You are on page 1of 9

b-Lactam antibiotic resistance: a current structural perspective

Mark S Wilke1, Andrew L Lovering1 and Natalie CJ Strynadka

Bacterial resistance to b-lactam antibiotics can be achieved by wall) for the preservation of cell shape and rigidity. This
any of three strategies: the production of b-lactam-hydrolyzing cell wall is comprised of a basic repeating unit of an
b-lactamase enzymes, the utilization of b-lactam-insensitive alternating disaccharide — N-acetyl glucosamine and
cell wall transpeptidases, and the active expulsion of b-lactam N-acetyl muramic acid. The latter sugar in this disacchar-
molecules from Gram-negative cells by way of efflux pumps. In ide is modified by a characteristic pentapeptide. This
recent years, structural biology has contributed significantly to varies amongst the Gram-negative and Gram-positive
the understanding of these processes and should prove species, but always terminates in two D-alanine residues.
invaluable in the design of drugs to combat b-lactam resistance The individual peptidoglycan units are produced inside
in the future. the cell, but their final cross-linking is catalyzed outside
the cytoplasmic membrane by a group of membrane-
Addresses anchored bacterial enzymes known as the cell-wall trans-
Department of Biochemistry and Molecular Biology, and the Center peptidases. In this cross-linking reaction, a peptide bond
for Blood Research, University of British Columbia, 2146 Health is formed between the penultimate D-alanine on one
Sciences Mall, Vancouver, BC, Canada V6T 1Z3
chain and the free amino end of a diamino pimelic acid
Corresponding author: Strynadka, Natalie CJ (Gram-negative) or an L-lysine (Gram-positive) residue
(natalie@byron.biochem.ubc.ca) on the other chain. The linkage is formed with the
1
Mark S Wilke and Andrew L Lovering contributed equally to this review. penultimate D-alanine, causing the terminal D-alanine
to be cleaved in the process.
Current Opinion in Microbiology 2005, 8:525–533
Transpeptidase enzymes utilize an active site serine and
This review comes from a themed issue on perform their catalytic cycle by way of an acylation/
Antimicrobials deacylation pathway. b-lactam antibiotics efficiently inhi-
Edited by Christopher Walsh and Malcolm GP Page
bit the bacterial transpeptidases, therefore these enzymes
Available online 29th August 2005 are often termed penicillin binding proteins or PBPs.
They are able to do this owing to the stereochemical
1369-5274/$ – see front matter similarity of the b-lactam moiety with the D-alanine–D-
# 2005 Elsevier Ltd. All rights reserved.
alanine substrate. In the presence of the antibiotic, the
DOI 10.1016/j.mib.2005.08.016 transpeptidases form a lethal covalent penicilloyl–
enzyme complex that serves to block the normal trans-
peptidation reaction. This results in weakly cross-linked
peptidoglycan, which makes the growing bacteria highly
Introduction susceptible to cell lysis and death.
The introduction of b-lactam antibiotics into the health
care system in the latter stages of World War II represents As with most antimicrobial agents, b-lactams are rendered
one of the most important contributions to medical inactive against bacteria by way of three primary mechan-
science in recent history. Today, b-lactams remain the isms of resistance (Figure 1). The most common mechan-
most widely utilized antibiotics owing to their compara- ism is the production of enzymes that degrade or modify
tively high effectiveness, low cost, ease of delivery and the antibiotic before it can reach the appropriate target
minimal side effects. b-lactams target transpeptidase site. In this case, the b-lactamase family of enzymes
enzymes that synthesize the bacterial cell wall. The degrade b-lactam antibiotics and are found widely dis-
desirable attributes of this class of antibiotic arise from seminated amongst Gram-positive and Gram-negative
the facts that these enzymes are localized to the outer bacteria. The second mechanism is alteration of the
leaflet of the bacterial cytoplasmic membrane (i.e. are antibiotic target site. In this case, the b-lactam-resistant
relatively accessible) and that they are specific to bacteria cell-wall transpeptidases perform this role; this is now a
(with no functional or structural counterpart in the human major cause of resistance in several pathogens including
host). In a practical sense, the low cost of production of the problematic Gram-positive Staphylococcal and Strepto-
b-lactam antibiotics allows for a wide availability; thus, it coccal species. The final mechanism is prevention of
is imperative that we preserve the power of this valuable access of the antibiotic to the target by way of altered
clinical resource. permeability or forced efflux. For example, this can be
performed by the MexA,B–OprM antibiotic efflux pump,
How do b-lactam antibiotics work? Bacteria of all species which is a major cause of resistance in Pseudomonas and in
rely on a heavily cross-linked peptidoglycan layer (cell other pathogenic Gram-negative species.

www.sciencedirect.com Current Opinion in Microbiology 2005, 8:525–533


526 Antimicrobials

Figure 1

Current Opinion in Microbiology 2005, 8:525–533 www.sciencedirect.com


b-Lactam antibiotic resistance Wilke, Lovering and Strynadka 527

In this review we describe seminal new discoveries in Classes B1 and B3 are able to bind one or two zinc ions [3],
which structural biology has provided new insights into whereas the class B2 enzymes appear to be mononuclear
these b-lactam antibiotic resistance phenomena as well as [4]. In the binuclear metallo-b-lactamases, the zinc ions
new strategies for drug development. are proximal to each other and are separated by a bridging
hydroxide that has been proposed to be the attacking
Antibiotic-modifying enzymes: nucleophile in b-lactam hydrolysis. The class B1 and B3
the b-lactamases metallo-b-lactamases can also function as mononuclear
The b-lactamases confer significant antibiotic resistance enzymes, in which a single zinc ion (that occupies the Zn1
to their bacterial hosts by hydrolysis of the amide bond of site) coordinates the nucleophilic hydroxide; this
the four-membered b-lactam ring. These enzymes are mechanism has been proposed to predominate in the
especially important in Gram-negative bacteria as they presence of substrate under physiological conditions
constitute the major defense mechanism against b-lac- [5]. The crystal structure of a class B2 metallo-b-lacta-
tam-based drugs. The spread of b-lactamase genes has mase (CphA from Aeromonas hydrophilia) has only been
been greatly exacerbated by their integration within published recently [4]. Its proposed catalytic mechan-
mobile genetic elements, such as plasmids or transposons, ism differs from the class B1 and B3 mononuclear
which facilitate the rapid transfer of genetic material mechanisms in that the zinc ion occupies the Zn2 site,
between microbes. Even more ominous is the organiza- a general base activates the nucleophilic water, and the
tion of b-lactamase genes within integrons as part of zinc ion forms a bond with the amine nitrogen of the
multi-drug resistance cassettes that bestow mechanisms hydrolyzed b-lactam amide (Figure 2).
for resistance not only to b-lactams but also to other
antibiotic classes such as aminoglycosides, macrolides, A new generation of b-lactamases
sulphonamides and chloramphenicol (for a recent review, The b-lactamases are ancient enzymes that were rela-
see [1]). Once expressed, b-lactamases are secreted into tively rare until b-lactam antibiotics were introduced into
the periplasmic space (in Gram-negative bacteria), bound medicine and agriculture half a century ago [6]. The
to the cytoplasmic membrane, or excreted (in Gram- widespread use of carbapenems, the monobactam aztreo-
positive bacteria). nam, cephamycins and oxyimino-cephalosporins in the
past few decades has led to the evolution of a new
Structure and mechanism generation of b-lactamases, which have an extended sub-
The >470 b-lactamases known to date [2] are typically strate spectrum (i.e. extended-spectrum b-lactamases or
organized into four classes (A to D) on the basis of ESBLs), as well as the development of novel carbapene-
sequence similarity. Crystal structures are currently avail- mases and plasmid-mediated AmpC b-lactamases (for
able for representatives of each class (for a recent review, recent reviews, see [2,7,8]). Common ESBLs include
see [2]). Classes A, C and D share a similar fold and all varieties from the class A b-lactamases TEM, SHV and
have a mechanism that involves creation of a serine CTX-M and the class D b-lactamase OXA. These
nucleophile by deprotonation of an active site serine with enzymes are typified by a broad substrate spectrum that
a general base, nucleophilic attack of the b-lactam ring to includes oxyimino-cephalosporins, aztreonam and, in the
form an acyl-enzyme intermediate, and hydrolysis of the case of some OXA and CTX-M enzymes, cefepime.
intermediate using a general base activated water mole-
cule. The differences between the catalytic mechanisms Recently, several CTX-M structures have been made
of the serine b-lactamase classes center around the type of available including inhibitor-bound structures, which pro-
general base residues used in acylation and deacylation. vide snapshots of two reaction cycle transition states, and
the acyl-enzyme intermediate, which can aid in the
The class B b-lactamases are zinc metalloenzymes and design of inhibitors [9] (Figure 3). In addition, several
are completely distinct from the serine b-lactamases in atomic resolution CTX-M structures demonstrate that
terms of sequence, fold and mechanism. There are three the enhanced ceftazidimase activity of these enzymes is a
subclasses of class B metallo-b-lactamases (B1 to B3). result of the increased active site flexibility; however, this

(Figure 1 Legend) Structural depiction of proteins involved in b-lactam resistance. The proteins responsible for resistance are sub-divided
into the b-lactamase, PBP and efflux pump groups. Also shown are two of the repressors for resistance operons — BlaI and MexR (genes are
shown as solid arrows). The structures of the glycosyltransferase domain of the class A PBPs and the cytoplasmic protease domain of BlaR that
are not yet determined are replaced by blue and green solid shapes, respectively. The diagram also shows the interaction of b-lactams with
these subgroups — the b-lactam-dependent signaling of BlaR, ring hydrolysis by the b-lactamases, acylation of the PBPs (poor in PBP2x and 2a),
and proton antiport of the antibiotics by the efflux pump systems. The recently solved structure of S. pneumoniae PBP1b is used in place of the
closely-related resistance determinant PBP1a, and the structures of MexB and OprM are used here as models derived from the co-ordinates
of their respective homologues, AcrB and TolC (docked together manually). Details of the precise interaction of MexA with MexB–OprM remain
unknown, as does the nature of the effector responsible for terminating MexR repression. Abbreviations: gm +/ , Gram-positive/negative; IM,
inner membrane; OM, outer membrane; PG, peptidoglycan. CTXM9 and CphA are shown as representatives of the >40 available serine and
metallo-b-lactamase structures.

www.sciencedirect.com Current Opinion in Microbiology 2005, 8:525–533


528 Antimicrobials

Figure 2 Figure 3

The fold and active site of cefoxitin-acylated CTXM9 (PDB code 1YMX).
(a) Ribbon representation of the CTXM9 fold. (b) Stick representation of
several active site residues, colored by atom type (C atoms, yellow; O,
red; N, blue; and S, orange). The covalently-bound cefoxitin is shown
with purple carbon atoms to distinguish it from the protein carbons
The fold and active site of CphA bound by the substrate biapenem
displayed in yellow. An active site water molecule is indicated with a
(PDB code 1X8). (a) Ribbon representation of the CphA fold. (b) Stick
cyan sphere.
representation of several active site residues, colored by atom type
(C atoms, yellow; O, red; N, blue; and S, orange). Biapenem is
displayed with purple carbon atoms to distinguish it from the protein
carbons shown in yellow. A single catalytic zinc and a water molecule have successfully restored the antibiotic activity of
are shown as dark grey and cyan spheres, respectively. Hydrogen b-lactam drugs against resistant pathogens. Although
bonds are represented by green dashes.
outside the scope of this review, several novel compounds
have been reported in the past two years that have
increase in flexibility is at the cost of protein stability [10]. inhibitory activity against class A, B and C b-lactamases;
Carbapenemases are derived from classes A, B and D and these molecules include peptides [11], succinic acid
they provide resistance to carbapenems as well as to derivatives [12], substituted penam sulfones [13], mer-
oxyimino-cephalosporins and cephamycins. The class B captomethyl-penicillinates [14], bridged bicyclic octa-
metallo-b-lactamase CphA (mentioned above) is a carba- nones [15], thiophene-carboxy derivatives [16], and
penamase. Its crystal structure in complex with the car- tricylclic 6-methylidene penems [17].
bapenem substrate biapenem (Figure 2) has been
determined; this might prove useful in the design of Regulation of resistance
inhibitors or of non-hydrolyzable antibiotics [4]. b-lactamase expression is often induced by b-lactams
through a novel regulation system that consists of the
Historically, combination therapies in which the action of repressor BlaI and the receptor BlaR (Figure 1). A com-
b-lactams is supplemented with b-lactamase inhibitors plete description of the induction process is currently

Current Opinion in Microbiology 2005, 8:525–533 www.sciencedirect.com


b-Lactam antibiotic resistance Wilke, Lovering and Strynadka 529

unavailable, but recent determination of the structures of Altered antibiotic targets: the cell wall
BlaI [18,19] and the extracellular PBP-like sensor transpeptidases (PBPs)
domain of BlaR [20–22] have characterized important PBPs are divided into two subgroups: low molecular
features of the b-lactamase regulation machinery. mass (LMM) and high molecular mass (HMM) enzymes.
The HMM enzymes are further subdivided into the
BlaR is a transmembrane protein composed of an extra- bifunctional class A enzymes (which possess a b-lac-
cellular sensor domain that is acylated by b-lactam anti- tam-insensitive transglycosylase domain and the tradi-
biotics, a transmembrane domain (consisting of four tional b-lactam-sensitive transpeptidase domain) and the
membrane-spanning helices) that transduces the b-lac- monofunctional transpeptidase class B enzymes. The
tam-binding signal across the membrane, and an intra- soluble LMM PBPs have no identified role in b-lactam
cellular zymogenic zinc metalloprotease domain that is resistance and are primarily involved in carboxypeptidase
activated by autoproteolysis upon acylation of the sensor reactions and peptidoglycan trimming. Despite their low
domain. Interestingly, the b-lactam sensor domain of sequence identity with the more essential HMM counter-
BlaR resembles the b-lactamases in terms of its fold parts, the LMM PBPs have been a common vehicle for
(especially the class D b-lactamases) as well as its use inhibitor studies owing to their soluble nature. Inhibitors
of a serine nucleophile to form an acyl-enzyme inter- that have been most recently used include peptidoglycan-
mediate. However, unlike the b-lactamases and akin to mimetic b-lactams and b-sultams [27,28]. By contrast,
the PBPs, the covalent b-lactam adduct is stabilized as it inhibitor studies of the membrane-anchored HMM PBPs
appears to lack a functional general base residue capable have been hindered by their under-representation in the
of deacylation. No significant conformational changes Protein Data Bank (PDB) structural database, primarily
accompany acylation of the sensor domain [21] and it owing to the difficulty of working with membrane pro-
has been proposed that the large extracellular BlaR loop teins and the scarce lipid II substrate. However, struc-
designated L2 that is positioned adjacent to the sensor tures of the major determinants in class B PBP-associated
domain in the BlaR receptor might mediate the b-lactam- b-lactam resistance have recently become available to
binding signal to the transmembrane domain through an increase understanding of these phenomena, for example,
interaction with the BlaR sensor [23]. PBP2x from the penicillin-resistant Streptococcus pneumo-
niae (PRSP) [29–31], PBP2a from MRSA [32] and
The cytosolic repressor BlaI forms a dimer and binds an PBP5fm from the naturally resistant Enterococcus faecium
operator in the bla divergon that encodes BlaI, BlaR and the [33]. The transpeptidase domain of the class A S. pneu-
b-lactamase structural gene. Once activated, the BlaR moniae PBP1b has also been solved [34] and might
metalloprotease domain elicits cleavage of BlaI within provide a basis on which to model mutations in the
its dimerization domain, which prevents operator binding resistant PBP1a enzyme.
and permits b-lactamase expression. The BlaI monomer
consists of an amino-terminal winged helix DNA-binding There are several PBP-mediated mechanisms of b-lactam
domain and a carboxy-terminal dimerization domain, resistance, including: acquisition of a ‘new’ less-sensitive
which are separated by a flexible hinge region. This flex- enzyme, mutation of an endogenous PBP to lessen the
ibility allows the BlaI dimer to not only bind the bla reaction with b-lactams (while maintaining some trans-
operator but bind the operator sequence for the mec diver- peptidase activity), or upregulation of PBP expression.
gon as well, allowing BlaR and BlaI to additionally mediate The latter option appears to be the least effective,
the expression of PBP2a (the protein encoded by mecA) although the converse (lowering expression of some
[19]. PBP2a expression in methicillin-resistant Staphylo- resistant PBPs) might prove to be useful in the control
coccus aureus (MRSA) is controlled by a highly similar of b-lactam-resistant bacteria [35].
regulation system that consists of the repressor MecI and
receptor MecR. Although many MRSA strains have MecI PBP2x structures
deletions that inactivate PBP2a repression, these strains The mutation of PBP2x in PRSP has been studied
appear to be co-regulated by BlaR–BlaI [24]. Indeed, the extensively [29–31] (Figure 4). Resistance occurs in a
bla or mec regulatory genes appear to be required for the mosaic pattern, with many mutations occurring in differ-
maintenance and expression of mecA, which suggests that ent clinical isolates. This can cause problems in isolation
PBP2a expression incurs a fitness cost on the host [25]. of the main determinants of resistance and also presents
Therefore, the bla and mec regulation machinery might the requirement for screening crystallization conditions
prove useful as targets for the design of inhibitors to counter anew. The structures of PBP2x from several resistant
b-lactamase and PBP2a-mediated resistance, particularly if strains have been determined [29–31].
further structural details are forthcoming. Alternatively,
the creation of non-b-lactam lead compounds that circum- Strain sp328 harbors the most clinically important muta-
vent the activation of such regulatory mechanisms are a tion, T338A, which was found to result in the loss of an
logical step in inhibitor design, as highlighted recently by important active site water molecule, thus weakening the
Tondi et al. [16] and Zervosen et al. [26]. hydrogen bonding network that stabilizes the acyl-

www.sciencedirect.com Current Opinion in Microbiology 2005, 8:525–533


530 Antimicrobials

Figure 4
enzyme complex [31]. The S389L and N514H mutations
that are also present in this strain were found to sterically
hinder favorable interactions with the b-lactam, reducing
the acylation rate. An additional mutation, M339F, con-
fers higher-level resistance to strains that possess the
T338A mutation. The structure of this variant was found
to re-orientate the S337 nucleophile and lower the reac-
tion rate by 4–10-fold [29].

PBP2x from the PRSP strain sp5259 has also been struc-
turally characterized [30] and appears to offer an alter-
native mechanism of resistance that shares features with
other class B enzymes. The mutation of Q552 to gluta-
mate introduces a negative charge near the edge of the
active site; this might act like a similarly positioned
residue in PBP5fm, disfavoring interaction with nega-
tively charged b-lactams. Other mutations in this strain
act in a similar way to residues from PBP2a, altering the
conformation of b-strand 3 so that an energetically costly
rearrangement is required for acylation. However, the
observation that resistant class B PBPs share these fea-
tures might not assist in the development of new broad-
spectrum antibiotics. Molecular dynamics simulations on
a group of enzymes that share the transpeptidase fold
raised the possibility that there are different mechanisms
of acylation dependent on the particular combination of
enzyme and antibiotic [36]; this leaves bacteria many
possibilities to develop resistance against new b-lactams.

Other PBP structures


It is also prudent to note that PBP1a and PBP2b are
involved in PRSP resistance. Sanbongi et al. [37]
sequenced six S. pneumoniae PBPs, all of which are pre-
sent in 40 clinical isolates, and thereby confirmed the
correlation of mutations in these proteins to b-lactam
minimum inhibitory concentrations (MICs). Mutations
in PBP2b resulted in higher MICs for penicillins and
carbapenems, whereas MIC increases for cephalosporins
were associated with PBP2x. Alteration of the class A
PBP1a conferred additional resistance to strains already
bearing mutations in PBPs 2b and 2x.

PBP2a of MRSA is encoded by the mecA gene, which is


believed to have arisen as a result of horizontal transfer
from an undisclosed species. When challenged with
b-lactams, MRSA will utilize the transglycosylase activity
of PBP2 (the only class A enzyme of S. aureus) and the
transpeptidase functionality of PBP2a to synthesize the
The fold and active site of S. pneumoniae PBP2x. (a) Surface cell wall. It has recently been shown that PBP2 is able to
representation overlaid with protein fold. The transpeptidase domain is
mutate to a resistant form in the laboratory [38], but
colored red and the other domains blue. (b) Detail of active site topology.
This shows the S337 nucleophile and residues responsible for the thankfully it is not responsible for the emergence of a
development of resistance to b-lactams. Residues are represented in second alternate form of MRSA in the environment. The
stick format and are colored according to atom type (C atoms, yellow; structure of PBP2a [32] suggested that the poor acylation
O, red; N, blue; and S, orange). The protein co-ordinates used are by b-lactams was caused by a b-strand 3 alteration, and a
from PDB 1RP5, with a molecule of cefuroxime (C atoms, yellow in
[a], magenta in [b]) placed in an equivalent position to that observed in
route to more effective antibiotics could be to increase the
the complex from PDB 1QMF. Note that because 1RP5 is used, the length of the b-lactam compound to improve non-cova-
native amino acid Q552 is mutated to a glutamate residue. lent interactions. Several such new compounds have been

Current Opinion in Microbiology 2005, 8:525–533 www.sciencedirect.com


b-Lactam antibiotic resistance Wilke, Lovering and Strynadka 531

described at length by Bush et al. [39], including tics that target the essential transglycosylase domain.
BAL9141, which is active against both PRSP and MRSA. Such inhibitors would undoubtedly work well in combi-
The use of specific chemical groups to mimic the differ- nation therapy with b-lactams, as shown by the essential
ences in peptidoglycan sub-structure between various nature of PBP2 and PBP2a co-operation in MRSA strains.
bacterial pathogens, or indeed to facilitate the conforma-
tional changes required for acylation, might also prove Altered antibiotic permeability and efflux:
fruitful, as suggested recently by Fuda et al. [40]. the Gram-negative efflux pumps
With the exception of some strains of the Streptococci,
In contrast to S. aureus and S. pneumoniae, the bacterium Enterococci and Staphylococci ‘superbugs’, Gram-negative
E. faecium is naturally resistant to b-lactam antibiotics. bacteria are generally more resistant to a large variety of
The PBP responsible for this resistance, PBP5fm, has antibiotics and chemotherapeutic agents than are Gram-
been structurally characterized [33], although no co-ordi- positive bacteria. It is now recognized that a major con-
nates for this enzyme are available in the PDB at present. tribution to antibiotic resistance in Gram-negative species
The reason for the endogenous low-level affinity for is the presence of broad-specificity drug-efflux pumps.
b-lactams in E. faecium isn’t immediately apparent, but
could be owing to the reduced active site accessibility and One of the best-characterized of these is the drug efflux
also to charge repulsion with the b-lactam carboxylate system MexAB–OprM (recently reviewed in [43]) of the
group. E. faecium can demonstrate higher-level resistance opportunistic pathogen, Pseudomonas aeruginosa. This tri-
by mutation of PBP5fm [41], including an insertion after partite pump (composed of the inner membrane RND
S466 that is present in a loop that shifts upon acylation. transporter ‘pump’ MexB, the outer membrane porin
OprM, and the soluble periplasmic MexA) acts on a wide
Bifunctional PBPs range of antibiotics, including tetracycline, chloramphe-
The first crystal structure of a transpeptidase domain from nicol, quinilones, novobiocin, macrolides and trimetho-
a class A enzyme — PBP1b from S. pneumoniae — has prim, as well as b-lactams and b-lactamase inhibitors such
recently been reported [34]. The role of PBP1b in as clavulanic acid. The past four years have seen a
b-lactam resistance is minor at best, but its structure tremendous increase in our understanding of the struc-
can be used as a model for mutational effects in the tural features of the individual components of the tripar-
resistant PBP1a enzyme (45% sequence identity between tite efflux pumps (for a recent review see [44]); the
the transpeptidase region of PBP1a and PBP1b). It is also orthologous outermembrane porin and innermembrane
noted that crystallization conditions for PBP1a have been pump components TolC and AcrB from E. coli have been
reported [42]. Like PBPs 2x, 2a and 5fm, the enzyme determined to 2.1 and 3.5 Å resolution, respectively, and
possesses a classical transpeptidase domain, which is the periplasmic component MexA from Pseudomonas aer-
flanked by regions of unknown function that might play uginosa to 3.5 Å resolution. A structure of the inner
a role in association with other cell wall modifying pro- membrane pump AcrB in the presence of several hydro-
teins. The postulation of multi-enzyme complexes for phobic small molecule compounds has also been deter-
HMM PBPs is particularly interesting when applied to mined [45], which implies a diverse binding mode for
this structure as the active site appears ‘closed’ and individual ligands, at least in this component of the efflux
activity might be regulated by interaction with other pump. Although these structures have provided a tre-
proteins. One consequence of open and closed active mendous new level of understanding of the distinct
sites in these enzymes is that resistant PBPs are expected architecture of the three proteins that make up these
to favor open conformations, allowing transpeptidation of pumps, there are still many unanswered questions with
the bulkier substrates that result from inhibition of the regard to the way in which these components interact to
LMM PBP carboxypeptidases by b-lactams. The resis- form a single path for extruded antibiotic ligands. These
tance-conferring mutation of T371S/A in PBP1a is ana- systems represent logical targets for novel antibiotic
logous to that of T338A described for PBP2x. Other design, and development of lead compounds in this area
mutations of b-lactam-insensitive PBP1a can be mapped is evolving rapidly (as reviewed recently by Kaatz [46]).
to a conserved proline near the classical SXN motif as well
as to a range of mutations along b-strands 4 and 5 [34]. Conclusions
Recent years have witnessed a substantial increase in
Given the structural data thus far and the requirement of our understanding of the mechanisms responsible for
conformational changes in catalysis of the HMM but not b-lactam resistance. The structures of the molecular
of the LMM PBPs, there is some concern about using the determinants of resistance — particularly in complex
latter as model systems for inhibitor design. It is therefore with antibiotics or inhibitors — are poised not only to
important to obtain structures of other HMM PBPs to explain resistance, but also to inspire novel methods of
better understand the commonality of this active site combating it. The b-lactam class of antibiotics has proven
plasticity. In addition, structures of the full-length class itself to be invaluable in the treatment of bacterial infec-
A enzymes might enable the generation of new antibio- tions, and structural biology will undoubtedly play a

www.sciencedirect.com Current Opinion in Microbiology 2005, 8:525–533


532 Antimicrobials

central role in ensuring that b-lactams remain therapeu- 13. Phillips OA, Reddy AV, Setti EL, Spevak P, Czajkowski DP,
Atwal H, Salama S, Micetich RG, Maiti SN: Synthesis and
tically effective. biological evaluation of penam sulfones as inhibitors of
beta-lactamases. Bioorg Med Chem 2005, 13:2847-2858.
Acknowledgements 14. Buynak JD, Chen H, Vogeti L, Gadhachanda VR, Buchanan CA,
We are grateful for support from the Howard Hughes Medical Institute Palzkill T, Shaw RW, Spencer J, Walsh TR: Penicillin-derived
(to NS), the Canadian Institute of Health Research (to NS and MW), and inhibitors that simultaneously target both metallo- and serine-
the Michael Smith Foundation for Health Research (to MW and AL). beta-lactamases. Bioorg Med Chem Lett 2004, 14:1299-1304.
15. Bonnefoy A, Dupuis-Hamelin C, Steier V, Delachaume C, Seys C,
References and recommended reading Stachyra T, Fairley M, Guitton M, Lampilas M: In vitro activity of
AVE1330A, an innovative broad-spectrum non-beta-lactam
Papers of particular interest, published within the annual period of
beta-lactamase inhibitor. J Antimicrob Chemother 2004,
review, have been highlighted as:
54:410-417.
 of special interest 16. Tondi D, Morandi F, Bonnet R, Costi MP, Shoichet BK: Structure-
 of outstanding interest based optimization of a non-beta-lactam lead results in
inhibitors that do not up-regulate beta-lactamase expression
in cell culture. J Am Chem Soc 2005, 127:4632-4639.
1. Weldhagen GF: Integrons and beta-lactamases — a novel
perspective on resistance. Int J Antimicrob Agents 2004, 17. Venkatesan AM, Gu Y, Dos Santos O, Abe T, Agarwal A,
23:556-562. Yang Y, Petersen PJ, Weiss WJ, Mansour TS, Nukaga M et al.:
Structure-activity relationship of 6-methylidene penems
2. Fisher JF, Meroueh SO, Mobashery S: Bacterial resistance to bearing tricyclic heterocycles as broad-spectrum beta-
 beta-lactam antibiotics: compelling opportunism, compelling lactamase inhibitors: crystallographic structures show
opportunity. Chem Rev 2005, 105:395-424. unexpected binding of 1,4-thiazepine intermediates.
An excellent and comprehensive review about the mechanisms of b- J Med Chem 2004, 47:6556-6568.
lactam resistance.
18. Melckebeke HV, Vreuls C, Gans P, Filee P, Llabres G, Joris B,
3. Heinz U, Adolph HW: Metallo-beta-lactamases: two binding Simorre JP: Solution structural study of BlaI: implications for
sites for one catalytic metal ion? Cell Mol Life Sci 2004, the repression of genes involved in beta-lactam antibiotic
61:2827-2839. resistance. J Mol Biol 2003, 333:711-720.
4. Garau G, Bebrone C, Anne C, Galleni M, Frere JM, Dideberg O: A 19. Safo MK, Zhao Q, Ko TP, Musayev FN, Robinson H, Scarsdale N,
 metallo-beta-lactamase enzyme in action: crystal structures  Wang AH, Archer GL: Crystal structures of the BlaI repressor
of the monozinc carbapenemase CphA and its complex with from Staphylococcus aureus and its complex with DNA:
biapenem. J Mol Biol 2005, 345:785-795. insights into transcriptional regulation of the bla and mec
This article reports the crystal structures of wild-type CphA, its N220G operons. J Bacteriol 2005, 187:1833-1844.
mutant, and a complex of N220G CphA with the carbapenem substrate This report describes the first crystal structures of BlaI in a free form and in
biapenem. As well as providing the first structures of a class B2 metallo- complex with the mec operator. Although the BlaI structures closely
b-lactamase, the authors propose a novel monozinc carbapenemase resemble the published structures of MecI, the authors describe an up-
mechanism based on the details of the CphA active site in complex with and-down model of repressor-binding to the mec operon (versus the
substrate. side-by-side model for the bla operon) and propose that proteolytic
cleavage of BlaI or MecI preferentially occurs in the DNA-bound form.
5. Wommer S, Rival S, Heinz U, Galleni M, Frere JM, Franceschini N,
Amicosante G, Rasmussen B, Bauer R, Adolph HW: Substrate- 20. Kerff F, Charlier P, Colombo ML, Sauvage E, Brans A, Frere JM,
activated zinc binding of metallo-beta-lactamases: Joris B, Fonze E: Crystal structure of the sensor domain of the
physiological importance of mononuclear enzymes. J Biol BlaR penicillin receptor from Bacillus licheniformis.
Chem 2002, 277:24142-24147. Biochemistry 2003, 42:12835-12843.
6. Palumbi SR: Humans as the world’s greatest evolutionary 21. Wilke MS, Hills TL, Zhang HZ, Chambers HF, Strynadka NC:
force. Science 2001, 293:1786-1790. Crystal structures of the Apo and penicillin-acylated forms of
the BlaR1 beta-lactam sensor of Staphylococcus aureus. J Biol
7. Poole K: Resistance to beta-lactam antibiotics. Cell Mol Life Sci Chem 2004, 279:47278-47287.
2004, 61:2200-2223.
22. Birck C, Cha JY, Cross J, Schulze-Briese C, Meroueh SO,
8. Jacoby GA, Munoz-Price LS: The new beta-lactamases. N Engl J Schlegel HB, Mobashery S, Samama JP: X-ray crystal structure
Med 2005, 352:380-391. of the acylated beta-lactam sensor domain of BlaR1 from
Staphylococcus aureus and the mechanism of receptor
9. Chen Y, Shoichet B, Bonnet R: Structure, function, and
activation for signal transduction. J Am Chem Soc 2004,
 inhibition along the reaction coordinate of CTX-M beta-
126:13945-13947.
lactamases. J Am Chem Soc 2005, 127:5423-5434.
The crystal structures of CTX-M-9 and/or CTX-M-14 are reported in 23. Hanique S, Colombo ML, Goormaghtigh E, Soumillion P, Frere JM,
complex with glycylboronic acid transition state analogues and with Joris B: Evidence of an intramolecular interaction between the
the inhibitor cefoxitin. These structures represent snapshots of CTX-M two domains of the BlaR1 penicillin receptor during the signal
progressing through b-lactam hydrolysis, (i.e. in an acylation transition transduction. J Biol Chem 2004, 279:14264-14272.
state, as an acyl-enzyme intermediate, and in a deacylation transition
state). To explain the inhibitory activity of cefoxitin, steric hindrance owing 24. Rosato AE, Kreiswirth BN, Craig WA, Eisner W, Climo MW,
to the 7a-group of the cefoxitin was implicated in blocking formation of Archer GL: mecA-blaZ corepressors in clinical Staphylococcus
the deacylation transition state. aureus isolates. Antimicrob Agents Chemother 2003,
47:1460-1463.
10. Chen Y, Delmas J, Sirot J, Shoichet B, Bonnet R: Atomic
resolution structures of CTX-M beta-lactamases: extended 25. Katayama Y, Zhang HZ, Hong D, Chambers HF: Jumping the
spectrum activities from increased mobility and decreased barrier to beta-lactam resistance in Staphylococcus aureus.
stability. J Mol Biol 2005, 348:349-362. J Bacteriol 2003, 185:5465-5472.
11. Sanschagrin F, Levesque RC: A specific peptide inhibitor of the 26. Zervosen A, Lu WP, Chen Z, White RE, Demuth TP Jr, Frere JM:
class B metallo-beta-lactamase L-1 from Stenotrophomonas Interactions between penicillin-binding proteins (PBPs) and
maltophilia identified using phage display. J Antimicrob two novel classes of PBP inhibitors, arylalkylidene rhodanines
Chemother 2005, 55:252-255. and arylalkylidene iminothiazolidin-4-ones. Antimicrob Agents
Chemother 2004, 48:961-969.
12. Moloughney JG, Thomas JD, Toney JH: Novel IMP-1 metallo-
beta-lactamase inhibitors can reverse meropenem resistance 27. Llinas A, Ahmed N, Cordaro M, Laws AP, Frere JM, Delmarcelle M,
in Escherichia coli expressing IMP-1. FEMS Microbiol Lett 2005, Silvaggi NR, Kelly JA, Page MI: Inactivation of bacterial DD-
243:65-71. peptidase by beta-sultams. Biochemistry 2005, 44:7738-7746.

Current Opinion in Microbiology 2005, 8:525–533 www.sciencedirect.com


b-Lactam antibiotic resistance Wilke, Lovering and Strynadka 533

28. Silvaggi NR, Josephine HR, Kuzin AP, Nagarajan R, Pratt RF, 35. Tajima Y: Polyoxotungstates reduce the beta-lactam
Kelly JA: Crystal structures of complexes between the R61 resistance of methicillin-resistant Staphylococcus aureus.
DD-peptidase and peptidoglycan-mimetic beta-lactams: a Mini Rev Med Chem 2005, 5:255-268.
non-covalent complex with a ‘‘perfect penicillin’’. J Mol Biol
2005, 345:521-533. 36. Oliva M, Dideberg O, Field MJ: Understanding the acylation
mechanisms of active-site serine penicillin-recognizing
29. Chesnel L, Pernot L, Lemaire D, Champelovier D, Croize J, proteins: a molecular dynamics simulation study. Proteins
Dideberg O, Vernet T, Zapun A: The structural modifications 2003, 53:88-100.
induced by the M339F substitution in PBP2x from
Streptococcus pneumoniae further decreases the 37. Sanbongi Y, Ida T, Ishikawa M, Osaki Y, Kataoka H, Suzuki T,
susceptibility to beta-lactams of resistant strains. J Biol Chem Kondo K, Ohsawa F, Yonezawa M: Complete sequences of
2003, 278:44448-44456. six penicillin-binding protein genes from 40 Streptococcus
pneumoniae clinical isolates collected in Japan. Antimicrob
30. Pernot L, Chesnel L, Le Gouellec A, Croize J, Vernet T, Dideberg O, Agents Chemother 2004, 48:2244-2250.
 Dessen A: A PBP2x from a clinical isolate of Streptococcus
pneumoniae exhibits an alternative mechanism for reduction 38. Leski TA, Tomasz A: Role of penicillin-binding protein 2 (PBP2)
of susceptibility to beta-lactam antibiotics. J Biol Chem 2004, in the antibiotic susceptibility and cell wall cross-linking of
279:16463-16470. Staphylococcus aureus: evidence for the cooperative
Following on from the work done on PRSP strain sp328 [31], the structure functioning of PBP2, PBP4, and PBP2A. J Bacteriol 2005,
of PBP2x from strain sp5259 was obtained. In this case, the mutation of 187:1815-1824.
residues serves more to mimic resistance effects observed in other
lactam-tolerant enzymes, including the re-arrangement of strain b3 (akin 39. Bush K, Macielag M, Weidner-Wells M: Taking inventory:
to PBP2a from S. aureus) and the introduction of negative charge near the antibacterial agents currently at or beyond phase 1. Curr Opin
active site (akin to PBP5fm from E. faecium). In tandem with the mutants Microbiol 2004, 7:466-476.
from sp328 that act to modify the environment around the active site
nucleophile, these results highlight the different techniques used to confer 40. Fuda C, Hesek D, Lee M, Morio K, Nowak T, Mobashery S:
resistance in PBP2x. Activation for catalysis of penicillin-binding protein 2a from
methicillin-resistant Staphylococcus aureus by bacterial cell
31. Dessen A, Mouz N, Gordon E, Hopkins J, Dideberg O: Crystal wall. J Am Chem Soc 2005, 127:2056-2057.
structure of PBP2x from a highly penicillin-resistant
Streptococcus pneumoniae clinical isolate: a mosaic 41. Rice LB, Bellais S, Carias LL, Hutton-Thomas R, Bonomo RA,
framework containing 83 mutations. J Biol Chem 2001, Caspers P, Page MG, Gutmann L: Impact of specific pbp5
276:45106-45112. mutations on expression of beta-lactam resistance in
Enterococcus faecium. Antimicrob Agents Chemother 2004,
32. Lim D, Strynadka NC: Structural basis for the beta lactam 48:3028-3032.
resistance of PBP2a from methicillin-resistant
Staphylococcus aureus. Nat Struct Biol 2002, 9:870-876. 42. Job V, Di Guilmi AM, Martin L, Vernet T, Dideberg O, Dessen A:
Structural studies of the transpeptidase domain of PBP1a
33. Sauvage E, Kerff F, Fonze E, Herman R, Schoot B, Marquette JP, from Streptococcus pneumoniae. Acta Crystallogr D Biol
Taburet Y, Prevost D, Dumas J, Leonard G et al.: The 2.4-A crystal Crystallogr 2003, 59:1067-1069.
structure of the penicillin-resistant penicillin-binding protein
PBP5fm from Enterococcus faecium in complex with 43. Poole K: Efflux-mediated antimicrobial resistance. J Antimicrob
benzylpenicillin. Cell Mol Life Sci 2002, 59:1223-1232. Chemother 2005, 56:20-51.
34. Macheboeuf P, Di Guilmi AM, Job V, Vernet T, Dideberg O, 44. Eswaran J, Koronakis E, Higgins MK, Hughes C, Koronakis V:
 Dessen A: Active site restructuring regulates ligand Three’s company: component structures bring a closer view
recognition in class A penicillin-binding proteins. Proc Natl of tripartite drug efflux pumps. Curr Opin Struct Biol 2004,
Acad Sci USA 2005, 102:577-582. 14:741-747.
The first structural information obtained for a class A PBP, both in the
unliganded form and when complexed with b-lactams. The structure is 45. Yu EW, McDermott G, Zgurskaya HI, Nikaido H, Koshland DE Jr:
representative of the classical PBP fold, but also provides information on Structural basis of multiple drug-binding capacity of the AcrB
the interaction with the transglycosylase domain and the regulation of multidrug efflux pump. Science 2003, 300:976-980.
enzymatic activity by way of active site opening. This work also discusses
the structural effects of mutations in the related enzyme PBP1a, which is 46. Kaatz GW: Bacterial efflux pump inhibition. Curr Opin Investig
implicated in b-lactam-resistant forms of S. pneumoniae. Drugs 2005, 6:191-198.

www.sciencedirect.com Current Opinion in Microbiology 2005, 8:525–533

You might also like