You are on page 1of 21

235

Molecular Mechanisms of Antibiotic


18  Resistance in Bacteria
Steven M. Opal and Aurora Pop-Vicas

MOLECULAR GENETICS OF multiple antibiotic resistance to a wide array of antibiotic classes.


ANTIBIOTIC RESISTANCE Aquatic environments are particularly rich with bacterial populations
Genetic variability is essential for microbial evolution to occur. The replete with antibiotic-resistance genes.12 Such environmental bacterial
fitness of a microorganism depends on its capacity to adapt to changing strains likely provide potential human pathogens with a source of
environmental conditions.1 Antimicrobial agents exert strong selective novel antibiotic-resistance genes.11,12 Selection pressures placed on
pressures on bacterial populations, favoring organisms that are capable microbial populations by antibiotics favor the expansion of strains
of resisting them.1,2 Genetic variability may occur by a variety of mech- that have the capacity to resist the inhibitory effects of antibiotics.
anisms. Point mutations may occur in a nucleotide base pair, which is These resistant populations proliferate and spread antibiotic-resistance
referred to as microevolutionary change. These mutations may alter genes vertically to subsequent generations and horizontally to suscep-
enzyme substrate specificity or the target site of an antimicrobial agent, tible strains of related bacteria, or even between species or different
interfering with its activity. Point mutations at crucial locations on genera.13 Although some antibiotic-resistance genes place a metabolic
“old” β-lactamase genes (e.g., genes for Temoneira-1 [TEM-1], sulfhy- “burden” on bacteria, many microorganisms evolved strategies to limit
dryl variable-1 [SHV-1]) are primarily responsible for the remarkable this cost by repressing gene expression when not needed or by phase
array of newly recognized extended-spectrum β-lactamases (ESBLs).3,4 variation. This allows favorable, but sometimes “costly,” antibiotic-
A second level of genomic variability in bacteria is referred to as a resistance genes to be held in reserve in the absence of antibiotic selec-
macroevolutionary change and results in whole-scale rearrangements tion pressure yet express their resistance potential upon reexposure to
of large segments of DNA as a single event. These rearrangements may antibiotics.14
include inversions, duplications, insertions, deletions, or transposition Continous exposure to foreign DNA within microbial communities
of large sequences of DNA from one location of a bacterial chromo- is so commonplace that many bacteria have evolved systems to defend
some or plasmid to another. These large-scale rearrangements of entire their genomes from exogenous DNA, phages, and plasmid insertion.
segments of the bacterial genome are frequently generated by special- This is accomplished by at least two mechanisms: (1) species and
ized genetic elements called integrons and transposons or insertion strain-specific DNA modifying enzymes (e.g., methylation of selected
sequences, which have the capacity to insert, rearrange, and move inde- sequences of host DNA into specific patterns) and restriction enzymes
pendently from the rest of the bacterial genome.2 that survey cellular host DNA and degrade foreign DNA that lacks
A third level of genetic variability in bacteria is created by the appropriate DNA modification seqences and (2) a type of adaptive
acquisition of large segments of foreign DNA carried by plasmids, defense system against foreign DNA known as CRISPR (clustered
bacteriophages, naked sequences of DNA, or specialized transposable regularly spaced short palindromic repeats).15 CRISPRs are detectable
genetic elements known as integrative and conjugative elements (ICE) in nearly 50% of all bacterial genomes, and this genetic element pro-
from other bacteria.2 These events are termed lateral or horizontal gene tects their genomes from attack by foreign DNA during transforma-
transfer and are now known to be a frequent event, especially for natu- tion, phage invasion, or plasmid insertion.
rally competent bacteria that can take up exogenous DNA from the The mechanism of protection is mediated by inserting small
environment. Inheritance of foreign DNA further contributes to the sequences of the mobilized invading DNA between palindromic
organism’s genetic variability and its capacity to respond to selection repeats within the CRISPR. Upon reexposure to similar DNA sequences
pressures imposed by antimicrobial agents.3 These mechanisms endow from phage or invading bacteria, the existing sequence within the
bacteria with the seemingly unlimited capacity to develop resistance CRISPR is transcribed into a small RNA (known as crRNA) that asso-
to any antimicrobial agent (Fig. 18-1). Examples of plasmid-mediated ciates with CRISPR-associated nucleases and prevents integration of
carbapenemase-producing Klebsiella pneumoniae,5 vancomycin- the targeted foreign DNA.
resistant and daptomycin-resistant Staphylococcus aureus,6,7 multidrug- Maintaining the fidelity of the host genome, while permiting
resistant Yersinia pestis,8 and transferable quinolone resistance in limited variation by microevolutionary and macroevolutionary
enterobacteria9 attest to the capacity of microorganisms to adapt to changes, allows pathogens to strike a balance between genomic stabil-
environmental stresses such as antibiotic exposure. When an antibiotic- ity and plasticity in rapidly changing microenvironments. Recent evi-
resistance gene evolves, this gene can spread between bacteria by trans- dence in the enterococci indicates that deletion of CRISPR elements
formation, transduction, conjugation, or transposition. Favored clones is inversely related to multiple antibiotic-resistance development.
of bacteria may proliferate in the microbiota of patients who receive CRISPR-deficient strains have been selected for, and are particularly
antibiotics.10 well-adapted for, health care–associated infections. These strains have
Antibiotic-resistance genes existed well before the introduction of significantly larger genomes as a result of insertion of large sequences
antimicrobials for treatment for human infections and can be found of DNA, including genes that mediate multiple antibiotic resistance.16
within bacteria in Arctic region permafrost samples untouched by
human hands for over 30,000 years.10 Antibiotics are often synthesized Plasmids
by antibiotic-producing bacteria when they are about to enter the late Extrachromosomal elements were present in bacteria before the advent
static growth phase and are about to become dormant or sporulate. of antibiotics.10,13 The introduction of antibiotics into clinical medicine
Antibiotic-producing bacteria are resistant to the antibiotics they in the 20th century created selection pressures, however, that favor the
produce. Therefore, antibiotics left in the microenvironment by dissemination of resistance genes via mobile genetic elements.2,3,13 Plas-
dormant bacteria are avoided as potentially toxic to competitors yet mids are particularly well adapted to serve as agents of genetic exchange
serve as a ready source of carbon (food) for the next generation of the and resistance-gene dissemination.1,3 Plasmids are autonomously rep-
antibiotic-producing bacterial strain once the growth phase begins licating genetic elements that generally consist of covalently closed,
again. Environmental levels of multiple classes of antimicrobial agents circular, double-stranded DNA molecules ranging from less than 10
are now so common in soil and water samples that multiple bacterial kilobase (kb) pairs to greater than 400 kb. They are extremely common
genera have strains that subsist entirely upon antibiotics as their sole in bacteria.13 Although multiple copies of a specific plasmid, or mul-
carbon source.11 These bacteria express remarkably high levels of tiple different plasmids, or both may be found in a single bacterial cell,
235
235.e1
KEYWORDS
antibiotic resistance; β-lactamases; carbapenemases; efflux pumps;
genomic rearrangement; integrons; lateral gene transfer; multidrug

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


resistance; plasmids; transposons
236
Donor replicate plasmid DNA. Plasmids must also retain a set of genes that
Nonconjugative facilitate their stable maintenance in host bacteria. The transfer of
plasmid plasmid DNA between bacterial species is a complex process, and the
Tet M genes needed for transfer (tra genes) make conjugative plasmids larger
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

bla
Transposon Chromosome than nonconjugative ones. Some small plasmids may be able to transfer
PBP' to other bacteria via the use of the conjugation apparatus provided by
co-resident conjugative plasmids or even conjugative transposons.
Conjugative
plasmid Many plasmid-encoded functions enable bacterial strains to persist in
the environment by resisting noxious agents, such as heavy metals.
Process Recipient
Mercury released from dental fillings may increase the number of
Transformation
antibiotic-resistant bacteria in the mouth.17 Compounds such as hexa-
chlorophene and quarternary ammonium compounds are used as

xx
PBP'
topical bacteriostatic agents, and plasmid-mediated resistance to these
PBP' agents has increased significantly.2
PBP'
Transposable Genetic Elements
A Transposons can translocate as a unit from one area of the bacterial
Transduction chromosome to another or between the chromosome and plasmid
Phage or bacteriophage DNA. Transposable genetic elements possess a spe-
bla cialized system of recombination that is independent of the general-
bla ized recombination system that classically permits recombination of
largely homologous sequences of DNA by crossover events (the recA
system of bacteria). The recA-independent recombination system
(“transposase”) of transposable elements usually occurs in a random
B fashion between nonhomologous sequences of DNA and results in
Tet M
Conjugation
whole-scale modifications of large sequences of DNA as a single event
Mating (Fig. 18-2).2,5
bridge There are two types of transposable genetic elements, called trans-
Mobilized posons and insertion sequences, that have similar characteristics. Evi-
Tet Mtransfer bla dence from whole-genome sequencing projects indicates that bacterial
chromosomes are replete with transposable elements.18 These mobile
C sequences probably play an important physiologic role in genetic varia-
Conjugative
tion and evolution in prokaryotic organisms. Transposons differ from
transposition
insertion sequences in that they encode functional genes that mediate
Tet M a recognizable phenotypic characteristic, such as an antibiotic-
resistance marker. Either element can translocate as an independent
unit. Both elements are usually flanked on either end by short identical
sequences of DNA in reverse order (inverted repeats). These inverted-
repeat DNA termini are essential to the transposition process. Trans-
D
posons (Tn) and insertion sequences (IS) are incapable of autonomous
FIGURE 18-1  Examples of recombination events and molecular self-replication and must exist on a replicon, such as the chromosome,
spread of antibiotic-resistance genes. The donor organism depicted bacteriophage, or plasmid, to be replicated and maintained in a bacte-
here has three antibiotic-resistance genes: the first on the chromosome,
rial population. Some transposons have the capability to move from
designated as PBP′, a low-affinity penicillin-binding protein; the second (a
β-lactamase gene labeled bla) on a small nonconjugative plasmid; and the one bacterium to another without being fixed within a plasmid or
third (Tet M, a tetracycline resistance determinant) on a transposon resid- bacteriophage. These elements are referred to as conjugative transpo-
ing on a large self-conjugative plasmid. A, Genetic exchange may occur sons, or integrative and conjugative elements (ICE). The ubiquitous
by transformation (naked DNA transfer for dying bacteria to a competent transposable element Tn916 and its derivatives are examples of conju-
recipient). This generally results in transfer of homologous genes located gative transposons and have been found primarily in aerobic and
on the chromosome by recombination enzymes (RecA). B, Transduction anaerobic gram-positive organisms, although they can also exist in
also may transfer antibiotic-resistance genes (usually from small plasmids) gram-negative bacteria.2,19,20
by imprecise packaging of nucleic acids by transducing bacteriophages.  Transposition usually results in the localized replication of the
C, Conjugation is an efficient method of gene transfer, requiring physical
transposable element from the original donor sequence of DNA and
contact between donor and recipient. Self-transferable plasmids mediate
direct contact by forming a mating bridge between cells. Smaller noncon- the insertion of a copy of the transposable element into the recipient
jugative plasmids might be mobilized in this mating process and be trans- sequence of DNA (replicative transposition).1,2 Transposition, similar
ported into the recipient. D, Transposons are specialized sequences of DNA to point mutation, is a continuous, ongoing process in bacterial popu-
that possess their own recombination enzymes (transposases), allowing lations. An example of this phenomenon is the spread of a tetracycline-
transposition (“hopping”) from one location to another, independent of resistance transposon among Neisseria gonorrhoeae, Mycoplasma
the recombination enzymes of the host (RecA-independent). They may hominis, and Ureaplasma urealyticum.21,22
transpose to nonhomologous sequences of DNA and spread antibiotic- An important variant of transposition is “one-ended” transposition,
resistance genes to multiple plasmids or genomic locations throughout the where only one end of the transposon is responsible for asymmetrical
host. Some transposons possess the ability to move directly from a donor
replication. This type of element is highly efficient in mobilizing resis-
to a recipient, independent of other gene transfer events (conjugative
transposons or integrative and conjugative elements). tance genes adjacent to its insertion site. These elements play a promi-
nent role, along with ICEs and integrons, in the evolution of large
regions of the chromosome where multiple resistance genes accumu-
late into one set of resistance gene cassettes known as resistance
closely related plasmids often cannot coexist in the same cell. This genomic islands.2 Some of these genomic islands are enormous in size
observation led to a classification scheme of plasmids based on incom- and scope. For example, the AbaR1 genomic island of Acinetobacter
patibility (Inc) groups.1,4 baumannii is 86 kb long and contains 46 different antimicrobial resis-
Plasmids may determine a wide range of functions besides antibi- tance genes to a wide swath of antimicrobials, antiseptics, and heavy
otic resistance, including virulence and metabolic capacities. All plas- metals.23 Once genomic resistance islands form, they often persist and
mids possess an origin of replication for DNA polymerase to bind and gain new genes over time. These islands serve as convenient depots for
237
Insertion sites for
5'-CS new gene cassettes 3'-CS

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


intl1 1
R1 R2 R3 qacE∆1 sul1 orf5
2

5' 3'
IRi attl1 IRt
59-bp elements
FIGURE 18-3  Organization of a hypothetical class I integron. The
5′ conserved sequence (5′-CS) contains a site-specific integrase (intI1); an
attachment site (attI1), which functions as a receptor for new gene cas-
settes; and two potential promoter sites (P1 and P2). The promoter is the
initiation site for the transcription of the multiple, potential, antibiotic gene
cassettes (labeled R1, R2, R3) that are inserted downstream from the
promoter. Repeated, variable-length, but usually 59-bp elements, flank the
central antibiotic-resistance gene cassettes. The conserved 3′ end of the
integron (3′-CS) usually consists of a gene for resistance to quaternary
ammonium compounds, a sulfonamide-resistance gene, and another open
reading frame (orf5). The outer boundaries of the integron structure are
flanked by a 25-bp inverted repeat sequence (noted as IRi and IRt).
A

TRANSPOSABLE Genetic exchange of antibiotic-resistance genes occurs between


DNA ELEMENT bacteria of widely disparate species and different genera.26 Identical
* aminoglycoside-resistance genes occur in streptococci and Campylo-
bacter,27 and enterococci apparently have acquired aminoglycoside28
* and β-lactam29 resistance from staphylococci. Given the highly variable
Central
Flanking Flanking environmental selection pressures created by the clinical, agricultural,
sequences
duplicated duplicated and industrial use of antibiotics and the plasticity of bacterial genomes,
recipient DNA Terminal repeat sequences recipient DNA the ongoing evolution of multiresistant species seems inevitable.29,30
on transposable element
B (direct or inverted) DNA Integration Elements
FIGURE 18-2  A, Characteristic appearance of a transposon by electron
The structural genes that mediate antibiotic resistance often are closely
microscopy showing the stem-loop configuration. The kanamycin resis- linked and may exist in tandem along the bacterial chromosome or
tance transposon Tn903 is inserted into a small plasmid (pSC105). After plasmid. Genetic analysis of sequences of DNA adjacent to antibiotic-
denaturation, intrastrand annealing of the complementary 1000-bp, resistance genes has revealed that unique integration units often exist
inverted repeat, terminal sequences of the transposon form the stem near promoter sites.31
structure. The kanamycin-resistance gene and the genes necessary for These integration elements, called integrons,32 function as recombi-
transposition are located in the central loop structure. B, Structure of a national “hot spots” for site-specific recombination events between
transposable element inserted into a recipient DNA sequence. The trans- largely nonhomologous sequences of DNA. Integrons facilitate the
poson (rectangles and wavy lines) consists of a central sequence containing lateral transfer and integration of antibiotic-resistance genes from
the phenotypic marker gene(s) (antibiotic-resistance gene) and the “trans-
posase” genes. The terminal-repeat sequences of the transposon flank the
mobile gene cassettes. The integron provides its own unique integrase
central sequences on both sides. Insertion of the transposon results in function33 that facilitates recA-independent recombination and a spe-
single-strand, staggered cuts in the recipient DNA (asterisks). Subsequent cialized attachment and integration site consisting of a variable length
gap-filling DNA synthesis and ligation results in duplication of a short (57 to 141 base pairs [bp]) but often a 59-bp spacer sequence of highly
sequence of recipient DNA at either end of the transposon. conserved DNA. This 59-bp element is preserved at the 3′ end of
inserted antibiotic-resistance genes.34,35
Although these integration elements differ structurally and func-
insertions of new DNA without the risk of insertion into a critical tionally from transposons,36 they seem to be widespread in bacterial
metabolic or structural gene of the bacterial genome. populations and play an important role in the dissemination of
The “fitness cost” of acquiring new resistance genes is minimized antibiotic-resistance genes.2,21,37,38 Integrons do not transpose indepen-
by depositing genes in genomic resistance islands.2 However, the meta- dently as a specific unit structure from one sequence of DNA to
bolic cost of retaining all these accessory, antibiotic-resistance genes another. This capability of autonomous movement of large sequences
by host bacteria remains. The extra metabolic burden is only worth it of DNA is reserved primarily for transposons, insertion sequence ele-
in the presence of repeated environmental antibiotic exposure as might ments, and bacteriophages. They may become flanked, however, by
exist in hospitals and special care areas, such as the critical care unit, transposable elements and become integrated into an existing transpo-
where antibiotic selection pressures favor multidrug-resistant organ- son. The principal role of integrons is to provide a convenient insertion
isms. Antibiotic-resistance genes, especially those existing on plas- site for antibiotic-resistance genes from foreign DNA sources.
mids, are in fact frequently disposed of when bacteria are removed There are five classes of integrons that encode antibiotic-resistance
from such environments and maintained in antibiotic-free media. genes, with type 1 integrons being the most common in pathogenic
Transposons also are essential in the evolution of resistance plas- microorganisms.38 A schematic representation of a class 1 integron is
mids that contain multiple antibiotic-resistance determinants.19 The shown in Figure 18-3. Integrons also serve as expression cassettes for
recent spread of carbepenem resistance from metallo-β-lactamases antibiotic-resistance genes in that an efficient promoter site is provided
among the Enterobacteriaceae in some European hospitals was facili- in close proximity to the 5′ end of the newly inserted DNA sequence.
tated by transposition of resistance genes on integrons to separate The frequency of transcription of integrated cassettes of antibiotic-
plasmids from different bacterial genera of gram-negative bacilli.24 resistance genes depends on the proximity of the gene to the promoter
High-level vancomycin resistance (vanA) in enterococci is mediated at the 5′ upstream end of the integron. The level of expression of a
by a composite transposon that encodes a series of genes needed to resistance gene diminishes as the distance between the promoter and
express vancomycin resistance.25 Single transposons may encode mul- the specific antibiotic-resistance gene cassette increases.33 Numerous
tiple antibiotic-resistance determinants within their inverted-repeat clusters of different antibiotic-resistance genes have been identified
termini as well.2 that have evolved through specific insertions into common integrons.34
238
Integrons have been found to possess five or more antibiotic-resistance H. influenzae.45 The extended-spectrum of activity for TEM-derived
genes lined up in a tandem sequence along a single, functioning ESBLs is obtained through changes in a single or a few amino acids
integron.39 that change the configuration of the enzyme at its active site, making
Complex integrons have recently been described that are typified by it more accessible to the bulky R1 oxymino side chains of third-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

a common open reading frame (orf 513) linked to the 3′ end of a typical generation cephalosporins (cefotaxime, cefpodoxime, ceftazidime, cef-
integron, followed by a series of inserted genes (often expanded- triaxone), and monobactam (aztreonam).51 The first TEM-derived
spectrum β-lactamases) after a duplication of the 3′ end of the integron. ESBL, TEM-3, was reported in 1988.52 There are now more than 200
This common 3′ end of type 1 integrons encodes genes for resistance TEM-derived ESBLs (see Lahey Clinic [Burlington, MA] website,
to quarternary ammonium compounds (qacE1), sulfonamide resistance www.lahey.org/Studies/temtable.asp). These enzymes are found pri-
(sul1), and an open reading frame of unknown function (orf5). These marily in E. coli and K. pneumoniae isolates but also in other Entero-
complex integrons can be mobilized and spread by adjacent insertion bacteriaceae, such as Enterobacter aerogenes, Morganella morganii,
sequences to disseminate among bacterial populations.40 Proteus spp., and Salmonella spp.45 The majority of TEM-derived
ESBLs remain susceptible to inhibition by clavulanic acid, although
MECHANISMS OF ANTIBIOTIC inhibitor-resistant TEM variants have also been described.53
RESISTANCE SHV-Derived.  The SHV-1 β-lactamase has a biochemical struc-
At least eight distinctive mechanisms of antibiotic resistance have been ture similar to that of TEM-1 (68% of amino acids are shared54), and
described in bacteria (Table 18-1). Examples of each of these mecha- its ESBL derivatives are also produced by point mutations (one or more
nisms are described in the following paragraphs. amino-acid substitutions) at its active site. SHV-type β-lactamases are
found primarily in K. pneumoniae strains. A three-dimensional image
Enzymatic Inhibition of TEM-type and SHV-type ESBLs is provided in Figure 18-5.
β-Lactamases CTX-M–Derived.  Cefotaxime-M (CTX-M) β-lactamases are not
Resistance to β-lactam antibiotics occurs primarily through produc- evolutionarily related to SHV and TEM families because they are
tion of β-lactamases, enzymes that inactivate these antibiotics by split- thought to have been acquired by plasmids from the chromosomal
ting the amide bond of the β-lactam ring. β-Lactamases have likely ampicillin C (AmpC) enzymes of Kluyvera spp., environmental gram-
coevolved with bacteria as mechanisms of resistance against natural negative rods of low pathogenic potential.55,56 In general, the CTX-M
antibiotics over time, and the selective pressure exerted by the wide- family hydrolyzes cefotaxime and ceftriaxone better than ceftazidime,
spread use of antimicrobial therapy in modern medicine may have and they are inhibited more by tazobactam than by clavulanic acid,45,54
accelerated their development and spread. although point mutations leading to increased activity against ceftazi-
β-Lactamases are encoded either by chromosomal genes or by dime can occur. CTX-M enzymes have disseminated rapidly and are
transferable genes located on plasmids and transposons. In addition, now among the most prevalent ESBLs worldwide.57 Recent reports of
β-lactamase genes (bla) frequently reside on integrons, which often community-acquired bloodstream infections with multiresistant,
carry multiple resistance determinants. If mobilized by transposable CTX-M Escherichia coli isolates from Spain and Israel have raised
elements, integrons can facilitate further dissemination of multidrug significant public health concerns.58,59 The ST131 (O25:H4) clone asso-
resistance among different bacterial species.41 ciated with the CTX-M-15 enzymes has emerged as an important
β-Lactamases can be classified according to their amino-acid struc- multidrug-resistant pathogen and may have been responsible for the
ture into four molecular classes, A through D (Table 18-2), as first majority of infections with multidrug-resistant E. coli infections in
suggested by Ambler. Alternatively, the Bush-Jacoby-Medeiros system Europe and the United States since 2007.60
classifies the enzymes according to their substrate profile and suscep- OXA-Derived.  Oxacillin (OXA)-type β-lactamases are also
tibility to β-lactamase inhibitors, such as clavulanic acid, into several plasmid derived and hydrolyze oxacillin and its derivatives very effec-
functional groups (Table 18-3).42 Class A, C, and D β-lactamases tively; they are poorly inhibited by clavulanic acid.42,45 OXA-derived
hydrolyze the β-lactam ring through a serine residue at their active site, ESBLs have been described mainly in P. aeruginosa, in which they
whereas class B enyzmes are metallo-β-lactamases that use zinc (Zn)2+ confer high-level resistance to oxymino-β-lactams.45
to break the amide bond (Fig. 18-4). AmpC Enzymes.  AmpC β-lactamases are primarily chromoso-
The first β-lactamase was described as a “penicillinase” capable mial enzymes that confer resistance to penicillins, narrow-spectrum
of hydrolyzing penicillin in Escherichia coli in 1940, about the same cephalosporins, oxymino-β-lactams, and cephamycins and are not sus-
time as the first clinical use of penicillin was reported in the literature.43 ceptible to β-lactamase inhibitors such as clavulanic acid (molecular
The next years witnessed the rapid spread of plasmid-encoded penicil- class C, functional group 1).61 Cefepime and aztreonam are usually
lin resistance among the majority of S. aureus clinical isolates.44,45 poor substrates, although modulation by point mutations at the R2
Among gram-negative organisms, the rise in ampicillin resistance in loop of the active site have been responsible for variants with increased
the 1960s was ascribed to the emergence of TEM-1, a plasmid-encoded ability to hydrolyze cefepime. AmpC production in gram-negative
β-lactamase named after a Greek patient, Temoniera, in whom the first bacilli is normally repressed. However, a transient increase in produc-
isolate was recovered. The family of TEM β-lactamases disseminated tion (10- to 100-fold) can occur in the presence of β-lactam antibiotics
worldwide through various Enterobacteriaceae, as well as Pseudomo- in the following species that possess inducible AmpC enzymes: Entero-
nas aeruginosa, Haemophilus influenzae, and N. gonorrhoeae.46,47 Simi- bacter, Citrobacter freundii, Serratia, M. morganii, Providencia, and P.
larly, both chromosomally encoded, as well as plasmid-mediated aeruginosa.62 AmpC β-lactamase production returns to low levels again
SHV-type β-lactamases, with a molecular structure related to TEM after antibiotic exposure is discontinued, unless spontaneous muta-
enzymes, became widely prevalent among E. coli and K. pneumoniae tions occur in the ampD locus of the gene, leading to permanent
isolates. hyperproduction (derepression) in these species. Third-generation
cephalosporin use in Enterobacter spp. infections can therefore select
Extended-Spectrum β-Lactamases for the overgrowth of these stably derepressed mutants, leading to the
The pharmaceutical industry’s development of third-generation cepha- emergence of antibiotic resistance during treatment.61,63
losporins, initially stable to the action of TEM- and SHV-type More than 20 plasmid-mediated AmpC enzymes, derived from
β-lactamases, was soon followed by the emergence and global spread chromosomally encoded genes in Enterobacteriaceae or Aeromonas
of ESBL, capable of hydrolyzing monobactam and broad-spectrum spp., have been described in E. coli, K. pneumoniae, Salmonella enterica,
cephalosporins.3,45,46,47,48 In addition, increasing reports of carbapene- and Proteus mirabilis. β-Lactam resistance attributable to this system
mase emergence and spread have raised concern over the currently appears to be increasing and confers a resistance phenotype similar to
limited antimicrobial arsenal against infections with multidrug- that of Enterobacter spp.47
resistant gram-negative bacteria.49,50 Carbapenemases.  Carbapenemases confer the largest antibiotic-
TEM-Derived.  TEM-1 is the most common β-lactamase in resistance spectrum because they can hydrolyze not only carbapenems
gram-negative bacteria, and it can hydrolyze penicillins and narrow- but also broad-spectrum penicillins, oxymino-cephalosporins, and
spectrum cephalosporins in Enterobacteriaceae, N. gonorrhoeae, and cephamycins. The K. pneumoniae carbapenemase (KPC) enzymes are
TABLE 18-1  Eight Major Mechanisms of Resistance by Antimicrobial Class
AMINO- CHLORAM- LINOSAMIDE;
β-LACTAM GLYCOSIDE PHENICOL MACROLIDE SULFONAMIDE TETRACYCLINE TRIMETHOPRIM QUINOLONE GLYCOPEPTIDE STREPTOGRAMIN RIFAMPIN
Enzymatic +++ +++ +++ + (gram- − − − + − − −
inactivation negative)
Decreased + (gram- + (gram- + (gram- ++ (gram- − + (gram-negative) + (gram-negative) + (gram- ++ (gram-negative) + (gram-negative) −
permeability negative) negative) negative) negative) negative)
Efflux + + + ++ − +++ − + − − −
Alteration of ++ ++ − +++ ++ + (Helicobacter +++ +++ +++ +++ +++
target site pylori)
Protection of − − − − ++ − + − − −
target site
Overproduce − − − − ++ − ++ − + − −
target
Bypass of − − − − + − + − − − −
inhibited
process
Bind up − − − − − − − − ++ − −
antibiotic
+++, most common mechanism; ++, common; + less common.
239

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


240

TABLE 18-2  Ambler Classification of β-Lactamases


CLASS ACTIVE SITE ENZYME TYPE SUBSTRATES EXAMPLE
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

A Serine Penicillinases:
Broad-spectrum Benzylpenicillin, aminopenicillins, PC1 in Staphylococcus aureus
carboxypenicillins, ureidopenicillins, TEM-1, SHV-1 in Escherichia coli, Klebsiella
narrow-spectrum cephalosporins pneumoniae, other gram-negative bacteria
Extended- Substrates of broad-spectrum plus In Enterobacteriaceae: TEM-derived, SHV-derived,
spectrum oxymino-β-lactams (cefotaxime, CTX-M–derived; PER-1, VEB-1, VEB-2, GES-1,
(β-lactamase) ceftazidime, ceftriaxone) and GES-2, IBC-2 in Pseudomonas aeruginosa
aztreonam
Carbapenemases Substrates of extended-spectrum plus KPC-1, KPC-2, KPC-3 in K. pneumoniae; NMC/IMI,
cephamycins and carbapenems SME family
B Metallo-β-lactamases Carbapenemases Substrates of extended-spectrum plus NDM-1 in Enterobacteriaceae, IMP, VIM, GIM, SPM,
(Zn2+) cephamycins and carbapenems SIM lineages in P. aeruginosa, Acinetobacter spp.
C Serine Cephalosporinases Substrates of extended-spectrum plus AmpC-type enzymes in Enterobacteriaceae,
cephamycins Acinetobacter spp.
D Serine Oxacillinases:
Broad-spectrum Aminopenicillins, ureidopenicillin, OXA-family in P. aeruginosa
cloxacillin, methicillin, oxacillin, and
some narrow-spectrum cephalosporins
Extended- Substrates of broad-spectrum plus OXA-derived in P. aeruginosa
spectrum oxymino-β-lactams and monobactams
Carbapenemases Substrates of extended-spectrum plus OXA-derived in Acinetobacter spp.
cephamycins and carbapenems
AmpC, ampicillin C; CTX-M, cefotaxime-M; GES-1, -2; Guyana extended-spectrum β-lactamase-1, -2; GIM, German imipenemase; IBC-2, integron-born cephalosporinase;
IMI, imipenem hydrolyzing; IMP, imipenem; KPC-1, -2, -3, K. pneumoniae carbapenemase-1, -2, -3; NDM-1, New Delhi metallo-β-lactamase-1; NMC, not metalloenzyme
carbapenamase; OXA, oxacillin; PC1, penicillin 1; PER-1, Pseudomonas extended resistance-1; SHV-1, sulfhydryl variable-1; SIM, Seoul imipenemase; SME, Serratia
marcescens extended-spectrum β-lactamase; SPM, Sao Paulo metallo-β-lactamase; TEM-1, Temoneira-1; VEB-1, -2, Vietnam extended-spectrum β-lactamase-1, -2; VIM,
Verona integron-encoded metallo-β-lactamase.

TABLE 18-3  Bush-Jacoby-Medeiros Functional Classification Scheme for β-Lactamases


INHIBITION BY MOLECULAR NO. OF
GROUP ENZYME TYPE CLAVULANATE CLASS ENZYMES EXAMPLES*
1 Cephalosporinase No C 57 Enterobacter cloacae P99 (C), MIR-1 (P)
2a Penicillinase Yes A 20 Bacillus cereus I, Staphylococcus aureus (B)
2b Broad-spectrum Yes A 16 SHV-1 (B), TEM-1 (P)
2be Extended-spectrum Yes A 81 Klebsiella oxytoca K1 (C), TEM-3 (P), SHV-2 (P)
2br† Inhibitor-resistant Diminished A 13 TEM-30 (IRT-2) (P)
2c Carbenicillinase Yes A 15 AER-1 (C), PSE-1 (P), CARB-3 (P)
2d Cloxacillinase Yes D or A 21 Streptomyces cacaoi (C), OXA-1 (P)
2e Cephalosporinase Yes A 19 Proteus vulgaris (C), FEC-1 (P)
2f† Carbapenemase Yes A 3 IMI-1 (C), NMC-A (C), KPC (P), Sme-1 (C)
3 Carbapenemase No B 15 Stenotrophomonas maltophilia L1 (C), NDM-1 (P), IMP-1 (P)
4 Penicillinase No 7 Burkholderia cepacia (C), SAR-2 (P)
*B, both; C, chromosomal; P, plasmid.

New groups; derived from Bush K, Jacoby GA, Medeiros AA. A functional classification scheme for β-lactamases and its correlation with molecular structure. Antimicrob
Agents Chemother. 1995;39:1211-1233.
AER-1, Aeromonas-1; CARB-3, carbenicillin-3; FEC-1, isolated from feces; IMI-1, imipenem hydrolyzing; IMP-1, imipenem-1; IRT-2, inhibitor-resistant TEM-2; KPC, K.
pneumoniae carbapenemase; MIR-1, Miriam Hospital–1; NDM-1, New Delhi metallo-β-lactamase-1; NMC-A, not metalloenzyme carbapenamase-A; OXA-1, oxacillin-1;
PSE-1, Pseudomonas-specific enzyme; SAR-2, southern Africa–related enzyme; SHV-1, sulfhydryl variable-1; TEM-3, Temoneira-3.

currently the most important class A serine carbapenemases. Since P. aeruginosa, Acinetobacter, other gram-negative nonfermenters, and
initially reported from K. pneumoniae isolates in several northeastern enteric bacterial pathogens.24,69
U.S. outbreaks,64-67 KPCs have been found worldwide in multiple other The New Delhi metallo-β-lactamase–1 (NDM-1) has received the
gram-negative species, such as E. coli, Citrobacter, Enterobacter, Salmo- most attention recently. Originally described in a K. pneumoniae
nella, Serratia, and P. aeruginosa.5 isolate from India in 2008, NDM-1 enzymes have since been reported
Class B metallo-β-lactamases (MBLs) use a Zn2+ cation for hydro- in the United States, the United Kingdom, and a number of other
lysis of the β-lactam ring; are susceptible to ion chelators, such as countries, primarily in connection with travel to India or Pakistan.70,71
ethylenediaminetetraacetic acid (EDTA); and resistant to clavulanic These enzymes confer resistance to all β-lactams except aztreonam.
acid, tazobactam, and sulbtactam. They confer resistance to all β-lactam However, most metallo-β-lactamases reside on mobile gene cassettes
antibiotics except monobactams. Chromosomally encoded MBLs are inserted into integrons that harbor additional antibiotic-resistance
primarily found in environmental isolates of Aeromonas, Chryseobac- genes to other antimicrobial classes; this multidrug resistance can be
terium, and Stenotrophomonas spp. and are of usually low pathogenic transferred to other species via transposons and plasmids, severely
potential.68 Most clinically important MBLs belong to five different limiting therapeutic options in serious infections.72,73
families (imipenem [IMP], Verona integron-encoded metallo-β- Finally, class D carbapenemases have been described among
lactamase [VIM], Sao Paulo metallo-β-lactamase [SPM], German imi- four subfamilies of OXA-type β-lactamases (OXA-23, OXA-24,
penemase [GIM], and Seoul imipenemase [SIM]), typically transmitted OXA-58, and OXA-146), primarily in A. baumanii. In the latter, the
by mobile gene elements inserted into integrons and spread through intrinsically weaker carbapenemase activity is augmented by coupling
241
Group β-lactamase production with an additional resistance mechanism,
Class C such as decreased membrane permeability or increased active efflux.74
Citrobacter freundii 1
LAT-1 1 Gram-Positive Bacteria

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


Enterobacter cloacae 1 Among gram-positive bacteria, staphylococci are the major pathogens
ACT-1 1 that produce β-lactamase. Staphylococcal β-lactamases preferentially
Morganella morganii 1 hydrolyze penicillins. Most are inducible and are excreted extracellu-
FOX-1 1 larly.3 The genes that determine staphylococcal β-lactamases usually
are carried on small plasmids or transposons. Larger plasmids encod-
MOX-1 1 ing β-lactamase and other resistances also exist and can transfer by
conjugation, not only between strains of S. aureus but also between S.
Class D OXA-10 2d aureus and Staphylococcus epidermidis.75
OXA-11 2d Enterococci produce a plasmid-determined β-lactamase that seems
to be of staphylococcal origin.76 Since the appearance of the first strain
Class A Klebsiella oxytoca 2be in Texas in 1981, β-lactamase–producing enterococci have been found
throughout the United States and in South America.77 The genes often
Proteus vulgaris 2e coexist with genes that determine high-level resistance to gentamicin
and may occur on transposons and on plasmids. These transposons are
NMC-A 2f
similar to staphylococcal β-lactamase transposons and may be derived
SHV-1 2b from them.78
SHV-5 2be

TEM-12 2be Anaerobic Bacteria


TEM-1 2b β-Lactamases also contribute to the resistance of anaerobic bacteria to
TEM-30 (IRT-2) 2br β-lactam antibiotics.79,80 The β-lactamases of fusobacteria and clostridia
are principally penicillinases.81,82 The β-lactamases produced by Bacte-
PSE-1 (CARB-2) 2c roides fragilis are predominantly cephalosporinases, some of which
have been found to hydrolyze cefoxitin and imipenem and may be
Class B Bacteroides fragilis 3 transferable.83,84 Most of the cephalosporinases are inhibited by clavu-
3 lanate, sulbactam, or tazobactam. Some isolates of Bacteroides spp.
IMP-1
produce carbapenemases, metalloenzymes inhibited by EDTA but not
Bacillus cereus 3 clavulanate, that confer resistance to imipenem.

FIGURE 18-4  Correlation between amino-acid sequences (Ambler Contribution of β-Lactamases to β-Lactam
classes) and functional properties of β-lactamases (Bush-Jacoby- Antibiotic Resistance
Medeiros groups). ACT-1, AmpC type-1; CARB-2, carbenicillin-2; The level of antibiotic resistance mediated by a particular β-lactamase
FOX-1, cefoxitin-1; IMP-1, active on imipenem; IRT-2, inhibitor-resistant in a population of bacteria is determined by at least five variables. The
TEM-2; LAT-1, latamoxef; MOX-1, moxicillin-1; NMC-A, not metalloen- efficiency of the β-lactamase in hydrolyzing an antibiotic depends on
zyme carbapenemase-A; OXA, oxacillin; PSE-1, Pseudomonas-specific (1) its rate of hydrolysis and (2) its affinity for the antibiotic. Other
enzyme; SHV, sulfhydryl variable; TEM, Temoneira. (Modified from Philip- variables are (3) the amount of β-lactamase produced by the bacterial
pon A, Dusart J, Doris B, Frère JM. The diversity, structure and regulation
cell, (4) the susceptibility of the target protein (penicillin-binding
of β-lactamases. Cell Mol Life Sci. 1998;54:341-346.)
protein [PBP]) to the antibiotic, and (5) the rate of diffusion of the
antibiotic into the periplasm of the cell.
Within the bacterial cell, β-lactamases contribute to antibiotic resis-
tance in several ways. The simplest model is that of penicillinase-
TEM SHV producing staphylococci, in which the bacteria, on exposure to
penicillin, begin to produce β-lactamase, which they excrete extracel-
lularly. Two events then take place concurrently: (1) penicillin lyses
bacteria and (2) β-lactamase hydrolyzes penicillin. If viable bacterial
cells remain after the level of penicillin has declined to less than the
Ser
minimal inhibitory concentration (MIC), regrowth of bacteria occurs.39
Ser Another model is exemplified by gram-negative bacteria, which (1)
produce a β-lactamase that remains trapped in the periplasmic space
and (2) have no barrier to antibiotic penetration. An example is H.
influenzae strains that produce the TEM-1 β-lactamase.85 In this model
and the first one discussed, a marked inoculum effect occurs in that
the MIC for a large inoculum (106 organisms/mL) may be 1000-fold
greater than that for a small inoculum (102 organisms/mL). Lysis of the
organism by ampicillin releases the trapped β-lactamase into the
microenvironment, providing partial protection to adjacent bacteria
residing in the same location. If a high inoculum exists before ampicil-
A B lin exposure occurs, the release of all the periplasmic β-lactamase
enzymes in a confined space might be sufficient to protect some of
FIGURE 18-5  Ribbon diagrams of TEM β-lactamases (A), and SHV the remaining viable bacteria of the original population of micro­
β-lactamases (B). α-Helices are shown in yellow, β-strands in pink, and organisms. However, the low level of resistance of single cells makes it
turns in gray. The (ser) serine residue (marked by white arrow) involved in possible for ampicillin to cure some infections caused by β-lactamase–
the hydrolysis of the β-lactam antibiotic ring is shown in ball-and-stick
producing strains of H. influenzae when the initial inoculum of infect-
mode in the active site at the center of each molecule. The surrounding
atoms, shown in stick mode, represent various sites of amino-acid substitu- ing bacteria is low.
tions (point mutations) that yield an extended-spectrum β-lactamase Another model is exemplified by ampicillin resistance of E. coli
phenotype. SHV, sulfhydryl variable; TEM, Temoneira. (Modified from strains that produce the TEM-1 β-lactamase. These bacteria have a
Jacoby GA, Munoz-Price SL. The new beta-lactamases. N Engl J Med. barrier to entry of β-lactam molecules (the outer membrane), and they
2005;352:380-391.) produce a β-lactamase that remains localized to the periplasmic space.
242

TABLE 18-4  Mechanisms of Resistance to TABLE 18-5  Aminoglycoside-Modifying Enzymes


β-Lactam Antibiotics USUAL ANTIBIOTICS COMMON
I. Alter target site (PBP) ENZYMES MODIFIED GENERA
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

A. Decrease affinity of PBP for β-lactam antibiotic Phosphorylation


1. Modify existing PBP
a. Create mosaic PBP APH(2″) K, T, G SA, SR
Insert nucleotides obtained from neighboring bacteria (e.g., APH(3′)-I K E, PS, SA, SR
penicillin-resistant Streptococcus pneumoniae)
APH(3′)-III K , ±A E, PS, SA, SR
Mutate structural gene of PBP(s) (e.g., ampicllin-resistant
β-lactamase–negative Haemophilus influenzae) Acetylation
2. Import new PBP, e.g., mecA in methicillin-resistant Staphylococcus AAC(2′) G PR
aureus
II. Destroy β-lactam antibiotic AAC(3′)-I ±T, G E, PS
A. Increase production of β-lactamase AAC(3′)-III, -IV, or -V K, T, G E, PS
1. Acquire more efficient promoter
AAC(6′) K, T, (A) E, PS, SA
a. Mutate existing promoter
b. Import new one Adenylation
2. Deregulate control of β-lactamase production ANT(2″) K, T, G E, PS
a. Mutate regulator genes (e.g., ampD in “stably derepressed”
Enterobacter cloacae) ANT(4′) K, T, A SA
B. Modify structure of resident β-lactamase Bifunctional Enzymes
1. Mutate its structural gene (e.g., extended-spectrum β-lactamases in
Klebsiella pneumoniae) AAC(6′)APH(2″) G, Ar SA, Ent
C. Import new β-lactamase(s) with different spectrum of activity AAC(6′)-Ib cr G, K, T, FQ* E
III. Decrease concentration of β-lactam antibiotic inside cell
A, amikacin; AAC, aminoglycoside acetyltransferase; ANT, aminoglycoside
A. Restrict its entry (loss of porins)
nucleotidyltransferase; APH, aminoglycoside phosphotransferase; cr, ciprofloxacin
B. Pump it out (efflux mechanisms)
resistance; Ar, arbekacin, E, Enterobacteriaceae; Ent, enterococci, *FQ,
PBP, penicillin-binding protein. fluoroquinolone (acetylates the piperazine ring in some fluroquinolones), G,
gentamicin; K, kanamycin; PR, Providencia-Proteus; PS, pseudomonads; SA,
staphylococci; SR, streptococci; T, tobramycin.
In this model, the kinetics are more complicated. The enzyme is situ-
ated strategically between the barrier to antibiotic penetration (outer
membrane) and the antibiotic targets (PBPs on the cytoplasmic mem- group. The nomenclature for these enzymes lists the molecular site
brane). In this position, the enzyme can destroy antibiotic molecules where the modification occurs after the type of enzymatic activity. An
sequentially as they make their way through the barrier, analogous to aminoglycoside acetyltransferase (AAC) that acts at the 3′ site is des-
a sharpshooter with abundant ammunition who aims at targets passing ignated AAC(3′) (Table 18-5). There may be more than one enzyme
through a single entry point. As a consequence, high levels of resistance that catalyzes the same reaction, however, and Roman numerals may
occur with single bacterial cells, in contrast to the previous example.39 be necessary (e.g., AAC[3′]-IV).
Variations on this model occur when the amount of β-lactamase Enzymatic aminoglycoside resistance is achieved by modification
produced increases with exposure to a β-lactam (induction), as occurs of the antibiotic in the process of transport across the cytoplasmic
in Enterobacter and Pseudomonas spp. High levels of β-lactamase are membrane.88 Resistance to a particular aminoglycoside is a function of
produced only after a period of exposure to the inducing antibiotic, two different rates—that of drug uptake versus that of drug inactiva-
and resistance may be expressed late. When Enterobacter strains are tion. An important factor in determining the level of resistance is the
exposed to two β-lactam antibiotics, one of which is a potent inducer affinity of the modifying enzyme for the antibiotic. If an enzyme has a
(e.g., cefamandole), antagonism between the two antibiotics may high affinity for the specific aminoglycoside, drug inactivation can
result.82 occur at very low concentrations of the enzyme.
Table 18-4 lists mechanisms of resistance to β-lactam antibiotics. The differences in the worldwide distribution of aminoglycoside-
Often these mechanisms work in concert and may accumulate in a modifying enzymes may be partially a function of antibiotic selection
single patient. An example is a 19-month-old child with aplastic pressures and may have had profound implications on the choice of
anemia who over 3 months had nine blood isolates of E. coli, all derived antibiotics used at specific medical centers. Aminoglycoside phos-
from a common ancestor, despite multiple courses of antibiotics, photransferase (APH)(3′) and APH(3″) are distributed widely among
including ceftazidime.86 The first isolate produced a TEM-1 β-lactamase gram-positive and gram-negative species worldwide and have led to
but was susceptible to ceftazidime (MIC, 0.25 µg/mL). A subsequent decreased use of kanamycin and streptomycin. The gene for amino­
isolate became resistant (MIC of ceftazidime, 32 µg/mL) by acquiring glycoside nucleotidyltransferase (ANT)(2″) has been associated with
a new plasmid-determined β-lactamase (SHV-1) linked to an efficient multiple nosocomial outbreaks in the 1990s across the United States.
promoter and turning off production of an outer membrane porin. An The gene for aminoglycoside acetyltransferase AAC(6′)-I has been
even higher level of resistance (MIC of ceftazidime ≥128 µg/mL) found to be more prevalent in enteric bacteria and in staphylococci in
occurred when the SHV-1 β-lactamase mutated to form the ESBL, East Asia.89 The AAC(3′) group of enzymes have been responsible for
SHV-8, which hydrolyzes ceftazidime much more rapidly. By turning outbreaks of antibiotic resistance in South America, western Europe,
off porin production to slow the rate of entry of ceftazidime into the and the United States. Although each outbreak of aminoglycoside-
periplasmic space and producing an extended-spectrum, ceftazidime- resistant Enterobacteriaceae has its own pattern, the most typical
inactivating β-lactamase, the infecting E. coli used two mechanisms manner of spread has been the appearance of a plasmid-carrying,
synergistically to achieve a high level of resistance to ceftazidime.86 aminoglycoside-resistant strain of K. pneumoniae, usually carrying
the ANT(2″) gene, with subsequent dissemination to other strains of
Aminoglycoside Resistance–Modifying the species and further spread later to other species and genera of
Enzymes Enterobacteriaceae.88
Among aerobic bacteria, aminoglycoside resistance is most commonly Major increases in plasmid-mediated aminoglycoside resistance
due to enzymatic inactivation through aminoglycoside-modifying have been noted among enterococci,90 initially in the developing
enzymes. These may be coded by genes on plasmids or chromosomes. world91 but increasingly in the United States and Europe.92,93 Their
Several aminoglycoside-modifying enzymes have been shown to be clinical impact is exacerbated by the frequent co-transmission of
carried on transposons.87 β-lactamases, resulting in a loss of synergy when combination therapy
Aminoglycoside-modifying enzymes confer antibiotic resistance is used for serious enterococcal infections. S. aureus and S. epidermidis
through three general reactions: N-acetylation, O-nucleotidylation, have become increasingly resistant to aminoglycosides because of the
and O-phosphorylation. For each of these general reactions, there are interspecies and intraspecies dissemination of plasmid-mediated,
several different enzymes that attack a specific amino or hydroxyl aminoglycoside-modifying enzymes.94
243
The two most interesting developments in aminoglycoside- TABLE 18-6  Mechanisms of Tetracycline
modifying ennzymes have been the discovery of bifunctional enzymes. Resistance
The first example is the AAC(6′)APH(2″) enzyme that has two func-
tioning active sites, one for acetylation and the other for phosphoryla- RESISTANCE COMMON TET COMMON

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


tion of aminoglycosides. This bifunctional enzyme probably arose MECHANISM DETERMINANTS BACTERIAL SPECIES
from a fusion event of the genes for these two enzymes. This enzyme Drug efflux Tet A-L, P*, V, Y, Z, Enterobacteriaceae,
otrB, tcr(3), Tet 30 Pseudomonas,
is now widespread in staphylococci and enterococci, frequently resid- Streptomyces,
ing on a common transposon Tn4001 found on the chromosome and Staphylococcus,
on transferable plasmids. The gene aac(6′)aph(2″) accounts for most Streptococcus spp.
of the high level gentamicin and arbekacin resistance observed in Ribosomal protection Tet M, O, P*, Q, S, T, Gram-positive and
methicillin-resistant S. aureus (MRSA) and enterococcal isolates in W, otrA gram-negative
anaerobes, Neisseria,
many countries worldwide.95,96 Haemophilus,
Recently, another major discovery with aminoglycoside-modifying Enterococcus.
enzymes finds evidence of variant enzymes that can modify the struc- Staphylococcus,
ture of an entirely different class of antimicrobial agent. The first Streptococcus spp.
bifunctional enzyme that can modify aminoglycosides and a fluoro- Enzymatic inactivation Tet X, Tet 34, Tet 37 Bacteroides, Vibrio spp.
quinolone (ciprofloxacin) was described in 2006.97 This enzyme, des- Unknown mechanism Tet U, otrC Mycobacterium,
ignated as AAC(6′)–Ib-cr, not only acetylates kanamycin, gentamicin, Enterococcus spp.
and tobramycin, but also acetylates the piperazinyl side group of cip- Altered ribosomal target — Helicobacter pylori
rofloxacin. This acetylated quinolone is fourfold less active than the *Tet P has two different genes mediating different mechanisms of resistance.
parent compound and may lead to clinically significant resistance in
enteric bacteria, particularly in strains that possess other mechanisms
for diminished quinolone activity. This enzyme has two important
mutations in the gene for the basic AAC(6′) enzyme (W102R and
D179Y) and 12 unique base pairs at its 5′ end that are essential to not against gram-negative bacilli.107 This difference in susceptibility to
alter substrate specificity and allow ciprofloxacin acetylation function penicillin is due in large part to the presence of the outer membrane
for this enzyme.98 The widespread use of ciprofloxacin and other in gram-negative bacilli, a thick lipopolysaccharide layer that acts as a
fluoroquinolones over the past 2 decades has changed the selection barrier to the penetration of many antibiotics into the cell.108 Situated
pressures on bacterial populations promoting quinolone-resistance outside the peptidoglycan cell wall of gram-negative bacteria, this
development.98 outer membrane is absent in gram-positive bacteria. The lipopolysac-
charide is made up of tightly bound hydrocarbon molecules that
Chloramphenicol Acetyltransferase impede the entry of hydrophobic antibiotics, such as nafcillin or eryth-
Resistance to chloramphenicol in gram-positive and gram-negative romycin. Agents that disrupt the integrity of the lipopolysaccharide
organisms is mediated primarily by the inactivating enzyme chloram- layer, such as polymyxin, or mutations that lead to the production of
phenicol acetyltransferase. This is an intracellular enzyme that inacti- defective lipopolysaccharides, result in increased permeability of
vates the drug by 3-O-acetylation99 and is encoded by plasmid-borne hydrophobic antibiotics.109
or chromosomal genes. Despite homology at the active site of this The passage of hydrophilic antibiotics through this outer mem-
enzyme, there is considerable diversity between chloramphenicol acet- brane is facilitated by the presence of porins, proteins that are arranged
yltransferase enzymes isolated from gram-positive and gram-negative so as to form water-filled diffusion channels through which antibiotics
organisms.100 may traverse.110 Bacteria usually produce many porins; approximately
105 porin molecules are present in a single cell of E. coli. Bacteria are
Macrolide-, Lincosamide-, Streptogramin- able to regulate the relative number of different porins in response to
Inactivating Enzymes the osmolarity of the surrounding media. In hyperosmolar media, E.
Although resistance to erythromycin and other macrolides is fre- coli may repress production of the larger porins (OmpF) while con-
quently the result of alteration in the ribosomal target site or efflux tinuing to express smaller ones (OmpC).111
pumps, several substrate-inactivating enzymes have been character- The rate of diffusion of antibiotics through this outer membrane is
ized.101 Erythromycin esterases have been isolated from E. coli that a function not only of the numbers and properties of the porin chan-
hydrolyze the lactone ring of the antibiotic and result in its inac­ nels but also of the physicochemical characteristics of the antibiotic.
tivation. This is a plasmid-mediated resistance determinant that is In general, the larger the antibiotic molecule, the more negatively
constitutively produced and results in high-level resistance to erythro- charged, and the greater the degree of hydrophobicity, the less likely it
mycin (MIC >2000 µg/mL).102 This type of high-level macrolide- will penetrate through the outer membrane.112 Small hydrophilic mol-
resistance gene is often linked with the carbapenemase-producing, ecules with a zwitterionic charge, such as imipenem, are highly perme-
NDM-1–bearing plasmids.70 These resistance genes limit the utility able. Conversely, larger highly charged molecules, such as carbenicillin,
of oral erythromycin or other macrolides in reducing the aerobic are much less permeable.
gram-negative flora of the intestinal tract before gastrointestinal surgi- Mutations resulting in the loss of specific porins can occur in clini-
cal procedures. Other plasmid-mediated resistance genes generate cal isolates and determine increased resistance to β-lactam antibiotics.
specific inactivating enzymes in Streptococcus haemolyticus and S. Resistance to aminoglycosides and carbapenems emerging during
aureus that adenylate103 lincosamides or acetylate104 or hydrolyze105 therapy has been associated with a lack of production of outer mem-
streptogramins. brane proteins.113 For example, emergence of imipenem resistance
during therapy, observed in up to 25% of P. aeruginosa infections,114
Tetracycline Inactivation has been ascribed to mutational loss of its OprD protein (also known
A tetracycline-inactivating enzyme called TetX has been described as the D2 porin).113-116
rarely in Bacteroides spp.106 Tetracycline resistance is principally medi- Resistance to nalidixic acid and other quinolones has been
ated by other mechanisms, including efflux and ribosomal protection associated with alterations of outer membrane proteins in Serratia
(Table 18-6). marcescens117 and P. aeruginosa. Single-step, high-level mutational
resistance to nalidixic acid by aerobic gram-negative bacilli occurs with
Decreased Permeability of Bacterial a 10−7 frequency, whereas only low-level resistance to the newer qui-
Membranes nolones (<10 × MIC) usually is obtained with a single-step selection
Outer Membrane Permeability of less than 10−9.118 Plasmid-mediated chloramphenicol resistance
Alexander Fleming recognized early in the history of antibiotic devel- resulting from decreased cell membrane permeability has been shown
opment that penicillin is effective against gram-positive bacteria but in E. coli.119
244
Inner Membrane Permeability (designated as Tet) or resistance gene products for oxytetracycline
The rate of entry of aminoglycoside molecules into bacterial cells is derivatives (Otr). The primary mechanism for the decreased accumu-
a function of their binding to a usually nonsaturable anionic trans- lation of tetracycline is through the active efflux of the antibiotic across
porter, whereupon they retain their positive charge and subsequently the cell membrane.126 Decreased uptake of tetracycline from the extra-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

are “pulled” across the cytoplasmic membrane by the internal negative cellular environment also accounts for decreased accumulation of tet-
charge of the cell.120 This process requires energy and a threshold racycline inside resistant cells. These resistance determinants may be
minimal level of internal negative charge of the cell that has to be found on the chromosome or plasmids and frequently are found on
present before significant transport occurs (proton motive force).121 transposable genetic elements. Tetracycline-resistance genes are gener-
The level of the internal charge that is required may depend on the ally inducible by subinhibitory concentrations of tetracycline. There
actual aminoglycoside concentration at a given time. The energy are now over 40 recognized tetracycline-resistance determinants, most
generation or the proton motive force that is required for substrate of which mediate drug efflux.127 The determinants have been desig-
transport into the cell may be altered in mutants resistant to nated in the past by letters (e.g., Tet A, Tet B). Because there are now
aminoglycosides. more determinants than letters in the English alphabet, new tet genes
These aminoglycoside-resistant isolates with altered proton motive now are designated by numbers.128
force occur rarely but can develop in the course of long-term amino-
glycoside therapy.121 These isolates usually have a “small colony” phe- Macrolides and Streptogramins
notype resulting from their reduced rate of growth. They may be In some strains of Streptococcus pneumoniae, Staphylococcus pyogenes,
unstable and revert to a sensitive phenotype in the absence of selective S. aureus, and S. epidermidis, an active efflux mechanism causes resis-
aminoglycoside pressure. The clinical significance of these isolates is tance to macrolides, streptogramins, and azalides.129 This efflux mecha-
not clear. They may retain some virulence122 and rarely may cause fatal nism is mediated by the mef (for macrolide efflux) genes in streptococci
bacteremia.123 Because oxidative metabolism is essential for aminogly- and msr (for macrolide streptogramin resistance) genes in staphylo-
coside uptake action and cell growth and development, Pseudomonas cocci.130 A similar efflux system, encoded by a gene called mreA (for
mutants have been found that have been deficient in specific cyto- macrolide resistance efflux), has been described in group B strepto-
chromes. Resistant mutants with defective electron transport systems cocci.131 This mechanism of resistance is prevalent in community-
have been described in E. coli, S. aureus, and Salmonella spp. Faculta- acquired infections,132 and dissemination of these resistance genes
tive organisms grown anaerobically are resistant to aminoglycoside among important bacterial pathogens is a considerable threat to the
actions because of a lack of a proton motor force and marked reduction usefulness of macrolide antibiotics (Table 18-7).
of drug uptake.124
β-Lactams
Promotion of Antibiotic Efflux Active efflux mechanisms also may contribute to the full expression of
Tetracyclines β-lactam resistance in P. aeruginosa. Multidrug efflux pumps in the
Active efflux of antimicrobial agents is recognized increasingly as a inner and outer membrane of P. aeruginosa act in concert with peri-
common mechanism of resistance in many clinically relevant patho- plasmic β-lactamases and membrane permeability components to
gens. Some strains of E. coli, Shigella spp., and other enteric organisms protect the bacterium from β-lactam agents.133,134
express a membrane transporter system that leads to multidrug resis-
tance by drug efflux.125 Many of these are multicomponent, regulated, Fluoroquinolones
energy-dependent transporter systems that promote the active efflux Active efflux of fluoroquinolones has been detected in enteric bacte-
of multiple classes of antibiotics. Specific efflux pumps also exist that ria135,136 and staphylococci.132 This efflux may be related to a multiple
promote the egress of single classes of antimicrobial agents. antibiotic-resistance transporter132 (i.e., NorA) or a specific quinolone
The major mechanism of resistance to tetracyclines found in enteric efflux pump (i.e., EmrAB, AcrAB, or plasmid-mediated qepA gene
gram-negative organisms results from the decreased accumulation of efflux system).137 This mechanism of limiting access of high levels of
tetracycline (see Table 18-6). This reduced uptake is an energy- fluoroquinolones works in concert with other mechanisms (point
dependent process that is related to the generation of an inner mem- mutations of DNA gyrases, gyrase protection, permeability barriers,
brane protein produced by the tetracycline-resistance determinant and acetylation) for full expression of quinolone resistance.

TABLE 18-7  Resistance Mechanisms against the Macrolides, Lincosamides, and Streptogramins
RESISTANCE PATTERN
14- or
BACTERIAL GENE RESISTANCE 15-Membered 16-Membered STREPTOGRAMIN
SPECIES DESIGNATION PHENOTYPE MECHANISM Ring Ring CLINDAMYCIN B
Streptococci, erm (A, B) MLSB−inducible Ribosomal (s) I or R (s) I or R (s) I or R (s) I or R
Enterococci methylation
erm (A, B) MLSB−constitutive Ribosomal R R R R
methylation
mef (A or E) M Efflux I or R S S S
L4/L22 mut M Ribosomal R R S S
mutation
inu (B) L Inactivation S S S-I S
Staphylococci erm (A, C) MLSB− inducible Ribosomal R (s) (s) (s)
methylation
erm (A, C) MLSB− Ribosomal R R R R
constitutive methylation
msr (A or B) MSB Efflux R S S R
vgb, vgbB SB Inactivation S S S R
ere (A or B) M Inactivation R R S S
inu (A) L Inactivation S S S-I S
14- or 15-membered ring structures, erythromycin, clarithromycin, azithromycin; 16-membered ring structures, spiramycin; I, intermediate susceptibility; L, lincosamides;  
M, macrolides; MLSB, macrolides, lincosamides, and streptogramin B; R, resistant; (s), appears susceptible in vitro but may select resistant clones in vivo; S, sensitive.
245
Altered Target Sites Ribosomal resistance often is associated with decreased intracellular
Alteration of Ribosomal Target Sites accumulation of the drug.147
Macrolides, Lincosamides, Streptogramins
Resistance to a wide variety of antimicrobial agents, including tetracy- Ketolides

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


clines, macrolides, lincosamides, streptogramins, and the aminoglyco- Ketolides have seen limited use clinically, and yet clinical isolates of S.
sides, may result from alteration of ribosomal binding sites. Failure of pneumoniae resistant to telithromycin have already been reported.
the antibiotic to bind to its target site or sites on the ribosome disrupts Resistance is due to constitutive expression of the erm gene, mutations
its ability to inhibit protein synthesis and cell growth. For macrolides, in domains II and V of the 23S rRNA binding sites, as well as mutations
lincosamides, and streptogramin B (MLS), this is the principal mecha- within ribosomal proteins L4 and L22.148
nism of multiple-agent resistance among aerobic and anaerobic gram-
positive organisms.130 Resistance is mediated by the products of the Oxazolidinones
erm (erythromcyin ribosome methylation) gene, the variety of meth- Resistance to linezolid, the main oxazolidinone currently in clinical
ylase enzymes (MLSB− determinant) that dimethylate adenine residues use, has been described in gram-positive isolates (S. aureus, Enterococ-
on the 23S ribosomal RNA (rRNA) of the 50S subunit of the prokary- cus faecium, Enterococcus faecalis, S. epidermidis) as a result of point
otic ribosome, disrupting the binding of MLS to the ribosome (see mutations within the genes encoding the 23S rRNA of the 50S ribo-
Table 18-7). Different classes of this resistance determinant may be somal subunit, the antibiotic’s main binding site.149 Specifically, trans-
located on plasmids or on the bacterial chromosome. location of peptidyl–transfer RNA (tRNA) from the A site to the P site
MLSB resistance resulting from ribosomal methylation has been during translation from bacterial ribosomes is inhibited through
described in many species, including S. aureus, Streptococcus sanguinis, guanine (G) to uridine (U) substitutions at position 2576 or similar
B. fragilis, and Clostridium perfringens. MLS resistance may be consti- binding sites in the peptidyl transferase region of 23S rRNA.150 This
tutive or inducible by either older macrolides (e.g., erythromycin) or remains the most common mechanism of linezolid resistance among
newer azalides. In S. pneumoniae, resistance is encoded by the erm(B) clinical isolates. Resistance development has been observed during
gene, which is responsible for the methylation of loop V of the 23S prolonged linezolid therapy in vancomycin-resistant enterococci
ribosomal subunit.138,139 In addition, point mutations in ribosomal pro- (VRE) bacteremia.151 and a number of other pathogens, including
teins L4 and L22 of the 50S subunit have also been described that coagulase-negative staphylococci and S. aureus. Another mechanism
render S. pneumoniae resistant to macrolides.140 of resistance, mediated by the cfr gene, is the result of methylation of
Inducible resistance in streptococci is generated by a variety of adenosine at position 2503 of 23S rRNA by a methyl transferase
lincosamides and macrolides, resulting in cross-resistance to the MLSB reaction.152
antibiotics. In staphylococci, only 14 to 15 numbered macrolides
induce MLSB methylation, and the organisms express resistance to Alteration of Cell Wall Precursor Targets
macrolides only. Vancomycin and other glycopeptide antibiotics, such as teicoplanin,
bind to d-alanine-d-alanine (d-ala-d-ala), which is present at the
Tetracyclines termini of the stem peptide in peptidoglycan precursors. The large
Tetracycline resistance may be mediated by a variety of mechanisms, glycopeptide molecules prevent the incorporation of the precursors
the most common of which are efflux mechanisms and ribosomal into the cell wall. Resistance of enterococci to vancomycin has been
protection mechanisms (see Table 18-6).141 An additional, unusual classified as A through G based on genotype, type of target site altera-
mechanism of tetracycline resistance by altering the target site of action tions, and levels of resistance to vancomycin, and susceptibility or
has been found in Helicobacter pylori.142 This organism can possess a resistance to teicoplanin (Table 18-8).153,154
mutation in its 16S rRNA that limits tetracycline binding to its target Strains of E. faecium and E. faecalis with high-level resistance to
site at the 30S subunit of the bacterial ribosome. vancomycin and teicoplanin have class A resistance. Either vancomy-
cin or teicoplanin can induce resistance in these strains. Class A resis-
Aminoglycosides tance to glycopeptides transfers by conjugation from E. faecium to
Resistance to aminoglycosides is also mediated at the ribosomal level. other gram-positive bacteria,155 including E. faecalis,156 S. pyogenes, S.
In Enterobacteriaceae and nonfermenting gram-negative bacteria, sanguis, and Listeria monocytogenes. The vanA gene on the plasmid
methylation of the 16S rRNA (the site where aminoglycosides bind and encodes an inducible protein that is related to the d-ala-d-ala ligases
inhibit protein synthesis) by enzymes usually carried on plasmids is involved in cell wall synthesis in E. coli.157 This protein synthesizes
mediated by at least seven different genes (armA, rmtA, rmtB, rmtC, peptidoglycan precursors that have a depsipeptide terminus (d-alanine-
rmtD, rmtE, and npmA).143 This is now recognized as a major mecha- d-lactate) instead of the usual d-ala-d-ala. The modified peptidoglycan
nism of resistance to all parenteral aminoglycosides, which appears to binds glycopeptide antibiotics with reduced affinity, conferring resis-
be spreading globally.144,145 Mutations of the S12 protein of the 30S tance to vancomycin and teicoplanin.158,159
subunit have been shown to interfere with binding streptomycin to the Strains of E. faecium and E. faecalis with class B resistance
ribosome. Ribosomal resistance to streptomycin may be a significant have levels of resistance to vancomycin that range from high (MIC,
cause of streptomycin resistance among enterococcal isolates.146 Ribo- 1024 µg/mL) to low (MIC, 4 µg/mL) and are susceptible to teico-
somal resistance to the 2-deoxystreptamine aminoglycosides (genta- planin. Vancomycin, but not teicoplanin, can induce resistance to
micin, tobramycin, amikacin) seems to be uncommon and may require vancomycin and teicoplanin in these strains. The genes determining
multiple mutations in that these aminoglycosides seem to bind to the VanB phenotype are self-transferable by conjugation to other
several sites on the 30S and 50S subunits of the prokaryotic ribosome. Enterococcus strains.160,161

TABLE 18-8  Vancomycin Resistance in Enterococci and Staphylococci


A B C D E G
Vanco (µg/mL) (MIC) 64 to >500 4 to >500 2-32 64-128 16 12-16
Teico (µg/mL) (MIC) 16 to >500 0.5-2 0.5-2 4-64 0.5 0.5
Expression Inducible Inducible Constitutive, inducible Constitutive Inducible ND
Genetic location P, C P, C C C C C
Target alteration D-ala-D-lac D-ala-D-lac D-ala-D-ser D-ala-D-lac D-ala-D-ser D-ala-D-ser
Common species Enterococcus faecalis, E. faecium, E. faecalis, E. gallinarum (C-1) E. faecium E. faecalis E. faecalis
Staphylococcus aureus E. faecium E. casseliflavus (C-2)
E. flavescens (C-3)
C, chromosome; D-ala, D-alanine; D-lac, D-lactate; D-ser, D-serine; MIC, minimal inhibitory concentration; ND, not described; P, plasmid; Teico, teicoplanin; Vanco, vancomycin.
246
All isolates of Enterococcus gallinarum, Enterococcus casseliflavus, community-acquired MRSA strains, tend to be smaller in size, and
and Enterococcus flavescens possess low-level resistance to vanco­ more susceptible to antibiotics other than β-lactams.
mycin and are susceptible to teicoplanin (class C phenotype). The Expression of the methicillin-resistance gene is controlled by two
resistance is mediated by chromosomal genes known as vanC1, regulatory components of the mec gene: mecR1-mecI, and the
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

vanC2, or vanC3.162 The vanC gene complex gives rise to resistance to β-lactamase genes blaI, blaRI, and blaZ, which can downregulate mecA
vancomycin by synthesis of an alternative dipeptide, d-alanine-d- transcription. Although the mecA gene is present in all MRSA isolates,
serine, in which a serine replaces the terminal alanine. Other variant the phenotypic expression of methicillin resistance is more variable.
genes known as vanE and vanG have been found in enterococcal For example, S. aureus isolates grown at 32° C, rather than at 37° C, are
species that also mediate various levels of glycopeptide resistance (see more likely to express methicillin resistance.184 In addition, the expres-
Table 18-8). sion of methicillin resistance seems to be modified also by auxiliary
Since 1987, reports from the United States and Japan have docu- genes, such as fem and aux, which are present in the staphylococcal
mented outbreaks of vancomycin-intermediate S. epidermidis,163 S. chromosome and affect various steps in peptidoglycan synthesis.185,186
haemolyticus,164 and S. aureus (VISA).165 The first high-level The PBPs of β-lactamase-negative, penicillin-resistant strains of N.
vancomycin-resistant S. aureus (VRSA) isolate in the United States was gonorrhoeae, Neisseria meningitidis, and H. influenzae have shown
recovered in 2002 from a foot ulcer in a diabetic patient on chronic reduced penicillin-binding affinity.187-190 Their PBPs seem to be encoded
hemodyalisis, concurrently with a VRE strain from the same wound.166 by hybrid genes containing segments of DNA scavenged from resistant
DNA sequencing revealed identical vanA genes in both isolates, and strains of related species, similar to penicillin-resistant pneumococci.191
further molecular analysis revealed plasmid-mediated transfer of resis- Mutations leading to a loss of outer membrane proteins also may be
tance through the Tn1546 genetic element encoding the vanA gene, associated with the acquisition of penicillin resistance in non–
from the enterococcus (VRE) strain into the vancomycin-susceptible penicillinase-producing strains of N. gonorrhoeae, suggesting that
MRSA strain recovered from the same patient, rendering it vancomy- altered permeability also may contribute to the resistance.192 Progres-
cin resistant (VRSA).166,167 sive loss of β-lactam activity through multiple mechanisms in N. gon-
Vancomycin-intermediate S. aureus (VISA) expresses unusually orrhoeae, coupled with its remarkable capacity to acquire other
thick peptidoglycan cell walls that are less completely cross-linked.168 resistance genes by transformation and plasmid transfer, can give rise
The cell wall in some strains of vancomycin-intermediate S. aureus to pan-resistant, untreatable gonorrhea.193
contains nonamidated glutamine precursors that provide an increased Permeability changes and decreased affinity of PBPs are mecha-
number of false binding sites to vancomycin.169,170 The vancomycin nisms found jointly in clinical isolates of P. aeruginosa194 and in non–β-
molecules are absorbed to these excess binding sites, preventing the lactamase-producing strains of H. influenzae.195 Multiple mutations
antibiotic from reaching its target and allowing peptidoglycan synthe- may be necessary to effect this type of resistance.
sis in the cytoplasmic membrane to continue uninhibited.170 The
specter of increasing outbreaks of vancomycin-resistant staphylococci Quinolones
has led the Centers for Disease Control and Prevention to develop DNA gyrase (also called bacterial topoisomerase II) is necessary for the
vigorous interim guidelines to mitigate the spread of this serious noso- supercoiling of chromosomal DNA in bacteria to have efficient cell
comial problem.171 division.196 Another related enzyme, topoisomerase IV, also is required
for segregation of bacterial genomes into two daughter cells during cell
Alteration of Target Enzymes division. These enzymes consist of two A subunits encoded by the gyrA
β-Lactams gene and two B subunits encoded by the gyrB gene (or parC and parE
β-Lactam antibiotics inhibit bacteria by binding covalently to PBPs in for topoisomerase IV). Although spontaneous mutation in the gyrA
the cytoplasmic membrane. These target proteins catalyze the synthesis locus is the most common cause of resistance to multiple fluoroquino-
of the peptidoglycan that forms the cell wall of bacteria.172 Alterations lones in enteric bacteria, B-subunit alterations also may affect resis-
of PBPs can lead to β-lactam antibiotic resistance.173 tance to these drugs. Quinolone resistance may also occur from a
In gram-positive bacteria, resistance to β-lactam antibiotics may be combination of decreased cell wall permeability, efflux, or enzyme
associated either with a decrease in the affinity of the PBP for the protection mechanisms.9,137
antibiotic174 or with a change in the amount of PBP produced by the DNA gyrase is the primary site of action in gram-negative bacteria,
bacterium.175 Multiple mechanisms seem to be present in some clinical whereas topoisomerase IV is the principal target of quinolones in
isolates. Penicillin-resistant strains of S. pneumoniae isolated in South gram-positive bacteria, including S. aureus. Mutations in a variety of
Africa have shown several changes in PBPs (i.e., decreased affinity of chromosomal loci have been described that resulted in altered DNA
some PBPs, loss of others, and appearance of PBPs not present in the gyrases resistant to nalidixic acid and the newer fluoroquinolones in
more susceptible cells).176 The genes that encode these PBPs are Enterobacteriaceae and P. aeruginosa.197 Many of these mutations
mosaics, composed of segments from susceptible pneumococci and involve the substitution of single amino acids at the quinolone-
segments from resistant commensal streptococci.177 In S. aureus178-180 resistance determining region (QRDR, located between amino acids
and E. faecium,181 additional PBPs may be inducible (i.e., their produc- 67 and 106 in the gyrase A subunit) that is involved in the generation
tion is stimulated by exposure of the microorganism to the β-lactam of the DNA gyrase–bacterial DNA complex.198 Clinical isolates of C.
antibiotic). These inducible PBPs have a lower affinity for β-lactam freundii in Japan have been found to be highly resistant to the newer
antibiotics, making them less susceptible to inhibition by low concen- quinolones via alterations in the DNA gyrase.199
trations of the drug. Changes in the types of PBPs observed in suscep- Plasmid-mediated quinolone resistance has been found in various
tible and resistant strains also have been seen with the viridans Enterobacteriaceae and is conferred by qnr-encoded proteins that bind
streptococcal species Streptococcus mitis.182 to the DNA gyrase antibiotic target and protect it from quinolone
action. Although fluoroquinolone resistance associated with plasmid-
MRSA Resistance borne qnr genes is low-level resistance, these genes are usually linked
In S. aureus, methicillin resistance is conferred by the expression of the to other antibiotic-resistance determinants carried on the same mobile
mecA gene, which encodes PBP2a, a protein with low affinity for element, and have been associated with clinical phenotypes of multi-
β-lactam antibiotics, conferring resistance to methicillin, nafcillin, drug resistance.9,137,200,201 Another plasmid-derived quinolone-
oxacillin, and cephalosporins. The mecA gene is the structural com­ resistance determinant, encoded by the aac(6′)-Ib-cr gene and derived
ponent of the mec gene cassette and is inserted into the larger staphy- by mutation of a plasmid-contained aminoglycoside-modifying
lococcal cassette chromosome mec (SCCmec), which appears to enzyme, appears widely disseminated among E. coli isolates in the
have been acquired by horizontal transfer from a coagulase-negative United States, mediating low-level ciprofloxacin resistance.92,137
Staphylococcus species.183 At least five different SCCmec types of
various genetic sequences and size have been described: types I to Sulfonamides
III, associated with health care–associated MRSA strains, tend to be There are two common genes that mediate resistance to sulfa drugs in
larger and multidrug resistant; type IV and V, associated with pathogenic bacteria: sul1 and sul2. These genes give rise to altered
247
forms of the target enzyme for sulfonamide, dihydropteroate synthase overproduction of the synthetic enzyme DHPS. The gene responsible
(DHPS).202 This enzyme is essential for folic acid synthesis in suscep- for DHPS is felP, and strains of bacteria that produce excess DHPS can
tible bacteria. The altered DHPS enzymes mediated by the sulfonamide- overwhelm sulfa inhibition.202 Trimethoprim resistance may occur in
resistance genes no longer bind to sulfa, yet continue to synthesize a similar fashion, by making excess amounts of DHFR from the bacte-

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


dihydropteroate from para-aminobenzoic acid substrate. The ubiqui- rial chromosomal gene folA.203,204
tous sul1 gene is part of the class 1 integron family, giving rise to
widespread resistance to sulfonamides.38 Bypass of Antibiotic Inhibition
Another mechanism for acquiring resistance to specific antibiotics is
Trimethoprim by the development of auxotrophs, which have growth factor require-
Trimethoprim is a potent inhibitor of bacterial dihydrofolate reductase ments different from those of the wild-type strain. These mutants
(DHFR). Many altered DHFR enzymes with loss of inhibition by require substrates that normally are synthesized by the target enzymes,
trimethoprim have been described from genes found primarily on and if the substrates are present in the environment, the organisms are
resistance plasmids. These altered DHFR genes are widespread in able to grow despite inhibition of the synthetic enzyme. Enterococci
gram-negative bacteria and are found in staphylococci (dfrA gene).203,204 can be folate auxotrophs requiring environmental acquisition of folic
acid for growth. They become intrinsically resistant to the folic acid
Protection of Target Site inhibitors (sulfa drugs or trimethoprim) in the process. In addition,
Tetracyclines bacteria with mutations in the enzyme thymidylate synthetase can
Tetracycline resistance also may occur by a mechanism that interferes retain viability but become “thymine dependent.” They require exog-
with the ability of tetracycline to bind to the ribosome. Resistance enous supplies of thymidine to synthesize thymidylate via salvage path-
genes such as tetM and others (see Table 18-6) protect the ribosome ways and are highly resistant to sulfa drugs and trimethoprim.205
from tetracycline action. The Tet M determinant is dispersed widely in A compilation of the most frequent mechanisms of resistance used
gram-positive organisms in addition to Mycoplasma,127 Ureaplasma,20 by common bacterial pathogens to inhibit the actions of antibiotics is
Campylobacter,127 and Neisseria spp.19 The tetM gene generates a provided in Table 18-9. Multiple mechanisms are increasingly in oper-
protein with elongation factor–like activity that stabilizes ribosome ation at the same time within individual bacterial cells. The problem
transfer RNA interactions in the presence of tetracycline molecules. of multiple antibiotic-resistance expression is considered in the follow-
ing sections.
Fluoroquinolones
The newly recognized plasmid-mediated, antibiotic-resistance gene Resistance to Newer, Older, and
mediating quinolone resistance seems to function as a target protection Miscellaneous Agents
system.9 The resistance mechanism seems to protect DNA gyrase from The major mechanisms of bacterial resistance to newer agents or older
binding to quinolones, allowing the bacterium to resist quinolone agents, such as polymyxin B and colistin, which have experienced
inhibitory effects. When this low-level resistance determinant is renewed interest in response to progressive antibiotic resistance, are
expressed in concert with other quinolone-resistance genes, such as summarized in Table 18-10.
DNA gyrase mutations or efflux pumps, clinical failures with the use
of fluoroquinolones can result.137 Oxazolidinones
Linezolid resistance mechanisms are primarily related to alterations in
Overproduction of Target the 23S rRNA-binding sites that inhibit linezolid actions upon bacterial
Sulfonamides and Trimethoprim translation.206 Efflux mechanisms may also contribute to reduced activ-
Sulfonamides compete with para-aminobenzoic acid to bind the ity of linezolid in some bacterial species.207
enzyme DHPS and halt the generation of pteridines and nucleic acids. The lipopeptide daptomycin is now widely used for treatment of
Sulfonamide resistance may be mediated in some bacteria by the MRSA and glycopeptide-resistant S. aureus (GRSA) infections. The

TABLE 18-9  Resistance Mechanisms Found in Common Bacterial Pathogens


PATHOGEN RESISTANCE PHENOTYPE MAJOR RESISTANCE MECHANISM
Streptococcus β-Lactams Alteration of target enzymes (PBPs)
pneumoniae Macrolides, lincosamides, streptogramin B Alteration of ribosomal target sites (methylation of adenine residue in domain V of 23S
rRNA—ermB); efflux (mefE)
Tetracycline Protection of ribosomal target site (tetM)
Trimethoprim and sulfonamides Alteration of target enzymes (dihydrofolate reductase–trimethoprim; dihydropteroate synthase—
sul1, sul2 in sulfonamines)
Fluoroquinolones Alteration of target enzymes (DNA gyrase—gyrA mutations; topoisomerase IV—parC mutations)
Staphylococcus β-Lactams Penicillin Enzymatic inhibition (penicillinase production)
aureus Methicillin, oxacillin, nafcillin, Alteration of target enzyme—PBP2a (mecA)
and cephalosporins
(MRSA)
Glycopeptide GISA Alteration of cell wall precursor targets (thickened cell wall binds drug, preventing it from
reaching its target)
GRSA Alteration of cell wall precursor targets (plasmid-mediated transfer of vanA genes from VRE,
resulting in D-ala-D-lac peptidoglycan precursors)
Enterococci β-Lactams (ampicillin) Alteration of target enzymes (PBP5 in Enterococcus faecium); enzymatic inhibition–rare
(penicillinase in E. faecalis)
Aminoglycosides Altered ribosomal target site mutations, enzymatic inhibition (high-level resistance:
aminoglycoside-modifying enzymes)
Vancomycin Alteration of cell wall precursor targets (high-level resistance: VanA, B, D phenotypes; low-level
resistance: VanC, E, G phenotypes)
Linezolid Alteration of ribosomal target sites (G2576U mutation in domain V of 23S rRNA)
Quinupristin-dalfopristin Enzymatic inhibition; efflux; target modification (Enterococcus faecium)
Continued
248

TABLE 18-9  Resistance Mechanisms Found in Common Bacterial Pathogens—cont’d


PATHOGEN RESISTANCE PHENOTYPE MAJOR RESISTANCE MECHANISM
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

Neisseria Penicillins PPNG: enzymatic inhibition (plasmid-acquired penicillinase); CRNG: altered target enzymes (PBPs)
gonorrhoeae Fluoroquinolones Alteration of target enzymes (DNA gyrase; topoisomerase IV); efflux (MtrR-CDE efflux system)
Tetracycline Protection of ribosomal target (tetM gene)
Macrolides Efflux; alteration in ribosomal targets (C2611T mutation in domain V of the 23S rRNA)
MDR Efflux (MtrR-CDE system: penicillin, tetracycline, macrolides)
Pseudomonas β-Lactams Enzymatic inhibition (AmpC cephalosporinases, extended-spectrum β-lactamases, metallo-β-
aeruginosa lactamases); active efflux (MexAB); reduced outer membrane permeability (loss of OprD
channel)
Aminoglycosides Enzymatic inhibition (aminoglycoside-modifying enzymes); efflux (MexXY); alteration of ribosomal
targets (ribosomal methylation)
Fluoroquinolones Efflux (MexAB, CD, EF, XY, GH, VW); alteration of target enzymes (DNA gyrase mutations—gyrA)
MDR Overexpression of the MexA-MexB-OprM active efflux system (resistance to quinolones,
tetracyclines, and trimethoprim)
Acinetobacter β-Lactams Enzymatic inhibition (AmpC cephalosporinases, plasmid-acquired β-lactamases of the TEM, SHV,
baumannii CTX-M, PER, VEB families, metallo-β-lactamases of the IMP, VIM, SIM families, and OXA-type
serine carbapenemases); alteration of target enzymes (PBPs); reduced outer membrane
permeability; efflux pumps
Aminoglycosides Enzymatic inhibition (aminoglycoside-modifying enzymes); efflux pumps
Quinolones Efflux pumps
Tigecycline Efflux pumps
Stenotrophomonas β-Lactams Impermeable outer membrane
maltophilia Enzymatic inhibition (inducible metallo-β-lactamases L1, L2)
TMP-SMX Alteration in sulfonamide target enzymes (sul1, sul2 genes—associated with plasmids or class 1
integrons)
Fluoroquinolones Alteration of target enzymes (DNA gyrase mutations); efflux pumps
MDR MDR efflux pump (smeDEF confers resistance to tetracycline, erythromycin, chloramphenicol,
norfloxacin, ofloxacin)
Klebsiella β-Lactams Enzymatic inhibition (constitutive expression of penicillinases; extended-spectrum β-lactamases;
pneumoniae KPC, NDM-1 carbapenemases); decreased outer membrane permeability
Fluoroquinolones Alteration of target enzymes (DNA gyrase mutations—gyrA); efflux; protection of target site
(plasmid-mediated qnr genes)
Aminoglycosides Enzymatic inhibition (aminoglycoside-modifying enzymes); alteration of ribosomal targets
(ribosomal methylation)
Bacteroides spp. β-Lactams Enzymatic inhibition (chromosomally encoded CepA cephalosporinases; metallo-β-lactamases);
efflux (homologues of RND-pumps); alteration in drug targets (PBPs)
Macrolides, lincosamides, streptogramin B Alteration of ribosomal targets
Tetracycline Protection of ribosomal target (tetQ); efflux
Quinolones Alteration of target enzymes (DNA gyrase mutations—gyr A); efflux
CRNG, chromosomally resistant N. gonorrhoeae; CTX-M, cefotaxime-M; GISA, glycopeptide intermediate S. aureus; GRSA, glycopeoptide-resistant S. aureus; IMP, imipenem;
KPC, K. pneumoniae carbapenemase; MDR, multidrug resistance; MRSA, methicillin-resistant S. aureus; MtrR, multiple transferable resistance; NDM-1; New Delhi
metallo-β-lactamase–1; PBPs, penicillin-binding proteins; PER, Pseudomonas extended resistance; PPNG, penicillinase-producing N. gonorrhoeae; RND, resistance-nodulation-
cell division; rRNA, ribosomal RNA; SHV, sulfhydryl variable; SIM, Seoul imipenemase; TEM, Temoneira; TMP-SMX, trimethoprim-sulfamethoxazole; VEB, Vietnam extended-
spectrum β-lactamase; VIM, Verona integron-encoded metallo-β-lactamase; VRE, vancomycin-resistant enterococci.

TABLE 18-10  Resistance Mechanisms of Newer, Older, and Other Antimicrobial Agents
QUINUPRISTIN-
POLYMYXIN DAPTOMYCIN LINEZOLID DALFOPRISTIN METRONIDAZOLE TIGECYCLINE
Enzymatic inactivation − − − ++ − −
Decreased permeability − + (gram-negative) + (gram-negative) − − +
Efflux + − + + − ++
Alteration of target site ++ ++ ++ + − −
Protection of target site − + − − − −
Overproduce target − − − − − −
Bypass of inhibited process − − − − ++ −
Bind up antibiotic ++ − − − − −
+++, most common mechanism; ++, common; + less common.

drug induces permeability changes and loss of intracellular potassium mutations in the gene LiaF, involved in the bacterial cell envelope
in susceptible gram-positive bacteria. Resistance is often associated response to antibiotics and antimicrobial peptides, and the gaped gene,
with the abnormally thick cell wall characteristic of vancomycin- which generates an enzyme likely involved in the cell membrane phos-
intermediate S. aureus strains.208,209 Accumulation of mutations, espe- pholipid metabolism, have been suggested as mechanisms of daptomy-
cially with the gene mprF (encoding lysylphosphatidylglycerol cin resistance.211
synthetase) indicates the alterations in potential cell membrane bind- Tigecycline is a glycylcycline antibiotic with a mechanism of action
ing sites account for reduced daptomycin activity.210 In enterococci, of tetracylines, but remarkable resistance to many of the standard
249
resistance mechanisms. Recent evidence indicates that resistance is
Outer
associated with some unusual efflux pumps expressed in some multi-
membrane
resistant gram-negative bacilli.212,213
Quinupristin-dalfopristin is a combination of synergistic strepto-

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


TolC
gramins that inhibit protein synthesis by binding to the 23S rRNA in
the 50S ribosomal subunit. Resistance in gram-positive bacteria can
develop through enzymatic modification (vatD and vatE genes encod-
ing for acetyltransferases that inactivate dalfopristin), active efflux, and
target alteration.214 AcrA
Polymyxins are cationic peptide antibiotics that alter the permeabil-
ity of the outer membrane of gram-negative bacteria. The use of poly-
myxins has been rekindled in light of the limited number of therapeutic
options available to treat multidrug-resistant bacilli. Resistance is AcrB
attributable to binding of the agent to the polysaccharide capsule,215
alterations in the affinity of binding to the outer membrane lipid target
of polymyxin B (the pmr operon),216 or increased expression of the
efflux pump (the metAB oprM gene).217
Inner
Metronidazole resistance is related to loss of activity of reduced membrane
nicotinamide adenine dinucleotide (NADPH) nitroreductase via
mutations of the synthetic gene rdxA. This enzyme activity is essential FIGURE 18-6  Proposed structural model for the multidrug-
resistance AcrAB-TolC efflux pump in Escherichia coli. The AcrAB-TolC
to convert metronidazole to its active metabolite.218 These mechanisms
efflux system is the most important resistance-nodulation-cell division
are summarized in Table 18-10. (RND) transporter in E. coli, and is composed of three interconnected ele-
ments: (1) the transmembrane AcrB transporter, which protrudes from the
MULTIDRUG-RESISTANCE inner membrane into the periplasm; (2) the outer membrane channel TolC,
MECHANISMS AMONG BACTERIA which crosses from the periplasm through the outer membrane, providing
Bacteria can express more than one mechanism of antibiotic resistance, the exit route for substrates into the extracellular medium; (3) the AcrA
leading to phenotypes of multidrug resistance (MDR), or even pan- periplasmic accessory protein, which stabilizes the complex. The pump
resistance. For example, molecular analysis of P. aeruginosa isolates recognizes a wide variety of substrates, including hydrophobic organic
from a nosocomial outbreak in Belgium revealed the convergence of solvents and lipids, as well as anionic, cationic, and zwitterionic antimicro-
bials, yielding a multidrug-resistant phenotype. (Modified from Lomovos-
several strategies for antibiotic resistance: (1) overexpression of AmpC
kaya O, Zgurskaya HI, Totrov M, et al. Waltzing transporters and “the
chromosomal β-lactamases, conferring resistance to multiple β-lactam dance macabre” betweens humans and bacteria. Nat Rev Drug Discov.
antibiotics; (2) mutational loss of OprD porin, conferring resistance to 2007;6:56-65.)
imipenem; (3) upregulation of the MexXY efflux system (a member of
the resistance-nodulation-cell division [RND] family), which exports
fluoroquinolones, tetracycline, aminoglycosides, and antipseudomonal
β-lactam agents.219 CONTROL OF ANTIBIOTIC
In general, multiple antibiotic resistance in gram-negative bacteria RESISTANCE
often starts with the relatively limited outer membrane permeability to Although the emergence of antibiotic-resistant bacteria generally has
many antibiotic agents, coupled with the overexpression of MDR efflux been correlated with the rise and fall of specific antibiotic use in clinical
pumps, which can export multiple unrelated antibiotics.220 In addition, practice, the chain of causality is not always clear-cut.227 Bacterial
by reducing the intracellular concentration of the antimicrobial below strains contain complex aggregations of genes that may be linked
the MIC required for bacterial killing, efflux mechanisms may allow together. The use of one antibiotic may select for the emergence of
bacterial survival for longer periods of time, facilitating the accumula- resistance to another. Mobile genetic elements and rapidly evolving
tion of new antibiotic-resistance mutations (i.e., those encoding topoi- integron cassettes with multiple antibiotic-resistance genes endow bac-
somerase IV or DNA gyrase targets, rendering fluoroquinolones teria with a remarkable capacity to resist antibiotics.33-36,37,38,39,40,41,226
ineffective).221 Although the development of antibiotic resistance may be inevitable,
The clinically important MDR efflux pumps belong to several dif- the rate at which it develops may be diminished by the rational use of
ferent families: (1) the RND family, (2) the major facilitator superfam- antibiotics.137
ily (MFS), (3) the staphylococcal multiresistance (SMR) family, and (4) The ability to track antibiotic-resistance genes with molecular tech-
the multidrug and toxic compound extrusion (MATE) family. Such niques has enhanced the ability to track the spread of antibiotic resis-
efflux pumps are widespread among prokaryotes (see Fig. 18-6) and tance. With the appropriate computerized surveillance, a hospital
are responsible for the export of toxic substances, allowing survival in laboratory may be able to detect rapidly the emergence of a new type
a noxious environment, such as the biliary system for enteric bacteria, of resistance or the presence of a new microbial strain within a specific
for example.222 They may also play a role in mediating bacterial adher- unit or patient population. Techniques such as restriction endonucle-
ence to host tissues and exporting virulence determinants, as has been ase digestion analyses of microbial genomes, rapid genomics, and
described for P. aeruginosa.223 genetic probes of antibiotic-resistance genes by polymerase chain reac-
Bacteria can also acquire multidrug resistance through sequential tion make it possible to confirm the presence of new genes in the
transfer of multiple resistance determinants located on mobile genetic environment. This information may be correlated with the phenotypic
elements. For example, conjugative transposons such as Tn916, con­ measures determined by the clinical microbiology surveillance system
ferring resistance to tetracycline and chloramphenicol, can easily (Fig. 18-7). Use of molecular techniques greatly augments surveillance
disseminate between bacterial species.224 Transposons often coexist data because large data sets may obscure subtle changes (“mini-
with other genetic elements, such as plasmids that may carry addi- epidemics”) that may be more amenable to the institution of stringent
tional antibiotic-resistant determinants. For example, analysis of a infection control measures.
plasmid encoding the bla-CTX-15 gene responsible for resistance Some bacterial strains have the ability to hypermutate in stressful
to extended-spectrum cephalosporins in an E. coli outbreak in environments, increasing the risk of acquisition of resistance muta-
Toronto, Canada, revealed a large MDR region, encoding multiple tions.228,229 Because prokaryotic organisms all contribute to a common
transposons, and numerous other resistance genes, such as bla(OXA- “gene pool,” the total complement of favorable genes mediating antibi-
1), bla(TEM-1), tetA, and aminoglycoside-resistance genes aac(6′)-Ib otic resistance (the metagenomic resistome) may disseminate among
and aac(3′)-II.225 The ability of bacteria to capture multiple antibiotic- bacterial populations. Reports of the rapidly spreading NDM-type
resistance genes into existing integrons, with their seemingly endless carbapenemases suggest that common, invasive, microbial pathogens
recombination potential, is astonishing.226 may become refractory to any chemotherapeutic agent in the future.
250

Kb subclones existing within a large population of bacteria can survive a


single, therapeutic dose of a bactericidal antibiotic. These subpopula-
tions include (1) bacterial persisters (antibiotic sensitive when growing
but refractory when metabolically dormant); (2) rare, relatively resis-
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

tant subpopulations within large populations; and (3) mutator strains


(high baseline mutation rate clones), which can all be selected for, and
lead to, in vivo acquisition of resistance during or after antibiotic
therapy. These small subpopulations are present in insignificant
numbers (<10−8) and are readily eliminated by host antimicrobial
defenses under normal circumstances. Their very existence goes unno-
~44 — ticed during antibacterial treatment for most infectious diseases.
However, these relatively resistant subpopulations can survive initial
low doses of antibiotics, regrow, and become a source of in vivo devel-
opment of resistance when treating infections with large microbial
loads, infections adherent to foreign bodies or nonviable tissues, and
6.1— in the absence of adequate host defenses (e.g., large populations of
5.6—
pathogens in undrained abscesses, infected joint prostheses, severe
immuncompromised states).
Dosing strategies that follow pharmacokinetic and pharmacody-
namic principles, and dosing regimens that consider mutant protection
concentration (MPC), rather than just the MIC, should become part
of antibiotic stewardship programs. The MPC can be as much as 10- to
20-fold higher than the MIC for many classes of antibiotics.137 Larger
concentrations of antibiotics can eliminate resistant subpopulations
A B that express one or two resistance mutations, allowing them to persist
at concentrations just above the MIC. These surviving subpopulations
1 2 3 4 5 6 1 2 3 4 5 6 are selected for during antibacterial treatment. If they can accumulate
FIGURE 18-7  A, Agarose gel of EcoRI-digested plasmids derived from additional resistance capacity by hypermutation or acquisition of genes
four isolates that contain a nosocomial trimethoprim-resistance plasmid from neighboring bacteria, clinical failures with in vivo development
(called pBWH10) from a Boston hospital (lanes 1 to 4). Another nosocomial of resistance might result. If the initial dose of antibiotic is high enough
plasmid from the same hospital that does not contain trimethoprim- to eradicate even these resistant subpopulations (above the MPC),
resistance genes (lane 5) and one in which the trimethoprim-resistance treatment can succeed and avoid this problem. Regrettably, the MPC
and -sensitive plasmids are present in the same isolate (lane 6) are shown. is not easily calculated in most clinical laboratories, and the MPCs for
B, To show that the “fingerprints” from the trimethoprim-resistance plas- the antibacterials might be difficult to achieve without toxicity.137 Early
mids in lanes 1 through 4 and 6 contain the same gene, DNA-DNA recognition and treatment before large bacterial loads accumulate,
hybridization of the same six plasmids was performed using a type II
dihydrofolate reductase probe. The probe and the restriction endonuclease
short courses of adequate doses of antibiotics to limit resistance devel-
analyses helped pinpoint the location and genetic homology of this opment, and restricting antimicrobial agents to only patients who
trimethoprim-resistance gene. actually need them are all important in the prevention of antibiotic
resistance.
Antibiotics are a precious commodity, and we should do what we
can to preserve the activity of antimicrobials to treat human infections.
Rational antibiotic usage policies suggest the curtailment of the unnec- Dosing regimens should be chosen for both clinical efficacy and for
essary use of antibiotics in situations such as animal husbandry. The the prevention of resistance development. Academic national and
causal link between the use of antibiotics for animal growth promotion international organizations, such as World Alliance against Antibiotic
and their augmentation of the resistance in human pathogens acquired Resistance (WAAAR) are now advocating for the commonsense mea-
from food is now reasonably well established.230-233 sures to limit progressive antibiotic resistance in clinical and nonclini-
Unfortunately, the paucity of new antimicrobials in development cal use and to promote research into new antibiotics and nonantibiotic
and the rapid spread of multidrug-resistant pathogens, especially therapies to treat infections.233 The best hope for the future is the
among gram-negative bacilli, offer limited therapeutic options in development of a greater understanding of how antimicrobial resis-
severe infections. Resistance can develop even during therapy with tance spreads, intelligent use and development of improved bacterial
seemingly adequate doses of antibacterial agents to which the pathogen vaccines, antibiotic stewardship, and the implementation of effective
appears to be susceptible by standard sensitivity testing. Three types of infection control strategies.234

Key References 17. Summers AO, Wireman J, Vimy MJ, et al. Mercury released
from dental silver fillings provokes an increase in mercury-
23. Fornier PE, Vallenet D, Barbe V, et al. Comparative genom-
ics of multidrug resistance in Acinetobacter baumannii.
resistant and antibiotic-resistant bacteria in oral and intes- PLoS Genet. 2006;2:e7.
The complete reference list is available online at Expert Consult.
tinal floras of primates. Antimicrob Agents Chemother. 24. Tato M, Coque TM, Ruiz-Garbajosa P, et al. Complex
1993;37:825-834. clonal and plasmid epidemiology in the first outbreak of
1. Rice W, Chippindale A. Sexual recombination and the
18. Blattner FR, Plunkett G 3rd, Bloch CA, et al. The complete Enterobacteriaceae infection involving VIM-1 metallo-
power of natural selection. Science. 2001;294:555-559.
genome sequence of Escherichia coli K-12 [comment]. beta-lactamase in Spain: toward endemicity? Clin Infect
2. Toleman MA, Walsh TR. Combinatorial events of insertion
Science. 1997;277:1453-1474. Dis. 2007;45:1171-1178.
sequences and ICE in gram-negative bacteria. FEMS
19. El Solh N, Allignet J, Bismuth R, et al. Conjugative transfer 25. Arthur M, Reynolds P, Courvalin P. Glycopeptide resis-
Microbiol Rev. 2011;35:912-935.
of staphylococcal antibiotic resistance markers in the tance in enterococci. Trends Microbiol. 1996;4:401-407.
3. Medeiros AA. Evolution and dissemination of β-lactamases
absence of detectable plasmid DNA. Antimicrob Agents 26. Brisson-Noel A, Arthur M, Courvalin P. Evidence for
accelerated by generations of β-lactam antibiotics. Clin
Chemother. 1986;30:161-169. natural gene transfer from gram-positive cocci to Esche-
Infect Dis. 1997;24:S19-S45.
20. Salyers AA, Shoemaker NB, Stevens AM, et al. Conjugative richia coli. J Bacteriol. 1988;170:1739-1745.
4. Gold HS, Moellering RC Jr. Antimicrobial-drug resistance.
transposons: an unusual and diverse set of integrated gene 27. Papadopoulou B, Courvalin P. Dispersal in Campylobacter
N Engl J Med. 1996;335:1445-1453.
transfer elements. Microbiol Rev. 1985;49:679-690. spp. of aphA-3, a kanamycin resistance determinant from
6. Chang S, Sievert DM, Hageman JE, et al. Infection with
21. Morse SA, Johnson SR, Biddle JW, Roberts MC. High-level gram-positive cocci. Antimicrob Agents Chemother. 1988;
vancomycin-resistant Staphylococcus aureus containing the
tetracycline resistance in Neisseria gonorrhoeae is result of 32:945-948.
vanA resistance gene. N Engl J Med. 2003;348:1342-1347.
acquisition of streptococcal tetM determinant. Antimicrob 28. Courvalin P, Carlier C, Collatz E. Plasmid-mediated resis-
10. D’Costa VM, King CE, Kalan L, et al. Antibiotic resistance
Agents Chemother. 1986;30:664-670. tance to aminocyclitol antibiotics in group D streptococci.
is ancient. Nature. 2011; 477:457-461.
22. Roberts MC, Kenny GE. Dissemination of the tetM tetra- J Bacteriol. 1980;143:541-551.
11. Dantas G, Sommer MOA, Oluwasegun RD, Church GM.
cycline resistance determinant to Ureaplasma urealyticum. 29. Cohen ML. Epidemiology of drug resistance: implications
Bacteria subsisting on antibiotics. Science. 2008;320:
Antimicrob Agents Chemother. 1986;29:350-352. for a post-antimicrobial era. Science. 1992;257:1050-1055.
100-103.
251
30. Levy SB. Antibiotic resistance: consequences of inaction. 79. Appelbaum PC. Patterns of resistance and resistance mech- 193. Unemo M, Nicholas RA. Emergence of multidrug resistant,
Clin Infect Dis. 2001;3:S124-S129. anisms in anaerobes. Clin Microbiol Newslett. 1992;14: extensively drug resistant and untreatable gonorrhea.
32. Stokes HW, Hall RM. A novel family of potentially mobile 49-53. Future Microbiol. 2012;7:1401-1422.
DNA elements encoding site-specific gene-integration 89. Barada K, Hanaki H, Ikeda S, et al. Trends in the gentamcin 200. Martinez-Martinez L, Pascual A, Jacoby GA. Quinolone

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


functions: integrons. Mol Microbiol. 1989;3:1669-1683. and arbenkacin susceptibility of methicillin-resistant Staph- resistance from a transferable plasmid. Lancet. 1998; 351:
37. Hall MAL, Block HEM, Donders RT, et al. Multidrug resis- ylococcus aureus and the enzymes encoding aminoglycoside 797-799.
tance among enterobacteriaceae is strongly associated with modifiying enzymes. J Infect Chemother. 2007;13:74-78. 201. Luo Y, Li J, Meng Y, et al. Joint effects of topoisomerase
the presence of integrons and is independent of species or 90. Mederski-Samoraj BD, Murray BE. High-level resistance to alteration and plasmid-mediated, quinolone-resistant
isolate origin. J Infect Dis. 2003;187:251-259. gentamicin in clinical isolates of enterococci. J Infect Dis. determinants in Salmonella enterica Typhimurium. Microb
40. Fluit AC, Schmitz F-J. Resistance integrons and super- 1983;147:751-757. Drug Resist. 2011;17:1-5.
integrons. Clin Microbiol Infect. 2004;10;274-288. 98. Robicsek A, Strahilevitz J, Jacoby EA, et al. Fluoroquinolone- 208. Longzhu C, Tominaga E, Neoh H, Hiramatsu K. Correla-
42. Bush K, Jacoby GA, Medeiros AA. A functional classifica- modifying enzyme: a new adaptation of a common amino- tion between reduced daptomycin susceptibility and
tion scheme for β-lactamases and its correlation with glycoside acetyltransferase. Nat Med. 2006;12:83-88. vancomycin resistance in vancomycin-intermediate Staph-
molecular structure. Antimicrob Agents Chemother. 1995; 110. Nikaido H, Vaara M. Molecular basis of bacterial outer ylococcus aureus. Antimicrob Agents Chemother. 2006;50:
39:1211-1233. membrane permeability. Microbiol Rev. 1985;49:1-32. 1079-1082.
43. Abraham EP, Chain E. An enzyme from bacteria able to 118. Hooper DC, Wolfson JS, Ng EY, Swartz MN. Mechanisms 209. Sakoulas G, Alder J, Thauvin-Eliopoulos C, et al. Induction
destroy penicillin. Nature. 1940;1446:837. of action of and resistance to ciprofloxacin. Am J Med. of daptomycin heterogeneous susceptibility in Staphylococ-
47. Babic M, Hujer AM, Bonomo RA. What’s new in antibiotic 1987;82:12-20. cus aureus by exposure to vancomycin. Antimicrob Agents
resistance? Focus on beta-lactamases. Drug Resist Updat. 125. Williams JB. Drug efflux as a mechanism of resistance. Br Chemother. 2006;50:1581-1585.
2006;9:142-156. J Biomed Sci. 1996;53:290-293. 211. Arias CA, Panesso D, McGrath DM, et al. Genetic basis for
49. Cornaglia G, Giamarellou H, Rossolini GM. Metallo-beta- 130. Leclercq R. Mechanisms of resistance to macrolides and in vivo daptomycin resistance in enterococci. N Engl J Med.
lactamases: a last frontier for beta-lactams? Lancet Infect lincosamides: nature of the resistance elements and their 2011;365:892-900.
Dis. 2011;11:381-393. clinical implications. Clin Infect Dis. 2002;34:482-492. 212. Iredell J, Thomas L, Power D, Mendes E. Tigecycline resis-
50. Gupta N, Limbago BM, Patel JB, Kallen AJ. Carbapenem- 145. Doi Y, Arakawa Y. 16S ribosomal RNA methylation: tance in Australian antibiotic-resistant gram-negative bac-
resistant Enterobacteriaceae: epidemiology and preven- emerging resistance mechanism against aminoglycosides. teria. J Antimicrob Chemother. 2007;59:816-818.
tion. Clin Infect Dis. 2011;53:60-67. Clin Infect Dis. 2007;45:88-94. 217. Pamp SJ, Gjermansen M, Johansen HK, Tolker-Nielsen T.
54. Jacoby GA, Munoz-Price SL. The new beta-lactamases. N 149. Meka VG, Gold HS. Antimicrobial resistance to linezolid. Tolerance to the antimicrobial peptide colistin in Pseudo-
Engl J Med. 2005;352:380-391. Clin Infect Dis. 2004;39:1010-1015. monas aeruginosa bioflims is linked to metabolically active
57. Canton R, Coque TM. The CTX-M beta-lactamase pan- 157. Centers for Disease Control and Prevention. Vancomycin- cells and depends upon pmr and metAB oprM genes. Mol
demic. Curr Opin Microbiol. 2006;9:466-475. resistant Staphylococcus aureus—Pennsylvania, 2002. Microbiol. 2008;68:223-240.
63. Chow JW, Fine MJ, Shlaes DM, et al. Enterobacter bac­ MMWR Morb Mortal Wkly Rep. 2002;51:902. 219. Deplano A, Denis O, Poirel L, et al. Molecular character­
teremia: clinical features and emergence of antibiotic 167. Courvalin P. Vancomycin resistance in gram-positive ization of an epidemic clone of panantibiotic-resistant
resistance during therapy. Ann Intern Med. 1991;115: cocci. Clin Infect Dis. 2006;42(suppl 1):S25-S34. Pseudomonas aeruginosa. J Clin Microbiol. 2005;43:
585-590. 170. Cui L, Iwamoto A, Lian JO, et al. Novel mechanism 1198-2004.
64. Bratu S, Landman D, Haag R, et al. Rapid spread of of antibiotic resistance originating in vancomycin- 221. Piddock L. Clinically relevant chromosomally encoded
carbapenem-resistant Klebsiella pneumoniae in New York intermediate Staphylococcus aureus. Antimicrob Agents multidrug resistance efflux pumps in bacteria. Clin Micro-
City: a new threat to our antibiotic armamentarium. Arch Chemother. 2006;50:428-438. biol Rev. 2006;19:382-402.
Intern Med. 2005;165:1430-1435. 171. Hospital Infection Control Practices Advisory Committee 223. Hirakata Y, Srikumar R, Poole K, et al. Multidrug efflux
68. Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo- (HICPAC). Recommendations for preventing the spread systems play an important role in the invasiveness of Pseu-
beta-lactamases: the quiet before the storm? Clin Microbiol of vancomycin resistance. Infect Control Hosp Epidemiol. domonas aeruginosa. J Exp Med. 2002;196:109-118.
Rev. 2005;18:305-325. 1995;16:105-113. 229. Tompkins JD, Nelson JL, Hazel JC, et al. Error-prone poly-
72. Bonomo RA, Szabo D. Mechanisms of multidrug resistance 178. Hartman BJ, Tomasz A. Low-affinity penicillin-binding merase, DNA polymerase IV, is responsible for transient
in Acinetobacter species and Pseudomonas aeruginosa. Clin protein associated with beta-lactam resistance in Staphylo- hypermutation during adaptive mutation in Escherichia
Infect Dis. 2006;43(suppl 2):49-56. coccus aureus. J Bacteriol. 1984;158:513-516. coli. J Bacteriol. 2003;185:3469-3472.
251.e1
References 31. Recchia GD, Hall RM. Origins of the mobile gene cassettes
found in integrons. Trends Microbiol. 1997;5:389-394.
59. Pitout JDD, Laupland KB. Extended-spectrum
β-lactamase-producing Enterobacteriaceae: an emerging
1. Rice W, Chippindale A. Sexual recombination and the
32. Stokes HW, Hall RM. A novel family of potentially mobile public-health concern. Lancet Infect Dis. 2008;8:159-166.
power of natural selection. Science. 2001;294:555-559.
DNA elements encoding site-specific gene-integration 60. Johnson JR, Johnston B, Clabots C, et al. Escherichia coli
2. Toleman MA, Walsh TR. Combinatorial events of insertion

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


functions: integrons. Mol Microbiol. 1989;3:1669-1683. sequence type ST131 as the major cause of serious
sequences and ICE in gram-negative bacteria. FEMS
33. Collis CM, Hall RM. Expression of antibiotic resistance multidrug-resistant E. coli infections in the United States.
Microbiol Rev. 2011;35:912-935.
genes in the integrated cassettes of integrons. Antimicrob Clin Infect Dis. 2010;51:286-294.
3. Medeiros AA. Evolution and dissemination of β-lactamases
Agents Chemother. 1995;39:155-162. 61. Livermore DM. Clinical significance of beta-lactamase
accelerated by generations of β-lactam antibiotics. Clin
34. Ouellette M, Bissonnette L, Roy PH. Precise insertion of induction and stable derepression in gram-negative rods.
Infect Dis. 1997;24:S19-S45.
antibiotic resistance determinants into Tn21-like transpo- Eur J Clin Microbiol. 1987;6:439-445.
4. Gold HS, Moellering RC Jr. Antimicrobial-drug resistance.
sons: nucleotide sequence of the OXA-1 beta-lactamase 62. Jones RN. Important and emerging beta-lactamase-
N Engl J Med. 1996; 335:1445-1453.
gene. Proc Natl Acad Sci U S A. 1987;84:7378-7382. mediated resistances in hospital-based pathogens: the
5. Landman D, Bratu S, Kochar S, et al. Evolution of antimi-
35. Paulsen IT, Littlejohn TG, Radstrom P, et al. The 3′ con- Amp C enzymes. Diagn Microbiol Infect Dis. 1998;31:
crobial resistance among Pseudomonas aeruginosa, Aci-
served segment of integrons contains a gene associated 461-466.
netobacter baumannii, and Klebsiella pneumoniae in
with multidrug resistance to antiseptics and disinfectants. 63. Chow JW, Fine MJ, Shlaes DM, et al. Enterobacter bactere-
Brooklyn, NY. J Antimicrob Chemother. 2007;60:78-82.
Antimicrob Agents Chemother. 1993;35:761-768. mia: clinical features and emergence of antibiotic resistance
6. Chang S, Sievert DM, Hageman JE, et al. Infection with
36. Messier N, Roy PH. Integron integrases possess a unique during therapy. Ann Intern Med. 1991;115:585-590.
vancomycin-resistant Staphylococcus aureus containing the
additional domain necessary for activity. J Bacteriol. 64. Bratu S, Landman D, Haag R, et al. Rapid spread of
vanA resistance gene. N Engl J Med. 2003;348:1342-1347.
2001;183:6699-6706. carbapenem-resistant Klebsiella pneumoniae in New York
7. Julian K, Kosowska-Shick K, Whitener C, et al. Character-
37. Hall MAL, Block HEM, Donders RT, et al. Multidrug resis- City: a new threat to our antibiotic armamentarium. Arch
ization of a daptomycin-nonsusceptible vancomycin-
tance among enterobacteriaceae is strongly associated with Intern Med. 2005;165:1430-1435.
intermediate Staphylococcus aureus strain in a patient with
the presence of integrons and is independent of species or 65. Smith Moland E, Hanson ND, Herrera VL, et al. Plasmid-
endocarditis. Antimicrob Agents Chemother. 2007;51:
isolate origin. J Infect Dis. 2003;187:251-259. mediated, carbapenem-hydrolysing beta-lactamase,
3445-3448.
38. Recchia GD, Hall RM. Gene cassettes: a new class of mobile KPC-2, in Klebsiella pneumoniae isolates. J Antimicrob Che-
8. Galimand M, Guiyoule A, Gerbaud G, et al. Multidrug
element. Microbiology. 1995;141:3015-3027. mother. 2003;51:711-714.
resistance in Yersinia pestis mediated by a transferable
39. Naas T, Mikami Y, Imai T, et al. Characterization of In53, 66. Bradford PA, Bratu S, Urban C, et al. Emergence of
plasmid. N Engl J Med. 1997;337:677-680.
a class 1 plasmid- and composite transposon-located inte- carbapenem-resistant Klebsiella species possessing the class
9. Tran JH, Jacoby GA. Mechanism of plasmid-mediated qui-
gron of Escherichia coli which carries an unusual array of A carbapenem-hydrolyzing KPC-2 and inhibitor-resistant
nolone resistance. Proc Natl Acad Sci U S A. 2002;99:
gene cassettes. J Bacteriol. 2001;183:235-249. TEM-30 beta-lactamases in New York City. Clin Infect Dis.
5638-5642.
40. Fluit AC, Schmitz F-J. Resistance integrons and super- 2004;39:55-62.
10. D’Costa VM, King CE, Kalan L, et al. Antibiotic resistance
integrons. Clin Microbiol Infect. 2004;10;274-288. 67. Yigit H, Queenan AM, Anderson GJ, et al. Novel
is ancient. Nature. 2011; 477:457-461.
41. Weldhagen GF. Integrons and beta-lactamases—a novel carbapenem-hydrolyzing beta-lactamase, KPC-1, from a
11. Dantas G, Sommer MOA, Oluwasegun RD, Church GM.
perspective on resistance. Int J Antimicrob Agents. 2004;23: carbapenem-resistant strain of Klebsiella pneumoniae.
Bacteria subsisting on antibiotics. Science. 2008;320:
556-562. Antimicrob Agents Chemother. 2001;45:1151-1161.
100-103.
42. Bush K, Jacoby GA, Medeiros AA. A functional classifica- 68. Walsh TR, Toleman MA, Poirel L, Nordmann P. Metallo-
12. Cattoir V, Poirel L, Aubert C, et al. Unexpected occurrence
tion scheme for β-lactamases and its correlation with beta-lactamases: the quiet before the storm? Clin Microbiol
of plasmid-mediated quinolone resistance determinants in
molecular structure. Antimicrob Agents Chemother. 1995; Rev. 2005;18:305-325.
environmental Aeromonas spp. Emerg Infect Dis. 2008;14:
39:1211-1233. 69. Walsh TR. The emergence and implications of metallo-
231-237.
43. Abraham EP, Chain E. An enzyme from bacteria able to beta-lactamases in gram-negative bacteria. Clin Microbiol
13. Barlow M, Reik RA, Jacobs SD, et al. High rate of mobiliza-
destroy penicillin. Nature. 1940;1446:837. Infect. 2005;11(suppl 6):2-9.
tion for blaCTX-Ms. Emerg Infect Dis. 2008;14:423-428.
44. Hawkey PM. Molecular epidemiology of clinically signifi- 70. Yong D, Toleman MA, Giske CG, et al. Characterization of
14. Massey RC, Buckling A, Peacock SJ. Phenotypic switching
cant antibiotic resistance genes. Br J Pharmacol. 2008;153: a new metallo-beta-lactamase gene and a novel erythromy-
of antibiotic resistance circumvents permanent costs in
S406-S413. cin esterase gene carried on a unique genetic structure in
Staphylococcus aureus. Curr Biol. 2001;11:1810-1814.
45. Bradford PA. Extended-spectrum beta-lactamases in the Klebsiella pneumoniae sequence type 14 from India. Anti-
15. Jorth P, Whiteley M. An evolutionary link between natural
21st century: characterization, epidemiology, and detec- microb Agents Chemother. 2009;53:5046-5054.
transformation and CRISPR adaptive immunity. MBio.
tion of this important resistance threat. Clin Microbiol Rev. 71 Nordmann P, Poirel L, Walsh TR, Livermore DM. The
2012;3:e00309-12.
2001;14:933-951. emerging NDM carbapenemases. Trends Microbiol. 2011;
16. Palmer KL, Gilmore MS. Mutlidrug-resistant enterococci
46. Brunton J, Clare D, Meier MA. Molecular epidemiology of 19:58-95.
lack CRISPR-cas. MBio. 2010;1:e00227-10.
antibiotic resistance plasmids of Haemophilus species and 72. Bonomo RA, Szabo D. Mechanisms of multidrug resistance
17. Summers AO, Wireman J, Vimy MJ, et al. Mercury released
Neisseria gonorrhoeae. Rev Infect Dis. 1986;8:713-724 in Acinetobacter species and Pseudomonas aeruginosa. Clin
from dental silver fillings provokes an increase in mercury-
47. Babic M, Hujer AM, Bonomo RA. What’s new in antibiotic Infect Dis. 2006;43(suppl 2):49-56.
resistant and antibiotic-resistant bacteria in oral and intes-
resistance? Focus on beta-lactamases. Drug Resist Updat. 73. Logan LK. Carbapenem-resistant enterobacteriaceae: an
tinal floras of primates. Antimicrob Agents Chemother.
2006;9:142-156. emerging problem in children. Clin Infect Dis. 2012;55:
1993;37:825-834.
48. Knothe H, Shah P, Kremery V, et al. Transferable resistance 852-859.
18. Blattner FR, Plunkett G 3rd, Bloch CA, et al. The complete
to cefotaxime, cefoxitin, cefamandole and cefuroxime in 74. Walther-Rasmussen J, Hoiby N. OXA-type carbapene-
genome sequence of Escherichia coli K-12 (comment).
clinical isolates of Klebsiella pneumoniae and Serratia marc- mases. J Antimicrob Chemother. 2006;57:373-383.
Science. 1997;277:1453-1474.
escens. Infection. 1983;11:315-317. 75. McDonnell RW, Sweeney HM, Cohen S. Conjugational
19. El Solh N, Allignet J, Bismuth R, et al. Conjugative transfer
49. Cornaglia G, Giamarellou H, Rossolini GM. Metallo-beta- transfer of gentamicin resistance plasmids intra- and inter-
of staphylococcal antibiotic resistance markers in the
lactamases: a last frontier for beta-lactams? Lancet Infect specifically in Staphylococcus aureus and Staphylococcus
absence of detectable plasmid DNA. Antimicrob Agents
Dis. 2011;11:381-393. epidermidis. Antimicrob Agents Chemother. 1983;23:
Chemother. 1986;30:161-169.
50. Gupta N, Limbago BM, Patel JB, Kallen AJ. Carbapenem- 151-160.
20. Salyers AA, Shoemaker NB, Stevens AM, et al. Conjugative
resistant Enterobacteriaceae: epidemiology and preven- 76. Zscheck KK, Murray BE. Genes involved in the regulation
transposons: an unusual and diverse set of integrated gene
tion. Clin Infect Dis. 2011;53:60-67. of beta-lactamase production in enterococci and staphylo-
transfer elements. Microbiol Rev. 1985;49:679-690.
51. Philippon A, Labia R, Jacoby G. Extended-spectrum cocci. Antimicrob Agents Chemother. 1993;37:1966-1970.
21. Morse SA, Johnson SR, Biddle JW, Roberts MC. High-level
beta-lactamases. Antimicrob Agents Chemother. 1989;33: 77. Murray BE. Beta-lactamase-producing enterococci. Anti-
tetracycline resistance in Neisseria gonorrhoeae is result of
1131-1136. microb Agents Chemother. 1992;36:2355-2359.
acquisition of streptococcal tetM determinant. Antimicrob
52. Sougakoff W, Goussard S, Courvalin P. The TEM-3- 78. Rice LB, Marshall SH. Evidence of incorporation of the
Agents Chemother. 1986;30:664-670.
lactamase, which hydrolyzes broad-spectrum cephalo­ chromosomal beta-lactamase gene of Enterococcus
22. Roberts MC, Kenny GE. Dissemination of the tetM tetra-
sporins, is derived from the TEM-2 penicillinase by two faecalis-CH19 into a transposon derived from staphylo-
cycline resistance determinant to Ureaplasma urealyticum.
amino acid substitutions. FEMS Microbiol Lett. 1988;56: cocci. Antimicrob Agents Chemother. 1992;36:1843-1846.
Antimicrob Agents Chemother. 1986;29:350-352.
343-348. 79. Appelbaum PC. Patterns of resistance and resistance mech-
23. Fornier PE, Vallenet D, Barbe V, et al. Comparative genom-
53. Lefon-Guibout V, Speldooren V, Heym B, Nicolas- anisms in anaerobes. Clin Microbiol Newslett. 1992;14:
ics of multidrug resistance in Acinetobacter baumannii.
Chanoine M-H. Epidemiological survey of amoxicillin- 49-53.
PLoS Genet. 2006;2:e7.
clavulanate resistance and corresponding molecular 80. Appelbaum PC, Spangler SK, Pankuch GA, et al. Charac-
24. Tato M, Coque TM, Ruiz-Garbajosa P, et al. Complex
mechanisms in Escherichia coli isolates in France: new terization of a beta-lactamase from Clostridium clostridio-
clonal and plasmid epidemiology in the first outbreak of
genetic features of blaTEM genes. Antimicrob Agents Che- forme. J Antimicrob Chemother. 1994;33:33-40.
Enterobacteriaceae infection involving VIM-1 metallo-
mother. 2000;44:2709-2714. 81. Tuner K, Lindqvist L, Nord CE. Purification and properties
beta-lactamase in Spain: toward endemicity? Clin Infect
54. Jacoby GA, Munoz-Price SL. The new beta-lactamases. N of a novel beta-lactamase from Fusobacterium nucleatum.
Dis. 2007;45:1171-1178.
Engl J Med. 2005;352:380-391. Antimicrob Agents Chemother. 1985;27:943-947.
25. Arthur M, Reynolds P, Courvalin P. Glycopeptide resis-
55. Poirel L, Kampfer P, Nordmann P. Chromosome-encoded 82. Sanders CC, Sanders WE Jr, Goering RV. In vitro antago-
tance in enterococci. Trends Microbiol. 1996;4:401-407.
Ambler class A beta-lactamase of Kluyvera georgiana, a nism of beta-lactam antibiotics by cefoxitin. Antimicrob
26. Brisson-Noel A, Arthur M, Courvalin P. Evidence for
probable progenitor of a subgroup of CTX-M extended- Agents Chemother. 1982;21:968-975.
natural gene transfer from gram-positive cocci to Esche-
spectrum beta-lactamases. Antimicrob Agents Chemother. 83. Hedberg M, Edlund C, Lindqvist L, et al. Purification and
richia coli. J Bacteriol. 1988;170:1739-1745.
2002;46:4038-4040. characterization of an imipenem hydrolysing metallo-beta-
27. Papadopoulou B, Courvalin P. Dispersal in Campylobacter
56. Humeniuk C, Arlet G, Gautier V, et al. β-lactamases of lactamase from Bacteroides fragilis. J Antimicrob Che-
spp. of aphA-3, a kanamycin resistance determinant from
Kluyvera ascorbata, probable progenitors of some plasmid- mother. 1992;29:105-113.
gram-positive cocci. Antimicrob Agents Chemother. 1988;
encoded CTX-M types. Antimicrob Agents Chemother. 84. Jacobs MR, Spangler SK, Appelbaum PC. Beta-lactamase
32:945-948.
2002;46:3045-3049. production and susceptibility to US and European anaero-
28. Courvalin P, Carlier C, Collatz E. Plasmid-mediated resis-
57. Canton R, Coque TM. The CTX-M beta-lactamase pan- bic gram-negative bacilli to beta-lactams and other agents.
tance to aminocyclitol antibiotics in group D streptococci.
demic. Curr Opin Microbiol. 2006;9:466-475. Eur J Clin Microbiol Infect Dis. 1992;11:1081-1093.
J Bacteriol. 1980;143:541-551.
58. Ben-Ami R, Schwaber MJ, Navon-Venezia S, et al. Influx 85. Moxon ER, Medeiros AA, O’Brien TF. Beta-lactamase
29. Cohen ML. Epidemiology of drug resistance: implications
of extended-spectrum β-lactamase-producing Entero­ effect on ampicillin treatment of Haemophilus influenzae B
for a post-antimicrobial era. Science. 1992;257:1050-1055.
bacteriaceae into the hospital. Clin Infect Dis. 2006;42: bacteremia and meningitis in infant rats. Antimicrob Agents
30. Levy SB. Antibiotic resistance: consequences of inaction.
925-934. Chemother. 1977;12:461-464.
Clin Infect Dis. 2001;3:S124-S129.
251.e2
86. Rasheed JK, Jay C, Metchock B, et al. Evolution of lactam and aminoglycoside antibiotics in a strain of Ser- 139. Shortridge VD, Doern GV, Brueggemann AB, et al.
extended-spectrum beta-lactam resistance (SHV-8) in a ratia marcescens. Ann Microbiol. 1983;134A:329-337. Pre­valence of macrolide resistance mechanisms in
strain of Escherichia coli during multiple episodes of bac- 114. Quinn JP, Dudek EJ, DiVincenzo CA, et al. Emergence of Streptococcus pneumoniae isolates from a multicenter anti-
teremia. Antimicrob Agents Chemother. 1997;41:647-653. resistance to imipenem during therapy for Pseudomonas biotic resistance surveillance study conducted in the United
Part I  Basic Principles in the Diagnosis and Management of Infectious Diseases

87. Shimizu K, Kumada T, Hsieh WC, et al. Comparison of aeruginosa infections. J Infect Dis. 1986;154:289-293. States in 1994-1995. Clin Infect Dis. 1999;29:1186-1188.
aminoglycoside resistance patterns in Japan, Formosa, and 115. Macia MD, Blanquer D, Togores B, et al. Hypermutation is 140. Tait-Kamradt X, Davies T, Appelbaum PC, et al. Two new
Korea, Chile, and the United States. Antimicrob Agents Che- a key factor in development of multiple antimicrobial resis- mechanisms of macrolide resistance in clinical strains
mother. 1985;28:282-288. tance in Pseudomonas aeruginosa strains causing lung infec- of Streptococcus pneumoniae from Eastern Europe and
88. John JF Jr, Twitty JA. Plasmids as epidemiologic markers tion. Antimicrob Agents Chemother. 2005;49:3382-3386. North America. Antimicrob Agents Chemother. 2000;44:
in nosocomial gram-negative bacilli: Experience at a uni- 116. Livermore DM. Interplay of impermeability and chromo- 3395-3401.
versity and review of the literature. Rev Infect Dis. 1986;8: somal beta-lactamase activity in imipenem-resistant Pseu- 141. Roberts MC. Update on acquired tetracycline resistance
693-704. domonas aeruginosa. Antimicrob Agents Chemother. 1992; genes. FEMS Microbiol Lett. 2005;245:195-203.
89. Barada K, Hanaki H, Ikeda S, et al. Trends in the gentamcin 36:2046-2048. 142. Trieber GA, Taylor DE. Mutations in the 16S rRNA genes
and arbenkacin susceptibility of methicillin-resistant 117. Sanders BC, Sanders WE Jr, Goering RV, Verner V. Selec- of Helicobacter pylori mediate resistance to tetracycline.
Staphylococcus aureus and the enzymes encoding amino- tion of multiple antibiotic resistance by quinolones, beta- J Bacteriol. 2002;184:2131-2140.
glycoside modifiying enzymes. J Infect Chemother. 2007;13: lactams, and aminoglycosides with special refercence to 143. Zhou Y, Yu H, Guo Q, et al. Distribution of 16S rRNA
74-78. cross resistance between unrelated drug classes. Antimicrob methylases among different species of gram-negative
90. Mederski-Samoraj BD, Murray BE. High-level resistance to Agents Chemother. 1984;306:797-801. bacilli with high-level resistance to aminoglycosides. Eur J
gentamicin in clinical isolates of enterococci. J Infect Dis. 118. Hooper DC, Wolfson JS, Ng EY, Swartz MN. Mechanisms Clin Microbiol Infect Dis. 2010;29:1349-1353.
1983;147:751-757. of action of and resistance to ciprofloxacin. Am J Med. 144. Yamane K, Wachino J, Doi Y, et al. Global spread of ami-
91. Murray BE, Tsao J, Panida J. Enterococci from Bangkok, 1987;82:12-20. noglycoside resistance genes. Emerg Infect Dis. 2005;11:
Thailand, with high-level resistance to currently available 119. Gaffney DF, Cundliffe E, Foster TJ. Chloramphenicol resis- 951-953.
aminoglycosides. Antimicrob Agents Chemother. 1983;23: tance that does not involve chloramphenicol acetyltrans- 145. Doi Y, Arakawa Y. 16S ribosomal RNA methylation:
799-802. ferase encoded by plasmids from gram-negative bacteria. emerging resistance mechanism against aminoglycosides.
92. Zervos MJ, Kauffman CA, Therasse PM, et al. Nosocomial J Gen Microbiol. 1981;125:113-121. Clin Infect Dis. 2007;45:88-94.
infection by gentamicin-resistant Streptococcus faecalis: an 120. Bryan LE, Kwan S. Roles of ribosomal binding, membrane 146. Eliopoulos GM, Farber BF, Murray BE, et al. Ribosomal
epidemiologic study. Ann Intern Med. 1987;106:687-691. potential, and ehectron transport in bacterial uptake of resistance of clinical enterococcal isolates to streptomycin
93. Leclercq R. Enterococci aquire new kinds of resistance. streptomycin and gentamicin. Antimicrob Agents Che- isolates. Antimicrob Agents Chemother. 1984;25:398-399.
Clin Infect Dis. 1997;24(suppl 1):880-884. mother. 1983;23:835-845. 147. Ahmad MH, Rechenmacher A, Bock A. Interaction
94. Hoffmann SA, Moellering RC Jr. The enterococcus: 121. Mates SM, Eisenberg ES, Mandel LJ, et al. Membrane between aminoglycoside uptake and ribosomal resistance
“Putting the bug in our ears.” Ann Intern Med. 1987;106: potential and gentamicin uptake in Staphylococcus aureus. mutations. Antimicrob Agents Chemother. 1980;18:
757-761. Proc Natl Acad Sci U S A. 1982;79:6693-6697. 798-806.
95. Lyon BR, Skurray R. Antimicrobial resistance of Staphylo- 122. Rusthoven JJ, Davies TA, Lerner SA. Clinical isolation and 148. Hisanaga T, Hoban DJ, Zhanel GG. Mechanisms of resis-
coccus aureus: genetic basis. Microbiol Rev. 1987;51: characterization of aminoglycoside-resistant small colony tance to telithromycin in Streptococcus pneumoniae. J Anti-
88-134. variants of Enterobacter aerogenes. Am J Med. 1979;67: microb Chemother. 2005;56:447-450.
96. Ichino K, Ishikawa J, Ikeda Y, Hopta K. Characterization of 702-706. 149. Meka VG, Gold HS. Antimicrobial resistance to linezolid.
a bifunctional aminoglycoside modifying enzyme with 123. Musher DM, Baughn RE, Merrell GL. Selection of small- Clin Infect Dis. 2004;39:1010-1015.
novel substrate specificity and its gene from a clinical colony variants of Enterobacteriaceae by in vitro exposure 150. Kloss P, Xiong L, Shinabarger DL, Mankin AS. Resistance
isolate of methicillin-resistant Staphylococcus aureus with to aminoglycosides: pathogenicity for experimental mutations in 23 S rRNA identify the site of action of the
high arbekacin resistance. J Antibiot. 2004;57:679-686. animals. J Infect Dis. 1979;140:209-214. protein synthesis inhibitor linezolid in the ribosomal pep-
97. Ardia N, Sareyyupoglu B, Ozyurt M, et al. Investigation of 124. Funada H, Hattori KI, Kosakai N. Catalase-negative Esch- tidyl transferase center. J Mol Biol. 1999;294:93-101.
aminoglycoside modifying enzyme genes in methicillin- erichia coli isolated from blood. J Clin Microbiol. 1978;7: 151. Raad II, Hanna HA, Hachem RY, et al. Clinical-use-asso-
resistant staphylococci. Microbiol Res. 2006;161:49-56. 474-478. ciated decrease in susceptibility of vancomycin-resistant
98. Robicsek A, Strahilevitz J, Jacoby EA, et al. Fluoroquinolone- 125. Williams JB. Drug efflux as a mechanism of resistance. Br Enterococcus faecium to linezolid: a comparison with
modifying enzyme: a new adaptation of a common amino- J Biomed Sci. 1996;53:290-293. quinupristin-dalfopristin. Antimicrob Agents Chemother.
glycoside acetyltransferase. Nat Med. 2006;12:83-88. 126. McMurry L, Petrucci RE Jr, Levy SB. Active efflux of tetra- 2004;48:3583-3585.
99. Gaffney DF, Foster TJ, Shaw WV. Chloramphenicol acetyl cycline encoded by four genetically different tetracycline 152. Quiles-Melero I, Gomez-Gil R, Romero-Gomez MP, et al.
transferases determined by R-plasmids from gram negative resistance determinants in Escherichia coli. Proc Natl Acad Mechanisms of linezolid resistance among staphylococci in
bacteria. J Gen Microbiol. 1978;109:351-358. Sci U S A. 1980;77:3974-3977. a teriary hospital. J Clin Microbiol. 2013;51:998-1001.
100. Davies J. General mechanisms of antimicrobial resistance. 127. Levy SB, McMurry LM, Barbosa TM, et al. Nomenclature 153. Dutka-Malen S, LeClercq R, Coutant V, et al. Phenotypic
Rev Infect Dis. 1979;1:23-29. for new tetracycline resistance determinants. Antimicrob and genotypic heterogeneity of glycopeptide resistance
101. LeClercq R, Courvalin P. Resistance to macrolides, Agents Chemother. 1999;43:1523-1524. determinants in gram-positive bacteria. Antimicrob Agents
azalides, and streptogramins. In: Neu HC, Young LS, 128. Chopra I, Roberts M. Tetracycline antibiotics: mode of Chemother. 1990;34:1875-1879.
Zinner SH, eds. The New Macrolides, Azalides, and Strepto- action, applications, molecular biology, and epidemiology 154. McKessar SJ, Barry AM, Bell JM, et al. Genetic character-
gramins. New York: Marcel Dekker; 1993:33-40. of bacterial resistance. Microbiol Mol Biol Rev. 2001;65: ization of vanG, a novel vancomycin resistance locus for
102. Andremont A, Gerbaud G, Courvalin P. Plasmid-mediated 232-260. Enterococcus faecalis. Antimicrob Agents Chemother. 2000;
high level resistance to erythromycin in Escherichia coli. 129. Sutcliffe J, Tait-Kamradt A, Wandrack L. Streptococcus 44:3224-3228.
Antimicrob Agents Chemother. 1986;29:515-518. pneumoniae and Streptococcus pyogenes resistant to macro- 155. LeClercq R, Derlot E, Weber M, et al. Transferable vanco-
103. Brisson-Noel A, Delrieu P, Samain D, Courvalin P. Inacti- lide but sensitive to clindamycin: a common resistance mycin and teicoplanin resistance in Enterococcus faecium.
vation of lincosamanide antibiotics in Staphylococcus: iden- pattern made by an efflux system. Antimicrob Agents Che- Antimicrob Agents Chemother. 1989;33:10-15.
tification of lincosaminide O-nucleotidyltransferases and mother. 1996;40:1817-1824. 156. Shlaes DM, Bouvet A, Devine C, et al. Inducible, transfer-
comparison of the corresponding resistance genes. J Biol 130. Leclercq R. Mechanisms of resistance to macrolides and able resistance to vancomycin in Enterococcus faecalis
Chem. 1988;263:15880-15887. lincosamides: nature of the resistance elements and their A256. Antimicrob Agents Chemother. 1989;33:198-203.
104. Le Goffic F, Capmau ML, Abbe J, et al. Plasmid-mediated clinical implications. Clin Infect Dis. 2002;34:482-492. 157. Centers for Disease Control and Prevention. Vancomycin-
pristinamycin resistance: PH 1A, a pristinamycin 1A 131. Clancy J, Dib-Hajj F, Petitpas JW, Yuan W. Cloning and resistant Staphylococcus aureus—Pennsylvania, 2002.
hydrolase. Ann Microbiol. 1977;128:417-474. characterization of a novel macrolide efflux gene, mreA, MMWR Morb Mortal Wkly Rep. 2002;51:902.
105. Allignet J, Loncle V, Mazodier P, el Solh N. Nucleotide from Streptococcus agalactiae. Antimicrob Agents Che- 158. Nicas TI, Cole CT, Preston DA, et al. Activity of glycopep-
sequence of a staphylococcal plasmid gene, vgb, ancoding mother. 1997;41:2719-2723. tides against vancomycin-resistant gram-positive bacteria.
a hydrolase inactivating the B components of virginiamycin- 132. Levy SB. Active efflux, a common mechanism for biocide Antimicrob Agents Chemother. 1989;33:1477-1481.
like antibiotics. Plasmid. 1988;20:271-275. and antibiotic resistance. J Appl Microbiol. 2002;92(suppl): 159. Bugg TD, Wright GD, Dutka-Malen S, et al. Molecular
106. Speer BS, Bedzyj L, Salyers AA. Evidence that a novel tet- S65-S71. basis for vancomycin resistance in Enterococcus faecium
racycline resistance gene found on two Bacteroides trans- 133. Srikumar R, Li XZ, Poole K. Inner membrane efflux com- BM4147: biosynthesis of a depsipeptide peptidoglycan pre-
posons encodes an NADPH-oxidoreductase. J. Bacteriol. ponents are responsible for β-lactam specificity of multi- cursor by vancomycin resistance proteins VanH and VanA.
1991;173:176-183. drug efflux pumps in Pseudomonas aeruginosa. J Bacteriol. Biochemistry. 1991;30:10408-10415.
107. Fleming A. On the antibacterial action of cultures of a 1997;179:7875-7881. 160. LeClercq R, Dutka-Malen S, Brissonnoel A, et al. Resis-
penicillium, with special reference to their use in the isola- 134. Masuda N, Sakagawa E, Ohya S, et al. Contribution of the tance of enterococci to aminoglycosides and glycopeptides.
tion of B. influenzae. Br J Exp Pathol. 1929;10:226-236. Mex-X-MexY-oprM efflux system to intrinsic resistance in Clin Infect Dis. 1992;15:495-501.
108. Nikaido H. Role of permeability barriers in resistance to Pseudomonas aeruginosa. Antimicrob Agents Chemother. 161. Quintiliani R, Evers S, Courvalin P. The vanB gene confers
beta-lactam antibiotics. Pharmacol Ther. 1985;27:197-231. 2000;44:2242-2246. various levels of self-transferable resistance to vancomycin
109. Vaara M. Polymyxin B nonapeptide complexes with lipo- 135. Ghosh AS, Ahamed J, Chauhan KK, Kundu M. Involve- in enterococci. J Infect Dis. 1993;167:1220-1223.
polysaccharide [letter]. FEMS Microbiol Lett. 1983;18: ment of an efflux system in high-level fluoroquinolone 162. Fluit AC, Visser MR, Schmitz FJ. Molecular detection of
117-121. resistance of Shigella dysenteriae. Biochem Biophys Res antimicrobial resistance. Clin Microbiol Rev. 2001;14:
110. Nikaido H, Vaara M. Molecular basis of bacterial outer Commun. 1998;242:54-56. 836-871.
membrane permeability. Microbiol Rev. 1985;49:1-32. 136. Cohen SP, Hooper DC, Wolfson JS, et al. Endogenous 163. Schwalbe RS, Stapleton JT, Gilligan PH. Emergence of van-
111. Hasegawa Y, Yamada H, Mizushima S. Interactions of outer active efflux of norfloxacin in susceptible Escherichia coli. comycin resistance in coagulase-negative staphylococci. N
membrane proteins 0-8 and 0-9 with peptidoglycan sac- Antimicrob Agents Chemother. 1988;32:1187-1191. Engl J Med. 1987;316:927-931.
culus of Escherichia coli K-12. J Biochem (Tokyo). 1976;80: 137. Canton R, Morosini M-I. Emergence and spread of antibi- 164. Biavasco F, Vignaroli C, Lazzarini R, Varaldo PE. Glyco-
1401-1409. otic resistance following exposure to antbiotics. FEMS peptide susceptibility profiles of Staphylococcus haemolyti-
112. Yoshimura F, Nikaido H. Diffusion of beta-lactam antibiot- Microbiol Rev. 2011; 35:977-991. cus blood stream isolates. Antimicrob Agents Chemother.
ics through the porin channels of Escherichia coli K-12. 138. Corso A, Severina EP, Petruk VF, et al. Molecular charac- 2000;44:3122-3126.
Antimicrob Agents Chemother. 1985;27:84-92. terization of penicillin-resistant Streptococcus pneumoniae 165. Centers for Disease Control and Prevention. Reduced sus-
113. Goldstein FW, Gutmann L, Williamson R, et al. In vivo and isolates causing respiratory disease in the United States. ceptibility of Staphylococcus aureus to vancomycin—Japan,
in vitro emergence of simultaneous resistance to both beta- Microb Drug Resist. 1998;4:325-327. 1996. MMWR Morb Mortal Wkly Rep. 1997;46:624-635.
251.e3
166. Clark NC, Weigel LM, Patel JB, Tenover FC. Com- 188. Mendelman PM, Chaffin DO, Kalaitzoglou G. Penicillin- 210. Friedman L, Alder JD, Silverman JA. Genetic changes that
parison of Tn1546-like elements in vancomycin-resistant binding proteins and ampicillin resistance in Haemophilus correlate with reduced susceptibility to daptomycin in
Staphylococcus aureus isolates from Michigan and Penn- influenzae. J Antimicrob Chemother. 1990;25:525-534. Staphylococcus aureus. Antimicrob Agents Chemother.
sylvania. Antimicrob Agents Chemother. 2005;49:470- 189. Dougherty TJ. Genetic analysis and penicillin-binding 2006;50:2137-2145.

Chapter 18  Molecular Mechanisms of Antibiotic Resistance in Bacteria


472. protein alterations in Neisseria gonorrhoeae with chromo- 211. Arias CA, Panesso D, McGrath DM, et al. Genetic basis for
167. Courvalin P. Vancomycin resistance in gram-positive somally mediated resistance. Antimicrob Agents Che- in vivo daptomycin resistance in enterococci. N Engl J Med.
cocci. Clin Infect Dis. 2006;42(suppl 1):S25-S34. mother. 1986;30:649-652. 2011;365:892-900.
168. Geisel R, Schmitz FJ, Fluit AC, et al. Emergence, mecha- 190. Mendelman PM, Campos J, Chaffin DO, et al. Relative 212. Iredell J, Thomas L, Power D, Mendes E. Tigecycline resis-
nism, and clinical implications of reduced glycopeptide penicillin G resistance in Neisseria meningitidis and tance in Australian antibiotic-resistant gram-negative bac-
susceptibility in Staphylococcus aureus. Eur J Clin Microbiol reduced affinity of penicillin-binding protein 3. Antimicrob teria. J Antimicrob Chemother. 2007;59:816-818.
Infect Dis. 2001;20:685-697. Agents Chemother. 1988;32:706-709. 213. Peleg AY, Potoski BA, Rea R, et al. Acinetobacter baumannii
169. Cui L, Murakami H, Kuwahara-Arai K, et al. Contribution 191. Spratt BG, Zhang Q-Y, Jones DM, et al. Recruitment of a bloodstream infection while receiving tigecycline: a cau-
of a thickened cell wall and its glutamine nonamidated penicillin-binding protein gene from Neisseria flavescens tionary report. J Antimicrob Chemother. 2007;59:128-131.
component to the vancomycin resistance expressed by during the emergence of penicillin resistance in Neisseria 214. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ.
Staphylococcus aureus M450. Antimicrob Agents Che- meningitidis. Proc Natl Acad Sci U S A. 1989;86:8988- Quinupristin-dalfopristin resistance in gram-positive bac-
mother. 2000;44:2276-2285. 8992. teria: mechanism of resistance and epidemiology. Clin
170. Cui L, Iwamoto A, Lian JO, et al. Novel mechanism of 192. Faruki H, Kohmescher RN, McKinney WP, Sparling PF. A Infect Dis. 2004;38:92.
antibiotic resistance originating in vancomycin- community-based outbreak of infection with penicillin- 215. Campos MA, Vargas MA, Regueiro V, et al. Capsule poly-
intermediate Staphylococcus aureus. Antimicrob Agents resistant Neisseria gonorrhoeae not producing penicillinase saccharide mediates bacterial resistance to antimicrobial
Chemother. 2006;50:428-438. (chromosomally mediated resistance). N Engl J Med. 1985; peptides. Infect Immun. 2004;72:7107-7114.
171. Hospital Infection Control Practices Advisory Committee 313:607-611. 216. Kwon DH, Lu CD. Polyamines induce resistance to cationic
(HICPAC). Recommendations for preventing the spread of 193. Unemo M, Nicholas RA. Emergence of multidrug resistant, peptide, aminoglycoside, and quinolone antibiotics in
vancomycin resistance. Infect Control Hosp Epidemiol. extensively drug resistant and untreatable gonorrhea. Pseudomonas aeruginosa PAO1. Antimicrob Agents Che-
1995;16:105-113. Future Microbiol. 2012;7:1401-1422. mother. 2006;50:1615-1622.
172. Waxman DJ, Strominger JL. Penicillin-binding proteins 194. Mirelman D, Nuchamowitz Y, Rubinstein E. Insensitivity 217. Pamp SJ, Gjermansen M, Johansen HK, Tolker-Nielsen T.
and the mechanism of action of beta-lactam antibiotics. of peptidoglycan biosynthetic reactions to beta-lactam Tolerance to the antimicrobial peptide colistin in Pseudo-
Annu Rev Biochem. 1983;52:825-869. antibiotics in a clinical isolate of Pseudomonas aeruginosa. monas aeruginosa bioflims is linked to metabolically active
173. Malouin F, Bryan LE. Modification of penicillin-binding Antimicrob Agents Chemother. 1981;19:687-695. cells and depends upon pmr and metAB oprM genes. Mol
proteins as mechanisms of beta-lactam resistance. Antimi- 195. Parr TR Jr, Bryan LE. Mechanism of resistance of Microbiol. 2008;68:223-240.
crob Agents Chemother. 1986;30:1-5. an ampicillin-resistant, beta-lactamase-negative clinical 218. van der Wouden EJ, Thijs JC, Kusters JG, et al. Mechanism
174. Williamson R. Resistance of Clostridium perfringens to isolate of Haemophilus influenzae type b to beta-lactam and clinical significance of metronidazole resistance in
beta-lactam antibiotics mediated by a decreased affinity of antibiotics. Antimicrob Agents Chemother. 1984;25: Helicobacter pylori. Scand J Gastroent Suppl. 2001;234:
a single essential penicillin-binding protein. J Gen Micro- 747-753. 10-15.
biol. 1983;129:2339-2342. 196. Wolfson JS, Hooper DC. The fluoroquinolones: structures, 219. Deplano A, Denis O, Poirel L, et al. Molecular characteriza-
175. Giles AF, Reynolds RE. Bacillus megaterium resistance to mechanisms of action and resistance, and spectra of activ- tion of an epidemic clone of panantibiotic-resistant Pseu-
cloxacillin accompanied by a compensatory change in ity in vitro. Antimicrob Agents Chemother. 1985;28: domonas aeruginosa. J Clin Microbiol. 2005;43:1198-2004.
penicillin binding proteins. Nature. 1979;280:167-168. 581-586. 220. Poole K. Outer membranes and efflux: the path to multi-
176. Hakenbeck R, Tarpay M, Tomasz A. Multiple changes of 197. Robillard NJ, Scarpa AL. Genetic and physiological char- drug resistance in gram-negative bacteria. Curr Pharm
penicillin-binding proteins in penicillin-resistant clinical acterization of ciprofloxacin resistance in Pseudomonas Biotechnol. 2002;3:77-98.
isolates of Streptococcus pneumoniae. Antimicrob Agents aeruginosa PAO. Antimicrob Agents Chemother. 1988;32: 221. Piddock L. Clinically relevant chromosomally encoded
Chemother. 1980;17:364-371. 535-539. multidrug resistance efflux pumps in bacteria. Clin Micro-
177. Spratt BG, Dowson CG, Zhang Q-Y, et al. Mosaic genes, 198. Schmitz FJ, Higgins P, Meyer S, et al. Activity of quinolones biol Rev. 2006;19:382-402.
hybrid penicillin-binding proteins, and the origins of peni- against gram-positive cocci: mechanisms of drug action 222. Lin J, Sahin O, Michael LO, Zhang Q. Critical role of mul-
cillin resistance in Neisseria meningitidis and Streptococcus and bacterial resistance. Eur J Clin Microbiol Infect Dis. tidrug efflux pump CmeABC in bile resistance and in vivo
pneumoniae. In: Pardee A, Campisi J, eds. Perspectives on 2002;21:647-659. colonization of Campylobacter jejuni. Infect Immun. 2003;
Cellular Regulation: From Bacteria to Cancer. New York: 199. Aoyama H, Fujimaki K, Sato K, et al. Clinical isolate of 71:4250-4259.
Wiley-Liss; 1991:73-83. Citrobacter freundii highly resistant to new quinolones. 223. Hirakata Y, Srikumar R, Poole K, et al. Multidrug efflux
178. Hartman BJ, Tomasz A. Low-affinity penicillin-binding Antimicrob Agents Chemother. 1988;32:922-924. systems play an important role in the invasiveness of Pseu-
protein associated with beta-lactam resistance in Staphylo- 200. Martinez-Martinez L, Pascual A, Jacoby GA. Quinolone domonas aeruginosa. J Exp Med. 2002;196:109-118.
coccus aureus. J Bacteriol. 1984;158:513-516. resistance from a transferable plasmid. Lancet. 1998;351: 224. Rice LB. Tn916 family of conjugative transposons and dis-
179. Ubukata K, Yamashita N, Konno M. Occurrence of a beta- 797-799. semination of antimicrobial resistance determinants. Anti-
lactam-inducible penicillin-binding protein in methicillin- 201. Luo Y, Li J, Meng Y, et al. Joint effects of topoisomerase microb Agents Chemother. 1998;42:1871-1877.
resistant staphylococci. Antimicrob Agents Chemother. alteration and plasmid-mediated, quinolone-resistant 225. Boyd DA, Tyler S, Christianson S, et al. Complete nucleo-
1985;27:851-857. determinants in Salmonella enterica Typhimurium. Microb tide sequence of a 92-kilobase plasmid harboring the
180. Fontana R. Penicillin-binding proteins and the intrinsic Drug Resist. 2011;17:1-5. CTX-15 extended-spectrum beta-lactamase involved in an
resistance to beta-lactams in gram-positive cocci. J Antimi- 202. Enne VI, King A, Livermore DM, Hall LMC. Sulfonamide outbreak in long-term-care facilities in Toronto, Canada.
crob Chemother. 1985;16:412-416. resistance in Haemophilus influenzae mediated by acquisi- Antimicrob Agents Chemother. 2004;48:3758-3764
181. Fontana R, Grossato A, Rossi L, et al. Transition from resis- tion of sul2 or a short insertion in chromosomal folP. Anti- 226. Bennet PM. Plasmid encoded antibiotic resistance: acquisi-
tance to hypersusceptibility to beta-lactam antibiotics microb Agents Chemother. 2002;46:1934-1939. tion and transfer of antibiotic resistance genes in bacteria.
associated with loss of a low-affinity penicillin-binding 203. Huovinen P. Trimethoprim resistance. Antimicrob Agents Br J Pharmacol. 2008;153: S347-S357.
protein in a Streptococcus faecium mutant highly resistant Chemother. 1987;31:1451-1456. 227. Whitney CG, Farley MM, Hadler J, et al. Increasing preva-
to penicillin. Antimicrob Agents Chemother. 1985;28: 204. Steen R, Skold O. Plasmid-borne or chromosomally medi- lence of multidrug-resistant Streptococcus pneumoniae in
678-683. ated resistance by Tn7 is the most common response to the United States. N Engl J Med. 2000;343:1917-1924.
182. Farber BF, Eliopoulos GM, Ward JI, et al. Multiply resistant ubiquitous use of trimethoprim. Antimicrob Agents Che- 228. Rainey PB, Moxon ER. When being hyper keeps you fit.
viridans streptococci: susceptibility to beta-lactam antibi- mother. 1985;27:933-937. Science. 2000;288:1186-1187.
otics and comparison of penicillin-binding protein pat- 205. Maskell R, Okubadejo OA, Payne RH, Pead L. Human 229. Tompkins JD, Nelson JL, Hazel JC, et al. Error-prone poly-
terns. Antimicrob Agents Chemother. 1977;3(suppl C): infections with thymine-requiring bacteria. J Med Micro- merase, DNA polymerase IV, is responsible for transient
47-51. biol. 1978;11:33-45. hypermutation during adaptive mutation in Escherichia
183. Lambert PA. Bacterial resistance to antibiotics: modified 206. Zhu W, Tenover FC, Limor J, et al. Use of pyrosequencing coli. J Bacteriol. 2003;185:3469-3472.
target sites. Adv Drug Deliv Rev. 2005;57:1471-1485. to identify point mutations in domain V of 23S rRNA genes 230. White DG, Shao S, Sudler R, et al. The isolation of
184. Sabath LD. Chemical and physical factors influencing of linezolid-resistant Staphylococcus aureus and Staphylo- antibiotic-resistant Salmonella from retail ground meats. N
methicillin resistance of Staphylococcus aureus and Staphy- coccus epidermidis. Eur J Clin Microbiol Infect Dis. 2006; Engl J Med. 2001;345:1147-1154.
lococcus epidermidis. J Antimicrob Chemother. 1977;3 26:161-165. 231. Sørenson TL, Blom M, Monnet DL, et al. Transient intes-
(suppl C):47-51. 207. Escribano I, Rodríguez JC, Llorca B, et al. Importance of tinal carriage after ingestion of antibiotic-resistant Entero-
185. Song MD, Wachi M, Doi M, et al. Evolution of an inducible the efflux pump systems in the resistance of Mycobacterium coccus faecium from chicken and pork. N Engl J Med. 2001;
penicillin-target protein in methicillin-resistant Staphylo- tuberculosis to fluoroquinolones and linezolid. Chemo- 345:1161-1166.
coccus aureus by gene fusion. FEBS Lett. 1987;221: therapy. 2007;53:397-401. 232. McDonald LC, Rossiter S, Mackinson C, et al. Quinupristin-
167-171. 208. Longzhu C, Tominaga E, Neoh H, Hiramatsu K. Correla- dalfopristin-resistant Enterococcus faecium on chicken and
186. Berger-Bachi B, Barberis-Maino L, Strassle A, Kayser FH. tion between reduced daptomycin susceptibility and in human stool specimens. N Engl J Med. 2001;345:
FemA, a host-mediated factor essential for methicillin vancomycin resistance in vancomycin-intermediate Staph- 1155-1160.
resistance in Staphylococcus aureus: molecular cloning and ylococcus aureus. Antimicrob Agents Chemother. 2006;50: 233. Carlet J, Collignon P, Goldman D, et al. Society’s failure to
characterization. Mol Gen Genet. 1989;219:263-269. 1079-1082. protect a precious resource: antibiotics. Lancet. 2011;378:
187. Dougherty TJ, Koller AE, Tomasz A. Penicillin-binding 209. Sakoulas G, Alder J, Thauvin-Eliopoulos C, et al. Induction 369-371.
proteins of penicillin-susceptible and intrinsically resistant of daptomycin heterogeneous susceptibility in Staphylococ- 234. McGowan JE Jr, Tenover FC. Control of antimicrobial
Neisseria gonorrhoeae. Antimicrob Agents Chemother. 1980; cus aureus by exposure to vancomycin. Antimicrob Agents resistance in the health care system. Infect Dis Clin North
18:730-737. Chemother. 2006;50:1581-1585. Am. 1997;11:297-311.

You might also like