You are on page 1of 22

REVIEW ARTICLE

A global view of antibiotic resistance


Jose Luis Martinez1,2, Alicia Fajardo1,2, Leonor Garmendia1,2, Alvaro Hernandez1,2, Juan Francisco
Linares1,2, Laura Martı́nez-Solano1,2 & Marı́a Blanca Sánchez1,2
1
Departamento de Biotecnologı́a Microbiana, Centro Nacional de Biotecnologı́a, CSIC, Cantoblanco, Madrid, Spain; and 2CIBERESP, Spain

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


Correspondence: Jose Luis Martinez, Abstract
Departamento de Biotecnologı́a Microbiana,
Centro Nacional de Biotecnologı́a, CSIC,
Antibiotic resistance is one of the few examples of evolution that can be addressed
Darwin 3, Cantoblanco, 28049-Madrid, experimentally. The present review analyses this resistance, focusing on the
Spain. Tel.: +34 915854542; fax: +34 networks that regulate its acquisition and its effect on bacterial physiology. It is
915854506; e-mail: jlmtnez@cnb.csic.es widely accepted that antibiotics and antibiotic resistance genes play fundamental
ecological roles – as weapons and shields, respectively – in shaping the structures of
Present address: Juan Francisco Linares, microbial communities. Although this Darwinian view of the role of antibiotics is
Department of Molecular Oncogenesis, still valid, recent work indicates that antibiotics and resistance mechanisms may
Genome Research Institute, University of
play other ecological roles and strongly influence bacterial physiology. The
Cincinnati College of Medicine, Cincinnati,
OH 45229, USA.
expression of antibiotic resistance determinants must therefore be tightly regulated
and their activity forms part of global metabolic networks. In addition, certain
Received 3 July 2008; revised 10 October 2008; bacterial modes of life can trigger transient phenotypic antibiotic resistance under
accepted 10 October 2008. some circumstances. Understanding resistance thus requires the analysis of the
First published online 19 November 2008. regulatory networks controlling bacterial evolvability, the physiological webs
affected and the metabolic rewiring it incurs.
DOI:10.1111/j.1574-6976.2008.00142.x

Editor: Victor de Lorenzo

Keywords
antibiotic resistance; bacterial evolution; fitness
cost; hypermutation; horizontal gene transfer;
HGT.

procedures once previously taken for granted could be


Introduction conceivably consigned to medical limbo. The repercussions
Bacterial pathogens, once susceptible to antimicrobial are almost unimaginable’ (World Health Organization,
agents, are becoming increasingly resistant. In addition, 2000).
hospitals are now facing problems caused by new opportu- Nonetheless, antibiotic resistance is one of the few
nistic pathogens (from nonclinical environments) that show examples of evolution that can be studied in real time
little susceptibility to antibiotics (Quinn, 1998; Gaynes & (Martinez et al., 2007; Courvalin, 2008); this is the focus of
Edwards, 2005). Increasing antibiotic resistance poses im- the present review [the danger to health and the basic
portant risks to human health (Cohen, 1992; Levy, 1998; mechanisms of resistance have been reviewed elsewhere
Levy & Marshall, 2004); it can affect the course of infectious (Cohen, 1992; Levy, 1998; Levy & Marshall, 2004)]. This
disease (World Health Organization, 1999) and increase the paper does not, therefore, use the clinical definition of
danger associated with immunosuppression (e.g. in trans- resistance based on minimal inhibitory concentration
plantation and anticancer chemotherapy), intubation, ca- breakpoints (Turnidge & Paterson, 2007), but rather adopts
theterization and other common procedures, all of which the wider view that it is represented by any mechanism that
rely on antibiotics to overcome the infections with which reduces susceptibility to antibiotics. Antibiotic resistance
they are commonly associated. As stated by the World can come about either as the consequence of genetic change
Health Organization, the spread of antibiotic-resistant (mutations) (Martinez & Baquero, 2000) or by the acquisi-
bacteria at hospitals ‘means that commonplace medical tion of antibiotic resistance genes (Davies, 1997) through


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 45

horizontal gene transfer (HGT). Some organisms have a pathogens of resistance genes that originated in antibiotic-
characteristic phenotype of low susceptibility to antibiotics producing microorganisms [perhaps even via DNA contam-
(Bonomo & Szabo, 2006; Fajardo et al., 2008), acquired ination of antibiotic preparations (Webb & Davies, 1993)]
before the use of antibiotics in medicine. still affords a compelling explanation for the rise of anti-
A global analysis of the phenomenon of antibiotic resis- biotic resistance.
tance, understood as an example of bacterial adaptation to If the only roles of antibiotics and their resistance
stressful conditions (Hamilton-Miller, 2004), requires that elements are to serve as weapons and shields, respectively,
the different features of this process be addressed. These the study of antibiotic resistance would not require a global
include the physiological role of resistance determinants, the approach, an idea this review reiterates throughout. Most of
regulatory networks that modulate genetic events leading to the several thousand works so far published on antibiotic
resistance (namely mutation, recombination and HGT), the resistance are based on the implicit belief that resistance is a

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


overall effect on the bacterial physiology of the acquisition of very specific phenomenon, in which any given mechanism
resistance and the existence of global metabolic networks of resistance is simply the defence response to the presence
that mediate bacterial susceptibility to antibiotics. Non- of a given antibiotic. However, recent work indicates that the
pathogenic bacteria may contain many determinants that situation is more complex. Whereas on some occasions the
can afford resistance, and yet just a few are encountered in weapon/shield role is still a reasonable explanation for the
pathogen populations (Martinez et al., 2007). Understand- function of antibiotics and resistance determinants in nature
ing the evolutionary process behind resistance requires a (Pang et al., 1994), alternative functional roles for resistance
global comprehension of the genetic causes and physiologi- elements are now being proposed.
cal consequences of its acquisition (Martinez & Baquero, Antibiotics have been sought as compounds capable of
2002; Martinez et al., 2007). inhibiting the growth of microbial pathogens (Waksman &
Woodruff, 1940), and on some occasions they likely play this
role in natural environments (de Lorenzo et al., 1984;
Functional role of antibiotics and
Raaijmakers et al., 2002; Haas & Defago, 2005). However, it
antibiotic resistance genes in natural should be remembered that nearly any compound can be
environments toxic when in a sufficiently high concentration. Because the
Although all environments may be said to be equally concentrations of antibiotics used for therapy can be much
natural, this review uses the terms ‘natural environments’ higher than those encountered in natural environments, it
or ‘environmental bacteria’ to describe those with no hu- may be that, like other toxic compounds, they have a
man-associated (infective or commensal) evolutionary link. hormetic effect (Fig. 1), i.e. at low concentrations they
Rapid evolution towards resistance to antibiotics (the con- induce responses in their target organisms that are actually
sequence of their use in the treatment of human infections) beneficial, but become toxic at high concentrations.
is unlikely in environmental bacteria; this contrasts sharply It has been suggested that antibiotics might serve as
with the situation for human pathogens. signalling molecules at the low concentrations in which they

Antibiotics as signals
The search for antibiotics in natural ecosystems was origin-
ally based on the idea that the latter contained compounds
that inhibited the growth of pathogens; certainly, pathogens
are rarely found in the environment (Waksman & Woodruff,
1940). The extraordinary success of this approach, plus the
fact that several of the antibiotics used for treating infections
are synthesized by soil microorganisms, led to the proposi-
tion that, in natural environments, antibiotics have the
function of inhibiting the growth of competitors – an
example of the Darwinian idea of ‘the struggle for life’. Fig. 1. Hormesis and the effect of antibiotics on bacterial physiology.
Conversely, antibiotic resistance genes must be elements that Hormesis has been defined as the property of a compound that is
beneficial at low concentrations and toxic at high concentrations. At
evolved to avoid the activity of antibiotics; work on the
low concentrations, antibiotics can trigger the expression of a specific set
molecular basis of antibiotic production has clearly shown of genes that may be beneficial to bacteria. However, at higher
that antibiotic producers usually possess determinants that concentrations, stress responses are induced and bacterial growth is
afford them resistance to the antimicrobial agents they inhibited. The white box indicates the region of beneficial, adaptive
produce (Benveniste & Davies, 1973). The acquisition by responses and the grey box indicates the region of harmful effects.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
46 J.L. Martinez et al.

are usually found in natural habitats (Davies, 2006; Yim (Alonso et al., 1999b; Alonso & Martinez, 2001; Sanchez
et al., 2006a, b, 2007; Fajardo & Martinez, 2008). This et al., 2004). In bacteria, it has been shown that besides
suggestion is based on the fact that low concentrations of offering resistance to antibiotics and other toxic compounds
antibiotics trigger specific transcriptional changes (Tsui (Silver & Phung, 1996; Hernandez et al., 1998; Ramos et al.,
et al., 2004; Yim et al., 2006a, b; Fajardo & Martinez, 2008) 2002; Sanchez et al., 2005), they contribute to virulence
that are independent of the microbial networks involved in (Piddock, 2006a, b), to maintaining homeostasis (Lewinson
the general stress response (Goh et al., 2002), that these & Bibi, 2001) and to the detoxification of intracellular
responses can be beneficial for the microorganisms involved metabolites (Aendekerk et al., 2005), among other func-
(Linares et al., 2006; Dietrich et al., 2008; Fajardo & tions. Mutations leading to the constitutive expression of
Martinez, 2008; Gerber et al., 2008) and that some com- these transporters afford antibiotic resistance (Alonso &
pounds previously characterized as bona fide signalling Martinez, 1997; Kohler et al., 1997; Ziha-Zarifi et al., 1999;

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


molecules also have antimicrobial activity (Ji et al., 1997; Sanchez et al., 2002a, b) and produce global changes in
Deziel et al., 2004; Kaufmann et al., 2005). bacterial metabolism specific for each MDR pump. It is
noteworthy that some of these MDR pumps can efflux signal
compounds (Evans et al., 1998; Kohler et al., 2001), indicat-
Functional roles of antibiotic resistance genes in
ing that signalling networks may be important in triggering
their original hosts
antibiotic resistance.
If antibiotics have functions other than the killing of Any enzyme capable of modifying a metabolite with a
competitors, antibiotic resistance genes may have functions structure similar to a given antibiotic might modify the
other than providing resistance. Even in antibiotic produ- antibiotic itself and confer resistance. This is the case of
cers, the presence of an antibiotic resistance gene does not chromosomal 2 0 -N-acetyltransferase in Providencia stuartii.
necessarily imply that its original role was to help resist the This enzyme, which modifies bacterial peptidoglycan, can
action of the antibiotic produced by the host (Nodwell, inactivate gentamicin because the latter’s structure is similar
2007). For instance, Streptomyces coelicolor encodes proteins to that of the enzyme’s original substrate (Macinga &
similar in sequence and indeed in mode of action to those Rather, 1999). The fact that elements involved in bacterial
involved in resistance to vancomycin in human pathogens metabolism can be important in antibiotic resistance is
(Hong et al., 2002), even though this microorganism does further shown in that a large number of Pseudomonas
not produce glycopeptide antibiotics. aeruginosa genes contribute to its characteristic low-suscept-
One example of antibiotic resistance determinants with a ibility phenotype (Fajardo et al., 2008). It has also been
functional role other than affording resistance is provided by suggested that b-lactamases, which have a structure similar
multidrug resistance (MDR) efflux pumps (Fig. 2). These to penicillin-binding proteins (PBPs, the targets of b-lactam
transporters are present in all organisms (Saier & Paulsen, antibiotics), might have originally been PBPs themselves,
2001; Piddock, 2006a, b; Lubelski et al., 2007), and the same involved in the synthesis of peptidoglycan. Their activity
MDR pumps are present in all strains of a given species against b-lactam antibiotics may have been a side effect
of their original function (Massova & Mobashery, 1998;
Meroueh et al., 2003).
In summary, the fact that antibiotics trigger specific
transcriptional responses, together with the role that deter-
minants involved in signal trafficking or bacterial metabo-
lism might have in resistance, supports the idea that
antibiotic resistance should be examined from a more global
viewpoint. Only then can the bacterial networks involved in
resistance, as well as the changes in bacterial physiology
associated with a phenotype, be addressed. These changes
might be important in the context of human infections and
in the survival and dissemination of resistant bacteria.

Regulation of evolvability and acquired


Fig. 2. Structure of a tripartite efflux pump. The tripartite efflux pumps antibiotic resistance
of Gram-negative bacteria are formed by an integral membrane protein,
whose activity is commonly linked to the membrane proton motive force, Resistance to antibiotics can be broadly classified as either
an outer membrane protein and a periplasmic protein. IM, inner intrinsic (Hogan & Kolter, 2002; Fajardo et al., 2008; Tamae
membrane; OM, outer membrane. et al., 2008) (due to the mechanisms present in all the strains


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 47

of a given bacterial species) or acquired (Davies, 1994; rates (Gomez-Gomez et al., 1997; Bjedov et al., 2003; Foster,
Alekshun & Levy, 2007) (due to the acquisition of a specific 2005; Saint-Ruf & Matic, 2006). Recently, the faster growth
mechanism of resistance, the consequence of mutation, of resistant mutants has been suggested to be the cause of
recombination or HGT). Studies of experimental evolution increased mutation frequencies with respect to rifampicin
using Metazoans as models are virtually impossible because resistance in ‘ageing’ bacterial colonies (Wrande et al.,
of the time and population sizes required (Navas et al., 2008).
2007), but the acquisition of antibiotic resistance by bacteria Key components of the mismatch repair system are
provides an example of an evolutionary process that can be downregulated under the control of the general-stress sigma
readily addressed experimentally. Learning about the me- factor RpoS (Tsui et al., 1997). In addition, the SOS response
chanisms that lead to resistance has implications for under- to DNA damage (Erill et al., 2007) triggers the transcription
standing evolution as a whole. Because genome changes are of several genes, one of which is dinB, which codes for the

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


the basis of any evolutionary process, the probability of such error-prone DNA polymerase IV (a Y family DNA polymer-
events occurring represents the first point of control in the ase) (Kim et al., 1997; Godoy et al., 2007). This enzyme is
development of resistance. In recent years, it has become able to bypass DNA lesions that block chain elongation by
evident that the ability to evolve [i.e. evolvability – see replicative DNA polymerase. In addition, PolIV shows less
Kirschner & Gerhart (1998) for an in-depth review of this fidelity than proofreading polymerases when operating on
concept] is a regulated phenomenon that must be under- undamaged DNA (Wagner et al., 1999). When PolIV is
stood from a global viewpoint (Pepper, 2003; Colegrave & overproduced, it introduces mutations (which may afford
Collins, 2008; Isalan et al., 2008; Pigliucci, 2008; Schlichting, antibiotic resistance) at a high rate (Kim et al., 1997). It has
2008). Some aspects of the networks regulating evolvability been shown that dinB expression forms part of a large
that can lead to the acquisition of antibiotic resistance are regulatory network that includes the LexA-controlled SOS
discussed below. regulon (Kim et al., 1997; Godoy et al., 2007), RpoS
polymerase (Layton & Foster, 2003), the histone-like protein
HU (Williams & Foster, 2007) and the polyphosphate kinase
Regulation of mutation rates
(Stumpf & Foster, 2005), among others.
Bacterial populations tend to have low mutation rates, Bacterial mutation rates (and thus the probability of
which helps preserve their genomes and avoid lethal muta- mutation-driven resistance) are therefore subject to com-
tions. DNA error correction mechanisms bring bacterial plex regulation that we are now just beginning to under-
evolution speeds down by around 1000-fold below their stand. Because this regulation can be triggered by
theoretical natural speed limit (Zeldovich et al., 2007). Some environmental or metabolic signals (Bjedov et al., 2003), it
authors report, however, that the mutation rate (and thus is important to know which situations might increase the
the speed of evolution) of some bacterial subpopulations is mutation rate and thus the likelihood of acquiring resis-
higher than normal (LeClerc et al., 1996; Matic et al., 1997; tance. Quinolones (Phillips et al., 1987) and b-lactams
Oliver et al., 2000; Baquero et al., 2004, 2005), mainly due to (Miller et al., 2004) induce the SOS response, and could
defects in the DNA repair systems (Oliver et al., 2002). therefore increase the mutation rate. Recent work has shown
Bacterial hypermutators are more commonly found in popu- that PBP3 inhibition by ceftazidime induces transcription of
lations under stress, including chronic infections (Oliver the error-prone polymerase PolIV and induces mutagenesis
et al., 2000), or in the presence of strong selective elements in P. aeruginosa (Blazquez et al., 2006). Although the SOS
such as antibiotics (Macia et al., 2005) or bacteriophages (Pal system regulates the expression of dinB, other elements are
et al., 2007). The presence of hypermutators increases the also involved. For instance, studies on Escherichia coli have
probability of acquiring resistance (Macia et al., 2005; Hen- shown that the induction of dinB by b-lactams occurs via a
richfreise et al., 2007), and antibiotics select for hypermuta- pathway independent of the SOS response (Perez-Capilla
tors (Mao et al., 1997) in such a way that they enhance the et al., 2005), highlighting that different regulatory networks
probability of resistance appearing (Blazquez et al., 2002). can modulate bacterial mutability. This view is further
The modulation of mutation frequencies may also be supported by recent work showing that subinhibitory con-
important in the development of resistance. Some works centrations of quinolones and aminoglycosides may result
report increased numbers of antibiotic-resistant mutants in in increased mutability in Streptococcus pneumoniae, inde-
bacterial populations under stress (Alonso et al., 1999a), and pendent of DinB activity (Henderson-Begg et al., 2006).
that the number of mutants in colonies of nongrowing
bacteria can increase. Two mechanisms have been proposed
Regulation of recombination
to explain this (Roth et al., 2006): one based on gene
amplification (Andersson et al., 1998; Pettersson et al., Recombination plays a major role in HGT (Thomas &
2005) and the other on the regulated increase of mutation Nielsen, 2005). Both the acquisition of DNA through

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
48 J.L. Martinez et al.

transformation and the combination of new elements (e.g. like mutation rates, change as a consequence of the
antibiotic resistance genes) into transferable replicons (e.g. structure of microbial populations (hypermutators) and as
plasmids) require recombination among DNA sequences. a response to external inputs, such as antibiotics (Hastings
Studies on E. coli have shown that efficient recombination et al., 2004).
occurs in plasmids when homologous sequences of 25 bp are
present. Increasing the length of homology enhances re-
Regulation of DNA uptake
combination (Lovett et al., 2002). Recombination is there-
fore favoured among closely related organisms or elements Transformation and conjugation usually involve the trans-
involved in HGT with similar structural features. location of single-stranded DNA through the bacterial
Some particularly important features that favour the envelope (Chen & Dubnau, 2004; Chen et al., 2005). After
spread of antibiotic resistance should be mentioned at this gaining entry, general mechanisms such as DNA modifica-

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


point. Firstly, gene-capture units exist that favour the tion/restriction (Tock & Dryden, 2005) or plasmid exclusion
acquisition of exogenous DNA and its incorporation into (Garcillan-Barcia & de la Cruz, 2008) must be bypassed by
a single element. The clearest examples are the integrons the exogenous DNA if it is to be maintained. The mechan-
(Fig. 3), genetic platforms that contain small repetitive isms that regulate bacterial DNA uptake and maintenance
sequences (typically 59 bp) that favour recombination and have been reviewed elsewhere (Chen et al., 2005; Thomas &
gene recruitment (Hall & Collis, 1995; Rowe-Magnus & Nielsen, 2005); only those aspects concerning resistance are
Mazel, 2001; Holmes et al., 2003; Mazel, 2006). These are discussed here. Natural competence involves the induction
very important in the dissemination of antibiotic resistance of a complex system (the com regulon) usually regulated by
genes (Carattoli, 2001; Rowe-Magnus & Mazel, 2002; Rowe- quorum sensing (QS) (Pestova et al., 1996; Cheng et al.,
Magnus et al., 2002). Secondly, the need for homologous 1997; Luo et al., 2003). Notably, it has recently been found
sequences to trigger recombination can be relaxed under that the same peptides that induce competence in closely
some circumstances. For instance, some hypermutator related members of the streptococci family can kill other
strains, particularly those with defects in the methyl-direc- members of the same genus, making large amounts of DNA
ted DNA-repair genes (such as mutS), recombine at high available and increasing the efficiency of lateral gene transfer
frequencies with divergent DNA (Matic et al., 2000). (Johnsborg et al., 2008). This provides an example of the
Thirdly, antibiotics such as ciprofloxacin may stimulate dual antimicrobial and signalling role of some metabolites
recombination among different sequences (Lopez et al., (discussed above) (Fajardo & Martinez, 2008).
2007). All this indicates that recombination rates, The simultaneous induction of competence in microor-
ganisms and their predation in the same environment is
particularly important for S. pneumoniae, which acquires
resistance via HGT from DNA released by related commen-
sal species (Sibold et al., 1994; Reichmann et al., 1997;
Hakenbeck, 1998). Streptococcus pneumoniae lacks an SOS-
like system, but possesses the recA gene, whose induction by
DNA-damaging agents is strictly dependent on the ability of
these compounds to trigger competence. It has been sug-
gested that the com regulon of S. pneumoniae might have
features similar to those of the SOS response regulon in
E. coli (Prudhomme et al., 2006). Because the SOS system is
Fig. 3. Structure of an integron. Integrons are formed by a gene coding induced by some antibiotics, it might be possible for
for a site-specific recombinase known as integrase (intl1), an attachment
antibiotics to induce competence in S. pneumoniae. Indeed,
site (attI) and a strong promoter that drives the expression of the genes
located downstream from the attachment site (P). Some conserved
it has been demonstrated that aminoglycosides and fluor-
genes are present at the 3 0 -end of integrons of the same type, suggest- oquinolones induce competence, and that an intact compe-
ing a common origin. The figure shows a Type I integron containing qac, tence regulatory cascade is needed (induction is lost in comA
which encodes for a protein involved in resistance to quaternary mutants) (Prudhomme et al., 2006).
ammonium disinfectants, sul1 that encodes sulphonamide resistance Antibiotics can thus accelerate DNA acquisition in
and orf5, which has no known function. Antibiotic resistance genes can S. pneumoniae by two different mechanisms: by killing
be recruited by site-specific recombination (a) driven by the integrase
susceptible neighbouring cells and increasing genetic trans-
(Intl1). After gene recruitment (b) the recombination sites are recon-
structed, allowing the incorporation of new gene cassettes. This struc-
formability (Prudhomme et al., 2006). If the bacteria that
ture allows the formation of integrons containing arrangements of anti- are resistant to a different type of antibiotic are killed, and
biotic resistance genes that can be acquired simultaneously by bacterial their DNA is released, this would increase the probability of
pathogens. S. pneumoniae acquiring that resistance.


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 49

Building blocks in HGT-acquired antibiotic pathogens. For instance, E. coli strains isolated before the era
resistance and the founder effect of antibiotics contain the same groups of plasmids as those
isolated after the introduction of antibiotics into medicine,
The elements of bacterial mobilomes are reviewed in an-
the only difference between them being the recruitment of
other article of this special issue; this paper only analyses the
resistance genes in the latter (Datta & Hughes, 1983).
features concerned with antibiotic resistance. The acquisi-
Similarly, the diversity of transposons and integrons asso-
tion and dissemination of resistance by HGT follows a
ciated with resistance is not as high as might be predicted
Chinese-box scheme. Inspection of the elements producing
from the diversity of these elements in natural ecosystems.
resistance at hospitals (Baquero et al., 2002) shows that there
Modularity is a common characteristic of biological
are predominant clones that may contain predominant
systems (Kashtan & Alon, 2005; Pereira-Leal et al., 2006).
plasmids, which in turn contain predominant elements
The rules that govern the combination of modules (Stadler
(transposons, integrons, etc.) in which antibiotic resistance

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


et al., 2001) have been analysed in different types of
genes are enclosed (Fig. 4). This hierarchical scheme of the
biological networks (Force et al., 2005; Pereira-Leal et al.,
elements involved in resistance dissemination has been
2006; Mete et al., 2008) and, more recently, in the structure
discussed in detail elsewhere (Baquero, 2004).
of bacterial genomes (Dagan et al., 2008). It has been
Although the number and variability of the elements
suggested that, like for other modular systems, there are
present in natural bacterial populations that might contri-
rules (grammar) governing the association of the blocks
bute to resistance is huge, only a few are present in human
required for building a transferable resistance element
(Baquero, 2004). This grammar has different components:
(1) functional elements: each replicon can make use of the
transcriptional and metabolic machineries of certain bacter-
ial groups but not of others; they therefore have host
specificity; (2) structural features: DNA structures that
favour the combination of some elements but not others.
On many occasions, homologous recombination is required,
so that only those elements with a high degree of sequence
identity are able to combine; (3) ecological aspects: first,
only those building blocks coexisting in the same environ-
ment can combine; second, the most abundant elements
may be favoured; and third, in the presence of strong
selection, the first element conferring a selective advantage
(antibiotic resistance) has the highest probability of spread-
ing and predominating (Fig. 5) in the bacterial population
under antibiotic threat.
The integrons (Stokes & Hall, 1989) are modular struc-
tures with a major role in the development of antibiotic
resistance. These gene-recruitment determinants (Fig. 3)
have a high impact in clustering antibiotic resistance genes,
which may later be transferred to pathogens. Although
integrase sequences in natural environments show wide
variability (Nield et al., 2001; Elsaied et al., 2007), that of
the different integrases involved in integrons containing
resistance genes in pathogenic bacteria is low. The Type 1
integrons are the most important (Walsh, 2006). If environ-
Fig. 4. Building blocks in the acquisition of HGT-mediated antibiotic ment-driven selection of integrons occurs, it may be that
resistance. The elements involved in HGT have common backbones that this type of integrase is the best adapted to infective
can incorporate different modules. Antibiotic resistance can be trans- environments. However, Type 1 integrases are found in a
ferred by plasmids that acquire transposons, which in turn can contain wide range of pathogens from E. coli to P. aeruginosa, which
integrons that recruit antibiotic resistance genes. The modular structure
come from different habitats. The founder effect explains
of the elements involved in HGT allows for rapid evolution when under
antibiotic threat. IS, insertion sequences; tra, transference genes; psk,
the dissemination of the most common types of integrons in
postsegregational killing; mrs, multimer resolution; rep, replication; cop, human pathogens. For instance, Type 1 integrons contain a
copy number control; par, partitioning. The figure of the plasmid is conserved gene coding for resistance to sulphonamides
modified from Thomas (2000). (Stokes & Hall, 1989), a family of antimicrobial agents

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
50 J.L. Martinez et al.

whereas the variability of HGT-acquired resistance determi-


nants in human pathogens is low. A founder effect (Fig. 5)
also explains the presence of some antibiotic resistance genes
but not of others in bacterial pathogens. Under stable,
strong selective pressure, the first determinant conferring
resistance to this pressure would predominate and spread in
the population. Only after the selective forces change does
diversification of the antibiotic resistance determinants
usually occur. This is likely the situation with plasmid-
encoded b-lactamases. The first b-lactamase described in
Fig. 5. The founder effect in the acquisition of antibiotic resistance
bacterial pathogens was TEM-1. This enzyme, which inacti-

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


genes. Natural environments (grey box) contain a large number of vates the first class of b-lactams, has been prevalent for
antibiotic resistance genes (letters). Some (Greek letters) cannot be several years in resistant enteric Gram-negative bacilli (Roy
transferred to bacterial pathogens (white box) because they do not share et al., 1983), whereas another b-lactamase, SHV-1, is pro-
the same ecosystem or because they have not been incorporated into duced by the vast majority of resistant Klebsiella pneumoniae
gene-transfer elements compatible with human pathogens (a). Although strains (Roy et al., 1983; Livermore, 1995). After the
several resistance genes (Roman letters) can be transferred, only a few
introduction of b-lactamase inhibitors and new generations
are currently present in infectious bacteria (darker grey box in b). If the
selective pressure does not change, the same antibiotic resistance genes
of b-lactams, a burst in TEM-1- and SHV-1-derived en-
are maintained and spread in the population (c), impeding the entrance zymes occurred (Paterson & Bonomo, 2005) – an example
of others that could play a similar role (founder effect). If the selective of accelerated evolution under stress (Medeiros, 1997). In
forces change (e.g. the appearance of new antibiotics belonging to the addition, novel families of b-lactamases (Paterson & Bono-
same family, darkest grey box in d), the original resistance gene evolves mo, 2005) began to be acquired by human pathogens,
into more efficient variants (white ‘a’ letters in d) with different indicating that a change in selective force is required for the
specificities.
diversification of antibiotic resistance determinants (Fig. 5).

widely used in the first half of the 20th century. It might be


suggested that, under strong selective pressure, the first
Effect of resistance in bacterial physiology
integron containing a sulphonamide resistance gene was It is generally accepted that the acquisition of antibiotic
selected, and that this became the ancestor of the integrons resistance produces a global burden on bacterial metabo-
now found in clinical settings. It is important to note that lism (fitness cost), making resistant bacteria less competitive
sulphonamide resistance is still widespread in the United than their susceptible partners (Andersson & Levin, 1999;
Kingdom despite national prescribing restrictions (Enne Andersson, 2006). This idea is based on the fact that muta-
et al., 2001), probably because of the success of these tions leading to resistance occur in genes that are important
integrons in recruiting other antibiotic resistance genes. for bacterial physiology (lethal targets, transporters, etc.). A
The full sequences of different plasmids of the same modification of the target proteins should cause deadapta-
family can provide new clues regarding the evolution of the tion from their normal functional role, leading to less
elements involved in the HGT of antibiotic resistance genes. efficient bacterial metabolism. Similarly, acquisition of a
The sequences of five IncW plasmids show that two different resistance gene by HGT, for instance due to plasmid transfer,
pathways were involved in the acquisition of resistance genes should be associated with a metabolic cost, the consequence
by the common plasmid backbone. Three plasmids contain of the replication, transcription and translation of the genes
a single Integron I platform, in the same position in each present in the new replicon (Bouma & Lenski, 1988). These
plasmid, with different gene cassettes. In the other two fitness costs will be reflected in a reduced growth rate. Thus,
plasmids, resistance arose because of the insertion of several most studies on fitness costs have involved in vitro analyses
transposons containing antibiotic resistance genes (Revilla using defined growth media.
et al., 2008). The best explanation for this is that, in the Nevertheless, some authors indicate that the effect of
former three plasmids, the integron was inserted once into antibiotic resistance on bacterial physiology is more com-
the IncW backbone, followed by diversification of the plex and more specific than previously thought. For in-
integron arrangements, while it evolved independently from stance, it has been shown that the mutations that
an IncW ancestor in the other two plasmids (Revilla et al., compensate for fitness defects are different in vivo and in
2008). vitro (Bjorkman et al., 2000), and that the same antibiotic
The number of antibiotic resistance genes in nonclinical resistance mutation can produce opposite effects in two
environments is huge (Alonso et al., 2001; D’Acosta et al., different strains of the same bacterial species (Luo et al.,
2006; Wright, 2007; Martinez, 2008; Sanchez et al., 2008), 2005). To understand in depth the effect of resistance on


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 51

bacterial physiology, the concepts and tools used in the field rifampin resistance likely led to greater fitness of the
of system biology must be used. Current models for analys- resistant mutants (compared with the wild-type strains) for
ing bacterial infections are based on the use of bacterial the soil environment.
strains that are susceptible to antibiotics. With the increase Enhanced fitness of antibiotic-resistant mutants for a
in resistance in bacterial populations, models using resistant given environment is not as rare as previously thought.
strains – if their physiological properties (including viru- Bacteria can acquire compensatory mutations that reduce
lence) are different – should be used as well. fitness costs (Bjorkman et al., 2000; Andersson, 2006), but
sometimes resistance confers an advantage, even in the
absence of selective pressure. For example, high-level quino-
Alterations in metabolic networks as the
lone resistance is mainly due to mutations in the topoi-
consequence of antibiotic resistance mutations
somerase genes, which encode the targets of this family of

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


In this section, and for the rest of this review, the term antibiotics (Martinez et al., 1998; Piddock, 1999). Because
‘mutation’ is used in its broadest sense, and includes point topoisomerases regulate DNA supercoiling, and DNA super-
mutations, deletions, insertions and intrachromosomal re- coiling may regulate gene expression (Galan & Curtiss, 1990;
combination. As stated above, mutations that produce Aleixandre et al., 1991; Dorman, 1991), it is likely that
resistance to antibiotics occur in genes that usually play mutations in the topoisomerases lead to changes in the
important roles in bacterial physiology. The products of bacterial transcriptome. Some mutations in S. pneumoniae
those genes form part of complex and highly integrated topoisomerases are associated with fitness costs when mea-
metabolic and regulatory networks. Alterations in their sured in in vitro tests, whereas other mutations have no
activities likely alter the cell’s entire metabolism. Recent associated costs (Johnson et al., 2005; Rozen et al., 2007;
work has shown, however, that the rewiring of E. coli Balsalobre & de la Campa, 2008). This is consistent with the
metabolic networks is usually well tolerated (Isalan et al., fact that different mutations in the topoisomerases alter
2008). Thus, metabolic alterations arising from antibiotic their activity by different degrees, changing global bacterial
resistance are not necessarily manifested as generally re- metabolism in different ways. It might be predicted that only
duced bacterial fitness for all possible environments. On the those mutants with low costs should predominate, which is
contrary, a certain degree of specificity and even gain of often the case. However, the situation is generally more
fitness might be expected. For instance, a Stenotrophomonas complex. Work involving an in vivo model of chicken
maltophilia mutant that overproduces the MDR efflux colonization by Campylobacter jejuni has shown that a
pump SmeDEF (Alonso & Martinez, 2000) shows impaired single-point quinolone-resistance mutation in the gyrA gene
virulence in a Dyctiostelium discoideum model, but is more can produce enhanced persistence in the avian host (Luo
proficient in the utilization of sugars such as gentibiose, et al., 2005). Although the basis for this increased fitness has
dextrin or mannose as well as the C1 carbon source formic not been studied in depth, it is conceivable that transcrip-
acid (Alonso et al., 2004). This indicates that the rewiring of tional changes caused by alterations in the activity of the
metabolism has occurred rather than the appearance of any topoisomerase GyrA affect the expression of the C. jejuni
general burden (fitness cost). Some recent data on the genes involved in chicken colonization. This enhanced
metabolic profiling of antibiotic-resistant mutants suggest fitness is host specific. In fact, the same mutation in two
that this may be common. For example, resistance to different C. jejuni strains belonging to the same clonal
rifampin is due to mutations in the gene rpoB that encodes complex had opposite effects: enhanced persistence for one
the RNA polymerase b subunit (Trinh et al., 2006). Some and reduced fitness for the other (Luo et al., 2005). Because
authors indicate that changes in the rifampin-binding the metabolic and regulatory networks of different strains of
pocket result in alterations in the interactions of the RNA the same bacterial species can have subtle differences with
polymerase of Bacillus subtilis with promoters and tran- effects on their interaction with the host (Carilla-Latorre et al.,
scriptional regulators, leading to important changes in 2008), network rewiring might also have different conse-
phenotypes for sporulation, germination and competence quences. This type of strain-specific response to an external
in DNA-mediated transformation (Maughan et al., 2004). In input is manifested by the very different transcriptomic
the latter work, some rifampin-resistant mutants, but not changes shown by two strains of P. aeruginosa in response to
others, showed reduced growth rates in a rich medium. the same type of oxidative stress (Salunkhe et al., 2005).
However, the mutants were found to be capable of using Two conclusions can be drawn from the above studies: (1)
b-glucosides, nutrients present in soil [produced from de- the effect of high-level quinolone resistance on bacterial
composing plant matter through the breakdown of celluloses physiology is specific for the mutation and strain involved
and hemicelluloses (Zhang et al., 2007)], more effectively and (2) small genetic changes, such as single-point muta-
than their susceptible counterparts (Perkins & Nicholson, tions leading to antibiotic resistance, may produce dramatic,
2008). Therefore, the metabolic rewiring produced by yet specific, changes in bacterial physiology.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
52 J.L. Martinez et al.

The specificity of bacterial responses to mutations leading infections (Jalal et al., 2000; Henrichfreise et al., 2007).
to resistance is perhaps best explained by studying mutants Further, experimental antibiotic therapy with ciprofloxacin
that overproduce MDR efflux pumps. Pseudomonas aerugi- in P. aeruginosa-infected mice selects MexCD-OprJ over-
nosa has several genes coding for them. The expression of producers (Macia et al., 2006). Although these results are
MDR pumps is commonly downregulated (Grkovic et al., not fully conclusive, they suggest that strains of P. aeruginosa
2002), but mutations in their transcriptional regulators may overproducing MexCD-OprJ might be more fit than other
lead to their constitutive expression and hence reduced antibiotic-resistant mutants with respect to the production
susceptibility to several antibiotics. Because MDR pumps of chronic infection.
efflux a wide range of substrates it is conceivable that their Because antibiotic resistance may be linked to global
constitutive expression might produce a global, nonspecific bacterial metabolism, genome-scale reconstructions of me-
metabolic burden – the consequence of the noncontrolled tabolic networks can provide insights into resistance. This

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


release of metabolites from bacterial cytoplasm. Some has been addressed in the small-colony-variants (SCVs) of
authors report, however, that the effect of MDR overpro- Staphylococcus aureus. SCVs are a type of mutant produced
duction is specific for each MDR pump. For instance, by different bacterial species (Proctor et al., 2006) and are
constitutive overexpression of MexAB-OprM or MexEF- characterized by slow growth, changes in their susceptibility
OprN is associated with defects in P. aeruginosa QS because to antibiotics and sometimes changes in their virulence
these MDR pumps efflux QS autoinducers (Evans et al., properties. SCVs are frequently found in biofilms (Haussler,
1998; Kohler et al., 2001). On the other hand, overexpres- 2004; Allegrucci & Sauer, 2007) and in chronic infections
sion of either MexEF-OprN or MexCD-OprJ, but not of (Haussler et al., 1999; von Gotz et al., 2004). The genetic
MexXY or MexAB-OprN, reduces P. aeruginosa Type 3 causes of SCVs are variable, but on some occasions, the
secretion (T3S) and cytotoxicity independent of the QS phenotype is linked to the interruption of the electron
response (Linares et al., 2005). Finally, mutants overexpres- transport chain. To analyse the effect of the acquisition of
sing either MexAB-OprM or MexCD-OprJ produce more an SCV phenotype, an S. aureus mutant lacking the haeme-
biofilm in in vitro static assays than their parental wild-type requiring cytochrome-b oxidase was constructed and its
strain (Sanchez et al., 2002b). phenotype was analysed in the context of the reconstructed
The above results show that the effect of overproducing S. aureus metabolic network (Heinemann et al., 2005).
one MDR pump or another is rather specific and not the Under anaerobic conditions, wild-type S. aureus produces
consequence of a general metabolic burden. Work at our energy from glucose via fermentation, whereas under aero-
laboratory (J.F. Linares et al., unpublished data) indicates biosis it uses glycolysis, the pentose phosphate pathway and
that the overexpression of MDR pumps can lead to global the triacarboxylic acid cycle. The predictions of the model,
changes in the P. aeruginosa transcriptome, and that these confirmed by experimental data, were that the SCV hemB
changes show a high degree of specificity. An important mutant would have an essentially fermentative metabolism
question to address is the effect of such changes on even in the presence of oxygen. Some amino acids such as
P. aeruginosa virulence. It was earlier stated that overpro- glutamate, arginine or glutamine enhanced growth by
duction of MDR pumps reduces this (Cosson et al., 2002; affording additional energy production, while isoleucine,
Hirakata et al., 2002), at least in models that resemble acute leucine, valine and lysine enhance growth because they can
infection. However, P. aeruginosa is a common cause of be used directly in protein synthesis. A third group, includ-
chronic infection and the virulence factors required for ing glycine and alanine, is not used at all by these bacteria
producing chronic and acute infections differ (Goodman (Heinemann et al., 2005). This evidence supports the idea
et al., 2004; Ventre et al., 2006). Isolates from acute infec- that resistance in SCV isolates is associated with the rewiring
tions are commonly cytotoxic and produce large amounts of of S. aureus metabolism.
proteases and siderophores but little in the way of biofilms.
In contrast, the evolution of P. aeruginosa during chronic
Alterations in metabolic networks as the
infection renders strains with the opposite phenotype
consequence of acquiring foreign antibiotic
(Martinez-Solano et al., in press). It should be noted that a
resistance genes
mutant overproducing MexCD-OprJ has low cytotoxicity,
produces only small amounts of protease and is impaired in Whereas chromosomal mutations leading to resistance
T3S (and thus cytotoxicity) – the phenotypes observed in likely alter bacterial metabolism, the situation may not be
P. aeruginosa strains obtained from chronic infections. It has the same with respect to acquired genes. Although on some
been shown that the causes of quinolone resistance in occasions the expression of HGT-acquired antibiotic resis-
P. aeruginosa involved in acute infections usually involve tance genes is regulated (Depardieu et al., 2007), they are
mutations in the topoisomerase genes, whereas overproduc- sometimes under the control of constitutive promoters
tion of MDR pumps is common in isolates from chronic (Fig. 3). In other words, these genes do not take part in the


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 53

same regulatory networks as in their original hosts. Further, activities of PBPs. For instance, PBP5 is a low-affinity PBP
the proteins they encode find themselves in a different responsible for resistance to ampicillin and third-generation
metabolic context, sometimes without their original bio- cephalosporins. The induction of VanA-mediated glycopep-
chemical substrates or without the protein partners required tide resistance increases susceptibility to b-lactam antibio-
for their activity. Under such circumstances, their only tics, indicating that the D,D-transpeptidase PBP5 does not
function is resistance (if the corresponding antibiotic is function properly with peptidoglycan ending in D-Lac.
present); such situations are said to represent ‘functional
shifting’ (Martinez, 2008). Under such circumstances, a Antibiotic-dependent bacterial growth
global metabolic burden can be expected; in fact, it has been
The existence of bacterial mutants dependent on the pre-
shown that the acquisition of a plasmid produces a meta-
sence of an antibiotic for their growth was recognized soon
bolic burden that is rapidly compensated by mutations in
after antibiotics became available in medicine (Gocke &

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


both the plasmid and the chromosome (Bouma & Lenski,
Finland, 1950; Barber, 1953; Worthington et al., 1999). Such
1988).
dependence does not mean that bacteria use antibiotics as a
However, on some occasions, HGT-acquired antibiotic
food resource (Dantas et al., 2008). Because dependence is
resistance determinants may encounter a metabolic sub-
usually linked to resistance, the underlying reason might be
strate/protein partner in the new host, so that specific
the metabolic rewiring produced by the resistance mechan-
changes in the host’s metabolism become possible. For
ism now only allowing the growth of the bacterium in the
instance, it has been found that the expression of the b-
presence of the antibiotic. The causes of antibiotic-depen-
lactamase AmpC by a plasmid leads to a reduction in the
dent growth have been studied in vancomycin-resistant
growth rate and reduced invasiveness in Salmonella enterica
enterococci (Baptista et al., 1997; Sifaoui & Gutmann,
serotype Typhimurium. Changes in colony morphology are
1997; Van Bambeke et al., 1999). As stated above, resistance
also seen (Morosini et al., 2000). Such changes are not
to glycopeptides involves the formation of a D-Ala-D-Lac
observed when the plasmid also contains the ampC repres-
depsipeptide. Under strong selective pressure and in the
sor AmpR, indicating that they are not the consequence of
presence of the vancomycin-resistance cluster, the activity of
the metabolic load caused by plasmid replication. Nor are
the host D-Ala : D-Ala ligase is not required because the
such alterations seen when another b-lactamase is expressed,
bacteria make use of an alternative ligase involved in
indicating that the observed effects are not the consequence
resistance (VanA or VanB) that ligates D-Lac to D-Ala. Thus,
of metabolic load caused by plasmid translation. It is thus
the ddl gene encoding the host D-Ala : D-Ala ligase accumu-
clear that AmpC production reduces the invasiveness of
lates mutations and bacteria can only grow in the presence
S. enterica in a specific manner. Although not reported, the
of the antibiotic because it induces expression of the
fact that AmpC may be involved in peptidoglycan synthesis,
alternative, antibiotic-resistance-linked ligase (Baptista
together with the evidence that protein–peptidoglycan in-
et al., 1997; Sifaoui & Gutmann, 1997; Van Bambeke et al.,
teractions modulate the assembly of the T3S translocon in
1999).
Salmonella (Pucciarelli & Garcia-del Portillo, 2003), suggests
that AmpC overproduction might produce subtle altera-
Remodelling bacterial structure as the
tions in the bacterial envelope that might alter T3S.
consequence of antibiotic resistance
Resistance to glycopeptides in Gram-positive bacteria,
mediated by the vanA gene cluster, is a well-studied example The first careful biochemical studies on the changes in
of the integration of a resistance determinant into bacterial bacterial structure as the consequence of antibiotic resis-
metabolism (for a review, see Mainardi et al., 2008). This tance examined the effect of penicillin resistance on the
cluster, present in the transposon Tn1546 (Arthur et al., structure of pneumococcal cell walls (Garcia-Bustos et al.,
1993), encodes VanH dehydrogenase, which reduces pyru- 1988; Garcia-Bustos & Tomasz, 1990). Penicillin resistance
vate to form D-Lac, and VanA ligase, an enzyme that in this bacterial species is due to the molecular remodelling
catalyses the formation of an ester bond between D-Ala and of the cell wall synthesis enzymes (PBPs), and consequently
D-Lac. This pathway forms the depsipeptide D-Ala-D-Lac, a different biosynthetic activity of the cell wall. It is reported
which is used for the biosynthesis of peptidoglycan with a that, while the peptidoglycan of antibiotic-susceptible
structure that ends in D-Lac instead of D-Ala (Arthur et al., strains contains monomeric and oligomeric forms of pri-
1992; Handwerger et al., 1992). The resistance mechanisms marily linear stem peptides with the sequence L-Ala-D-iGln-
are therefore integrated into the physiology of the bacteria. L-Lys-D-Ala (where iGln is isoglutamine), the major peptide
Nevertheless, this integration somehow alters microbial species of the cell walls of resistant bacteria are branched-
metabolism. The substitution of D-Ala by D-Lac is well stem peptides carrying Ala–Ser or Ala–Ala dipeptides on the
tolerated by the enzymes involved in the peptidoglycan E-amino groups of lysine residues (Garcia-Bustos & Tomasz,
assembly, but the modified precursor challenges the in vivo 1990). This remodelling of the cell wall structure has been

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
54 J.L. Martinez et al.

observed for other antibiotics targeting peptidoglycan bio- best-studied bacterial networks involved in antibiotic resis-
synthesis and has been recently reviewed in Mainardi et al. tance. Since its first description, the mar system has been
(2008). Resistance to two classes of antibiotics, namely identified in several bacterial genera, mainly belonging to
glycopeptides and b-lactams, strongly modifies the structure Enterobacteriaceae (Cohen et al., 1993a, b; Sulavik et al.,
of the peptidoglycan in Gram-positive bacteria. 1997; Kunonga et al., 2000; Udani & Levy, 2006). The
The main conclusion of these studies is that antibiotic operon marRAB encodes MarR, a repressor that down-
resistance, rather than being a general metabolic burden, regulates marRAB expression, MarA, a global activator that
produces specific changes in the metabolism and in the regulates the expression of a large number of genes includ-
structure of bacterial pathogens that can only be fully ing marA, and marB, which encodes MarB, a small protein
understood from a global perspective. for which an independent effect has not been demonstrated
but that increases antibiotic resistance when present along-

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


Global regulatory networks and antibiotic side MarA (White et al., 1997). MarA regulates, directly or
resistance indirectly, the expression of several genes (Barbosa & Levy,
2000) that encode proteins with different functions in
Intrinsic antibiotic resistance is a specific feature of any
general metabolism, DNA repair and translation, pH and
given bacterial species. It is thus conceivable that, like other
superoxide stress responses, and low-level resistance to
characteristic phenotypes, it is integrated into global regula-
antibiotics, disinfectants and organic solvents (Cohen et al.,
tory networks. A good example of the need to study
1993a, b; Martin et al., 1996; Alekshun & Levy, 1997, 1999;
antibiotic resistance from a global perspective is that pro-
Pomposiello et al., 2001). Two other regulatory proteins
vided by the protein IgaA. A study of mucoid mutants of
homologous to MarA, SoxS and Rob are able to activate the
S. enterica resistant to mecillinam revealed that mutations in
Mar regulon directly or indirectly.
the gene mucM, leading to RcsCDB overexpression, pro-
The use of bioinformatics, genomics and molecular
duced mucoidity and antibiotic resistance (Costa & Anton,
genetics in the analysis of transcriptomic data led to the
2001). Independent work undertaken by another group
conclusion that these three transcriptional activators di-
(Cano et al., 2001) characterized the protein IgaA as an
rectly activate a common set of 40 promoters in E. coli
attenuator of intracellular growth required for virulence in
(Martin & Rosner, 2002). Each activator is regulated by
S. enterica. It has since been shown (Mariscotti & Garcia-Del
different signals: aromatic weak acids such as salicylate
Portillo, 2008) that mucM and igaA are the same gene, and
increase marRAB transcription (Cohen et al., 1993a, b),
that it is simultaneously involved in resistance and virulence
superoxides trigger soxS transcription (Amabile-Cuevas &
in S. enterica. Further work showed that mutations leading
Demple, 1991; Wu & Weiss, 1991) and bile salts, decanoate
to unstable forms of IgaA induce the RcsCDB system,
and bipyridyl activate Rob (Rosenberg et al., 2003).
leading to overproduction of colanic acid (Mariscotti &
The marA/soxS/rob regulon therefore encompasses a
Garcia-Del Portillo, 2008), and that RcsCDB overactivation
complex network that produces a global bacterial response
in IgaA-defective strains attenuates the acute virulence of
to external signals. The upregulation of the MarA, SoxS and
S. enterica (Tierrez & Garcia-del Portillo, 2004). Later it was
Rob activators increases antibiotic efflux (acrAB-tolC), DNA
shown that RcsCDB provides a phospho-relay system con-
repair (nfo) and superoxide resistance (zwf, fpr and sodA),
sisting of a hybrid sensor kinase, a phosphotransferase and a
and reduces outer membrane permeability (ompF via micF).
response regulator. RcsCDB regulates a large number of
It is important to note here that the main resistance system
bacterial genes (Majdalani & Gottesman, 2007), including
regulated by MarA is the efflux pump AcrAB-TolC. This
those involved in capsule production, which are important
MDR pump can efflux antibiotics and bile salts (Ma et al.,
in intrinsic resistance to b-lactams such as mecillinam
1995; Thanassi et al., 1997; Prouty et al., 2004). Because bile
(Laubacher & Ades, 2008). This example shows how two
salts are common compounds in the natural habitat of
independent but convergent studies, one focusing on viru-
Enterobacteriaceae it is likely that the original function of
lence and the other on resistance, concluded the involve-
AcrAB-TolC was the resistance of these detergents. It has
ment of the same elements, illustrating the need to make
now been shown that AcrAB-TolC is required for chicken
studies on these topics more integrated. Below, two exam-
colonization and virulence by S. enterica (Buckley et al.,
ples of networks that simultaneously modulate several
2006), highlighting that this MDR determinant has other
different bacterial phenotypes, including resistance, are
functions besides resistance.
discussed.
The rules that govern the differential expression of the
members of this network are beginning to be understood. It
The mar regulon
has been shown recently that both the abundance and the
The mar (from multiple antibiotic resistance) regulon type of activator trigger different responses in the network
(George & Levy, 1983; Cohen et al., 1989) is one of the (Martin et al., 2008). For instance, both MarA and SoxS can


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 55

saturate in vivo the marRAB and sodA promoters. However, Altogether, this indicates that the antibiotic resistance of
saturation with MarA leads to more marRAB and less sodA S. aureus is under the control of global regulation networks
expression than does SoxS saturation. The extent to which that involve virulence determinants as well as basal meta-
genes of this operon are activated is a function of the MarA bolic or structural genes.
concentration, a phenomenon termed ‘commensurable reg-
ulon activation,’ because it allows E. coli to elicit a propor-
tional response to stress signals (the activation of the Metabolic control of antibiotic resistance
minimum number of genes required to evade this signal) It has already been shown that antibiotic resistance can
(Martin et al., 2008). It was further suggested that this type produce specific changes in bacterial metabolism and that
of activation allows bacteria to mount different defensive resistance is integrated into global regulatory networks. It is
responses depending on the type of signal received, the thus conceivable that resistance might be controlled by the

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


amplitude of the signal and its duration. This regulon thus metabolic condition of bacteria. The clearest example of this
provides a flexible response to different kinds and level of noninherited phenotypic resistance (Martinez et al., 1994;
threat, including the presence of antibiotics. Levin & Rozen, 2006) is the lack of effect of several
antibiotics against cells that are not actively dividing (dor-
mant cells). Known as ‘antibiotic indifference,’ this phenom-
The mgrA regulon
enon was recognized soon after the first use of antibiotics
MgrA was simultaneously described by different groups as a (Lee et al., 1944; McDermott, 1958). It is of no importance
regulator of the expression of the MDR pump NorA of in infections produced by actively growing bacteria, but
S. aureus (Truong-Bolduc et al., 2003), of the autolysis reservoirs of nongrowing bacteria can be important in the
process in this bacterial species (Ingavale et al., 2003) and persistence and reappearance of some infections. The ex-
as a general regulator affecting the transcription of S. aureus istence of dormant cells helps to explain the presence of
a-toxin, nuclease and protein A, among others (Luong et al., persistent subpopulations in antibiotic-susceptible bacterial
2003). Since then, it has been shown that around 350 genes populations, and in the phenotypic resistance shown by
are regulated by MgrA (Luong et al., 2006). These genes bacterial biofilms. The global regulation shown by bacteria
include several MDR pumps (Truong-Bolduc et al., 2003, in response to particular modes of life is also important in
2005) and virulence factors (Ingavale et al., 2005). Notably, noninherited resistance (Levin & Rozen, 2006). This is
the expression of several metabolic genes is regulated by discussed below.
MgrA, highlighting the crossconnection between resistance,
virulence and the global metabolism of bacterial pathogens.
Swarming motility and global regulation
Although it has been shown that MgrA binds some
promoter regions (Truong-Bolduc et al., 2003), several of Swarming is a specific type of motility across a semisolid
the observed effects are likely indirect. Moreover, the fact surface commonly demonstrated by flagellated bacteria
that some genes are upregulated by MgrA in one growth (Fraser & Hughes, 1999; Jarrell & McBride, 2008). It is a
phase while they are downregulated in another indicates that multicellular phenomenon dependent on bacterial cell den-
there must be different pathways of regulation of the mgrA sity, nutrients conditions and surface moistness.
regulon (Luong et al., 2006). This indirect effect can be Swarmer cells show structural changes, including an
mediated by the activity of MgrA in the regulation of other increase in their number of flagellae or in cell length. They
regulators, as seen in S. aureus autolysis. also show substantial alterations in their metabolism, in-
Murein hydrolases in staphylococci have important roles dicating that swarming is a global lifestyle adaptation in
in cell division, including daughter cell separation and response to specific conditions rather than just a form of
peptidoglycan recycling. Their expression is tightly con- locomotion. In Salmonella, swarming is associated with
trolled because, if they are expressed at too high a level, they phenotypic antibiotic resistance (Kim & Surette, 2003).
destroy the cell wall and cell lysis ensues. It has been shown Recent work has shown that swarming in P. aeruginosa is
that MgrA regulates expression of the autolytic regulators influenced by a large number of cooperating genes (Overhage
lysSR, lgrAB and arlRS (Ingavale et al., 2005). Mutations in et al., 2007), and that swarmer P. aeruginosa cells show
mgrA result in enhanced autolysis of S. aureus and increased altered levels of expression for 407 genes. Several virulence
susceptibility to detergents and penicillin. determinants and one MDR efflux pump are also upregu-
As mentioned above, MgrA regulates the expression of lated in swarmer cells, indicating that this phenomenon is a
several MDR pumps in S. aureus. However, its effect on global response that might be useful in host colonization in
susceptibility to penicillin is also linked to its role in the presence of several antibiotics. One characteristic feature
regulating penicillin-induced autolysis, indicating that mul- of swarmer cells is their enhanced resistance to polymyxin B,
tiple layers exist in the regulation of resistance by MgrA. gentamicin and ciprofloxacin; it has been suggested that this

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
56 J.L. Martinez et al.

is due to metabolic differentiation during swarming and due and persister cells that occurred during growth. This switch
to changes in the cell envelope (Overhage et al., 2008). can be mathematically described in the framework of a
simple two-state model (Balaban et al., 2004). Recent work
indicates that the dormancy shown by Type I persistence is
Persistence
not completely attributable to a stationary-phase state.
Persistence is a phenomenon by which bacterial subpopula- Single-cell analysis has shown that protein production takes
tions enter a refractory state in the presence of different place over a small time window (around 1.5 h for E. coli) in
antibiotics (Balaban et al., 2004; Levin, 2004), and has been Type I persisters following their exit from the stationary
described in several species. Around one in every million of phase (Gefen et al., 2008). This suggests that the dormancy
cells in the exponential growth phase, and around one in that protects persisters from antibiotics does not occur
every hundred in the stationary growth phase possess a during the stationary phase but rather develops after this

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


persistence phenotype. Persistent cells can grow when the phase is over. Notably, during this time window, persistent
antibiotic disappears, but, like the susceptible wild-type cells are as susceptible as normal cells to antibiotics, indicat-
population, they are killed if the antibiotic is added again ing that, even for Type I persisters, this phenotype implies a
(Fig. 6). This indicates that persistence is not the conse- metabolic switch in a subset of the bacterial population
quence of mutations. It has been suggested that persistence (Gefen et al., 2008).
provides a means of survival in changing environments Compelling evidence supports the notion that persistence
(Kussell et al., 2005). is highly regulated. Persistence might well be explained by
It was first suggested that, in asynchronous cultures, the activity of toxin/antitoxin systems (Keren et al., 2004;
persistence was the consequence of the presence of a Lewis, 2008), whose actions maintain a fraction of the
bacterial subpopulation still in the lag period. These cells population in a nongrowing state. Recent work confirms
are in a dormant state and thus refractory to antibiotics. this, and indicates that persistence is due to several mechan-
Single-cell analysis using microfluidic devices allowed two isms involving the participation of a number of global
types of persisters to be defined. Type I is represented by the regulators and metabolic enzymes (Hansen et al., 2008).
preexisting population of growing cells generated during the
stationary phase that did not resume growth. Type II
persisters are the consequence of a switch between normal Biofilms
Most studies in microbiology are performed with bacteria
growing planktonically, but bacterial populations frequently
grow attached to surfaces where they form biofilms (Cost-
erton et al., 1999; Hall-Stoodley et al., 2004; Kolter &
Greenberg, 2006; Hansen et al., 2007). There is still some
debate as to whether a specific program exists for triggering
biofilm formation (O’Toole et al., 2000; Stoodley et al.,
2002) or whether growth in biofilms is simply due to the
physicochemical characteristics of the cell envelopes and
substrate, and due to the fluid dynamics (including bacterial
motility) of the system (van Loosdrecht et al., 2002).
However, it is clear that bacteria growing in biofilms
experience changes in microbial metabolism (Whiteley
et al., 2001; Sauer et al., 2002; Waite et al., 2005, 2006),
including the development of greater resistance to antibio-
tics than planktonically growing cells (Drenkard & Ausubel,
2002; Davies, 2003). Several hypotheses have been proposed
to explain this more resilient phenotype (Stewart, 2002),
some of them involving the structure of the biofilm and
others involving the metabolic state of biofilm-growing
bacteria. For instance, the structure of the biofilm itself
Fig. 6. Bacterial persistence to antibiotics. Persistence in the presence of
might preclude the entrance of antibiotics through the
an antibiotic is characterized by the presence of a small bacterial
subpopulation refractory to the antimicrobial agent. If the antibiotic is
matrices (e.g. oligosaccharides or DNA) that form part of
removed (arrow), this population resumes growth. If it is added again, biofilms (Suci et al., 1994). Because biofilms are structured
growing bacteria are killed, indicating that persistence is a noninherited environments (Lawrence et al., 1991) containing regions
resistance phenotype. with different amounts of oxygen and nutrients, the


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 57

metabolic activity in different parts of the biofilm is likely process will help in the fight against noninherited antibiotic
different (Huang et al., 1995; Sternberg et al., 1999); thus, resistance.
biofilms are home to bacterial populations with different
metabolic statuses. Owing to the scarcity of nutrients, it has
been suggested that subpopulations of bacteria forming
biofilms enter a dormant state, similar to that seen in
Acknowledgements
antibiotic persisters. Work in our laboratory is supported by grants BIO2005-
The study of the response of mature, preformed biofilms 04278 and BIO2008-00090 from the Spanish Ministerio de
to antibiotics may provide more insight into noninherited Educacion y Ciencia, and LSHM-CT-2005-518152 and
resistance. Resistance in biofilms is dependent on the LSHM-CT-2005-018705 from the European Union. A.F. is
structure of the biofilm and on the type of antibiotic used the recipient of a fellowship from the Spanish Ministerio de

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


(Folkesson et al., 2008). Escherichia coli in mature biofilms Educacion y Ciencia. We thank Adrian Burton for help with
are resistant to the antibiotic peptide colistin but not to the the English manuscript.
fluoroquinolone ciprofloxacin. Increased survival in the
presence of polymyxin is linked to modifications in bacterial
lipopolysaccharide that favour sequestration of the antibio-
tic. These modifications are the consequence of upregula-
References
tion of the yfbE operon mediated by the two-component Aendekerk S, Diggle SP, Song Z, Hoiby N, Cornelis P, Williams P
system basR/basS. This specific response is produced before & Camara M (2005) The MexGHI-OpmD multidrug efflux
polymyxin is added to the culture. Thus, it is not a classically pump controls growth, antibiotic susceptibility and virulence
induced resistance mechanism but the consequence of the in Pseudomonas aeruginosa via 4-quinolone-dependent cell-to-
metabolic changes experienced by a bacterial subpopulation cell communication. Microbiology 151: 1113–1125.
in response to as yet ignored signals in given locations of the Aleixandre V, Herrera G, Urios A & Blanco M (1991) Effects of
biofilm structure (Folkesson et al., 2008). It is important to ciprofloxacin on plasmid DNA supercoiling of Escherichia coli
topoisomerase I and gyrase mutants. Antimicrob Agents Ch 35:
note that the Salmonella typhimurium operon pmrH, which
20–23.
is homologous to yjbE, is induced during swarming (Kim &
Alekshun MN & Levy SB (1997) Regulation of chromosomally
Surette, 2003), a phenomenon during which reduced sus-
mediated multiple antibiotic resistance: the mar regulon.
ceptibility to antibiotics is also shown. Antimicrob Agents Ch 41: 2067–2075.
Recent studies have shown that MDR efflux pumps can Alekshun MN & Levy SB (1999) The mar regulon: multiple
specifically contribute to phenotypic resistance in biofilms resistance to antibiotics and other toxic chemicals. Trends
(Zhang & Mah, 2008). The metabolically heterogeneous Microbiol 7: 410–413.
nature of biofilms is highlighted by a study showing that Alekshun MN & Levy SB (2007) Molecular mechanisms of
P. aeruginosa growing in biofilms is tolerant to ciprofloxacin antibacterial multidrug resistance. Cell 128: 1037–1050.
or tetracycline in the deeper areas, whereas tolerance to Allegrucci M & Sauer K (2007) Characterization of colony
colistin is restricted to the upper regions (Pamp et al., 2008). morphology variants isolated from Streptococcus pneumoniae
Cells in the deepest regions of biofilms show low metabolic biofilms. J Bacteriol 189: 2030–2038.
activity, consistent with their refractory state. However, the Alonso A & Martinez JL (1997) Multiple antibiotic resistance in
upper layer of a biofilm contains metabolically active Stenotrophomonas maltophilia. Antimicrob Agents Ch 41:
bacteria; thus, resistance to colistin cannot be due to 1140–1142.
dormancy. It is reported that tolerance to colistin in Alonso A & Martinez JL (2000) Cloning and characterization of
P. aeruginosa biofilms is dependent on the pmr operon, SmeDEF, a novel multidrug efflux pump from
Stenotrophomonas maltophilia. Antimicrob Agents Ch 44:
which mediates lipopolysaccharide modification, and on the
3079–3086.
MDR pump MexAB-OprM (Pamp et al., 2008). Notably,
Alonso A & Martinez JL (2001) Expression of multidrug efflux
this efflux pump does not contribute to intrinsic resistance
pump SmeDEF by clinical isolates of Stenotrophomonas
to colistin in planktonic P. aeruginosa cells, although it seems
maltophilia. Antimicrob Agents Ch 45: 1879–1881.
to be important to subpopulations surviving the presence of Alonso A, Campanario E & Martinez JL (1999a) Emergence of
colistin within P. aeruginosa biofilms (Pamp et al., 2008). All multidrug-resistant mutants is increased under antibiotic
this indicates that the tolerance of biofilms to antibiotics is selective pressure in Pseudomonas aeruginosa. Microbiology
mediated by several different elements in bacterial subpo- 145: 2857–2862.
pulations with different metabolic signatures. Alonso A, Rojo F & Martinez JL (1999b) Environmental and
It is thus clear that different bacterial ways of life can clinical isolates of Pseudomonas aeruginosa show pathogenic
trigger the expression of specific sets of genes that reduce and biodegradative properties irrespective of their origin.
susceptibility to antibiotics. Understanding this type of Environ Microbiol 1: 421–430.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
58 J.L. Martinez et al.

Alonso A, Sanchez P & Martinez JL (2001) Environmental Barbosa TM & Levy SB (2000) Differential expression of over 60
selection of antibiotic resistance genes. Environ Microbiol 3: chromosomal genes in Escherichia coli by constitutive
1–9. expression of MarA. J Bacteriol 182: 3467–3474.
Alonso A, Morales G, Escalante R, Campanario E, Sastre L & Benveniste R & Davies J (1973) Aminoglycoside antibiotic-
Martinez JL (2004) Overexpression of the multidrug efflux inactivating enzymes in actinomycetes similar to those present
pump SmeDEF impairs Stenotrophomonas maltophilia in clinical isolates of antibiotic-resistant bacteria. P Natl Acad
physiology. J Antimicrob Chemoth 53: 432–434. Sci USA 70: 2276–2280.
Amabile-Cuevas CF & Demple B (1991) Molecular Bjedov I, Tenaillon O, Gerard B et al. (2003) Stress-induced
characterization of the soxRS genes of Escherichia coli: two mutagenesis in bacteria. Science 300: 1404–1409.
genes control a superoxide stress regulon. Nucleic Acids Res 19: Bjorkman J, Nagaev I, Berg OG, Hughes D & Andersson DI
4479–4484. (2000) Effects of environment on compensatory mutations to
Andersson DI (2006) The biological cost of mutational antibiotic

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


ameliorate costs of antibiotic resistance. Science 287:
resistance: any practical conclusions? Curr Opin Microbiol 9: 1479–1482.
461–465. Blazquez J, Oliver A & Gomez-Gomez JM (2002) Mutation and
Andersson DI & Levin BR (1999) The biological cost of antibiotic evolution of antibiotic resistance: antibiotics as promoters of
resistance. Curr Opin Microbiol 2: 489–493. antibiotic resistance? Curr Drug Targets 3: 345–349.
Andersson DI, Slechta ES & Roth JR (1998) Evidence that gene Blazquez J, Gomez-Gomez JM, Oliver A, Juan C, Kapur V &
amplification underlies adaptive mutability of the bacterial lac Martin S (2006) PBP3 inhibition elicits adaptive responses in
operon. Science 282: 1133–1135. Pseudomonas aeruginosa. Mol Microbiol 62: 84–99.
Arthur M, Molinas C, Bugg TD, Wright GD, Walsh CT & Bonomo RA & Szabo D (2006) Mechanisms of multidrug
Courvalin P (1992) Evidence for in vivo incorporation of
resistance in Acinetobacter species and Pseudomonas
D-lactate into peptidoglycan precursors of vancomycin-
aeruginosa. Clin Infect Dis 43(Suppl 2): S49–S56.
resistant enterococci. Antimicrob Agents Ch 36: 867–869.
Bouma JE & Lenski RE (1988) Evolution of a bacteria/plasmid
Arthur M, Molinas C, Depardieu F & Courvalin P (1993)
association. Nature 335: 351–352.
Characterization of Tn1546, a Tn3-related transposon
Buckley AM, Webber MA, Cooles S, Randall LP, La Ragione RM,
conferring glycopeptide resistance by synthesis of depsipeptide
Woodward MJ & Piddock LJ (2006) The AcrAB-TolC efflux
peptidoglycan precursors in Enterococcus faecium BM4147.
system of Salmonella enterica serovar Typhimurium plays a
J Bacteriol 175: 117–127.
role in pathogenesis. Cell Microbiol 8: 847–856.
Balaban NQ, Merrin J, Chait R, Kowalik L & Leibler S (2004)
Cano DA, Martinez-Moya M, Pucciarelli MG, Groisman EA,
Bacterial persistence as a phenotypic switch. Science 305:
Casadesus J & Garcia-Del Portillo F (2001) Salmonella enterica
1622–1625.
serovar Typhimurium response involved in attenuation of
Balsalobre L & de la Campa AG (2008) Fitness of Streptococcus
pathogen intracellular proliferation. Infect Immun 69:
pneumoniae fluoroquinolone-resistant strains with
topoisomerase IV recombinant genes. Antimicrob Agents Ch 6463–6474.
Carattoli A (2001) Importance of integrons in the diffusion of
52: 822–830.
Baptista M, Depardieu F, Reynolds P, Courvalin P & Arthur M resistance. Vet Res 32: 243–259.
(1997) Mutations leading to increased levels of resistance to Carilla-Latorre S, Calvo-Garrido J, Bloomfield G et al. (2008)
glycopeptide antibiotics in VanB-type enterococci. Mol Dictyostelium transcriptional responses to Pseudomonas
Microbiol 25: 93–105. aeruginosa: common and specific effects from PAO1 and PA14
Baquero F (2004) From pieces to patterns: evolutionary strains. BMC Microbiol 8: 109.
engineering in bacterial pathogens. Nat Rev Microbiol 2: Chen I & Dubnau D (2004) DNA uptake during bacterial
510–518. transformation. Nat Rev Microbiol 2: 241–249.
Baquero F, Coque TM & Canton R (2002) Allodemics. Lancet Chen I, Christie PJ & Dubnau D (2005) The ins and outs of DNA
Infect Dis 2: 591–592. transfer in bacteria. Science 310: 1456–1460.
Baquero MR, Nilsson AI, Turrientes Mdel C et al. (2004) Cheng Q, Campbell EA, Naughton AM, Johnson S & Masure HR
Polymorphic mutation frequencies in Escherichia coli: (1997) The com locus controls genetic transformation in
emergence of weak mutators in clinical isolates. J Bacteriol 186: Streptococcus pneumoniae. Mol Microbiol 23: 683–692.
5538–5542. Cohen ML (1992) Epidemiology of drug resistance: implications
Baquero MR, Galan JC, del Carmen Turrientes M, Canton R, for a post-antimicrobial era. Science 257: 1050–1055.
Coque TM, Martinez JL & Baquero F (2005) Increased Cohen SP, McMurry LM, Hooper DC, Wolfson JS & Levy SB
mutation frequencies in Escherichia coli isolates harboring (1989) Cross-resistance to fluoroquinolones in multiple-
extended-spectrum beta-lactamases. Antimicrob Agents Ch 49: antibiotic-resistant (Mar) Escherichia coli selected by
4754–4756. tetracycline or chloramphenicol: decreased drug accumulation
Barber M (1953) Penicillin-resistant and penicillin-dependent associated with membrane changes in addition to OmpF
staphylococcal variants. J Gen Microbiol 8: 111–115. reduction. Antimicrob Agents Ch 33: 1318–1325.


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 59

Cohen SP, Hachler H & Levy SB (1993a) Genetic and functional community behavior in divergent bacteria. Science 321:
analysis of the multiple antibiotic resistance (mar) locus in 1203–1206.
Escherichia coli. J Bacteriol 175: 1484–1492. Dorman CJ (1991) DNA supercoiling and environmental
Cohen SP, Levy SB, Foulds J & Rosner JL (1993b) Salicylate regulation of gene expression in pathogenic bacteria. Infect
induction of antibiotic resistance in Escherichia coli: activation Immun 59: 745–749.
of the mar operon and a mar-independent pathway. J Bacteriol Drenkard E & Ausubel FM (2002) Pseudomonas biofilm
175: 7856–7862. formation and antibiotic resistance are linked to phenotypic
Colegrave N & Collins S (2008) Experimental evolution: variation. Nature 416: 740–743.
experimental evolution and evolvability. Heredity 100: Elsaied H, Stokes HW, Nakamura T, Kitamura K, Fuse H &
464–470. Maruyama A (2007) Novel and diverse integron integrase
Cosson P, Zulianello L, Join-Lambert O et al. (2002) Pseudomonas genes and integron-like gene cassettes are prevalent in deep-sea
aeruginosa virulence analyzed in a Dictyostelium discoideum hydrothermal vents. Environ Microbiol 9: 2298–2312.

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


host system. J Bacteriol 184: 3027–3033. Enne VI, Livermore DM, Stephens P & Hall LM (2001)
Costa CS & Anton DN (2001) Role of the ftsA1p promoter in the Persistence of sulphonamide resistance in Escherichia coli in
resistance of mucoid mutants of Salmonella enterica to the UK despite national prescribing restriction. Lancet 357:
mecillinam: characterization of a new type of mucoid mutant. 1325–1328.
FEMS Microbiol Lett 200: 201–205. Erill I, Campoy S & Barbe J (2007) Aeons of distress: an
Costerton JW, Stewart PS & Greenberg EP (1999) Bacterial evolutionary perspective on the bacterial SOS response. FEMS
biofilms: a common cause of persistent infections. Science 284: Microbiol Rev 31: 637–656.
1318–1322. Evans K, Passador L, Srikumar R, Tsang E, Nezezon J & Poole K
Courvalin P (2008) Predictable and unpredictable evolution of (1998) Influence of the MexAB-OprM multidrug efflux system
antibiotic resistance. J Intern Med 264: 4–16. on quorum sensing in Pseudomonas aeruginosa. J Bacteriol 180:
D’Acosta VM, McGrann KM, Hughes DW & Wright GD (2006) 5443–5447.
Sampling the antibiotic resistome. Science 311: 374–377. Fajardo A & Martinez JL (2008) Antibiotics as signals that trigger
Dagan T, Artzy-Randrup Y & Martin W (2008) Modular specific bacterial responses. Curr Opin Microbiol 11: 161–167.
networks and cumulative impact of lateral transfer in Fajardo A, Martinez-Martin N, Mercadillo M et al. (2008) The
prokaryote genome evolution. P Natl Acad Sci USA 105: neglected intrinsic resistome of bacterial pathogens. PLoS ONE
10039–10044. 3: e1619.
Dantas G, Sommer MO, Oluwasegun RD & Church GM (2008) Folkesson A, Haagensen JA, Zampaloni C, Sternberg C & Molin S
Bacteria subsisting on antibiotics. Science 320: 100–103. (2008) Biofilm induced tolerance towards antimicrobial
Datta N & Hughes VM (1983) Plasmids of the same Inc groups in peptides. PLoS ONE 3: e1891.
Enterobacteria before and after the medical use of antibiotics. Force A, Cresko WA, Pickett FB, Proulx SR, Amemiya C & Lynch
Nature 306: 616–617. M (2005) The origin of subfunctions and modular gene
Davies D (2003) Understanding biofilm resistance to regulation. Genetics 170: 433–446.
antibacterial agents. Nat Rev Drug Discov 2: 114–122. Foster PL (2005) Stress responses and genetic variation in
Davies J (1994) Inactivation of antibiotics and the dissemination bacteria. Mutat Res 569: 3–11.
of resistance genes. Science 264: 375–382. Fraser GM & Hughes C (1999) Swarming motility. Curr Opin
Davies J (2006) Are antibiotics naturally antibiotics? J Ind Microbiol 2: 630–635.
Microbiol Biot 33: 496–499. Galan JE & Curtiss R III (1990) Expression of Salmonella
Davies JE (1997) Origins, acquisition and dissemination of typhimurium genes required for invasion is regulated by
antibiotic resistance determinants. Ciba F Symp 207: 15–27; changes in DNA supercoiling. Infect Immun 58: 1879–1885.
discussion 27–35. Garcia-Bustos J & Tomasz A (1990) A biological price of
de Lorenzo V, Martinez JL & Asensio C (1984) Microcin- antibiotic resistance: major changes in the peptidoglycan
mediated interactions between Klebsiella pneumoniae and structure of penicillin-resistant pneumococci. P Natl Acad Sci
Escherichia coli strains. J Gen Microbiol 130: 391–400. USA 87: 5415–5419.
Depardieu F, Podglajen I, Leclercq R, Collatz E & Courvalin P Garcia-Bustos JF, Chait BT & Tomasz A (1988) Altered
(2007) Modes and modulations of antibiotic resistance gene peptidoglycan structure in a pneumococcal transformant
expression. Clin Microbiol Rev 20: 79–114. resistant to penicillin. J Bacteriol 170: 2143–2147.
Deziel E, Lepine F, Milot S, He J, Mindrinos MN, Tompkins RG & Garcillan-Barcia MP & de la Cruz F (2008) Why is entry exclusion
Rahme LG (2004) Analysis of Pseudomonas aeruginosa an essential feature of conjugative plasmids? Plasmid 60: 1–18.
4-hydroxy-2-alkylquinolines (HAQs) reveals a role for Gaynes R & Edwards JR (2005) Overview of nosocomial
4-hydroxy-2-heptylquinoline in cell-to-cell communication. infections caused by gram-negative bacilli. Clin Infect Dis 41:
P Natl Acad Sci USA 101: 1339–1344. 848–854.
Dietrich LE, Teal TK, Price-Whelan A & Newman DK (2008) Gefen O, Gabay C, Mumcuoglu M, Engel G & Balaban NQ (2008)
Redox-active antibiotics control gene expression and Single-cell protein induction dynamics reveals a period of

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
60 J.L. Martinez et al.

vulnerability to antibiotics in persister bacteria. P Natl Acad Hansen SK, Rainey PB, Haagensen JA & Molin S (2007)
Sci USA 105: 6145–6149. Evolution of species interactions in a biofilm community.
George AM & Levy SB (1983) Gene in the major cotransduction Nature 445: 533–536.
gap of the Escherichia coli K-12 linkage map required for the Hastings PJ, Rosenberg SM & Slack A (2004) Antibiotic-induced
expression of chromosomal resistance to tetracycline and lateral transfer of antibiotic resistance. Trends Microbiol 12:
other antibiotics. J Bacteriol 155: 541–548. 401–404.
Gerber M, Walch C, Loffler B, Tischendorf K, Reischl U & Haussler S (2004) Biofilm formation by the small colony variant
Ackermann G (2008) Effect of sub-MIC concentrations of phenotype of Pseudomonas aeruginosa. Environ Microbiol 6:
metronidazole, vancomycin, clindamycin and linezolid on 546–551.
toxin gene transcription and production in Clostridium Haussler S, Tummler B, Weissbrodt H, Rohde M & Steinmetz I
difficile. J Med Microbiol 57: 776–783. (1999) Small-colony variants of Pseudomonas aeruginosa in
Gocke TM & Finland M (1950) Development of

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


cystic fibrosis. Clin Infect Dis 29: 621–625.
chloramphenicol-resistant and chloramphenicol-dependent
Heinemann M, Kummel A, Ruinatscha R & Panke S (2005) In
variants of a strain of Klebsiella pneumoniae. P Soc Exp Biol
silico genome-scale reconstruction and validation of the
Med 74: 824–829.
Staphylococcus aureus metabolic network. Biotechnol Bioeng
Godoy VG, Jarosz DF, Simon SM, Abyzov A, Ilyin V & Walker GC
92: 850–864.
(2007) UmuD and RecA directly modulate the mutagenic
Henderson-Begg SK, Livermore DM & Hall LM (2006) Effect of
potential of the Y family DNA polymerase DinB. Mol Cell 28:
subinhibitory concentrations of antibiotics on mutation
1058–1070.
Goh EB, Yim G, Tsui W, McClure J, Surette MG & Davies J (2002) frequency in Streptococcus pneumoniae. J Antimicrob Chemoth
Transcriptional modulation of bacterial gene expression by 57: 849–854.
subinhibitory concentrations of antibiotics. P Natl Acad Sci Henrichfreise B, Wiegand I, Luhmer-Becker I & Wiedemann B
USA 99: 17025–17030. (2007) Development of resistance in wild-type and
Gomez-Gomez JM, Blazquez J, Baquero F & Martinez JL (1997) hypermutable Pseudomonas aeruginosa strains exposed to
H-NS and RpoS regulate emergence of Lac Ara1 mutants of clinical pharmacokinetic profiles of meropenem and
Escherichia coli MCS2. J Bacteriol 179: 4620–4622. ceftazidime simulated in vitro. Antimicrob Agents Ch 51:
Goodman AL, Kulasekara B, Rietsch A, Boyd D, Smith RS & Lory 3642–3649.
S (2004) A signaling network reciprocally regulates genes Hernandez A, Mellado RP & Martinez JL (1998) Metal
associated with acute infection and chronic persistence in accumulation and vanadium-induced multidrug resistance by
Pseudomonas aeruginosa. Dev Cell 7: 745–754. environmental isolates of Escherichia hermannii and
Grkovic S, Brown MH & Skurray RA (2002) Regulation of Enterobacter cloacae. Appl Environ Microb 64: 4317–4320.
bacterial drug export systems. Microbiol Mol Biol R 66: Hirakata Y, Srikumar R, Poole K et al. (2002) Multidrug efflux
671–701. systems play an important role in the invasiveness of
Haas D & Defago G (2005) Biological control of soil-borne Pseudomonas aeruginosa. J Exp Med 196: 109–118.
pathogens by fluorescent pseudomonads. Nat Rev Microbiol 3: Hogan D & Kolter R (2002) Why are bacteria refractory to
307–319. antimicrobials? Curr Opin Microbiol 5: 472–477.
Hakenbeck R (1998) Mosaic genes and their role in penicillin- Holmes AJ, Gillings MR, Nield BS, Mabbutt BC, Nevalainen KM
resistant Streptococcus pneumoniae. Electrophoresis 19: & Stokes HW (2003) The gene cassette metagenome is a basic
597–601.
resource for bacterial genome evolution. Environ Microbiol 5:
Hall RM & Collis CM (1995) Mobile gene cassettes and integrons:
383–394.
capture and spread of genes by site-specific recombination.
Hong HJ, Paget MS & Buttner MJ (2002) A signal transduction
Mol Microbiol 15: 593–600.
system in Streptomyces coelicolor that activates the expression
Hall-Stoodley L, Costerton JW & Stoodley P (2004) Bacterial
of a putative cell wall glycan operon in response to vancomycin
biofilms: from the natural environment to infectious diseases.
Nat Rev Microbiol 2: 95–108. and other cell wall-specific antibiotics. Mol Microbiol 44:
Hamilton-Miller JM (2004) Antibiotic resistance from two 1199–1211.
perspectives: man and microbe. Int J Antimicrob Ag 23: Huang CT, Yu FP, McFeters GA & Stewart PS (1995) Nonuniform
209–212. spatial patterns of respiratory activity within biofilms during
Handwerger S, Pucci MJ, Volk KJ, Liu J & Lee MS (1992) The disinfection. Appl Environ Microb 61: 2252–2256.
cytoplasmic peptidoglycan precursor of vancomycin-resistant Ingavale S, van Wamel W, Luong TT, Lee CY & Cheung AL (2005)
Enterococcus faecalis terminates in lactate. J Bacteriol 174: Rat/MgrA, a regulator of autolysis, is a regulator of virulence
5982–5984. genes in Staphylococcus aureus. Infect Immun 73: 1423–1431.
Hansen S, Lewis K & Vulic M (2008) Role of global regulators and Ingavale SS, Van Wamel W & Cheung AL (2003) Characterization
nucleotide metabolism in antibiotic tolerance in Escherichia of RAT, an autolysis regulator in Staphylococcus aureus. Mol
coli. Antimicrob Agents Ch 52: 2718–2726. Microbiol 48: 1451–1466.


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 61

Isalan M, Lemerle C, Michalodimitrakis K et al. (2008) Laubacher ME & Ades SE (2008) The Rcs phosphorelay is a cell
Evolvability and hierarchy in rewired bacterial gene networks. envelope stress response activated by peptidoglycan stress and
Nature 452: 840–845. contributes to intrinsic antibiotic resistance. J Bacteriol 190:
Jalal S, Ciofu O, Hoiby N, Gotoh N & Wretlind B (2000) 2065–2074.
Molecular mechanisms of fluoroquinolone resistance in Lawrence JR, Korber DR, Hoyle BD, Costerton JW & Caldwell DE
Pseudomonas aeruginosa isolates from cystic fibrosis patients. (1991) Optical sectioning of microbial biofilms. J Bacteriol
Antimicrob Agents Ch 44: 710–712. 173: 6558–6567.
Jarrell KF & McBride MJ (2008) The surprisingly diverse ways Layton JC & Foster PL (2003) Error-prone DNA polymerase IV is
that prokaryotes move. Nat Rev Microbiol 6: 466–476. controlled by the stress-response sigma factor, RpoS, in
Ji G, Beavis R & Novick RP (1997) Bacterial interference caused Escherichia coli. Mol Microbiol 50: 549–561.
by autoinducing peptide variants. Science 276: 2027–2030. LeClerc JE, Li B, Payne WL & Cebula TA (1996) High mutation
Johnsborg O, Eldholm V, Bjornstad ML & Havarstein LS (2008) A frequencies among Escherichia coli and Salmonella pathogens.

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


predatory mechanism dramatically increases the efficiency of Science 274: 1208–1211.
lateral gene transfer in Streptococcus pneumoniae and related Lee SW, Foley EJ & Epstein JA (1944) Mode of action of
commensal species. Mol Microbiol 69: 245–253. penicillin: I. Bacterial growth and penicillin activity –
Johnson CN, Briles DE, Benjamin WH Jr, Hollingshead SK & Staphylococcus aureus FDA. J Bacteriol 48: 393–399.
Waites KB (2005) Relative fitness of fluoroquinolone-resistant Levin BR (2004) Microbiology. Noninherited resistance to
Streptococcus pneumoniae. Emerg Infect Dis 11: 814–820. antibiotics. Science 305: 1578–1579.
Kashtan N & Alon U (2005) Spontaneous evolution of Levin BR & Rozen DE (2006) Non-inherited antibiotic resistance.
modularity and network motifs. P Natl Acad Sci USA 102: Nat Rev Microbiol 4: 556–562.
13773–13778. Levy SB (1998) Multidrug resistance – a sign of the times. New
Kaufmann GF, Sartorio R, Lee SH et al. (2005) Revisiting quorum Engl J Med 338: 1376–1378.
sensing: discovery of additional chemical and biological Levy SB & Marshall B (2004) Antibacterial resistance worldwide:
functions for 3-oxo-N-acylhomoserine lactones. P Natl Acad causes, challenges and responses. Nat Med 10: S122–S129.
Sci USA 102: 309–314. Lewinson O & Bibi E (2001) Evidence for simultaneous binding
Keren I, Shah D, Spoering A, Kaldalu N & Lewis K (2004) of dissimilar substrates by the Escherichia coli multidrug
Specialized persister cells and the mechanism of multidrug transporter MdfA. Biochemistry 40: 12612–12618.
tolerance in Escherichia coli. J Bacteriol 186: 8172–8180. Lewis K (2008) Multidrug tolerance of biofilms and persister
Kim SR, Maenhaut-Michel G, Yamada M et al. (1997) Multiple cells. Curr Top Microbiol 322: 107–131.
pathways for SOS-induced mutagenesis in Escherichia coli: an Linares JF, Lopez JA, Camafeita E, Albar JP, Rojo F & Martinez JL
overexpression of dinB/dinP results in strongly enhancing (2005) Overexpression of the multidrug efflux pumps
mutagenesis in the absence of any exogenous treatment to MexCD-OprJ and MexEF-OprN is associated with a reduction
damage DNA. P Natl Acad Sci USA 94: 13792–13797. of type III secretion in Pseudomonas aeruginosa. J Bacteriol
Kim W & Surette MG (2003) Swarming populations of 187: 1384–1391.
Salmonella represent a unique physiological state coupled to Linares JF, Gustafsson I, Baquero F & Martinez JL (2006)
multiple mechanisms of antibiotic resistance. Biol Proced Antibiotics as intermicrobial signaling agents instead of
Online 5: 189–196. weapons. P Natl Acad Sci USA 103: 19484–19489.
Kirschner M & Gerhart J (1998) Evolvability. P Natl Acad Sci USA Livermore DM (1995) Beta-Lactamases in laboratory and clinical
95: 8420–8427. resistance. Clin Microbiol Rev 8: 557–584.
Kohler T, Michea-Hamzehpour M, Plesiat P, Kahr AL & Pechere Lopez E, Elez M, Matic I & Blazquez J (2007) Antibiotic-mediated
JC (1997) Differential selection of multidrug efflux systems by recombination: ciprofloxacin stimulates SOS-independent
quinolones in Pseudomonas aeruginosa. Antimicrob Agents Ch recombination of divergent sequences in Escherichia coli. Mol
41: 2540–2543. Microbiol 64: 83–93.
Kohler T, van Delden C, Curty LK, Hamzehpour MM & Pechere Lovett ST, Hurley RL, Sutera VA Jr, Aubuchon RH & Lebedeva
JC (2001) Overexpression of the MexEF-OprN multidrug MA (2002) Crossing over between regions of limited
efflux system affects cell-to-cell signaling in Pseudomonas homology in Escherichia coli. RecA-dependent and RecA-
aeruginosa. J Bacteriol 183: 5213–5222. independent pathways. Genetics 160: 851–859.
Kolter R & Greenberg EP (2006) Microbial sciences: the Lubelski J, Konings WN & Driessen AJ (2007) Distribution and
superficial life of microbes. Nature 441: 300–302. physiology of ABC-type transporters contributing to
Kunonga NI, Sobieski RJ & Crupper SS (2000) Prevalence of the multidrug resistance in bacteria. Microbiol Mol Biol R 71:
multiple antibiotic resistance operon (marRAB) in the genus 463–476.
Salmonella. FEMS Microbiol Lett 187: 155–160. Luo N, Pereira S, Sahin O, Lin J, Huang S, Michel L & Zhang Q
Kussell E, Kishony R, Balaban NQ & Leibler S (2005) Bacterial (2005) Enhanced in vivo fitness of fluoroquinolone-resistant
persistence: a model of survival in changing environments. Campylobacter jejuni in the absence of antibiotic selection
Genetics 169: 1807–1814. pressure. P Natl Acad Sci USA 102: 541–546.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
62 J.L. Martinez et al.

Luo P, Li H & Morrison DA (2003) ComX is a unique link Martinez JL, Blazquez J & Baquero F (1994) Non-canonical
between multiple quorum sensing outputs and competence in mechanisms of antibiotic resistance. Eur J Clin Microbiol 13:
Streptococcus pneumoniae. Mol Microbiol 50: 623–633. 1015–1022.
Luong TT, Newell SW & Lee CY (2003) Mgr, a novel global Martinez JL, Alonso A, Gomez-Gomez JM & Baquero F (1998)
regulator in Staphylococcus aureus. J Bacteriol 185: 3703–3710. Quinolone resistance by mutations in chromosomal gyrase
Luong TT, Dunman PM, Murphy E, Projan SJ & Lee CY (2006) genes. Just the tip of the iceberg? J Antimicrob Chemoth 42:
Transcription Profiling of the mgrA Regulon in Staphylococcus 683–688.
aureus. J Bacteriol 188: 1899–1910. Martinez JL, Baquero F & Andersson DI (2007) Predicting
Ma D, Cook DN, Alberti M, Pon NG, Nikaido H & Hearst JE antibiotic resistance. Nat Rev Microbiol 5: 958–965.
(1995) Genes acrA and acrB encode a stress-induced efflux Martinez-Solano L, Macia MD, Fajardo A, Oliver A & Martinez
system of Escherichia coli. Mol Microbiol 16: 45–55. JL (in press) Chronic infection by Pseudomonas aeruginosa in
Macia MD, Blanquer D, Togores B, Sauleda J, Perez JL & Oliver A chronic obstructive pulmonary disease. Clin Infect Dis, DOI:

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


(2005) Hypermutation is a key factor in development of 10.1086/593186.
multiple-antimicrobial resistance in Pseudomonas aeruginosa Massova I & Mobashery S (1998) Kinship and diversification of
strains causing chronic lung infections. Antimicrob Agents Ch bacterial penicillin-binding proteins and beta-lactamases.
49: 3382–3386. Antimicrob Agents Ch 42: 1–17.
Macia MD, Borrell N, Segura M, Gomez C, Perez JL & Oliver A Matic I, Radman M, Taddei F et al. (1997) Highly variable
(2006) Efficacy and potential for resistance selection of mutation rates in commensal and pathogenic Escherichia coli.
antipseudomonal treatments in a mouse model of lung Science 277: 1833–1834.
infection by hypermutable Pseudomonas aeruginosa. Matic I, Taddei F & Radman M (2000) No genetic barriers
Antimicrob Agents Ch 50: 975–983. between Salmonella enterica serovar Typhimurium and
Macinga DR & Rather PN (1999) The chromosomal 2 0 -N- Escherichia coli in SOS-induced mismatch repair-deficient
acetyltransferase of Providencia stuartii: physiological cells. J Bacteriol 182: 5922–5924.
functions and genetic regulation. Front Biosci 4: D132–D140. Maughan H, Galeano B & Nicholson WL (2004) Novel rpoB
Mainardi JL, Villet R, Bugg TD, Mayer C & Arthur M (2008) mutations conferring rifampin resistance on Bacillus subtilis:
Evolution of peptidoglycan biosynthesis under the selective global effects on growth, competence, sporulation, and
pressure of antibiotics in Gram-positive bacteria. FEMS germination. J Bacteriol 186: 2481–2486.
Microbiol Rev 32: 386–408. Mazel D (2006) Integrons: agents of bacterial evolution. Nat Rev
Majdalani N & Gottesman S (2007) Genetic dissection of Microbiol 4: 608–620.
signaling through the Rcs phosphorelay. Method Enzymol 423: McDermott W (1958) Microbial persistence. Yale J Biol Med 30:
349–362. 257–291.
Mao EF, Lane L, Lee J & Miller JH (1997) Proliferation of Medeiros AA (1997) Evolution and dissemination of beta-
mutators in a cell population. J Bacteriol 179: 417–422. lactamases accelerated by generations of beta-lactam
Mariscotti JF & Garcia-Del Portillo F (2008) Instability of the antibiotics. Clin Infect Dis 24(Suppl 1): S19–S45.
Salmonella RcsCDB signalling system in the absence of the Meroueh SO, Minasov G, Lee W, Shoichet BK & Mobashery S
attenuator IgaA. Microbiology 154: 1372–1383. (2003) Structural aspects for evolution of beta-lactamases
Martin RG & Rosner JL (2002) Genomics of the marA/soxS/rob from penicillin-binding proteins. J Am Chem Soc 125:
regulon of Escherichia coli: identification of directly activated 9612–9618.
promoters by application of molecular genetics and Mete M, Tang F, Xu X & Yuruk N (2008) A structural approach
informatics to microarray data. Mol Microbiol 44: 1611–1624. for finding functional modules from large biological networks.
Martin RG, Jair KW, Wolf RE Jr & Rosner JL (1996) BMC Bioinformatics 9(Suppl 9): S19.
Autoactivation of the marRAB multiple antibiotic resistance Miller C, Thomsen LE, Gaggero C, Mosseri R, Ingmer H & Cohen
operon by the MarA transcriptional activator in Escherichia SN (2004) SOS response induction by beta-lactams and
coli. J Bacteriol 178: 2216–2223. bacterial defense against antibiotic lethality. Science 305:
Martin RG, Bartlett ES, Rosner JL & Wall ME (2008) Activation of 1629–1631.
the Escherichia coli marA/soxS/rob regulon in response to Morosini MI, Ayala JA, Baquero F, Martinez JL & Blazquez J
transcriptional activator concentration. J Mol Biol 380: (2000) Biological cost of AmpC production for Salmonella
278–284. enterica serotype Typhimurium. Antimicrob Agents Ch 44:
Martinez JL (2008) Antibiotics and antibiotic resistance genes in 3137–3143.
natural environments. Science 321: 365–367. Navas A, Cobas G, Talavera M, Ayala JA, Lopez JA & Martinez JL
Martinez JL & Baquero F (2000) Mutation frequencies and (2007) Experimental validation of Haldane’s hypothesis on the
antibiotic resistance. Antimicrob Agents Ch 44: 1771–1777. role of infection as an evolutionary force for Metazoans. P Natl
Martinez JL & Baquero F (2002) Interactions among strategies Acad Sci USA 104: 13728–13731.
associated with bacterial infection: pathogenicity, epidemicity, Nield BS, Holmes AJ, Gillings MR, Recchia GD, Mabbutt BC,
and antibiotic resistance. Clin Microbiol Rev 15: 647–679. Nevalainen KM & Stokes HW (2001) Recovery of new


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 63

integron classes from environmental DNA. FEMS Microbiol Phillips I, Culebras E, Moreno F & Baquero F (1987) Induction of
Lett 195: 59–65. the SOS response by new 4-quinolones. J Antimicrob Chemoth
Nodwell JR (2007) Novel links between antibiotic resistance and 20: 631–638.
antibiotic production. J Bacteriol 189: 3683–3685. Piddock LJ (1999) Mechanisms of fluoroquinolone resistance: an
Oliver A, Canton R, Campo P, Baquero F & Blazquez J (2000) update 1994–1998. Drugs 58(Suppl 2): 11–18.
High frequency of hypermutable Pseudomonas aeruginosa in Piddock LJ (2006a) Multidrug-resistance efflux pumps – not just
cystic fibrosis lung infection. Science 288: 1251–1254. for resistance. Nat Rev Microbiol 4: 629–636.
Oliver A, Baquero F & Blazquez J (2002) The mismatch repair Piddock LJ (2006b) Clinically relevant chromosomally encoded
system (mutS, mutL and uvrD genes) in Pseudomonas multidrug resistance efflux pumps in bacteria. Clin Microbiol
aeruginosa: molecular characterization of naturally occurring Rev 19: 382–402.
mutants. Mol Microbiol 43: 1641–1650. Pigliucci M (2008) Is evolvability evolvable? Nat Rev Genet 9:

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


O’Toole G, Kaplan HB & Kolter R (2000) Biofilm formation as 75–82.
microbial development. Annu Rev Microbiol 54: 49–79. Pomposiello PJ, Bennik MH & Demple B (2001) Genome-wide
Overhage J, Lewenza S, Marr AK & Hancock RE (2007) transcriptional profiling of the Escherichia coli responses to
Identification of genes involved in swarming motility using a superoxide stress and sodium salicylate. J Bacteriol 183:
Pseudomonas aeruginosa PAO1 mini-Tn5-lux mutant library. 3890–3902.
J Bacteriol 189: 2164–2169. Proctor RA, von Eiff C, Kahl BC, Becker K, McNamara P,
Overhage J, Bains M, Brazas MD & Hancock RE (2008) Swarming Herrmann M & Peters G (2006) Small colony variants: a
of Pseudomonas aeruginosa is a complex adaptation leading to pathogenic form of bacteria that facilitates persistent and
increased production of virulence factors and antibiotic recurrent infections. Nat Rev Microbiol 4: 295–305.
resistance. J Bacteriol 190: 2671–2679. Prouty AM, Brodsky IE, Falkow S & Gunn JS (2004) Bile-salt-
Pal C, Macia MD, Oliver A, Schachar I & Buckling A (2007) mediated induction of antimicrobial and bile resistance in
Coevolution with viruses drives the evolution of bacterial Salmonella typhimurium. Microbiology 150: 775–783.
mutation rates. Nature 450: 1079–1081. Prudhomme M, Attaiech L, Sanchez G, Martin B & Claverys JP
Pamp SJ, Gjermansen M, Johansen HK & Tolker-Nielsen T (2008) (2006) Antibiotic stress induces genetic transformability in the
Tolerance to the antimicrobial peptide colistin in Pseudomonas human pathogen Streptococcus pneumoniae. Science 313:
aeruginosa biofilms is linked to metabolically active cells, and 89–92.
depends on the pmr and mexAB-oprM genes. Mol Microbiol Pucciarelli MG & Garcia-del Portillo F (2003) Protein-
68: 223–240. peptidoglycan interactions modulate the assembly of the
Pang Y, Brown BA, Steingrube VA, Wallace RJ Jr & Roberts MC needle complex in the Salmonella invasion-associated type III
(1994) Tetracycline resistance determinants in Mycobacterium secretion system. Mol Microbiol 48: 573–585.
and Streptomyces species. Antimicrob Agents Ch 38: 1408–1412. Quinn JP (1998) Clinical problems posed by multiresistant
Paterson DL & Bonomo RA (2005) Extended-spectrum beta- nonfermenting gram-negative pathogens. Clin Infect Dis
lactamases: a clinical update. Clin Microbiol Rev 18: 657–686. 27(Suppl 1): S117–S124.
Pepper JW (2003) The evolution of evolvability in genetic linkage Raaijmakers JM, Vlami M & de Souza JT (2002) Antibiotic
patterns. Biosystems 69: 115–126. production by bacterial biocontrol agents. Antonie Van
Pereira-Leal JB, Levy ED & Teichmann SA (2006) The origins and Leeuwenhoek 81: 537–547.
evolution of functional modules: lessons from protein Ramos JL, Duque E, Gallegos MT et al. (2002) Mechanisms of
complexes. Philos T Roy Soc B 361: 507–517. solvent tolerance in gram-negative bacteria. Annu Rev
Perez-Capilla T, Baquero MR, Gomez-Gomez JM, Ionel A, Microbiol 56: 743–768.
Martin S & Blazquez J (2005) SOS-independent induction of Reichmann P, Konig A, Linares J et al. (1997) A global gene pool
dinB transcription by beta-lactam-mediated inhibition of cell for high-level cephalosporin resistance in commensal
wall synthesis in Escherichia coli. J Bacteriol 187: 1515–1518. Streptococcus species and Streptococcus pneumoniae. J Infect Dis
Perkins AE & Nicholson WL (2008) Uncovering new metabolic 176: 1001–1012.
capabilities of Bacillus subtilis using phenotype profiling of Revilla C, Garcillan-Barcia MP, Fernandez-Lopez R et al. (2008)
rifampin-resistant rpoB mutants. J Bacteriol 190: 807–814. Different pathways to acquiring resistance genes illustrated by
Pestova EV, Havarstein LS & Morrison DA (1996) Regulation of the recent evolution of IncW plasmids. Antimicrob Agents Ch
competence for genetic transformation in Streptococcus 52: 1472–1480.
pneumoniae by an auto-induced peptide pheromone and a Rosenberg EY, Bertenthal D, Nilles ML, Bertrand KP & Nikaido H
two-component regulatory system. Mol Microbiol 21: 853–862. (2003) Bile salts and fatty acids induce the expression of
Pettersson ME, Andersson DI, Roth JR & Berg OG (2005) The Escherichia coli AcrAB multidrug efflux pump through their
amplification model for adaptive mutation: simulations and interaction with Rob regulatory protein. Mol Microbiol 48:
analysis. Genetics 169: 1105–1115. 1609–1619.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved
64 J.L. Martinez et al.

Roth JR, Kugelberg E, Reams AB, Kofoid E & Andersson DI from pbpX genes of a penicillin-sensitive Streptococcus oralis.
(2006) Origin of mutations under selection: the adaptive Mol Microbiol 12: 1013–1023.
mutation controversy. Annu Rev Microbiol 60: 477–501. Sifaoui F & Gutmann L (1997) Vancomycin dependence in a
Rowe-Magnus AD, Davies J & Mazel D (2002) Impact of vanA-producing Enterococcus avium strain with a nonsense
integrons and transposons on the evolution of resistance and mutation in the natural D-Ala-D-Ala ligase gene. Antimicrob
virulence. Curr Top Microbiol 264: 167–188. Agents Ch 41: 1409.
Rowe-Magnus DA & Mazel D (2001) Integrons: natural tools for Silver S & Phung LT (1996) Bacterial heavy metal resistance: new
bacterial genome evolution. Curr Opin Microbiol 4: 565–569. surprises. Annu Rev Microbiol 50: 753–789.
Rowe-Magnus DA & Mazel D (2002) The role of integrons in Stadler BM, Stadler PF, Wagner GP & Fontana W (2001) The
antibiotic resistance gene capture. Int J Med Microbiol 292: topology of the possible: formal spaces underlying patterns of
115–125. evolutionary change. J Theor Biol 213: 241–274.
Roy C, Foz A, Segura C, Tirado M, Fuster C & Reig R (1983)

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


Sternberg C, Christensen BB, Johansen T, Toftgaard Nielsen A,
Plasmid-determined beta-lactamases identified in a group of Andersen JB, Givskov M & Molin S (1999) Distribution of
204 ampicillin-resistant Enterobacteriaceae. J Antimicrob bacterial growth activity in flow-chamber biofilms. Appl
Chemoth 12: 507–510. Environ Microb 65: 4108–4117.
Rozen DE, McGee L, Levin BR & Klugman KP (2007) Fitness Stewart PS (2002) Mechanisms of antibiotic resistance in
costs of fluoroquinolone resistance in Streptococcus bacterial biofilms. Int J Med Microbiol 292: 107–113.
pneumoniae. Antimicrob Agents Ch 51: 412–416. Stokes HW & Hall RM (1989) A novel family of potentially
Saier MH Jr & Paulsen IT (2001) Phylogeny of multidrug mobile DNA elements encoding site-specific gene-integration
transporters. Semin Cell Dev Biol 12: 205–213.
functions: integrons. Mol Microbiol 3: 1669–1683.
Saint-Ruf C & Matic I (2006) Environmental tuning of mutation
Stoodley P, Sauer K, Davies DG & Costerton JW (2002) Biofilms
rates. Environ Microbiol 8: 193–199.
as complex differentiated communities. Annu Rev Microbiol
Salunkhe P, Topfer T, Buer J & Tummler B (2005) Genome-wide
56: 187–209.
transcriptional profiling of the steady-state response of
Stumpf JD & Foster PL (2005) Polyphosphate kinase regulates
Pseudomonas aeruginosa to hydrogen peroxide. J Bacteriol 187:
error-prone replication by DNA polymerase IV in Escherichia
2565–2572.
coli. Mol Microbiol 57: 751–761.
Sanchez MB, Hernandez A, Rodriguez-Martinez JM, Martinez-
Suci PA, Mittelman MW, Yu FP & Geesey GG (1994)
Martinez L & Martinez JL (2008) Predictive analysis of
Investigation of ciprofloxacin penetration into Pseudomonas
transmissible quinolone resistance indicates Stenotrophomonas
aeruginosa biofilms. Antimicrob Agents Ch 38: 2125–2133.
maltophilia as a potential source of a novel family of Qnr
Sulavik MC, Dazer M & Miller PF (1997) The Salmonella
determinants. BMC Microbiol 8: 148.
typhimurium mar locus: molecular and genetic analyses and
Sanchez P, Alonso A & Martinez JL (2002a) Cloning and
characterization of SmeT, a repressor of the Stenotrophomonas assessment of its role in virulence. J Bacteriol 179: 1857–1866.
Tamae C, Liu A, Kim K et al. (2008) Determination of antibiotic
maltophilia multidrug efflux pump SmeDEF. Antimicrob
Agents Ch 46: 3386–3393. hypersensitivity among 4,000 single-gene-knockout mutants
Sanchez P, Linares JF, Ruiz-Diez B, Campanario E, Navas A, of Escherichia coli. J Bacteriol 190: 5981–5988.
Baquero F & Martinez JL (2002b) Fitness of in vitro selected Thanassi DG, Cheng LW & Nikaido H (1997) Active efflux of bile
Pseudomonas aeruginosa nalB and nfxB multidrug resistant salts by Escherichia coli. J Bacteriol 179: 2512–2518.
mutants. J Antimicrob Chemoth 50: 657–664. Thomas CM (2000) Paradigms of plasmid organization. Mol
Sanchez P, Alonso A & Martinez JL (2004) Regulatory regions of Microbiol 37: 485–491.
smeDEF in Stenotrophomonas maltophilia strains expressing Thomas CM & Nielsen KM (2005) Mechanisms of, and barriers
different amounts of the multidrug efflux pump SmeDEF. to, horizontal gene transfer between bacteria. Nat Rev
Antimicrob Agents Ch 48: 2274–2276. Microbiol 3: 711–721.
Sanchez P, Moreno E & Martinez JL (2005) The biocide triclosan Tierrez A & Garcia-del Portillo F (2004) The Salmonella
selects Stenotrophomonas maltophilia mutants that membrane protein IgaA modulates the activity of the RcsC-
overproduce the SmeDEF multidrug efflux pump. Antimicrob YojN-RcsB and PhoP-PhoQ regulons. J Bacteriol 186:
Agents Ch 49: 781–782. 7481–7489.
Sauer K, Camper AK, Ehrlich GD, Costerton JW & Davies DG Tock MR & Dryden DT (2005) The biology of restriction and
(2002) Pseudomonas aeruginosa displays multiple phenotypes anti-restriction. Curr Opin Microbiol 8: 466–472.
during development as a biofilm. J Bacteriol 184: 1140–1154. Trinh V, Langelier MF, Archambault J & Coulombe B (2006)
Schlichting CD (2008) Hidden reaction norms, cryptic genetic Structural perspective on mutations affecting the function of
variation, and evolvability. Ann NY Acad Sci 1133: 187–203. multisubunit RNA polymerases. Microbiol Mol Biol R 70:
Sibold C, Henrichsen J, Konig A, Martin C, Chalkley L & 12–36.
Hakenbeck R (1994) Mosaic pbpX genes of major clones of Truong-Bolduc QC, Zhang X & Hooper DC (2003)
penicillin-resistant Streptococcus pneumoniae have evolved Characterization of NorR protein, a multifunctional regulator


c 2008 Federation of European Microbiological Societies FEMS Microbiol Rev 33 (2009) 44–65
Published by Blackwell Publishing Ltd. All rights reserved
Antibiotic resistance 65

of norA expression in Staphylococcus aureus. J Bacteriol 185: Walsh TR (2006) Combinatorial genetic evolution of
3127–3138. multiresistance. Curr Opin Microbiol 9: 476–482.
Truong-Bolduc QC, Dunman PM, Strahilevitz J, Projan SJ & Webb V & Davies J (1993) Antibiotic preparations contain DNA:
Hooper DC (2005) MgrA is a multiple regulator of two new a source of drug resistance genes? Antimicrob Agents Ch 37:
efflux pumps in Staphylococcus aureus. J Bacteriol 187: 2379–2384.
2395–2405. White DG, Goldman JD, Demple B & Levy SB (1997) Role of the
Tsui HC, Feng G & Winkler ME (1997) Negative regulation of acrAB locus in organic solvent tolerance mediated by
mutS and mutH repair gene expression by the Hfq and RpoS expression of marA, soxS, or robA in Escherichia coli.
global regulators of Escherichia coli K-12. J Bacteriol 179: J Bacteriol 179: 6122–6126.
7476–7487. Whiteley M, Bangera MG, Bumgarner RE, Parsek MR, Teitzel
Tsui WH, Yim G, Wang HH, McClure JE, Surette MG & Davies J GM, Lory S & Greenberg EP (2001) Gene expression in
(2004) Dual effects of MLS antibiotics: transcriptional Pseudomonas aeruginosa biofilms. Nature 413: 860–864.

Downloaded from https://academic.oup.com/femsre/article/33/1/44/2683937 by guest on 09 December 2020


modulation and interactions on the ribosome. Chem Biol 11: Williams AB & Foster PL (2007) The Escherichia coli histone-like
1307–1316. protein HU has a role in stationary phase adaptive mutation.
Turnidge J & Paterson DL (2007) Setting and revising Genetics 177: 723–735.
antibacterial susceptibility breakpoints. Clin Microbiol Rev 20: World Health Organization (1999) Removing obstacles for healthy
391–408. development. World Health Organization, Geneva.
Udani RA & Levy SB (2006) MarA-like regulator of multidrug World Health Organization (2000) Overcoming antibiotic
resistance in Yersinia pestis. Antimicrob Agents Ch 50: resistance. World Health Organization, Geneva.
2971–2975. Worthington T, White J, Lambert P, Adlakha S & Elliott T (1999)
Van Bambeke F, Chauvel M, Reynolds PE, Fraimow HS & Beta-lactam-dependent coagulase-negative staphylococcus
associated with urinary-tract infection. Lancet 354: 1097.
Courvalin P (1999) Vancomycin-dependent Enterococcus
Wrande M, Roth JR & Hughes D (2008) Accumulation of
faecalis clinical isolates and revertant mutants. Antimicrob
mutants in ‘‘aging’’ bacterial colonies is due to growth under
Agents Ch 43: 41–47.
selection, not stress-induced mutagenesis. P Natl Acad Sci USA
van Loosdrecht MC, Heijnen JJ, Eberl H, Kreft J & Picioreanu C
105: 11863–11868.
(2002) Mathematical modelling of biofilm structures. Antonie
Wright GD (2007) The antibiotic resistome: the nexus of
Van Leeuwenhoek 81: 245–256.
chemical and genetic diversity. Nat Rev Microbiol 5: 175–186.
Ventre I, Goodman AL, Vallet-Gely I et al. (2006) Multiple
Wu J & Weiss B (1991) Two divergently transcribed genes, soxR
sensors control reciprocal expression of Pseudomonas
and soxS, control a superoxide response regulon of Escherichia
aeruginosa regulatory RNA and virulence genes. P Natl Acad
coli. J Bacteriol 173: 2864–2871.
Sci USA 103: 171–176.
Yim G, de la Cruz F, Spiegelman GB & Davies J (2006a)
von Gotz F, Haussler S, Jordan D et al. (2004) Expression analysis
Transcription modulation of Salmonella enterica serovar
of a highly adherent and cytotoxic small colony variant of
Typhimurium promoters by sub-MIC levels of rifampin.
Pseudomonas aeruginosa isolated from a lung of a patient with J Bacteriol 188: 7988–7991.
cystic fibrosis. J Bacteriol 186: 3837–3847. Yim G, Wang HH & Davies J (2006b) The truth about antibiotics.
Wagner J, Gruz P, Kim SR, Yamada M, Matsui K, Fuchs RP & Int J Med Microbiol 296: 163–170.
Nohmi T (1999) The dinB gene encodes a novel E. coli DNA Yim G, Wang HH & Davies J (2007) Antibiotics as signalling
polymerase, DNA pol IV, involved in mutagenesis. Mol Cell 4: molecules. Philos T Roy Soc B 362: 1195–1200.
281–286. Zeldovich KB, Chen P & Shakhnovich EI (2007) Protein stability
Waite RD, Papakonstantinopoulou A, Littler E & Curtis MA imposes limits on organism complexity and speed of
(2005) Transcriptome analysis of Pseudomonas aeruginosa molecular evolution. P Natl Acad Sci USA 104: 16152–16157.
growth: comparison of gene expression in planktonic cultures Zhang L & Mah TF (2008) Involvement of a novel efflux system
and developing and mature biofilms. J Bacteriol 187: in biofilm-specific resistance to antibiotics. J Bacteriol 190:
6571–6576. 4447–4452.
Waite RD, Paccanaro A, Papakonstantinopoulou A, Hurst JM, Zhang L, Xu Z & Patel BK (2007) Bacillus decisifrondis sp. nov.,
Saqi M, Littler E & Curtis MA (2006) Clustering of isolated from soil underlying decaying leaf foliage. Int J Syst
Pseudomonas aeruginosa transcriptomes from planktonic Evol Micr 57: 974–978.
cultures, developing and mature biofilms reveals distinct Ziha-Zarifi I, Llanes C, Kohler T, Pechere JC & Plesiat P (1999) In
expression profiles. BMC Genomics 7: 162. vivo emergence of multidrug-resistant mutants of
Waksman SA & Woodruff HB (1940) The soil as a source of Pseudomonas aeruginosa overexpressing the active efflux
microorganisms antagonistic to disease-producing bacteria. system MexA-MexB-OprM. Antimicrob Agents Ch 43:
J Bacteriol 40: 581–600. 287–291.

FEMS Microbiol Rev 33 (2009) 44–65 


c2008 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

You might also like