How antibiotics kill bacteria: from targets to networks

Page created by Alfredo Barker
 
CONTINUE READING
REVIEWS

                                    How antibiotics kill bacteria:
                                    from targets to networks
                                    Michael A. Kohanski*‡, Daniel J. Dwyer* and James J. Collins*‡§
                                    Abstract | Antibiotic drug–target interactions, and their respective direct effects, are
                                    generally well characterized. By contrast, the bacterial responses to antibiotic drug
                                    treatments that contribute to cell death are not as well understood and have proven to be
                                    complex as they involve many genetic and biochemical pathways. In this Review, we discuss
                                    the multilayered effects of drug–target interactions, including the essential cellular
                                    processes that are inhibited by bactericidal antibiotics and the associated cellular response
                                    mechanisms that contribute to killing. We also discuss new insights into these mechanisms
                                    that have been revealed through the study of biological networks, and describe how these
                                    insights, together with related developments in synthetic biology, could be exploited to
                                    create new antibacterial therapies.

                                   Our understanding of how antibiotics induce bacte-                   structural integrity following treatment with inhibitors of
Bactericidal
Antimicrobial exposure that
                                   rial cell death is centred on the essential bacterial cell           cell wall synthesis6 , and with cellular energetics, ribosome
leads to bacterial cell death.     function that is inhibited by the primary drug–target                binding and protein mistranslation following treatment
                                   interaction. Antibiotics can be classified based on the              with inhibitors of protein synthesis7. In addition, recent
Bacteriostatic                     cellular component or system they affect, in addition to             evidence points towards a common mechanism of cell
Antimicrobial exposure that
                                   whether they induce cell death (bactericidal drugs) or               death involving disadvantageous cell responses to drug-
inhibits growth with no loss of
viability.                         merely inhibit cell growth (bacteriostatic drugs). Most              induced stresses that are shared by all classes of bacteri-
                                   current bactericidal antimicrobials — which are the                  cidal antibiotics, which ultimately contributes to killing
Cell envelope                      focus of this Review — inhibit DNA, RNA, cell wall or                by these drugs8. Specifically, treatment with lethal con-
Layers of the cell surrounding     protein synthesis1.                                                  centrations of bactericidal antibiotics results in the pro-
the cytoplasm that include lipid
membranes and peptidoglycans.
                                       Since the discovery of penicillin in 1929 (Ref. 2), other,       duction of harmful hydroxyl radicals through a common
                                   more effective antimicrobials have been discovered and               oxidative damage cell death pathway that involves altera-
                                   developed by elucidation of drug–target interactions and by          tions in central metabolism (that is, in the tricarboxylic
*Howard Hughes Medical             drug molecule modification. These efforts have greatly               acid (TCA) cycle) and iron metabolism8–10.
Institute and the Department
of Biomedical Engineering,
                                   enhanced our clinical armamentarium. Antibiotic-                         In this Review we describe our current knowledge of
Center for BioDynamics,            mediated cell death, however, is a complex process that              the drug–target interactions and the associated mecha-
and Center for Advanced            begins with the physical interaction between a drug                  nisms by which the main classes of bactericidal antibiot-
Biotechnology, Boston              molecule and its specific target in bacteria, and involves           ics kill bacteria. We also describe recent efforts in network
University, 44 Cummington
                                   alterations to the affected bacterium at the biochemical,            biology that have yielded new mechanistic insights into
Street, Boston,
Massachusetts 02215, USA.          molecular and ultrastructural levels. The increasing prev-           how bacteria respond to lethal antibiotic treatments, and
‡
 Boston University School of       alence of drug-resistant bacteria3, as well as the increased         discuss how these insights and related developments in
Medicine, 715 Albany Street,       means of gaining resistance, has made it crucial to bet-             synthetic biology could be used to develop new, effective
Boston, Massachusetts              ter understand the multilayered mechanisms by which                  means to combat bacterial infections.
02118, USA.
§
 Wyss Institute for
                                   currently available antibiotics kill bacteria, as well as to
Biologically Inspired              explore and find alternative antibacterial therapies.                Inhibition of DNA replication by quinolones
Engineering, Harvard                   Antibiotic-induced cell death has been associated                DNA synthesis, mRNA transcription and cell division
University, Boston,                with the formation of double-stranded DNA breaks                     require the modulation of chromosomal supercoiling
Massachusetts 02115, USA.
                                   following treatment with inhibitors of topoisomer-                   through topoisomerase-catalysed strand breakage and
Correspondence to J.J.C.
e‑mail: jcollins@bu.edu            ase II (also known as DNA gyrase)4, with the arrest of               rejoining reactions11–13. These reactions are exploited by
doi:10.1038/nrmicro2333            DNA-dependent RNA synthesis following treatment                      the synthetic quinolone class of antimicrobials, includ-
Published online 4 May 2010        with rifamycins5, with cell envelope damage and loss of              ing the clinically relevant fluoroquinolones, which target

NATuRe RevIeWS | Microbiology                                                                                                         vOluMe 8 | juNe 2010 | 423

                                                         © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

 Table 1 | Antibiotic targets and pathways
Drug type     Drug                         Derivation                           Species range                  Primary target       Pathways affected
Fluoroquinolones*
DNA synthesis Nalidixic acid,              Synthetic                            Aerobic Gram-positive Topoisomerase II              DNA replication, SOS
inhibitor     ciprofloxacin,                                                    and Gram-negative     (DNA gyrase),                 response, cell division,
              levofloxacin and                                                  species, some         topoisomerase IV              ATP generation, TCA
              gemifloxacin                                                      anaerobic                                           cycle, Fe–S cluster
                                                                                Gram-negative species                               synthesis, ROS formation,
                                                                                (C. perfringes) and                                 and envelope and
                                                                                M. tuberculosis                                     redox-responsive
                                                                                                                                    two-component systems
Trimethoprim–sulfamethoxazole
DNA synthesis Co-trimoxazole               Synthetic                            Aerobic Gram-positive          Tetrahydrofolic acid Nucleotide biosynthesis
inhibitor     (a combination of                                                 and Gram-negative              synthesis inhibitors and DNA replication
              trimethoprim and                                                  species
              sulfamethoxazole
              in a 1:5 ratio)
Rifamycins
RNA synthesis Rifamycins, rifampin         Natural and semi-synthetic           Gram-positive and      DNA-dependent                RNA transcription, DNA
inhibitor     and rifapentine              forms of ansamycins (derived         Gram-negative species, RNA polymerase               replication and SOS
                                           from S. mediterranei)                and M. tuberculosis                                 response
β-lactams*
Cell wall       Penicillins (penicillin,   Natural and semi-synthetic           Aerobic and anaerobic Penicillin-binding            Cell wall synthesis, cell
synthesis       ampicillin, oxacillin),    forms of carbonyl lactam             Gram-positive and     proteins                      division, autolysin activity
inhibitors      cephalosporins             ring-containing azetidinone          Gram-negative species                               (regulated by LytSR–VncRS
                (cefazolin, cefoxitin      molecules (from P. notatum,                                                              two-component system),
                ceftriaxone,               C. acremonium and S. cattleya)                                                           SOS response, TCA cycle,
                cefepime) and                                                                                                       Fe–S cluster synthesis,
                carbapenems                                                                                                         ROS formation,
                (imipenem)                                                                                                          and envelope and
                                                                                                                                    redox-responsive
                                                                                                                                    two-component systems
Glycopeptides and glycolipopeptides
Cell wall      Vancomycin;          Natural and semi-synthetic forms            Gram-positive species          Peptidoglycan        Cell wall synthesis,
synthesis      teicoplanin          of amino sugar-linked peptide                                              units (terminal      transglycosylation,
inhibitor                           chains (for glycopeptides) or                                              d-Ala-d-Ala          transpeptidation and
                                    of fatty acid-bearing, amino                                               dipeptide)           autolysin activation (VncRS
                                    sugar-linked peptide chains (for                                                                two-component system)
                                    glycolipopetides) derived from
                                    actinobacteria
Lipopeptides
Cell wall      Daptomycin and       Natural and semi-synthetic                  Gram-positive         Cell membrane                 Cell wall synthesis and
synthesis      polymixin B          forms of fatty acid-linked                  species (daptomycin),                               envelope two-component
inhibitors                          peptide chains (from                        Gram-negative species                               systems
                                    S. roseosporus and B. polymyxa)             (polymixins)
Aminoglycosides
Protein        Gentamicin,          Natural and semi-synthetic                  Aerobic Gram-positive          30S ribosome         Protein translation
synthesis      tobramycin,          forms of amino sugars (-mycins              and Gram-negative                                   (mistranslation by tRNA
inhibitors     streptomycin and     from Streptomyces spp. and                  species, and                                        mismatching), ETC, SOS
               kanamycin            -micins from Micromonospora                 M. tuberculosis                                     response, TCA cycle, Fe–S
                                    spp.)                                                                                           cluster synthesis, ROS
                                                                                                                                    formation, and envelope
                                                                                                                                    and redox-responsive
                                                                                                                                    two-component systems
Tetracyclines
Protein         Tetracycline and           Natural and semi-synthetic           Aerobic Gram-positive          30S ribosome         Protein translation
synthesis       doxycycline                forms of four-ringed                 and Gram-negative                                   (through inhibition of
inhibitors                                 polyketides (from                    species                                             aminoacyl tRNA binding
                                           S. aureofaciens and S. rimosus)                                                          to ribosome)
Macrolides
Protein         Erythromycin and           Natural and semi-synthetic       Aerobic and anaerobic 50S ribosome                      Protein translation
synthesis       azythromycin               forms of 14- and 16-membered Gram-positive and                                           (through inhibition
inhibitors                                 lactone rings (from S. erythraea Gram-negative species                                   of elongation and
                                           and S. ambofaciens)                                                                      translocation steps) and
                                                                                                                                    free tRNA depletion

424 | juNe 2010 | vOluMe 8                                                                                                       www.nature.com/reviews/micro

                                                    © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

 Table 1 (cont.) | Antibiotic targets and pathways
 Drug type         Drug                  Derivation                                    Species range                         Primary target        Pathways affected
 Streptogramins
 Protein           Pristinamycin,        Natural and semi-synthetic          Aerobic and anaerobic                           50S ribosome          Protein translation
 synthesis         dalfopristin and      forms of pristinamycin I (group B,  Gram-positive and                                                     (through inhibition of
 inhibitors        quinupristin          macrolactone ringed-peptides) and   Gram-negative species‡                                                initiation, elongation
                                         pristinamycin II (group A,                                                                                and translocation
                                         endolactone oxazole nucleus-bearing                                                                       steps) and free tRNA
                                         depsipeptides) (from Streptomyces                                                                         depletion
                                         spp.)
 Phenicols
 Protein           Chloramphenicol Natural and semi-synthetic forms of                 Some Gram-positive and                50S ribosome          Protein translation
 synthesis                         dichloroacetic acid with an aromatic                Gram-negative species,                                      (through inhibition of
 inhibitor                         nucleus and aminopropanediol chain                  including B. fragilis,                                      elongation step)
                                   (from S. venezuelae)                                N. meningitidis, H. influenzae
                                                                                       and S. pneumoniae
 * Drug efficacy can vary across species range based on drug generation. ‡When used as a combination of pristinamycin I and pristinamycin II. B. fragilis, Bacillus fragilis;
 B. polymyxa, Bacillus polymyxa; C. acremonium, Cephalosporium acremonium; ETC: electron transport chain; H. influenzae, Haemophilus influenzae; M. tuberculosis,
 Mycobacterium tuberculosis; N. meningitidis, Neisseria meningitidis; P. notatum, Penicillum notatum; ROS, reactive oxygen species; S. ambofaciens, Streptomyces ambofaciens;
 S. aureofaciens, Streptomyces aureofaciens; S. cattleya, Streptomyces cattleya; S. erythraea, Saccharopolyspora erythraea; S. mediterranei, Streptomyces mediterranei;
 S. pneumoniae, Streptococcus pneumoniae; S. rimosus, Streptomyces rimosus; S. roseosporus, Streptomyces roseosporus; S. venezuelae, Streptomyces venezuelae; TCA,
 tricarboxylic acid.

                                  DNA–topoisomerase complexes4,14,15. Quinolones are                      is to generate double-stranded DNA breaks that are
                                  derivatives of nalidixic acid, which was discovered as a                trapped by covalently (but reversibly) linked topo-
                                  byproduct of the synthesis of chloroquine (a quinine)                   isomerases, the function of which is compromised26–28.
                                  and was introduced in the 1960s to treat urinary tract                  As a result of quinolone–topoisomerase–DNA complex
                                  infections16. Nalidixic acid and other first generation                 formation, the DNA replication machinery becomes
                                  quinolones (for example, oxolinic acid) are rarely used                 arrested at blocked replication forks, leading to inhi-
                                  today owing to their toxicity 17. Second (ciprofloxacin),               bition of DNA synthesis, which immediately leads
                                  third (levofloxacin) and fourth (gemifloxacin) genera-                  to bacteriostasis and eventually cell death4 (fIG. 1). It
                                  tion quinolone antibiotics (TABLe 1) can be classified on               should be noted that the effects on DNA replication
                                  the basis of their chemical structure and of qualitative                that correlate with bacteriostatic concentrations of qui-
                                  differences between the killing mechanisms they                         nolones are thought to be reversible4,29. Nonetheless,
                                  use16,18.                                                               considering that topoisomerase II has been found to
                                      The quinolone class of antimicrobials interferes with               be distributed approximately every 100 kb along the
                                  the maintenance of chromosomal topology by target-                      chromosome30, inhibition of topoisomerase function
                                  ing topoisomerase II and topoisomerase Iv, trapping                     by quinolone antibiotics and the resulting formation of
                                  these enzymes at the DNA cleavage stage and prevent-                    stable complexes with DNA have substantial negative
                                  ing strand rejoining 4,19,20 (fIG. 1). Despite the general              consequences for the cell in terms of its ability to deal
                                  functional similarities between topoisomerase II and                    with drug-induced DNA damage31.
                                  topoisomerase Iv, their susceptibility to quinolones
                                  varies across bacterial species20 (TABLe 1). For exam-                  The role of protein expression in quinolone-mediated
                                  ple, several studies have shown that topoisomerase Iv                   cell death. The introduction of double-stranded
                                  is the primary target of quinolones in Gram-positive                    DNA breaks following topoisomerase inhibition by
                                  bacteria (for example, Streptococcus pneumoniae 21),                    quinolones induces the DNA stress response ( SOS
                                  whereas in Gram-negative bacteria (for example,                         response), in which RecA is activated by DNA dam-
                                  Escherichia coli 13 and Neisseria gonorrhoea22) their pri-              age and promotes self-cleavage of the lexA repres-
                                  mary target is topoisomerase II (and topoisomerase Iv                   sor protein, inducing the expression of SOS response
                                  is the secondary target).                                               genes such as DNA repair enzymes32. Notably, several
                                                                                                          studies have shown that preventing the induction of
                                  Introduction of DNA breaks and replication fork arrest.                 the SOS response enhances killing by quinolones
                                  The ability of quinolone antibiotics to kill bacteria is                (except in the case of nalidixic acid)8,33. Preventing
                                  a function of the stable interaction complex that is                    the activation of the SOS response has also been
SOS response
                                  formed between drug-bound topoisomerases and                            shown to reduce the formation of drug-resistant
The DNA stress response
pathway in E. coli, the           cleaved DNA4. On the basis of studies using DNA cleav-                  mutants by blocking the induction of error-prone
prototypical network of genes     age mutants of topoisomerase II23 and topoisomerase                     DNA polymerases34, homologous recombination20 and
of which is regulated by the      Iv24 that do not prevent quinolone binding, and stud-                   horizontal transfer of drug-resistance elements35,36.
transcriptional repressor LexA,   ies that have shown that strand breakage can occur in                       Together with studies revealing that co-treatment with
and is commonly activated by
the co-regulatory protein RecA,
                                  the presence of quinolones25, it is accepted that DNA                   quinolones and the protein synthesis inhibitor chloram-
which promotes LexA               strand breakage occurs after the drug has bound to the                  phenicol inhibits the ability of quinolones to kill bacte-
self-cleavage when activated.     enzyme. Therefore, the net effect of quinolone treatment                ria19,37, there seems to be a clear relationship between the

NATuRe RevIeWS | Microbiology                                                                                                                vOluMe 8 | juNe 2010 | 425

                                                          © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                             Quinolones

                                                 DNA polymerase                                                  Protein independent
                                                 complex

                             Replication                            Quinolones
                             fork

                                                                                                                  SOS       DNA repair                     Cell death

                                                                  Topoisomerase

                                                                                                                 Protein dependent

                             β-lactams
                              Gram negative                                                                     Gram positive
                                                              Outer
                                                              membrane
                                               PBP
                                                               β-lactam
                                                                                      Lysis and
                                                                                      cell death
                                                               Autolysin

                                                              Inner                                                                          Periplasmic
                                                              membrane                                                                       membrane

                             Aminoglycosides

                                   Aminoglycoside                                                                                         Outer
                                                            30s                                                                           membrane
                              mRNA
                                                                                                                                                           Cell death

                                                                        Misfolded                                                         Inner
                                Ribosome
                                                                        protein                                                           membrane
                                                            50s
                                                                                                                                           Increased
                                                                                                                                           aminoglycoside
                                                                                                                                           uptake
                             Figure 1 | Drug-target interactions and associated cell death mechanisms. Quinolone antibiotics interfere
                             with changes in DNA supercoiling by binding to topoisomerase II or topoisomerase IV. This leads    to the
                                                                                                                             Nature    formation
                                                                                                                                     Reviews      of
                                                                                                                                             | Microbiology
                             double-stranded DNA breaks and cell death in either a protein synthesis-dependent or protein synthesis-independent
                             manner. β-lactams inhibit transpeptidation by binding to penicillin-binding proteins (PBPs) on maturing peptidoglycan
                             strands. The decrease in peptidoglycan synthesis and increase in autolysins leads to lysis and cell death. Aminoglycosides
                             bind to the 30S subunit of the ribosome and cause misincorporation of amino acids into elongating peptides. These
                             mistranslated proteins can misfold, and incorporation of misfolded membrane proteins into the cell envelope leads
                             to increased drug uptake. This, together with an increase in ribosome binding, has been associated with cell death.

                             primary effects of quinolone–topoisomerase–DNA com-                    inhibition of DNA replication by quinolones, has a cata-
                             plex formation and the response of the bacteria (through               strophic effect on prokaryotic nucleic acid metabolism
                             the stress-induced expression of proteins) to these effects            and is a potent means of inducing bacterial cell death5.
                             in the bactericidal activity of quinolone antibiotics. For             Rifamycins inhibit DNA-dependent transcription by sta-
                             example, the contribution of reactive oxygen species                   bly binding with high affinity to the β-subunit (encoded
                             (ROS) to quinolone-mediated cell death has recently been               by rpoB) of a DNA-bound and actively transcribing
                             shown to occur in a protein synthesis-dependent man-                   RNA polymerase41–43 (TABLe 1). The β-subunit is located
                             ner38. Also, the chromosome-encoded toxin MazF has                     in the channel that is formed by the RNA polymerase–
                             been shown to be required under certain conditions for                 DNA complex, from which the newly synthesized RNA
                             efficient killing by quinolones owing to its ability to alter          strand emerges44. Rifamycins uniquely require RNA
                             protein carbonylation39, a form of oxidative stress40.                 synthesis to not have progressed beyond the addition
                                                                                                    of two ribonucleotides; this is attributed to the ability of
                             Inhibition of RNA synthesis by rifamycins                              the drug molecule to sterically inhibit nascent RNA
                             The inhibition of RNA synthesis by the rifamycin class                 strand initialization45. It is worth noting that rifamycins
                             of semi-synthetic bactericidal antibiotics, similarly to the           are not thought to act by blocking the elongation step of

426 | juNe 2010 | vOluMe 8                                                                                                           www.nature.com/reviews/micro

                                                     © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                  RNA synthesis, although a recently discovered class                  ring) is an analogue of the terminal d-alanyl-d-alanine
                                  of RNA polymerase inhibitors (based on the compound                  dipeptide of peptidoglycan and acts as a substrate for the
                                  CBR703) could inhibit elongation by allosterically                   PBP during the acylation phase of cross link formation.
                                  modifying the enzyme46.                                              Penicilloylation of the PBP active site blocks the hydro-
                                      Rifamycins were first isolated47 from the Gram-positive          lysis of the bond created with the now ring-opened drug,
                                  bacterium Amycolatopsis mediterranei (originally known               thereby disabling the enzyme59,60.
                                  as Streptomyces mediterranei) in the 1950s. Mutagenesis of               By contrast, most actinobacterium-derived glyco-
                                  this organism has led to the isolation and characterization          peptide antibiotics (for example, vancomycin) inhibit
                                  of more potent rifamycin forms48, including the clinically           peptidoglycan synthesis by binding peptidoglycan units
                                  relevant rifamycin Sv and rifampicin. Rifamycins are                 (at the d-alanyl-d-alanine dipeptide) and by blocking
                                  considered bactericidal against Gram-positive bacteria               transglycosylase and PBP activity 61. In this way, glyco-
                                  and bacteriostatic against Gram-negative bacteria, a dif-            peptides (whether free in the periplasm like vancomy-
                                  ference that has been attributed to drug uptake and not to           cin or membrane-anchored like teicoplanin62) generally
                                  affinity of the drug with the RNA polymerase β-subunit 49.           act as steric inhibitors of peptidoglycan maturation and
                                  Notably, rifamycins are among the first-line therapies               reduce the mechanical strength of the cell, although some
                                  used against mycobacteria because they efficiently induce            chemically modified glycopeptides have been shown to
                                  mycobacterial cell death50, although rifamycins are often            directly interact with the transglycosylase63. It is worth
                                  used in combinatorial therapies owing to the rapid nature            noting that β-lactams can be used to treat Gram-positive
                                  of resistance development49,51.                                      and Gram-negative bacteria, whereas glycopeptides are
                                      Interestingly, an interaction between DNA and                    effective against only Gram-positive bacteria owing to
                                  the hydroquinone moiety of RNA polymerase-bound                      low permeability (TABLe 1). In addition, antibiotics that
                                  rifamycin has been observed52, and this interaction has              inhibit the synthesis (for example, fosfomycin) and trans-
                                  been attributed to the location of the rifamycin mol-                port (for example, bacitracin) of individual peptidogly-
                                  ecule in relation to DNA in the DNA–RNA polymerase                   can units are also currently in use, as are lipopeptides (for
                                  complex 42. This proximity, coupled with the reported                example, daptomycin), which affect structural integrity by
                                  ability of rifamycin to cycle between a radical and non-             inserting themselves into the cell membrane and inducing
                                  radical form (rifamycin Sv and rifamycin S52,53), may                membrane depolarization.
                                  damage DNA through a direct drug–DNA interaction.                        Research into the mechanism of killing by peptido-
                                  This hypothesis could account for the observation that               glycan synthesis inhibitors has centred on the lysis event.
                                  rifamycin Sv can induce the SOS DNA damage response                  Initially, it was thought that inhibition of cell wall syn-
                                  in E. coli and that treatment of recA-mutant E. coli results in      thesis by β-lactams caused cell death when internal pres-
                                  cell death whereas treatment of wild-type E. coli leads              sure built up owing to cell growth outpacing cell wall
                                  to bacteriostasis8.                                                  expansion, resulting in lysis6. This unbalanced growth
                                                                                                       hypothesis was based in part on the notion that active pro-
                                  Inhibition of cell wall synthesis                                    tein synthesis is required for lysis to occur following the
                                  Lytic cell death. The bacterial cell is encased by layers of         addition of β-lactams.
                                  peptidoglycan (also known as murein), a covalently cross-                The lysis-dependent cell death mechanism, however,
                                  linked polymer matrix that is composed of peptide-linked             has proven to be much more complex, involving many
                                  β-(1-4)-N-acetyl hexosamine54. The mechanical strength               active cellular processes. Seminal work showed that
                                  afforded by this layer of the cell wall is crucial to a bacte-       S. pneumoniae deficient in amidase activity (possessed by
                                  rium’s ability to survive environmental conditions that can          peptidoglycan hydrolase or autolysins) did not grow or die
                                  alter prevailing osmotic pressures; of note, the degree of           following treatment with a lysis-inducing concentration
Lysis                             peptidoglycan cross-linking correlates with the structural           of a β-lactam, an effect known as antibiotic tolerance64.
Rupture of the cell envelope      integrity of the cell55. Maintenance of the peptidoglycan            Autolysins are membrane-associated enzymes that break
leading to the expulsion of
                                  layer is accomplished by the activity of transglycosylases           down bonds between and within peptidoglycan strands,
intracellular contents into the
surrounding environment with      and penicillin-binding proteins (PBPs; also known as                 making them important during normal cell wall turnover
eventual disintegration of the    transpeptidases), which add disaccharide pentapeptides               and maintenance of cell shape55. Autolysins have also been
cell envelope.                    to extend the glycan strands of existing peptidoglycan               shown to play a part in lytic cell death in bacterial species
                                  molecules and cross-link adjacent peptide strands of                 that contain numerous peptidoglycan hydrolases, such as
Peptidoglycan hydrolase
An enzyme that introduces
                                  immature peptidoglycan units, respectively 56.                       E. coli 65. In E. coli, a set of putative peptidoglycan hydro-
cuts between carbon–nitrogen          β-lactams and glycopeptides are among the classes of             lases (lytM domain factors) were shown to be important
non-peptide bonds while           antibiotics that interfere with specific steps in homeostatic        for rapid ampicillin-mediated lysis66. The discovery that
pruning the peptidoglycan         cell wall biosynthesis. Successful treatment with a cell wall        autolysins contribute to cell death expanded our under-
layer. It is important for
                                  synthesis inhibitor can result in changes to cell shape and          standing of lysis and showed that active degradation of
homeostatic peptidoglycan
turnover.                         size, induction of cell stress responses and ultimately cell         the peptidoglycan layer by peptidoglycan hydrolases, in
                                  lysis6 (fIG. 1). For example, β-lactams (including penicil-          conjunction with inhibition of peptidoglycan synthesis by
Autolysin                         lins, carbapenems and cephalosporins) block the cross-               a β-lactam antibiotic, triggers lysis64 (fIG. 1).
An enzyme that hydrolyses the     linking of peptidoglycan units by inhibiting the peptide
β-linkage between the
monosaccharide monomers in
                                  bond formation reaction that is catalysed by PBPs55,57,58.           Non-lytic cell death. S. pneumoniae lacking peptidoglycan
peptidoglycan units and can       This inhibition is achieved by penicilloylation of the PBP           hydrolase activity can still be killed by β-lactams, but at
induce lysis when in excess.      active site — the β-lactam (containing a cyclic amide                a slower rate than autolysin-active cells, indicating that

NATuRe RevIeWS | Microbiology                                                                                                         vOluMe 8 | juNe 2010 | 427

                                                        © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                    there is a lysis-independent mode of killing induced by              ofloxacin or pefloxacin83. This suggests that peptido-
                                    β-lactams64,67. evidence suggests that some of these non-            glycan turnover and the SOS response could have a role
                                    lytic pathways are regulated by bacterial two-component              in antibiotic-mediated lytic killing responses.
                                    systems68. For example, in S. pneumoniae, the vncSR two-
                                    component system controls the expression of the autolysin            Inhibition of protein synthesis
                                    lytA and regulates tolerance to vancomycin and penicil-              The process of mRNA translation occurs over three
                                    lin through lysis-dependent69 and lysis-independent70 cell           sequential phases (initiation, elongation and termina-
                                    death pathways.                                                      tion) that involve the ribosome and a range of cytoplas-
                                        In Staphylococcus aureus, the lytSR two-component                mic accessory factors84. The ribosome is composed of
                                    system can similarly affect cell lysis by regulating autolysin       two ribonucleoprotein subunits, the 50S and 30S, which
                                    activity 71. lytR activates the expression of lrgAB72, which         assemble (during the initiation phase) following the
                                    was found to inhibit autolysin activity and thereby lead             formation of a complex between an mRNA transcript,
                                    to antibiotic tolerance73. lrgA is similar to bacteriophage          N-formylmethionine-charged aminoacyl tRNA, several
                                    holin proteins73, which regulate the access of autolysins            initiation factors and a free 30S subunit 85. Drugs that
                                    to the peptidoglycan layer. Based on this information,               inhibit protein synthesis are among the broadest classes
                                    an additional holin-like system, cidAB, was uncovered                of antibiotics and can be divided into two subclasses: the
                                    in S. aureus and found to activate autolysins, render-               50S inhibitors and 30S inhibitors (TABLe 1).
                                    ing S. aureus more susceptible to β-lactam-mediated                      50S ribosome inhibitors include macrolides (for
                                    killing 74,75. Complementation of cidA into a cidA-null              example, erythromycin), lincosamides (for example,
                                    strain reversed the loss of autolysin activity but did not           clindamycin), streptogramins (for example, dalfopristin–
                                    completely restore sensitivity to β-lactams74.                       quinupristin), amphenicols (for example, chlorampheni-
                                                                                                         col) and oxazolidinones (for example, linezolid)86,87.
                                    Role of the SOS response in cell death by β-lactams.                 50S ribosome inhibitors work by physically blocking
                                    Treatment with β-lactams leads to changes in cell mor-               either initiation of protein translation (as is the case for
                                    phology that are associated with the primary drug–PBP                oxazolidinones88) or translocation of peptidyl tRNAs,
                                    interaction. Generally speaking, PBP1 inhibitors cause               which serves to inhibit the peptidyltransferase reac-
                                    cell elongation and are potent triggers of lysis, PBP2               tion that elongates the nascent peptide chain. A model
                                    inhibitors alter cell shape but do not cause lysis and               for the mechanism by which these drugs act has been
                                    PBP3 inhibitors influence cell division and can induce               formulated by studies of macrolides, lincosamides and
                                    filamentation76. Interestingly, β-lactam subtypes have dis-          streptogramins. The model involves blocking the access
                                    tinct affinities for certain PBPs, which correlate with the          of peptidyl tRNAs to the ribosome (to varying degrees),
                                    ability of these drugs to stimulate autolysin activity and           subsequent blockage of the peptidyltransferase elonga-
                                    induce lysis76,77. Accordingly, PBP1-binding β-lactams are           tion reaction by steric inhibition and eventually trigger-
                                    also the most effective inducers of peptidoglycan hydro-             ing dissociation of the peptidyl tRNA89,90. This model
                                    lase activity, and PBP2 inhibitors are the least proficient          also accounts for the phenomenon that these classes of
                                    autolysin activators77.                                              drugs lose their antibacterial activity when elongation has
                                        Filamentation can occur following the activation of the          progressed beyond a crucial length91.
                                    DNA damage-responsive SOS network of genes78 owing                       30S ribosome inhibitors include tetracyclines and
                                    to expression of SulA, a key component of the SOS net-               aminocyclitols. Tetracyclines work by blocking the access
                                    work that inhibits septation and leads to cell elongation            of aminoacyl tRNAs to the ribosome92. The aminocycli-
                                    by binding to and inhibiting polymerization of septation-            tol class comprises spectinomycin and aminoglycosides
                                    triggering FtsZ monomers79,80. Interestingly, β-lactams              (for example, streptomycin, kanamycin and gentamicin),
                                    that inhibit PBP3 and induce filamentation have been                 which bind the 16S rRNA component of the 30S ribos-
                                    shown to stimulate the DpiAB two-component system,                   ome subunit. Spectinomycin interferes with the stability
                                    which can activate the SOS response81. β-lactam lethality            of peptidyl tRNA binding to the ribosome by inhibiting
                                    can be enhanced by disrupting DpiAB signalling or by                 elongation factor-catalysed translocation, but does not
                                    knocking out sulA. This indicates that SulA may protect              cause protein mistranslation93–95. By contrast, the inter-
                                    against β-lactam killing by shielding FtsZ and limiting a            action between aminoglycosides and the 16S rRNA can
                                    division ring interaction among PBPs and peptidoglycan               induce an alteration in the conformation of the com-
                                    hydrolases. In support of this idea, SulA expression lim-            plex formed between an mRNA codon and its cognate
                                    its the lysis observed in a strain of E. coli that expresses         charged aminoacyl tRNA at the ribosome. This pro-
                                    FtsZ84 (a mutant of FtsZ that is active only under certain           motes tRNA mismatching, which can result in protein
                                    temperatures and media conditions) and lacks PBP4 and                mistranslation96–99.
                                    PBP7 (Ref. 82).                                                          Among ribosome inhibitors, naturally derived
                                        DNA-damaging antimicrobials that do not directly                 aminoglycosides are the only class that is broadly bac-
                                    disrupt peptidoglycan turnover, such as quinolones, also             tericidal. Macrolides, streptogramins, spectinomycin,
Two-component system                cause filamentation by activating the SOS response4.                 tetracyclines, chloramphenicol and macrolides are typi-
A two-protein signal relay          Interestingly, a mutant strain of E. coli that is deficient          cally bacteriostatic; however, they can be bactericidal in a
system composed of a sensor
histidine kinase and a cognate
                                    in diaminopimelic acid synthesis (E. coli W7), a key                 species- or treatment-specific manner. For example, chlo-
receiver protein, which is          building block of peptidoglycan, undergoes lysis follow-             ramphenicol has been shown to kill S. pneumoniae and
typically a transcription factor.   ing treatment with the fluoroquinolone antimicrobials                Neisseria meningitidis effectively 100, and chloramphenicol

428 | juNe 2010 | vOluMe 8                                                                                                          www.nature.com/reviews/micro

                                                          © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

  Box 1 | Drug synergy                                                                                      In E. coli aminoglycoside-mediated killing has
                                                                                                        been linked with alterations to the cell membrane
  Combinatorial antibiotic treatments can have diverse effects on bacterial survival.                   ultrastructure that ultimately increase drug uptake109,110.
  Antibiotics can be more effective as a combination treatment displaying either an                     Aminoglycosides can affect membrane composition
  additive effect (an effect equal to the sum of the treatments) or a synergistic effect
                                                                                                        through the incorporation of mistranslated membrane
  (an effect greater than the sum of the treatments). The combination can also be
                                                                                                        proteins into the cytoplasmic membrane, thereby increas-
  antagonistic — that is, the effect of the combination treatment is less than the
  effect of the respective single-drug treatments136. Technological advances have                       ing cell permeability, which allows increased access of the
  allowed high-throughput quantification of drug–drug interactions at the level of                      drug 103 (fIG. 1). Sufficient aminoglycoside uptake resulting
  cell survival and target binding, thereby opening the door for the systematic study                   in increased ribosome inhibition and cell death could also
  of synergistic and antagonistic drug combinations137.                                                 occur as a function of the changes in membrane integrity
     The exploration of the survival fitness landscape between drug combinations has                    owing to the incorporation of mistranslated membrane
  allowed the study of the mechanisms by which antibiotics work against bacteria138 and                 proteins103. An alteration in membrane permeability
  has also allowed a study of the evolution of drug resistance137. Further study of the synergy         owing to aminoglycoside-induced membrane damage is
  or antagonism between antibiotics will provide additional insight into the underlying cell            thought to be one of the mechanisms by which amino-
  death mechanisms for the individual classes of antibiotics. For example, the suppressive
                                                                                                        glycosides cooperate with β-lactams (see BOX 1 for more
  interaction between protein synthesis inhibitors and DNA synthesis inhibitors has been
                                                                                                        on drug synergy and antagonism).
  shown to be due to non-optimal ribosomal RNA regulation by DNA-inhibiting drugs139.
     The synergy between aminoglycosides and β-lactams has been attributed to                               Another consequence of mistranslated protein incorpo-
  β-lactam-mediated membrane damage leading to increased uptake of aminoglycosides140.                  ration into the bacterial membrane is the activation of enve-
  It will be interesting to see whether the synergy between these two drugs is also related             lope (Cpx) and redox-responsive (Arc) two-component
  to the induction of the envelope stress response that has been observed following                     systems. These intracellular signal relay systems regulate
  treatment with aminoglycosides10.                                                                     the expression of genes that are important for the main-
                                                                                                        tenance of membrane integrity and composition111, and
                                   and the macrolide azythromycin have exhibited bacteri-               membrane-coupled energy generation112,113, respectively.
                                   cidal activity against Haemophilus influenzae100,101. This           Disruption of Cpx or Arc two-component system sig-
                                   species-specific variability in ribosome inhibitor-medi-             nalling (through a series of single-gene knockouts) has
                                   ated cell death probably has to do with sequence differ-             recently been shown to reduce the killing efficacy of
                                   ences among bacterial species in the variable regions of             aminoglycosides, a result associated with findings link-
                                   the highly conserved ribosomal proteins and RNAs102. In              ing bactericidal antibiotic-induced cell death with drug
                                   addition, high concentrations of macrolides and combina-             stress-induced changes in metabolism. Interestingly, dis-
                                   tions of streptogramin group A and group B can behave in             ruption of Cpx or Arc two-component system signalling
                                   a bactericidal manner. For the rest of this section, however,        was also shown to reduce the lethality of β-lactam and
                                   we focus on aminoglycosides, which have the best-studied             quinolone antibiotics10. Together, these findings point
                                   mechanism of killing by ribosome inhibition.                         towards a broad role for the envelope stress-responsive
                                                                                                        and redox-responsive two-component systems in killing
                                   Aminoglycoside uptake and cell death. Binding of                     by bactericidal drugs (fIG. 2).
                                   aminoglycosides to the ribosome does not bring trans-
                                   lation to an immediate standstill. Instead, as noted                 Antibiotic network biology
                                   above, this class of drugs promotes protein mistransla-              As noted above, antibiotic-mediated cell death is a complex
                                   tion through the incorporation of inappropriate amino                process that only begins with the drug–target interaction
                                   acids into elongating peptide strands96; this phenotype              and the primary effects of these respective interactions.
                                   is specific for aminoglycosides and contributes to cell              The development of new antibiotics and the improvement
                                   killing (fIG. 1).                                                    of current antibacterial drug therapies would benefit from
                                       Respiration also has a crucial role in aminoglycoside            a better understanding of the specific sequences of events
                                   uptake and lethality 103. Following the initial step of drug         beginning with the binding of a bactericidal drug to its
                                   molecule adsorption (in Gram-negative species such                   target and ending in bacterial cell death.
                                   as E. coli) through electrostatic interaction, changes in                Bioinformatics approaches that use high-throughput
                                   membrane potential allow aminoglycosides to access the               genetic screening or gene expression profiling have
                                   cell. Respiration-dependent uptake relies on the activity            proven to be valuable tools to explore the response layers
                                   of membrane-associated cytochromes and maintenance of                of bacteria to different antibiotic treatments114. For exam-
                                   the electrochemical potential through the quinone pool104,105.       ple, recent screens for antibiotic susceptibility in a single-
                                   Accordingly, under anaerobic conditions aminoglyco-                  gene deletion library of non-essential genes in E. coli 115
                                   side uptake is severely limited in both Gram-positive and            and a transposon mutagenesis library in P. aeruginosa116
                                   Gram-negative bacteria106,107, although there is evidence            have provided important insights into the numbers and
                                   that aminoglycoside uptake can occur under certain                   types of genes that affect treatment efficiency (bactericidal
                                   anaerobic conditions by a mechanism that is sensitive                versus bacteriostatic effects), including those related to
                                   to nitrate levels. In E. coli and Pseudomonas aeruginosa,            drug molecule efflux, uptake or degradation. In addition,
                                   aminoglycoside uptake can take place when nitrate is                 monitoring global changes in gene expression patterns,
Quinone pool                       used as an electron acceptor in place of oxygen, and                 or signatures, resulting from antibiotic treatment over
Membrane-associated cyclic
aromatic-based compounds
                                   anaerobic bacteria that have quinones and cytochromes                a range of conditions, has advanced our understanding
that shuttle electrons along the   can take up aminoglycosides if sufficient anaerobic                  of the off-target effects elicited by primary drug–target
electron transport chain.          electron transport occurs108.                                        interactions114.

NATuRe RevIeWS | Microbiology                                                                                                          vOluMe 8 | juNe 2010 | 429

                                                         © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                   Aminoglycoside
                                                                                                                                                            β-lactam
                                                                                                 Quinolone
                                                                                                                                                                     PBP

                                                                                                                 Topoisomerase
                                                                                     NADH

                                                                                     mdh
                                                                  NAD+
                                                                                                                                       Cpx and Arc two
                                                  Metabolic                                                         acnB               component systems
                                                  feedback                                       TCA cycle          [Fe–S]

                                                                  NADH                                                         NADP+                S        S    Disulphide
                                                                                                                                                                  bond
                                                                                                                                                    HS      HS    formation

                                                                                      NAD+                      NADPH

                                                                     I         II                        IV              V

                                   Hyperactivation of
                                electron transport chain                                    Uq

                                                                                                                                              O2–   Superoxide
                                                                                                                                                     formation
                                                            NADH           NAD+
                                                                                                                   ADP + Pi    ATP

                                                                                                                                     Damage
                                                                                    Fenton
                                                                                    reaction                                                                 Fe–S cluster
                                                                         •OH
                                                                                                  Fe2+                                                         damage
                                               Hydroxyl radical
                                                 production                          Fe3+

                                                                                                                                                          Damage to DNA,
                                                                                                                                                         lipids and proteins

                                                                                                                  Cell death
                             Figure 2 | common mechanism of cell death induced by bactericidal antibiotics. The primary drug–target interactions
                             (aminoglycoside with the ribosome, quinolone with topoisomerase, and β-lactam with penicillin-binding proteins (PBPs))
                             stimulate the oxidation of NADH through the electron transport chain, which is dependent onNature
                                                                                                                             the tricarboxylic acid (TCA)
                                                                                                                                    Reviews | Microbiology
                             cycle. Hyperactivation of the electron transport chain stimulates superoxide (O2–) formation. Superoxide damages Fe–S
                             clusters, making ferrous iron available for oxidation by the Fenton reaction. The Fenton reaction leads to the formation of
                             hydroxyl radicals (•OH), which damage DNA, lipids and proteins. This contributes to antibiotic-induced cell death.
                             Quinolones, β-lactams and aminoglycosides also trigger hydroxyl radical formation and cell death through the envelope
                             (Cpx) and redox-responsive (Arc) two-component systems. It is also possible that redox-sensitive proteins, such as those
                             containing disulphides, contribute in undetermined way to the common mechanism (dashed lines). Figure modified, with
                             permission, from Ref. 8 © (2007) Elsevier Science. acnb, aconitase b; mdh, malate dehydrogenase; uq, ubiquinone.

                                A need also exists for the application of network biol-                       connections and biochemical pathway classifications,
                             ogy methods to discern and resolve the potential interplay                       to functionally enrich datasets and predict relationships
                             between genes and proteins coordinating bacterial stress                         that exist among genes under tested conditions. As such,
                             response pathways. Typically, such methods incorporate                           biological network studies of drug-treated bacteria can
                             gene expression profiling data and the results of high-                          be used to advance our understanding of how groups of
                             throughput genetic screens, along with the contents of                           genes interact functionally, rather than in isolation, when
                             databases detailing experimentally identified regulatory                         cells react to antibiotic stress117.

430 | juNe 2010 | vOluMe 8                                                                                                                 www.nature.com/reviews/micro

                                                   © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                                                                                                               Protein                                            Outer membrane
                                                                                             DegP            degradation

                                                                                                                                                                          Inner
                                                                                                                                                O2        NO3–
                                                                                                                                                                       membrane

                                                                                                                                       Uq   I        II   Uq IV    V
                                                                                                 P                  PP                 P

                                                                                                     CpxA

                                                                                                             CpxA

                                                                                                                                ArcA
                                                                                                                         ArcB
                                                                                                                                                                          ADP + Pi
                                                                  SecA         SecA
                                                                                                                                                                    ATP

                                                                                                 CpxR                    ArcA
                                  Aminoglycoside
                                                                                                                                                 TCA cycle
                                                                                                                          ArcA         P
                                                                                                CpxR          P

                                                                                                                                                                     Superoxide
                                                                                                                                                                      formation
                                           Mistranslation                             Envelope stress response                                                      and cell death
                                  Figure 3 | Aminoglycosides trigger hydroxyl radical-mediated cell death. The interaction between aminoglycosides and
                                  the ribosome causes mistranslation and misfolding of membrane proteins. Incorporation of mistranslated, misfolded proteins
                                                                                                                                     Nature Reviews | Microbiology
                                  into the cell membrane stimulates the envelope (Cpx) and redox-responsive (Arc) two-component systems. Activation of these
                                  systems perturbs cell metabolism and the membrane potential, resulting in the formation of lethal hydroxyl radicals. Figure
                                  modified, with permission, from Ref. 10 © (2008) Elsevier Science. TCA, tricarboxylic acid; uq, ubiquinone.

                                      To help address this problem, researchers have                        reaction123. under these conditions, the Fenton reac-
                                  developed methods to construct quantitative models of                     tion was found to be fuelled by superoxide-mediated
                                  regulatory networks118–122 and have recently used these                   destabilization of Fe–S cluster catalytic sites, repair
                                  reconstructed network models to identify the sets of                      of these damaged Fe–S clusters and related changes
                                  genes, associated functional groups and biochemical path-                 in iron-related gene expression9.
                                  ways that act in concert to mediate bacterial responses                       Building on this work, it was later shown that all
                                  to antibiotics8–10,119. Below we highlight some mechanistic               major classes of bactericidal antibiotics (including
                                  insights that have been obtained from antibiotic network                  β-lactams, aminoglycosides and quinolones) pro-
                                  biology, and discuss some opportunities and challenges                    mote the generation of lethal hydroxyl radicals in
                                  for this emerging area of research.                                       both Gram-negative and Gram-positive bacteria,
                                                                                                            despite the stark differences in their primary drug–
                                  A common mechanism for antibiotic-mediated cell                           target interactions8. Stress response network analysis
                                  death. As an example of the utility of studying bacterial                 methods used in this study suggested that antibiotic-
                                  stress responses at the systems level, biological network                 induced hydroxyl radical formation is the end prod-
                                  analysis methods were recently employed to identify                       uct of a common mechanism, in which alterations in
                                  new mechanisms that contribute to bacterial cell death                    central metabolism related to NADH consumption
                                  following topoisomerase II inhibition by the fluo-                        (increased TCA cycle and respiratory activity) are
                                  roquinolone antibiotic norfloxacin9. As noted above,                      crucial to superoxide-mediated iron–sulphur cluster
                                  quinolones are known to induce cell death through                         destabilization and stimulation of the Fenton reaction.
                                  the introduction of double-stranded DNA breaks fol-                       These predictions were validated by the results of addi-
                                  lowing arrest of topoisomerase function4. To identify                     tional phenotypic experiments, biochemical assays and
                                  additional contributions to cell death resulting from                     gene expression measurements, confirming that lethal
                                  topoisomerase II poisoning, reconstruction of stress                      levels of bactericidal antibacterials trigger a com-
                                  response networks was carried out following treatment                     mon oxidative damage cellular death pathway, which
                                  of E. coli with lethal concentrations of norfloxacin. This                contributes to killing by these drugs (fIG. 2).
                                  work identified an oxidative damage-mediated cell                             Most recently, the study of antibiotic-induced stress
                                  death pathway, which involves ROS generation and                          response networks has been aimed at determining
                                  a breakdown in iron regulatory dynamics following                         exactly how the primary effect of a given bactericidal
                                  norfloxacin-induced DNA damage. More specifically,                        triggers aspects of cell death that are common to all
Fenton reaction                   norfloxacin treatment was found to promote super-                         bactericidal drugs. For example, a comparative analy-
Reaction of ferrous iron (fe2+)
with hydrogen peroxide to
                                  oxide generation soon after topoisomerase II poison-                      sis of stress response networks, reconstructed using
produce ferric iron (fe3+) and    ing and to ultimately result in the generation of highly                  gene expression data from E. coli treated with amino-
a hydroxyl radical.               destructive hydroxyl radicals through the fenton                          cyclitols (spectinomycin, gentamicin and kanamycin),

NATuRe RevIeWS | Microbiology                                                                                                                        vOluMe 8 | juNe 2010 | 431

                                                        © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

                              Box 2 | Synthetic biology for antibacterial applications
                              The study of complex antibiotic-related cell death systems
                              can be aided by synthetic biology. Delivery of engineered
                              gene circuits that alter response network behaviour can serve
                                                                                                                              Synthetic
                              as a tool to experimentally examine antibiotic-mediated
                                                                                                                              perturbation
                              cell death pathways, as well as a means to enhance killing by
                              an antibiotic (see the figure).
                                Bacteriophages, which are bacterium-specific viruses,                      Antibiotic
                              show promise as an effective means to deliver network                    perturbed network
                              perturbations to bacteria to improve antibiotic
                                                                                                                                             Novel
                              lethality141,142. Bacteriophages have been used to enhance                                                     drug    Bacteriophage
                              killing of Escherichia coli by bactericidal antibiotics through                                                targets
                                                                                                            Network
                              the delivery of proteins that modify the oxidative stress                  reconstruction
                              response or inhibit DNA damage repair systems142.                            algorithms
                              Bacteriophages are species specific, so it may be possible
                              to use engineered bacteriophages to deliver antibiotic-
                              enhancing synthetic gene networks, therapeutic proteins
                                                                                                                            Antibiotics
                              or antimicrobial peptides that are highly specific for an              Increased antibiotic
                              infecting organism. This would allow efficient treatment of            susceptibility
                              a bacterial infection, while sparing the typical commensal
                              body flora (see the figure).

                                                                                                                                    Nature Reviews | Microbiology
                             was used to identify the incorporation of mistrans-                  in the same way to antibiotic treatment. Network biol-
                             lated proteins into the cell membrane as the trigger                 ogy approaches, which provide the field of antibiotic
                             for aminoglycoside-induced oxidative stress10 (fIG. 3).              research with an opportunity to view response mecha-
                             Interestingly, mistranslated membrane proteins were                  nisms of different bacterial species to various classes of
                             shown to stimulate radical formation by activating                   antibiotics, could be extended to the context of partic-
                             the Cpx and Arc two-component systems, ultimately                    ular infectious species, persistent infections or disease
                             altering TCA cycle metabolism; the TCA cycle had                     settings. As an example, it is generally accepted that
                             previously been implicated in bacterial susceptibility               Gram-negative bacteria are not susceptible to the glyco-
                             to aminoglycosides8,124.                                             peptide vancomycin or the depolarizing lipopeptide
                                 The discovery of the common oxidative damage                     daptomycin; however, a single gene, yfgL, was recently
                             cellular death pathway has important implications for                found that can make E. coli susceptible to glycolipid
                             the development of more effective antibacterial thera-               derivatives of vancomycin132. Gene expression profil-
                             pies. Specifically, it indicates that all major classes of           ing of daptomycin-treated S. aureus has revealed that
                             bactericidal drugs can be potentiated by inhibition                  daptomycin perturbs peptidoglycan synthesis through
                             of the DNA stress response network (that is, the SOS                 a mechanism involving the activation of cell wall stress
                             response), which plays a key part in the repair of                   systems and membrane depolarization133. Given these
                             hydroxyl radical-induced DNA damage. This may be                     findings, we might be able to combine our knowledge of
                             accomplished through the development of small mol-                   β-lactam- and aminoglycoside-induced gene signatures
                             ecules (for example, RecA inhibitors125) or synthetic                with the results of high-throughput screens at various
                             biology approaches (BOX 2).                                          drug doses to reconstruct drug-specific cell death net-
                                 ROS, such as superoxide and hydroxyl radicals, are               works that use Ygfl as a network anchor. Predicted func-
                             highly toxic and have deleterious effects on bacterial               tional and regulatory relationships between enriched
                             physiology 123,126,127, even under steady-state conditions.          genes could then be used to determine the secondary
                             There is still much to be learned about how oxidative                effects of lipopeptide antibiotics and gain insight into
                             stress-related changes in bacterial physiology affect                the different properties of this drug in Gram-negative
                             antibiotic-mediated cell death and the emergence of                  and Gram-positive bacteria.
                             resistance128,129. For example, it was recently discov-                  Moreover, the development of comparative net-
                             ered that endogenous nitric oxide produced by bac-                   work biology techniques will be essential to further
                             teria with nitric oxide synthases can protect against                our understanding of how species-specific differ-
                             ROS-mediated cell death130. In addition, considering                 ences manifest themselves in divergent drug-specific
                             bacteria have developed mechanisms to avoid ROS                      cell death networks and variations in physiological
                             produced by phagocytes of the immune system 131, it                  responses. These methods could be particularly use-
                             will be interesting to explore, from a systems-level per-            ful when examining pathogenic bacteria with sparse
                             spective, the relationship between immune-mediated                   systems-level data (such as Shigella or Salmonella spp.)
                             and drug-mediated cell death.                                        that are closely related to well-studied bacteria (such
                                                                                                  as E. coli). Through a greater understanding of the
                             Opportunities and challenges for antibiotic network                  biological networks that are related to an individual
                             biology. One of the more intriguing aspects of antibac-              drug target, we eventually might be able to search for
                             terial therapies is that not all bacterial species respond           meaningful network homologues among species in

432 | juNe 2010 | vOluMe 8                                                                                                       www.nature.com/reviews/micro

                                                   © 2010 Macmillan Publishers Limited. All rights reserved
REVIEWS

Antimicrobial peptide
                                    the same spirit as we currently search for gene homo-                        Concluding remarks
A short, naturally occurring        logues. Network-based efforts could also lead to the                         Drug-resistant bacterial infections are becoming more
cationic peptide that has           development of species-specific treatments, includ-                          prevalent and are a major health issue facing us today.
antibacterial properties            ing synthetic biology-derived therapies (BOX 2), which                       This rise in resistance has limited our repertoire of
through its ability to interfere
                                    could be useful in killing off harmful, invasive bacte-                      effective antimicrobials, creating a problematic situa-
with bacterial membranes.
                                    ria, while leaving our normal bacterial flora intact.                        tion that has been exacerbated by the small number of
                                        Finally, bacterial network analyses will also be use-                    new antibiotics introduced in recent years. The com-
                                    ful in the study of non-classical antibacterial agents                       plex effects of bactericidal antibiotics discussed in this
                                    that induce cell death. Antimicrobial peptides are short                     Review provide a large playing field for the develop-
                                    cationic peptides that are thought to kill through                           ment of new antibacterial compounds, as well as adju-
                                    interactions with the membrane that result in pore                           vant molecules and synthetic biology constructs, that
                                    formation134,135. However, the mode of action of many                        could enhance the potency of current antibiotics. It
                                    antimicrobial peptides could, in fact, be more complex,                      will be important to translate our growing understand-
                                    and cell death networks uncovered for existing anti-                         ing of antibiotic mechanisms into new clinical treat-
                                    biotics could be used as mechanistic templates to study                      ments and approaches so that we can effectively fight
                                    cellular responses induced by antimicrobial peptides.                        the growing threat from resistant pathogens.

1.  Walsh, C. Antibiotics: actions, origins, resistance (ASM   13. Gellert, M., Mizuuchi, K., O’Dea, M. H. & Nash, H. A.      25. Kampranis, S. C. & Maxwell, A. The DNA gyrase-
    Press, Washington, D.C., 2003).                                DNA gyrase: an enzyme that introduces superhelical             quinolone complex. ATP hydrolysis and the mechanism
2.  Fleming, A. On antibacterial action of culture of              turns into D.NA. Proc. Natl Acad. Sci. USA 73,                 of DNA cleavage. J. Biol. Chem. 273, 22615–22626
    penicillium, with special reference to their use in            3872–3876 (1976).                                              (1998).
    isolation of B. influenzae. Br. J. Exp. Pathol. 10,        14. Sugino, A., Peebles, C. L., Kreuzer, K. N. & Cozzarelli,       Reveals that quinolone antibiotic binding to the
    226–236 (1929).                                                N. R. Mechanism of action of nalidixic acid:                   topoisomerase II–DNA complex occurs before DNA
3.  Taubes, G. The bacteria fight back. Science 321,               purification of Escherichia coli nalA gene product and         strand breakage and that DNA cleavage can occur,
    356–361 (2008).                                                its relationship to DNA gyrase and a novel nicking-            albeit at a slower rate, in the presence of the drug
4.  Drlica, K., Malik, M., Kerns, R. J. & Zhao, X.                 closing enzyme. Proc. Natl Acad. Sci. USA 74,                  based on the results of ATP hydrolysis and DNA
    Quinolone-mediated bacterial death. Antimicrob.                4767–4771 (1977).                                              cleavage assays.
    Agents Chemother. 52, 385–392 (2008).                      15. Gellert, M., Mizuuchi, K., O’Dea, M. H., Itoh, T. &        26. Yoshida, H., Bogaki, M., Nakamura, M. & Nakamura, S.
5.  Floss, H. G. & Yu, T. W. Rifamycin-mode of action,             Tomizawa, J. I. Nalidixic acid resistance: a second            Quinolone resistance-determining region in the DNA
    resistance, and biosynthesis. Chem. Rev. 105,                  genetic character involved in DNA gyrase activity.             gyrase gyrA gene of Escherichia coli. Antimicrob.
    621–632 (2005).                                                Proc. Natl Acad. Sci. USA 74, 4772–4776 (1977).                Agents Chemother. 34, 1271–1272 (1990).
6.  Tomasz, A. The mechanism of the irreversible                   References 14 and 15 discuss the results of                27. Morais Cabral, J. H. et al. Crystal structure of the
    antimicrobial effects of penicillins: how the beta-            complementary in vivo and in vitro studies that                breakage-reunion domain of DNA gyrase. Nature 388,
    lactam antibiotics kill and lyse bacteria. Annu. Rev.          characterized the genetic locus (nalA, later gyrA)             903–906 (1997).
    Microbiol. 33, 113–137 (1979).                                 and the basic mechanism of quinolone antibiotic            28. Heddle, J. & Maxwell, A. Quinolone-binding pocket of
    This seminal review of β‑lactam‑mediated cell death            action (prevention of DNA duplex strand                        DNA gyrase: role of GyrB. Antimicrob. Agents
    discusses the intricacies of killing by various                rejoining yielding double‑stranded DNA breaks),                Chemother. 46, 1805–1815 (2002).
    members of this antibiotic class in terms of the               while postulating on the composition and                   29. Goss, W. A., Deitz, W. H. & Cook, T. M. Mechanism of
    specific drug‑inhibited protein targets and their              energetic requirements of DNA gyrase activity.                 action of nalidixic acid on Escherichia coli.II. Inhibition
    related cell wall maintenance functions.                   16. Hooper, D. C. & Rubinstein, E. Quinolone                       of deoxyribonucleic acid synthesis. J. Bacteriol. 89,
7.  Vakulenko, S. B. & Mobashery, S. Versatility of                antimicrobial agents (ASM Press, Washington, D.C.,             1068–1074 (1965).
    aminoglycosides and prospects for their future. Clin.          2003).                                                     30. Snyder, M. & Drlica, K. DNA gyrase on the bacterial
    Microbiol. Rev. 16, 430–450 (2003).                        17. Rubinstein, E. History of quinolones and their side            chromosome: DNA cleavage induced by oxolinic acid.
8.  Kohanski, M. A., Dwyer, D. J., Hayete, B., Lawrence,           effects. Chemotherapy 47 (Suppl. 3), 3–8 (2001).               J. Mol. Biol. 131, 287–302 (1979).
    C. A. & Collins, J. J. A common mechanism of cellular      18. Lu, T. et al. Enhancement of fluoroquinolone activity      31. Cox, M. M. et al. The importance of repairing stalled
    death induced by bactericidal antibiotics. Cell 130,           by C-8 halogen and methoxy moieties: action against            replication forks. Nature 404, 37–41 (2000).
    797–810 (2007).                                                a gyrase resistance mutant of Mycobacterium                32. Courcelle, J. & Hanawalt, P. C. RecA-dependent
    Reveals that treatment of Gram‑positive and                    smegmatis and a gyrase-topoisomerase IV double                 recovery of arrested DNA replication forks. Annu. Rev.
    Gram‑negative bacteria with lethal levels of                   mutant of Staphylococcus aureus. Antimicrob. Agents            Genet. 37, 611–646 (2003).
    bactericidal antibiotics induces the formation of              Chemother. 45, 2703–2709 (2001).                           33. Howard, B. M., Pinney, R. J. & Smith, J. T. Function of
    hydroxyl radicals through a common mechanism               19. Chen, C. R., Malik, M., Snyder, M. & Drlica, K. DNA            the SOS process in repair of DNA damage induced by
    involving drug‑induced changes in NADH                         gyrase and topoisomerase IV on the bacterial                   modern 4-quinolones. J. Pharm. Pharmacol. 45,
    consumption and central metabolism, notably the                chromosome: quinolone-induced DNA cleavage.                    658–662 (1993).
    TCA cycle.                                                     J. Mol. Biol. 258, 627–637 (1996).                         34. Cirz, R. T. et al. Inhibition of mutation and combating
9.  Dwyer, D. J., Kohanski, M. A., Hayete, B. & Collins,           Identifies topoisomerase IV as a second target of              the evolution of antibiotic resistance. PLoS Biol. 3,
    J. J. Gyrase inhibitors induce an oxidative damage             fluoroquinolone antibiotics in Gram‑negative                   e176 (2005).
    cellular death pathway in Escherichia coli. Mol. Syst.         bacteria and characterizes subtle but crucial              35. Guerin, E. et al. The SOS response controls integron
    Biol. 3, 91 (2007).                                            differences in the mechanism of killing by various             recombination. Science 324, 1034 (2009).
    Describes the physiological responses of E. coli               quinolone drugs.                                           36. Beaber, J. W., Hochhut, B. & Waldor, M. K. SOS
    following inhibition of topoisomerase by a                 20. Drlica, K. & Zhao, X. DNA gyrase, topoisomerase IV,            response promotes horizontal dissemination of
    fluoroquinolone and a peptide toxin, which include             and the 4-quinolones. Microbiol. Mol. Biol. Rev. 61,           antibiotic resistance genes. Nature 427, 72–74
    activation of the superoxide stress response and               377–392 (1997).                                                (2004).
    increased Fe–S cluster synthesis. These                    21. Munoz, R. & De La Campa, A. G. ParC subunit of DNA         37. Lewin, C. S., Howard, B. M. & Smith, J. T. Protein- and
    physiological changes result in hydroxyl radical               topoisomerase IV of Streptococcus pneumoniae is a              RNA-synthesis independent bactericidal activity of
    production, which contributes to cell death.                   primary target of fluoroquinolones and cooperates              ciprofloxacin that involves the A subunit of DNA
10. Kohanski, M. A., Dwyer, D. J., Wierzbowski, J.,                with DNA gyrase A subunit in forming resistance                gyrase. J. Med. Microbiol 34, 19–22 (1991).
    Cottarel, G. & Collins, J. J. Mistranslation of                phenotype. Antimicrob. Agents Chemother. 40,               38. Wang, X., Zhao, X., Malik, M., & Drlica, K.
    membrane proteins and two-component system                     2252–2257 (1996).                                              Contribution of reactive oxygen species to pathways of
    activation trigger antibiotic-mediated cell death. Cell    22. Belland, R. J., Morrison, S. G., Ison, C. & Huang,             quinolone-mediated bacterial cell death. J. Antimicrob.
    135, 679–690 (2008).                                           W. M. Neisseria gonorrhoeae acquires mutations in              Chemother. 65, 520–524 (2010).
    Shows that systems which facilitate membrane                   analogous regions of gyrA and parC in                      39. Kolodkin-Gal, I., Sat, B., Keshet, A. & Engelberg-Kulka,
    protein trafficking are central to aminoglycoside‑             fluoroquinolone-resistant isolates. Mol. Microbiol. 14,        H. The communication factor EDF and the toxin-
    induced oxidative stress and cell death. This occurs           371–380 (1994).                                                antitoxin module mazEF determine the mode of action
    by signalling through the redox‑ and the envelope          23. Critchlow, S. E. & Maxwell, A. DNA cleavage is not             of antibiotics. PLoS Biol. 6, e319 (2008).
    stress‑responsive two‑component systems.                       required for the binding of quinolone drugs to the         40. Dukan, S. et al. Protein oxidation in response to
11. Espeli, O. & Marians, K. J. Untangling intracellular           DNA gyrase-DNA complex. Biochemistry 35,                       increased transcriptional or translational errors. Proc.
    DNA topology. Mol. Microbiol. 52, 925–931                      7387–7393 (1996).                                              Natl Acad. Sci. USA 97, 5746–5749 (2000).
    (2004).                                                    24. Marians, K. J. & Hiasa, H. Mechanism of quinolone          41. Hartmann, G., Honikel, K. O., Knusel, F. & Nuesch, J.
12. Drlica, K. & Snyder, M. Superhelical Escherichia coli          action. A drug-induced structural perturbation of the          The specific inhibition of the DNA-directed RNA
    DNA: relaxation by coumermycin. J. Mol. Biol. 120,             DNA precedes strand cleavage by topoisomerase, IV.             synthesis by rifamycin. Biochim. Biophys. Acta 145,
    145–154 (1978).                                                J. Biol. Chem. 272, 9401–9409 (1997).                          843–844 (1967).

NATuRe RevIeWS | Microbiology                                                                                                                         vOluMe 8 | juNe 2010 | 433

                                                               © 2010 Macmillan Publishers Limited. All rights reserved
You can also read