional profiling of maceuticals from two Oslo city hospitals and a wastewater treatment works.
the Escherichia coli responses to superoxide stress and sodium salicylate. Journal Journal of Environmental Monitoring: JEM 9 (12), 1410–1418.
of Bacteriology 183 (13), 3890–3902. Thuy, H.T., Nga le, P., et al., 2011. Antibiotic contaminants in coastal wetlands from
Poole, K., 2001. Multidrug resistance in Gram-negative bacteria. Current Opinion in Vietnamese shrimp farming. Environmental Science and Pollution Research
Microbiology 4 (5), 500–508. International 18 (6), 835–841.
Poole, K., 2002. Mechanisms of bacterial biocide and antibiotic resistance. Journal of Tkachenko, O., Shepard, J., et al., 2007. A triclosan-ciprofloxacin cross-resistant
Applied Microbiology 92 (Suppl.), 55S–64S. mutant strain of Staphylococcus aureus displays an alteration in the expression of
Putman, M., Van Veen, H.W., et al., 2000. Antibiotic resistance: era of the multidrug several cell membrane structural and functional genes. Research in Microbiology
pump. Molecular Microbiology 36 (3), 772–773. 158 (8–9), 651–658.
Randall, L.P., Cooles, S.W., et al., 2007. Commonly used farm disinfectants can select Tumah, H.N., 2009. Bacterial biocide resistance. Journal of Chemotherapy 21 (1),
for mutant Salmonella enterica serovar Typhimurium with decreased suscep- 5–15.
tibility to biocides and antibiotics without compromising virulence. Journal of van de Sande-Bruinsma, N., Grundmann, H., et al., 2008. Antimicrobial drug use and
Antimicrobial Chemotherapy 60 (6), 1273–1280. resistance in Europe. Emerging Infectious Diseases 14 (11), 1722–1730.
Ren, L., Rahman, M.S., et al., 1999. Escherichia coli cells exposed to strep- Verlicchi, P., Galletti, A., et al., 2010. Management of hospital wastewaters: the
tomycin display a mutator phenotype. Journal of Bacteriology 181 (3), case of the effluent of a large hospital situated in a small town. Water Science
1043–1044. and Technology: A Journal of the International Association on Water Pollution
Richter, S.N., Frasson, I., et al., 2011. Transfer of KPC-2 carbapenemase from Klebsiella Research 61 (10), 2507–2519.
pneumoniae to Escherichia coli in a patient: first case in Europe. Journal of Clinical Wahlberg, C., Bjorlenius, B., et al., 2011. Fluxes of 13 selected pharmaceuticals in the
Microbiology 49 (5), 2040–2042. water cycle of Stockholm, Sweden. Water Science and Technology: A Journal of
Rooklidge, S.J., 2004. Environmental antimicrobial contamination from terraccumu- the International Association on Water Pollution Research 63 (8), 1772–1780.
lation and diffuse pollution pathways. The Science of the Total Environment 325 Wei, R., Ge, F., et al., 2011. Occurrence of veterinary antibiotics in animal wastewater
(1–3), 1–13. and surface water around farms in Jiangsu Province, China. Chemosphere 82
Salverda, M.L., De Visser, J.A., et al., 2010. Natural evolution of TEM-1 beta-lactamase: (10), 1408–1414.
experimental reconstruction and clinical relevance. FEMS Microbiology Review Weigel, L.M., Clewell, D.B., et al., 2003. Genetic analysis of a high-level vancomycin-
34 (6), 1015–1036. resistant isolate of Staphylococcus aureus. Science 302 (5650), 1569–1571.
Sanchez, P., Moreno, E., et al., 2005. The biocide triclosan selects Stenotrophomonas Wright, G.D., 2010. Antibiotic resistance in the environment: a link to the clinic?
maltophilia mutants that overproduce the SmeDEF multidrug efflux pump. Current Opinion in Microbiology 13 (5), 589–594.
Antimicrobial Agents and Chemotherapy 49 (2), 781–782. Yang, D., Guo, Y., et al., 2009. Combined porin loss and extended spectrum beta-
SCENIHR, 2009. Assessment of the antibiotic resistance effects of biocides, 19 January lactamase production is associated with an increasing imipenem minimal
2009. In: E.C. Scientific Committee on Emerging and Newly Identified Health inhibitory concentration in clinical Klebsiella pneumoniae strains. Current Micro-
Risks, Brussels. biology 58 (4), 366–370.
Seifrtova, M., Pena, A., et al., 2008. Determination of fluoroquinolone antibiotics in Yiruhan, Wang, Q.J., et al., 2010. Determination of four fluoroquinolone antibi-
hospital and municipal wastewaters in Coimbra by liquid chromatography with otics in tap water in Guangzhou and Macao. Environmental Pollution 158 (7),
a monolithic column and fluorescence detection. Analytical and Bioanalytical 2350–2358.
Chemistry 391 (3), 799–805. Yoshida, H., Bogaki, M., et al., 1990. Quinolone resistance-determining region in the
Sekiguchi, J., Asagi, T., et al., 2005. Multidrug-resistant Pseudomonas aeruginosa strain DNA gyrase gyrA gene of Escherichia coli. Antimicrobial Agents and Chemother-
that caused an outbreak in a neurosurgery ward and its aac(6 )-Iae gene cassette apy 34 (6), 1271–1272.
encoding a novel aminoglycoside acetyltransferase. Antimicrobial Agents and Ysern, P., Clerch, B., et al., 1990. Induction of SOS genes in Escherichia coli and mutage-
Chemotherapy 49 (9), 3734–3742. nesis in Salmonella typhimurium by fluoroquinolones. Mutagenesis 5 (1), 63–66.
Sekiguchi, J., Asagi, T., et al., 2007. Outbreaks of multidrug-resistant Pseudomonas Zou, S., Xu, W., et al., 2011. Occurrence and distribution of antibiotics in coastal water
aeruginosa in community hospitals in Japan. Journal of Clinical Microbiology 45 of the Bohai Bay, China: impacts of river discharge and aquaculture activities.
(3), 979–989. Environmental Pollution 159 (10), 2913–2920.

You might also like