You are on page 1of 16

Biosci. Biotechnol. Biochem.

, 70 (5), 1060–1075, 2006

Review
Antibiotic Resistance in Bacteria and Its Future
for Novel Antibiotic Development
Hiroshi YONEYAMAy and Ryoichi K ATSUMATA
Laboratory of Animal Microbiology, Department of Microbial Biotechnology,
Graduate School of Agricultural Science, Tohoku University,
1-1 Tsutsumi-dori Amamiya-machi, Aoba-ku, Sendai 981-8555, Japan

Since the first introduction of the sulfa drugs and dence that modern medicine would prevail against
penicillin into clinical use, large numbers of antibiotics infectious diseases.1) However, contrary to this opti-
have been developed and hence contributed to human mism, infectious diseases are still the second-leading
health. But extensive use of antibiotics has raised a cause of death worldwide and the third-leading cause of
serious public health problem due to multiantibiotic death in developed countries.2,3) The global public
resistant bacterial pathogens that inevitably develop health problem has been caused by emerging infections
resistance to every new drug launched in the clinic. not yet recognized, re-emerging infections experienced
Consequently, there is a pressing need to develop new previously that have reappeared in more virulent forms,
antibiotics to keep pace with bacterial resistance. Recent and antimicrobial-resistant bacterial infections.2) Anti-
advances in microbial genomics and X-ray crystallog- microbial resistance was recognized soon after the
raphy provide opportunities to identify novel antibacte- deployment of sulfonamides and penicillins, and it
rial targets for the development of new classes of now appears that the emergence of antibiotic-resistant
antibiotics and to design more potent antimicrobial bacteria is inevitable to most every new drug.1) To
compounds derived from existing antibiotics respective- prevent the emergence and dissemination of resistant
ly. To prevent and control infectious diseases caused by bacteria, continuing efforts to develop new antibacterial
multiantibiotic resistant bacteria, we need to under- agents are needed.4,5)
stand more about the molecular aspects of the patho- In this review, we focus on antibacterial agents,
gens’ physiology and to pursue ways to prolong the life resistance to them, and their future development.
of precious antibiotics.
I. Definition of Antibiotics
Key words: antibiotics; multiantibiotic resistance; bac-
teria; chemotherapy Antibiotics, literally ‘‘against life,’’ are chemical
compounds produced by actinomycetes, fungi, or bac-
The history of humankind can be regarded from a teria that interfere with some essential bacterial structure
medical point of view as a struggle against infectious or process with no effects on the eukaryotic host having
diseases. Although infectious diseases were the leading the infectious agents.6) Antibiotics exert two effects on
cause of death worldwide at the beginning of the bacteria: bacteriostatic and bactericidal. Bacteriostatic
twentieth century, the introduction of sulfa drugs and agents prevent the bacterial cells from growing, as
penicillin into clinical use in the 1930s and 1940s exemplified by the drug chloramphenicol, and bacter-
respectively had a striking impact on the treatment of icidal agents kill bacteria, as exemplified by penicillin.6)
infectious diseases and decreased mortality dramatical- Although the term ‘‘antibiotic’’ was first introduced
ly.1) After the clinical success of penicillin, intensive by Waksman in 1942 to denote chemical substances
screening for antibacterial agents led to the discovery produced by a microorganism, semisynthetic modifica-
and successful clinical development of streptomycin, tions of natural products have produced a large variety
chloramphenicol, tetracycline, erythromycin, rifamycin, of antibacterial agents, such as -lactam antibiotics and
vancomycin, and cephalosporin between 1940 and 1960. macrolides, that are also called antibiotics.7) This
‘‘The era of antibiotics’’ led to optimism that infectious definition distinguishes antibiotics from antibacterial
diseases can be controlled and prevented, and confi- agents totally synthesized by the chemist. At present,

y
To whom correspondence should be addressed. Tel: +81-22-717-8915; Fax: +81-22-717-8708; E-mail: yoneyama@bios.tohoku.ac.jp
Abbreviations: PBP, penicillin binding protein; MRSA, methicillin resistant Staphylococcus aureus; VRE, vancomycin resistant Enterococci; MFS,
major facilitator superfamily; RND, resistance-nodulation-division; SMR, small multidrug resistance; MATE, multidrug and toxic compounds
extrusion; ABC, ATP-binding cassette; MLSB , macrolides, lincosamides, and streptogramin B; ORF, open reading frame; IVET, in vitro expression
technology; DFI, differential fluorescence induction; STM, signature-tagged mutagenesis; PKS, polyketide synthase; PDF, peptide deformylase; FAS,
fatty acid synthase; DHP, dihydropteroate; DHF, dihydrofolate; THF, tetrahydrofolate; MCIPC, cloxacillin; DOC, deoxycholate; AC, acriflavine; TC,
tetracycline; Erm, erythromycin ribosome methylation; EM, erythyromycin; CAT, chloramphenicol acetyltransferase
Antibiotic Resistance in Bacteria 1061

PBP
Cytoplasmic membrane

Outer membrane
Cell wall biosynthesis
Cell wall
(peptidoglycan)

DNA/RNA biosynthesis

DNA gyrase DNA GTP

DNA
dTTP DHP
RNA mRNA
polymerase
dTMP DHF
ribosome
dUMP THF
mRNA
Folic acid
metabolism
Protein biosynthesis

Gram-negative Gram-positive

Fig. 1. Major Targets of Antibitoics.


PBP, penicillin binding protein; DHP, dihydropteroate; DHF, dihydrofolate; THF, tetrahydrofolate.

there are three classes of chemically synthesized anti- have a peptidoglycan layer as part of their cell-wall
bacterial agents in clinical use: the sulfa drugs intro- structure. The peptidoglycan layer of Gram-positive
duced in the 1930s, the quinolones introduced in the bacteria is generally much thicker than that of Gram-
1960s, and an oxazolidinone approved in the United negative bacteria, and constitutes the outermost layer.9)
States in 2000.4,7) These purely synthesized antibacterial The peptidoglycan undergoes crosslinking of the glycan
agents are also treated as antibiotics in this review. strands by the action of transglycosidase, and of the
peptide strands by the action of transpeptidase.6) Bifunc-
II. Targets of Antibiotics tional enzymes possessing both transglycosidase and
transpeptidase domains are the target sites of penicillins
Although a large number of antibiotics are used and cephaolosporins, collectively called -lactams.10) -
clinically, the variety of targets that they inhibit is lactams attack and acylate the active site of the
limited. Antibiotics are usually classified on the basis of transpeptidase, leading to inactivation of the enzymes,
their chemical structure and mode of action. To under- hence they are called penicillin-binding proteins or
stand how antibiotics work and why bacteria become PBPs.10)
resistance to them, a brief description of the targets of In addition to -lactams, vancomycin, considered a
the main classes of antibiotics is required. The main last resort among antibiotics, also targets peptidoglycan
classes of antibiotics inhibit four classical targets to block cell-wall biosynthesis.11) It binds to the D-Ala4 -
(Fig. 1): (i) cell wall biosynthesis, (ii) protein biosyn- D-Ala5 moiety of pentapeptides and thereby keeps the
thesis, (iii) DNA and RNA biosynthesis, and (iv) folate substrates away from transpeptidase and transglycosi-
biosynthesis.8) dase.11) This substrate sequestration leads to the failure
of peptidoglycan crosslinks, making the cell wall
1. Inhibition of bacterial cell wall biosynthesis susceptible to osmolysis.
Since bacteria have high intracellular pressure, they
must protect themselves from osmolysis with a rigid 2. Inhibition of protein biosynthesis
peptidoglycan, a meshwork of strands of glycan and Protein biosynthesis, a central reaction for cellular
peptide covalently crosslinked (Fig. 1). Gram-negative function, is catalyzed by ribosomes, composed of two
bacteria, such as Escherichia coli and Pseudomonas nucleoprotein subunits, 30S and 50S subunits with about
aeruginosa, and Gram-positive bacteria, such as Staph- two-thirds RNA and one-thirds protein, in prokaryotes
ylococcus aureus and Streptococcus pneumoniae, both (Fig. 1).12) This machinery of prokaryotes differs sub-
1062 H. YONEYAMA and R. KATSUMATA

stantially from the eukaryotic counterpart, which ex- rifamycin B, isolated from a strain of Amycolatopsis
plains the effectiveness and selective toxicity of many mediterranea (formerly known as Streptomyces medi-
clinically important antibiotics. The X-ray structure of terranea or Nocardia mediterranea). It is used princi-
30S and 50S subunits12–14) and of 70S ribosome15) pally in the treatment of tuberculosis and leprosy.22)
reveals the architecture of this giant protein biosynthesis Rifampicin inhibits RNA polymerase by binding to the
machinery in detail, and the peptidyltransferase activity  subunit of the enzyme at an allosteric site, not at the
deriving from the catalytic ribozyme domain of the 23S active site, as revealed by resistant mutations in clinical
rRNA with no obvious assistance from protein sub- isolates of Mycobacterium tuberculosis and M. leprae26)
units.12) Due to the large number of steps involved in and X-ray structural analysis of the Thermus aquaticus
protein biosynthesis, initiation, elongation, and termi- core RNA polymerase complexed with rifampicin.27)
nation, it is not surprising that there are many steps to be
blocked, or that many clinically useful antibiotics that 4. Other targets
interact with the conserved sequences of the 16S rRNA Folic acid metabolism. The longest-used antibacterial
of the 30S subunit (aminoglycosides and tetracyclines) agents are the sulfa drugs, or sulfonamides, first
and the 23S rRNA of the 50S subunit (macrolides, introduced in the 1930s.6,8) The current generation of
chloramphenicol, lincosamides, and quinupristin-dalfo- sulfa drugs is sulfamethoxazole, which is used in
pristin) have been found.16–18) combination with trimethoprim.22) Each antibacterial
agent is bacteriostatic when used individually, but they
3. Inhibition of DNA and RNA biosynthesis are bactericidal when used together. This effect makes
DNA replication is an essential process for all the combination recipe remains clinically relevant in the
organisms. Bacterial chromosomal DNA is packed in a treatment of many bacterial infections, such as urinary
highly twisted (supercoiling) state in the cell, and and respiratory tract infections.22) Each of the drugs
dynamically changes its structure during the replication inhibits distinct steps in folic acid metabolism (Fig. 1).
process.19) The enzymes involved in interconversion of a Sulfamethoxazole inhibits dihydropteroate synthase in
topologically different form of the DNA are called the biosynthetic pathway of folate in a competitive
topoisomerases. The DNA topoisomerases are classified manner with higher affinity for the enzyme than a
into type I and type II. Type I topoisomerase transiently natural substrate, p-aminobenzoic acid.22) The resulting
breaks either one strand at a time (also called a nick), dead-end product cannot be a substrate for the next
while type II topoisomerase breaks both strands at the enzyme, dihydrofolate synthase. Since humans do not
same time.20) Due to the essential character of DNA have dihydropteroate synthase, sulfa drugs achieve high
replication, it is natural that microorganisms such as selectivity against bacteria. Trimethoprim inhibits dihy-
streptomycetes produce metabolites such as novobiocin drofolate reductase, a key enzyme providing a one-
and coumermycin that kill their neighbors by inhibiting carbon carrier, tetrahydrofolate, for the synthesis of
the DNA replication process, the target of which has deoxythymidylate, purine nucleotides, and the amino
been found to be type II topoisomerase, specifically acids glycine and methionine.28) In contrast to dihy-
DNA gyrase.20) Nalidixic acid, the prototype quinolone, dropteroate synthase, dihydrofolate reductase is essen-
was discovered to possess antibacterial activity in tial for all organisms. Trimethoprim is a competitive
196221) and was approved for urinary tract infections inhibitor of dihydrofolate reductase, and is 50,000 to
in 1964.22) This finding led to more potent quinolone 100,000 times more active against bacterial enzymes
antibiotics, such as norfloxacin, ciprofloxacin, ofloxacin, than against the human counterparts, achieving high
and gatifloxacin, also called fluoroquinolone due to the selectivity.22) A combination of trimethoprim and sulfa
presence of a fluorine substituent. These quinolones drugs acting at distinct steps on the same biosynthetic
target the DNA gyrase and affect the double-strand pathway shows synergy and a reduced mutation rate for
cleavage/double-strand religation equilibrium in the resistance as compared to the use of each drug alone,
gyrase catalytic reaction, in such a way that the cleaved providing validation of combination chemotherapy.
complex accumulates by stabilization of the complex in Cell membranes. The integrity of cell membranes,
the presence of quinolones.20) A second type II top- including the outer membrane of Gram-negative bac-
oisomerase, known as topoisomerase IV, is also impli- teria, is essential to all organisms. Cationic peptides
cated in DNA replication and decatenation of knotted exemplified by the polymyxins, a group of cyclic
daughter chromosomes at the end of bacterial DNA peptides with a fatty acid chain attached to the peptide,
replication,23) and thus is an important target, probably attack the cytoplasmic membrane of Gram-positive and
the primary one in Staphylococcus aureus.24) Gram-negative bacteria, and the outer membrane of
Transcription is an essential process for decoding Gram-negative bacteria.29) The interaction of the cat-
genetic information from DNA to RNA in all organisms. ionic peptide and the membrane disrupts membrane
RNA polymerase of bacteria composed of different organization and causes increased permeability of cell
subunits with a stoichiometry of 2 0 ! to form the core components, which eventually leads to cell-killing.29)
enzyme catalyzes transcription.25) Rifampicin (known in The toxicity of polymyxin B due to its membrane-active
some countries as rifampin) is a semisynthetic version of nature restricts its use in topical applications.29)
Antibiotic Resistance in Bacteria 1063

A second set of antibacterial peptides is bacteriocins and avoparicin, pushed the European Union to ban the
produced by both Gram-positive and Gram-negative use of avoparcin in 1997 for growth promotion on the
bacteria to compete with neighboring bacterial cells.30) basis of the ‘‘Precautionary Principle.’’48)
With some exceptions, most of these bacteriocins, such To combat these life-threatening problems, continu-
as nisin, permeabilize target cell membranes.29,30) ing efforts to develop new antibiotics are obviously
Studies on the mechanism of action of nisin show that needed. Since the introduction of nalidixic acid in 1962,
it binds with high affinity to a docking molecule, lipid II, a totally new class of antibiotic oxazolidinones (line-
a precursor of cell-wall biosynthesis, leading to the zolid) and cyclic lipopeptides (daptomycin) were ap-
hypothesis of the dual functionality of nisin.31,32) Recent proved in 2000 and 2003 respectively for clinical use in
structural analysis of the nisin-lipid II complex reveals the United States.4,8) Although these new antibiotics are
that a novel lipid II recognition motif of nisin and related effective against vancomycin-resistant MRSA and En-
lantibiotics (lanthionine-containing antibiotic peptides) terococci, it is clear that the problematic pathogens will
binds to the pyrophosphate moiety of lipid II. This eventually develop resistance to these compounds too,
provides a basis for rational design of future anti- and that the therapeutic lifetime of the new antibiotics
biotics.33) will be shortened if we use them indiscriminately.
Indeed, clinical isolates of S. aureus and Enterococcus
III. Resistance to Antibiotics faecium showing linezolid resistance were first reported
in 2001, a year after launch.49,50)
Since sulfonamide and penicillin were introduced into Antibiotic resistance can be either intrinsic (natural)
clinical use in the 1930s and 1940s respectively, people or acquired. An example of intrinsic resistance is
were captured by the illusion that infectious diseases Pseudomonas aeruginosa, whose low membrane per-
were totally controlled by antibiotics. The widespread meability is likely to be a main reason for its innate
use of antibiotics, however, imposes strong selection resistance to many antibiotics.9,51) Acquired resistance is
pressure for the development of antibiotic resistance, a caused by acquisition of a plasmid and/or transposon
presentday major public health problem.4,34) We must that harbors determinants for resistance, or by chromo-
now accept that the occurrence of antibiotic resistance is some mutation.35,42) The major mechanisms of antibiotic
inevitable, that is, it is only a question of time.35) As the resistance are (i) prevention of accumulation of anti-
frequency of antibiotic use increases, the speed of biotics either by decreasing uptake or increasing efflux,
resistance development also increases, thereby reducing (ii) inactivation of antibiotics either by hydrolysis or by
the effectiveness of the antibiotics. Clinically significant modification, and (iii) qualitative alteration of the target
resistance appears after months to years whenever a new that reduces the affinity for antibiotics either by mutation
antibiotic is introduced into medical use.36,37) For or by target modification, or quantitatively by over-
example, penicillin resistance was detected a few years production of the target (Fig. 2).9,26,35,36)
after its clinical debut in 1942,38) and streptomycin, a
year after its discovery in 1944.35,39) In the exceptional
case of vancomycin, its resistance appeared almost 30 A
years (in 1987) after its clinical introduction.40) The long
delay was likely due in part to limited use of
vancomycin in the first 25 years, since other effective
antibiotics were available during the ‘‘antibiotic era’’
of the 1950s through the 1960s. But then indiscriminate
use of antibiotics resulted in the emergence of methi-
cillin-resistant Straphylococcus aureus (MRSA), which C
exhibits multiantibiotic resistance against many struc-
turally unrelated antibiotics.41) In turn, vancomycin- B
resistant Enterococci (VRE) appeared in 1986 due to
or
widespread use of vancomycin, which was regarded as
the antibiotic of last resort.11,42) Then the discovery of P
partially vancomycin-resistant strains of S. aureus in
1997 lead to the first case of fully vancomycin-resisitant
S. aureus.43)
More to the point, what makes the problem more
complicated is that about 50% of antibiotics are used for
prophylaxis, chemotherapy, and growth promotion in Fig. 2. Principle Mechanisms of Antibiotic Resistance.
A, Prevention of accumulation of antibiotics by decreasing uptake
animals.42,44–46) There is now evidence that antibiotic
or increasing efflux. B, Inactivation of antibiotics by hydrolysis or
use in animals plays a role in the development of modification, in this case, phosphorylation. C, Alteration of targets
antibiotic-resistance in human pathogens.47) A case of qualitatively to reduce the affinity for antibiotics or quantitatively by
causal relationship of the glycopeptides, vancomycin overproduction of the target.
1064 H. YONEYAMA and R. KATSUMATA

1. Efflux pumps
Prevention of antibiotic access to their targets results A
from either decreasing the influx or increasing the efflux
of antibiotics across biological membranes (Fig. 2).9,52)
Pseudomonas aeruginosa, an opportunistic pathogen,
exemplifies this mechanism of antibiotic resistance.
Mutants lacking an outer membrane porin channel,
OprD2, decrease the uptake of carbapenem, a -lactam
antibiotic, and increase resistance to this antibiotic due
to low accessibility of the drug to its target, penicillin-
binding protein.9,53) A mutation resulting in overexpres-
sion of a multidrug efflux pump, MexAB-OprM, renders
this organism resistant to a wide variety of structurally
unrelated antibiotics.54,55) Since a loss-of-function mu-
tation in the MexAB-OprM efflux pump causes hyper-
sensitivity toward various antibiotics, the intrinsic B Side view Vertical view
resistance of this organism appears to be due mainly
to an active efflux of antibiotics.54,56)
OprM
From the first indication that plasmid-coded tetracy- trimer
cline resistance in E. coli is due to energy-dependent
efflux of the drug,52) this mechanism of antibiotic
resistance, namely transmembrane efflux of antibiotics, MexA
attracted scientists’ attention, and led to the recognition
of the widespread presence of multidrug efflux pumps in
bacteria that create resistance to clinically important
drugs.9,51) The efflux pumps are grouped into five
families based on their mechanisms and primary
sequence homologies: (i) the major facilitator super- MexB
trimer
family (MFS), (ii) the resistance-nodulation-division
(RND) family, (iii) the small multidrug resistance
(SMR) family, (iv) the multidrug and toxic compounds
extrusion (MATE) family, and (v) the ATP-binding
cassette (ABC) superfamily.57) Each member of the first Fig. 3. 3D Structure of Multidrug Exporter.
A, Cutaway view displaying the solvent-accessible surface of
four groups is a secondary transporter that uses proton
AcrB. The front protomer is removed. Broken lines indicate the
motive force (or sodium ion motive force for the MATE putative membrane boundaries and the framework of the funnel and
family) to expel antibiotics from cells.57) Members of cavity. Broken arrows indicate the putative substrate translocation
the ABC superfamily are primary transporters that use pathway. Pore helices and transmembrane helices comprising a
energy liberated by ATP hydrolysis such as LmrA of groove are depicted in yellow and green respectively. MCIPC,
cloxacillin; DOC, deoxycholate; AC, acriflavine; TC, tetracycline.
Lactococcus lactis.58)
Adapted with permission from (67). B, Assembly model of the
The ubiquitous multidrug efflux pumps raised the MexAB-OprM effulx pump. The figure is based on the 3-fold
question how they recognize and transport multiple MexA-dimer model. Adapted with permission from (61).
structurally unrelated antibiotics, and the question what
their natural physiological role is. Recently, there has
been tremendous progress in structural analysis of stoichiometry of the pump is predicted to be 1:1:1, 1:1:2,
multidrug efflux pumps and their cognate regulators or 1:1:3 for MexB:OprM:MexA.61,62,65) Quantitative
that has provided significant insight into these ques- analysis of each subunit in intact cells supports the
tions.57,59) Efflux pumps of the RND family are 3-fold MexA-dimer model in which the functional pump
composed of an inner membrane proton antiporter, a is composed of MexB trimer, OprM trimer, and three
periplasmic membrane fusion protein, and an outer dimers of MexA (Fig. 3B).61,66) The mechanism of
membrane exit channel.9,51,57) The crystal structures multidrug efflux by the tripartite pump has been
of an inner membrane component, AcrB of E. coli suggested by the striking features of the AcrB struc-
(Fig. 3A),60) a periplasmic protein, MexA of P. aerugi- ture.60) Substrates are collected from the inner and outer
nosa (Fig. 3B),61,62) and outer membrane channels, TolC leaflet of the plasma membrane through vestibules,
of E. coli63) and OprM of P. aeruginosa (Fig. 3B),64) which are formed between AcrB protomers, captured on
have been solved. It has been suggested that the the wall of the central cavity, and then actively trans-
functional tripartite efflux pump of P. aeruginosa ported through the pore into the TolC channel
comprises MexB trimer, a homolog of AcrB, OprM (Fig. 3A).60,67,68) The wide substrate specificity of the
trimer, and three to nine protomers of MexA. Hence the multidrug efflux pump can be explained by the large
Antibiotic Resistance in Bacteria 1065
18)
central cavity that interacts with different substrates, teins. Chloramphenicol acetyltransferases, widely dis-
mainly through hydrophobic interaction, as well as tributed among bacterial pathogens of all genera,
electrostatic interaction.69) inactivate chloramphenicol by transferring the acetyl
group from acetyl CoA (Fig. 4C).77) This modification
2. Inactivation of antibiotics of chloramphenicol again results in the considerably
Enzymatic inactivation of antibiotics either by hy- lower affinity of the antibiotic for the rRNA.
drolysis or by modification is a major mechanism of Enzymatic inactivation of antibiotics is one of the
resistance in pathogenic bacteria to natural antibiotics most common mechanisms of antibiotic resistance.35)
such as -lactams (penicillins and cephalosporins), The antibiotic-resistant clinical isolates in most cases
aminoglycosides, and chloramphenicol.35,36) The classic inherit the antibiotic-resistance genes on R (resistance)
case is the hydrolysis of the four-membered -lactam plasmids.35) Although definitive evidence for the origin
ring in penicillins and cephalosporins by hydrolytic of the antibiotic-resistant determinants remains to be
enzymes, -lactamases, resulting in products that cannot found, such resistance determinants most probably are
inhibit their targets, penicillin binding proteins (PBPs) acquired by pathogenic bacteria from a pool of resist-
(Fig. 4A).35,36) A group of -lactamases, which have ance genes in other microbial genera, including anti-
serine residue at their active sites, form penicilloyl-O- biotic-producing organisms.35) -lactamase was identi-
Ser -lactamase covalent intermediates as in the fied in 1940, several years before the introduction of
catalytic cycle of PBPs, which also form penicilloyl- penicillin into clinical use, indicating that there has been
O-Ser PBPs.70) The different lifetimes of each penicil- a pool of antibiotic resistant genes in nature.78)
loyl-O-Ser enzyme intermediate result in totally differ-
ent outcomes (Fig. 4A). The long lifetime of penicilloyl- 3. Alteration of the target
O-Ser PBPs results in inactivation of PBP transpeptidase In contrast to enzymatic inactivation of natural
activity, leading to prevention of cross-linking.70,71) By antibiotics, no enzymes that hydrolyze or modify man-
contrast, the deacylation rate of penicilloyl-O-Ser - made antibiotics such as sulfonamides, trimethoprim,
lactamase is fast, resulting in hydrolyzed, ring-opened and quinolones have been found.26) The mechanism of
penicilloic acid that is deactivated and nonfunctional as antibiotic resistance for these synthetic antibiotics and
a PBP pseudosubstrate.36) The structural and mechanical those natural products such as rifampicin, where
similarities of the serine -lactamases to the PBPs inactivating enzymes have not been found, is commonly
suggest evolution of the -lactamase from PBPs.71,72) achieved by target alterations.26)
In contrast to -lactams, the aminoglycoside anti- The most common mechanism of target alteration is
biotics do not possess hydrolytically fragile groups. the acquisition of new genes, carried on plasmids or
Thus, the enzymatic inactivation strategy for amino- transposons, that result in enzymatic modification of the
glycoside-resistant bacteria is covalently to modify normal target so that it does not bind the antibiotics. The
the OH and NH2 groups of the aminoglycosides erythromycin family of macrolides and the streptogra-
(Fig. 4B).73,74) There are three types of aminoglyco- min B family exemplify this mechanism of antibiotic
side-modifying enzymes: aminoglycoside acetyltrans- resistance.36,79) In erythromycin-resistant bacteria, the
ferases, which transfer the acetyl group from acetyl exocyclic amino group of a specific adenine residue,
CoA, aminoglycoside phosphotransferases, which trans- A2058, in 23S rRNA is mono- or dimethylated by an
fer the phosphoryl group from ATP, and aminoglycoside Erm (erythromycin ribosome methylation) methyltrans-
adenylyltransferases, which transfer the AMP group ferase, resulting in cross-resistance to macrolides,
from ATP.73,74) Aminoglycosides bind specifically to lincosamides, and streptogramin B (MLSB phenotype)
regions near the A site for aminoacyl-tRNA binding of (Fig. 5A).80,81) This modification reduces the affinity of
16S rRNA on the 30S ribosome subunit by hydrogen the rRNA for the antibiotics but does not interfere with
bond network through the various hydroxyl and amino protein biosynthesis. The recent crystal structure of the
substitutions of the antibiotics.16,17) This high affinity 50S subunit of the ribosome complexed with erythro-
binding of aminoglycosides with rRNA inhibits protein mycin revealed the importance of hydrogen bonds
biosynthesis.16,17) Enzymatic modification of aminogly- between erythromycin and A2058 of 23S rRNA.18)
cosides disrupts the hydrogen bond network, resulting in Erm is the major resistance mechanism in antibiotic-
the considerably lower affinity of the drugs for the resistant Staphylococcus aureus and is present in
rRNA. erythomycin-producing organisms as a self-protection
Chloramphenicol has been found to inhibit protein mechanism.79,80)
biosynthesis by preventing binding of tRNA to the A site Another example of target alteration can be seen in
of the ribosome.75,76) Recent structural analysis of the VRE. Five clinical phenotypes of vancomycin resistance
bacterial 50S ribosome subunit complexed with chlo- have been identified so far, VanA, VanB, VanC, VanD,
ramphenicol has revealed that the antibiotic interacts and VanE.82) VanA, VanB, VanD, and VanE are
specifically with various nucleotides of the peptidyl acquired resistance phenotypes, and VanC is an intrinsic
transferase cavity of the 23S rRNA through hydrogen resistance phenotype.83) The enzymes in VanA, VanB,
bonds without any interactions with ribosomal pro- and VanD resistance are involved in reprogramming of
1066 H. YONEYAMA and R. KATSUMATA

A
R R S
S
H2 O
+ β−lactamase
O N O N
H H
O COOH HO COOH
rapid
Ser deacylation
β−lactamase
R β−lactamase
S
N
O
COOH PBP H2 O
R S
O N
H very slow
O deacylation
COOH
Ser

PBP

B O

HN HO
HO O O OH
N−Acetylation HO NH2
OH HO
HO
O O
H2 N NH 2

NH 2 HO NH 2 HO
HO O O OH O HO O O OH
HO NH2 O−Phosphorylation O NH2
OH HO P OH HO
HO HO HO
O O OH O O
H2 N NH2 H2 N NH2

NH2 HO
AMP O O O OH
O−Adenylation HO NH2
OH HO
HO
O O
H2 N NH2

C
O
C CH3
nonenzymatic
OH OH O reaction O
CAT
OH O C CH3 OH
HN CHCl2 HN CHCl2 HN CHCl2
O2N O2N O2 N
O Acetyl−CoA O O

Acetyl−CoA
CAT

O
C CH3
O O
O C CH3
HN CHCl2
O2N
O

Fig. 4. Inactivation of Antibiotics Either by Hydrolysis or Modification.


A, Inactivation of penicillin by -lactamases. During the inactivation of PBPs by penicillins, hydrolysis of the penicilloyl-O-Ser PBP
intermediate is very slow due to the inaccessibility of the active site to water. By contrast, free access of water to the penicilloyl-O-Ser -
lactamases hydrolyses the intermediate very rapidly, leading to the inactivation of penicillins. B, Inactivation of aminoglycosides by acetylation,
phosphorylation, and adenylation. Adapted with permission from (6). C, Inactivation of chloramphenicol by chloramphenicol acetyltransferase
(CAT). CAT catalyzes acetylation of chloramphenicol at C-3 to yield 3-acetylchloramphenicol. Next, nonenzymatic intramolecular transfer of
the acetyl group from C-3 to C-1 occurs to produce a mixture of 1- and 3-acetylchloramphenicol. Finally, the 1-acetylchloramphenicol is
acetylated again by CAT to yield 1,3-diacetylchloramphenicol. Both mono- and di-acetylchloramphenicol are inactive.
Antibiotic Resistance in Bacteria 1067

A
EM EM

CH3 CH3
NH2 N
Erm
N methyltransferase N
N N

N N N N

A2058 A2058

B
HO HO
NH2 NH2
O O
OH OH
O OH O OH
O O OH O O OH
Cl Cl
O O O O
OH OH
HO Cl O HO Cl O
O O H O O H
H N H N
O N N N O N N N
N N H H N N H H
H H H H
NH O O NH O O
HO O HO O
NH2 NH2
O OH O OH
OH OH
HO HO

O H O O O
N O
R N O R N O
H O H O
N−Acyl−D−Ala−D−Ala N−Acyl−D−Ala−D−Lactate

Fig. 5. Antibiotic Resistance Caused by Target Alterations.


A, Enzymatic dimethylation of the amino group of a specific adenine residue, A2058, in 23S rRNA disrupts interaction of the reactive groups
of erythromycin (EM) with the nucleotides of the peptidyl transferase cavity. B, Reprograming of the peptidoglycan termini from D-Ala-D-Ala
to D-Ala-D-lactate causes loss of one hydrogen bond between vancomycin and D-Ala-D-lactate moiety, leading to a 1,000-fold decrease in
binding affinity. Adapted with permission from (6).

the peptidoglycan termini from D-Ala-D-Ala to D-Ala- (also PBP2A), with low affinity for all -lactams.26,36)
D-lactate, which decreases the binding affinity of Although the source of the PBP20 gene is not known,
vancomycin to its target (D-Ala-D-Ala) by 1,000-fold, horizontal transmission of the mecA gene from other
leading to high vancomycin resistance (Fig. 5B).84) On Staphylococcus species has been postulated.26,92–94)
the other hand, VanC and VanE replace the terminal D-
Ala with D-Ser, leading to low-level resistance.85) IV. New Approaches to Development of
-lactam antibiotic resistance can be caused not only Novel Antibiotics
by -lactamase, but also by alteration (or mutation) of
PBPs that have low affinity for the -lactams, as well as There is a pressing need for new antibiotics due to the
by acquisition of new PBPs with low affinity for the inevitable development of resistance that follows the
antibiotic.26,36) Unlike many other pathogens, Strepto- introduction of antibiotics to the clinic. In the past 10–15
coccus pneumoniae does not employ -lactamases as the years, there has been a dramatic increase in the
major mechanism of resistance to -lactams. Its resist- emergence of antibiotic-resistant bacteria, including
ance to -lactams is due to alteration of PBPs.26) The multiantibiotic resistant strains in both community and
most highly -lactam-resistant clinical isolates produce hospital settings.95,96) The main strategy of the pharma-
altered forms of high-molecular weight (Mr) PBPs (1A, ceutical industry for development of new therapeutics
1B, 2A, 2B, and 2X) that have reduced affinity for - has been modification of existing antibiotics, such as the
lactams.86–89) It is now known that low-affinity forms of second- and third-generation molecules of the best-
high-Mr pneumococcal PBPs have arisen by interspecies selling antibiotic groups of -lactams, macrolides, and
homologous recombination,26,90,91) suggesting the oc- fluoroquinolones.8) Although this approach is effective,
currence of gene shuffling in nature. MRSA acquires the it has turned out to be increasingly difficult to launch
mecA gene, which encodes a new PBP, termed PBP20 new drugs to meet the needs of the community.96)
1068 H. YONEYAMA and R. KATSUMATA

Almost 40 years were required for the development of a natural products, such as erythromycin, are assembled
new structural class of synthetic antibiotics of broad by multifunctional polyketide synthases (PKSs) that
spectrum, the oxazolidinone linezolid, since the intro- contain modular arrangements of functional domains.104)
duction of nalidixic acid in 1962.8) Since the main A library of >50 macrolides that are not produced by
classes of antibiotic targets are cell wall, protein, DNA/ chemical methods have been generated by simultaneous
RNA, and folate biosynthetic pathways, expanding the manipulation of multiple catalytic centers of erythro-
number of potential targets is of great importance for mycin PKS genes.105)
discovery and development of new antibiotics to keep The high-resolution X-ray crystal structures of
pace with the emergence of antibiotic resistance. the 30S and 50S ribosomal subunits as well as whole
The explosion in microbial genome sequencing in the 70S ribosome complexed with antibiotics offer a
past decade has revealed over 200 complete bacterial good opportunity to develop new antibiotic com-
genome sequences, including important pathogens (270 pounds.16–18,106,107) As to macrolides, the 3D structures
as of January 8, 2006; http://gib.genes.nig.ac.jp/), which of the ligands adjacent to peptidyl-transferase center on
provides more opportunities to identify and validate new the 50S ribosomal subunit reveal how an inhibitor binds
antibacterial targets.8,96) The bioinformatics approach is to the target, thus facilitating computational analysis of
based on searches of open reading frames (ORFs) other factors that contribute to binding, such as shape
conserved among potential pathogenic bacteria, as well and electrostatic complementlity.18,106–108) This struc-
as ORFs found only in prokaryotes, not in eukar- ture-based drug-design approach produces promising
yotes.8,96) An example of this approach compared the potential macrolide derivatives against respiratory tract
4,289 E. coli genes against the genomes of Haemophilus pathogens resistant to macrolides and ketolides, pen-
influenzae, Mycoplasma, S. pneumoniae, Streptococci, icillin, chloramphenicol, and sulphamethoxazole.106)
Chlamydia pneumoniae, Klebsiella, and P. aeruginosa.
The process identified 246 conserved genes in all these V. New Targets
bacteria, of which 68 genes are not found in humans.97)
Next, loss-of-function tests showed that 18 genes were Historically, the main classes of antibiotics inhibit
essential, 16 genes nonessential, and 34 genes unknown four classical targets: (i) cell wall biosynthesis, (ii)
as to function. In this bioinformatic analysis, three genes protein biosynthesis, (iii) DNA and RNA biosynthesis,
were finally chosen as targets for new respiratory-tract and (iv) folate coenzyme biosynthesis.8) The recent
antibiotics.97) advance in bacterial genomics has changed the anti-
The key aspect of the potential targets is that they are bacterial therapeutic environment from target-poor to
essential for bacterial growth or viability and are target-rich. The genomics provide many molecular
expressed under the conditions of the infection proc- targets to be identified and effectively screened:5,96)
ess.96,98) There is now good evidence to suggest that the peptide deformylase (PDF),109) aminoacyl-tRNA syn-
expression pattern of pathogens during infection can be thetase,110) fatty acid biosynthesis,111) isoprenoid biosyn-
quite different from that in culture medium.3,96,99) In thesis,112) lipid A biosynthesis,113) efflux pump,114)
fact, traditional whole-cell screenings focus on targets aromatic amino acid biosynthesis,96) DNA replication
that are important for in vitro growth in rich media and (GyrB subunit inhibitor),115) protein secretion,116) cell
thus might not identify inhibitors of target systems that wall biosynthesis (sortase),117) proteasome,118) glyoxy-
are induced specifically in vivo and are relevant to the late cycle,119) and signal networks (two-component
infection process. systems and quorum sensing pathways).120,121)
Several new techniques detect and monitor gene
expression during infection. In vivo expression technol- 1. Peptide deformylase (PDF)
ogy (IVET) allows identification of genes specifically Although there have been few novel antibacterial lead
induced in the host. It has been applied successfully to compounds that produce commercially successful anti-
several pathogens.100,101) A similar technique, differ- biotics from these new approaches so far, PDF appears
ential fluorescence induction (DFI), utilizes promoter to have gained intense awareness recently as a promis-
fusions with green fluorescent protein to identify active ing target.109) Although the bacterial protein-synthesiz-
promoters in vivo.101,102) Signature-tagged mutagenesis ing machinery is similar to those of mammalian cells,
(STM) is another technique that identifies the genes there are sufficient differences to allow selective
required for the infection process and viability in inhibition of this process in bacteria. One significant
vivo.101,103) difference is the transformylation and subsequent de-
Combinatorial chemistry has a significant impact on formylation of initiating methionine in bacteria.122,123)
the discovery of new antibiotics.104) This new approach Unlike cytosolic protein synthesis in mammalian cells,
to high-throughput screening requires large, diverse protein synthesis in bacteria and mitochondria is
synthetic compounds and natural products. In nature, initiated with N-formylmethionine,124,125) which is
biosynthetic assembly lines of polyketides and non- formed by N-methionyl-tRNA transformylase.126) In
ribosomal peptides furnish an example of combinatorial bacteria, but not in mitochondria, the N-formylmethio-
libraries.104) The structures of complex polyketide nine of newly synthesized protein is removed sequen-
Antibiotic Resistance in Bacteria 1069

tially by PDF and a methionine amino peptidase to Recent structural analysis of bacterial fatty acid bio-
produce mature proteins.125,127) This essential role of synthetic enzymes complexed with ligand is also
bacterial PDF in protein biosynthesis provides a rational facilitating the lead optimization process.139–141)
basis for selective inhibition. PDF inhibitor-resistant
mutants appear easily like other antibiotics; the trans- 4. Bacterial virulence factors
formylase gene has changed in E. coli, H. influenzae, In the past decades, antibiotic discovery programs
and S. aureus.128–130) But resistant mutants defective in have focused on eliminating infection by bactericidal
the transformylase gene have a slow growth pheno- and/or bacteriostatic agents that target essential proc-
type,109,128) suggesting diminished virulence of the esses for bacterial viability in vitro. However, patho-
pathogenic bacteria. Indeed, a PDF inhibitor-resistant genic bacteria elaborate virulence factors, involved in
mutant of S. aureus showed weakened virulence in a adhesion, invasion, and host defense system evasion, to
murine thigh-abscess model,130) and hence this type of establish an infection142) the expression of which is
resistance might not emerge in a clinical situation.109) carefully regulated in response to environmental
changes.99) Recently developed techniques, such as
2. Non-mevalonate pathway IVET, DFI, and STM, have identified genes that are
Although the mevalonate pathway has been known involved in the infectious process of pathogenic bacteria
for several decades as the source of isoprenoids, an in the context of their expression.100–103) According to
alternative non-mevalonate pathway has recently been ‘‘Koch’s molecular postulates,’’ genetic disruption of an
found,112,131) in which the first intermediate 1-deoxy-D- essential virulence factor should reduce bacterial viru-
xylulose 5-phosphate, also an intermediate in thiamine lence.143) In fact, allelic replacement of individual genes
and pyridoxal biosynthesis, is formed by condensing causes attenuation of virulence in in vivo models,
glyceraldehyde 3-phosphate and pyruvate.112) In contrast confirming their product’s role in infection.99) They are
to animals that use the mevalonate pathway exclusively, global regulators of virulence factor expression and
many bacteria, including clinically important pathogens, sortase in S. aureus,117,144) the two-component system in
utilize the non-mevalonate pathway.112) Since isoprenoid S. pneumoniae145) and in S. aureus,146) the quorum
biosynthesis is essential and humans do not have sensing system in P. aeruginosa,147) host cell invasion
enzymes of the non-mevalonate pathway, all of the in S. enterica,148) S. flexneri,149) and L. monocyto-
seven enzymes in the non-mevalonate pathway are genes,150) and the glyoxylate cycle in M. tuberculo-
attractive antibacterial targets for the development of sis.119) These factors are unique to bacteria, and hence
novel antibiotics. In fact, the 2C-methyl-D-erythritol 4- the virulence factors can be a target for development of
phosphate synthase inhibitor fosmidomycin has been new antibiotics.99)
shown to possess antibacterial activity.132) On the other The current concept of antibiotic discovery is that
hand, bacterial bioinformatics suggests that genes broad-spectrum agents are preferred over narrow-spec-
encoding the mevalonate pathway are essential in trum ones, implying that targets should be conserved
S. aureus and other Gram-positive cocci. Hence inhib- among pathogenic bacteria.99) In this context, inhibitors
itors of the mevalonate pathway should work against targeting these virulence factor-related targets are
these Gram-positive pathogens.133,134) narrow-spectrum. Among them, sortase, which cova-
lently attaches cell-surface proteins to the peptidoglycan
3. Bacterial fatty acid synthesis layer in Gram-positive bacteria, is an attractive drug
Fatty acid biosynthesis is essential for bacterial target, since sortase has a common substrate specificity
growth.5,96) In mammals, fatty acid is synthesized by a (LPXTG)151–153) and should be easily accessible to drugs
single large polypeptide called fatty acid synthase due to its cell-surface location. In addition, the 3D
(FAS),135) whereas in the bacterial FAS system individ- structure of sortase has been determined by NMR
ual enzymes perform the individual synthetic steps,136) spectroscopy, enabling rational structure-based drug
suggesting that the differences in the systems are the design.154) Thus there is an increasing possibility of a
basis for selective inhibition by natural products, such as single sortase inhibitor with activity against many Gram-
cerulenin, thiolactomycin, and isoniazid.5) Recently, an positive pathogens.99)
antibacterial antiseptic, triclosan, was shown to inhibit
enoyl-ACP reductase (FabI) in E. coli,137) and isoniazid, VI. Conclusion
an important antituberculosis drug, was found to be
activated by mycobacterial KatG catalase to acylate The development of antibiotics is among the greatest
NAD bound in the active site of InhA, the homolog of achievements of medicine in human history, but it is
FabI of E. coli, and thus to inhibit the enzyme.138) Thus clear that infectious diseases are still of major medical
the fatty acid biosynthetic pathway is validated to and scientific concern.96,98) The ability of bacterial
provide good targets. Screening against FabI has pathogens to gain resistance to new antibiotics has been
identified two novel antibacterial leads (benzodiazepine proved since the early introduction of sulfonamides and
and imidazole compounds) that inhibit S. aureus en- penicillins into clinical use.8,35) To keep pace with the
zyme selectively, but not the human FAS system.111) development of antibiotic resistance in bacterial patho-
1070 H. YONEYAMA and R. KATSUMATA

gens, we must continue the development of novel 16) Carter, A. P., Clemons, W. M., Brodersen, D. E.,
antibiotics.4,8,155) In addition, it is of great importance to Morgan-Warren, R. J., Wimberly, B. T., and
rethink strategies for preserving and extending the useful Ramakrishnan, V., Functional insights from the struc-
life of antibiotics, such as rotating the use of anti- ture of the 30S ribosomal subunit and its interactions
with antibiotics. Nature, 407, 340–348 (2000).
biotics156) and combination antibiotic therapy.8,36) The
17) Brodersen, D. E., Clemons, W. M., Carter, A. P.,
challenges can be met only by understanding the Morgan-Warren, R. J., Wimberly, B. T., and
fundamental biology of bacterial pathogens and host- Ramakrishnan, V., The structural basis for the action
pathogen interactions, and by valuing the antibiotics as of the antibiotics tetracycline, pactamycin, and hy-
precious, finite resources.36) gromycin B on the 30S ribosomal subunit. Cell, 103,
1143–1154 (2000).
Acknowledgments 18) Schlunzen, F., Zarivach, R., Harms, J., Bashan, A.,
Tocilj, A., Albrecht, R., Yonath, A., and Franceschi, F.,
We are grateful to Professor Yoshiyuki Kamio of Structural basis for the interaction of antibiotics with
Tohoku University for making it possible for us to write the peptidyl transferase centre in eubacteria. Nature,
this article. 413, 814–821 (2001).
19) Sherratt, D. J., Bacterial chromosome dynamics.
Science, 301, 780–785 (2003).
References 20) Maxwell, A., DNA gyrase as a drug target. Trends
1) Cohen, M., Changing patterns of infectious disease. Microbiol., 5, 102–109 (1997).
Nature, 406, 762–767 (2000). 21) Lesher, G. Y., Forelich, E. D., Gruet, M. D., Bailey,
2) Fauch, A. S., Infectious diseases: considerations for the J. H., and Brundage, R. P., 1,8-Naphthyridone deriv-
21st century. Clin. Infect. Dis., 32, 675–685 (2001). atives. A new class of chemotherapeutic agents. J.
3) Nathan, C., Antibiotics at the crossroads. Nature, 431, Med. Pharm. Chem., 5, 1063–1068 (1962).
899–902 (2004). 22) Mascaretti, O. A., ‘‘Bacteria Versus Antibacterial
4) Leeb, M., A shot in the arm. Nature, 431, 892–893 Agents, an Integrated Approach,’’ ASM Press, Wash-
(2004). ington, DC, pp. 289–328 (2003).
5) Projan, S. J., New (and not so new) antibacterial 23) Pan, X., and Fisher, L. M., Targeting of DNA gyrase in
targets: from where and when will the novel drugs Streptococcus pneumoniae by sparfloxacin: selective
come? Curr. Opin. Pharmacol., 2, 513–522 (2002). targeting of gyrase or topoisomerase IV by quinolones.
6) Walsh, C., ‘‘Antibiotics, Action, Origins, Resistance,’’ Antimicrob. Agents Chemother., 41, 471–474 (1997).
ASM Press, Washington, DC, pp. 3–49 (2003). 24) Ng, E. Y., Trucksis, M., and Hooper, D. C., Quinolone
7) Mascaretti, O. A., ‘‘Bacteria Versus Antibacterial resistance mutations in topoisomerase IV: relationship
Agents, an Integrated Approach,’’ ASM Press, Wash- to the flqA locus and genetic evidence that topoisomer-
ington, DC, pp. 97–105 (2003). ase IV is the primary target and DNA gyrase is the
8) Walsh, C., Where will new antibiotics come from? Nat. secondary target of fluoroquinolones in Staphylococcus
Rev. Microbiol., 1, 65–70 (2003). aureus. Antimicrob. Agents Chemother., 40, 1881–
9) Nikaido, H., Prevention of drug access to bacterial 1888 (1996).
targes: permeability barriers and active efflux. Science, 25) Murakami, K. S., and Darst, S. A., Bacterial RNA
264, 382–388 (1994). polymerases: the whole story. Curr. Opin. Struct. Biol.,
10) Spratt, B. G., and Cromie, K. D., Penicillin-binding 13, 31–39 (2003).
proteins of gram-negative bacteria. Rev. Infect. Dis., 26) Spratt, B. G., Resistance to antibiotics mediated by
10, 699–711 (1988). target alterations. Science, 264, 388–393 (1994).
11) Williams, D. H., The glycopeptide story: how to kill 27) Campbell, E. A., Korzheva, N., Mustaev, A.,
the deadly ‘superbugs.’ Nat. Prod. Rep., 13, 469–477 Murakami, K., Nair, S., Goldfarb, A., and Darst, S.
(1996). A., Structural mechanism for rifampicin inhibition of
12) Ban, N., Nissen, P., Hansen, J., Moore, P. B., and bacterial RNA polymerase. Cell, 104, 901–912 (2001).
Steitz, T. A., The complete atomic structure of the 28) Stryer, L., ‘‘Biochemistry’’ 4th ed., W. H. Freeman and
large ribosomal subunit at 2.4 Å resolution. Science, Company, New York, pp. 719–721 (1995).
289, 905–920 (2000). 29) Hancock, R. E. W., and Chapple, D. S., Peptide
13) Wimberly, B. T., Brodersen, D. E., Clemons, W. M., antibiotics. Antimicrob. Agents Chemother., 43, 1317–
Jr., Morgan-Warren, R. J., Carter, A. P., Vonrhein, C., 1323 (1999).
Hartsch, T., and Ramakrishnan, V., Structure of the 30) Baba, T., and Schneewind, O., Instruments of micro-
30S ribosomal subunit. Nature, 407, 327–339 (2000). bial warfare: bacteriocin synthesis, toxicity and im-
14) Schluenzen, F., Tocilj, A., Zarivach, R., Harms, J., munity. Trends Microbiol., 6, 66–71 (1998).
Gluehmann, M., Janell, D., Bashan, A., Bartels, H., 31) Breukink, E., Wiedemann, I., van Kraaij, C., Kuuipers,
Agmon, I., Franceschi, F., and Yonath, A., Structure of O. P., Sahl, H.-G., and de Kruijff, B., Use of the cell
functionally activated small ribosomal subunit at 3.3 Å wall precursor lipid II by a pore-forming peptide
resolution. Cell, 102, 615–623 (2000). antibiotic. Science, 286, 2361–2364 (1999).
15) Yusupov, M. M., Yusupova, G. Z., Baucom, A., 32) Wiedemann, I., Breukink, E., van Kraaij, C., Kuipers,
Lieberman, K., Earnest, T. N., Cate, J. H. D., and O. P., Bierbaum, G., de Kruijff, B., and Sahl, H.-G.,
Noller, H. F., Crystal structure of the ribosome at 5.5 Å Specific binding of nisin to the peptidoglycan precursor
resolution. Science, 292, 883–896 (2001). lipid II combines pore formation and inhibition of cell
Antibiotic Resistance in Bacteria 1071
wall biosynthesis for potent antibiotic activity. J. Biol. 51) Nakae, T., Role of membrane permeability in deter-
Chem., 276, 1772–1779 (2001). mining antibiotic resistance in Pseudomonas aerugi-
33) Hsu, S.-T. D., Breukink, E., Tishenko, E., Lutters, M. nosa. Microbiol. Immunol., 39, 221–229 (1995).
A. G., de Kuijuff, B., Kaptein, R., Bonvin, A. M. J. J., 52) McMurry, L., Petrucci, R. E., Jr., and Levy, S. B.,
and van Nuland, N. A. J., The nisin-lipid II complex Active efflux of tetracycline encoded by four genet-
reveals a pyrophosphate cage that provides a blueprint ically different tetracycline resistance determinants in
for novel antibiotics. Nat. Struct. Mol. Biol., 11, 963– Escherichia coli. Proc. Natl. Acad. Sci. USA, 77, 3974–
967 (2004). 3977 (1980).
34) Palumbi, S. R., Humans as the world’s greatest 53) Yoneyama, H., and Nakae, T., Cloning of the protein
evolutionary force. Science, 293, 1786–1790 (2001). D2 gene of Pseudomonas aeruginosa and its functional
35) Davies, J., Inactivation of antibiotics and the dissem- expression in the imipenem-resistant host. FEBS Lett.,
ination of resistance genes. Science, 264, 375–382 283, 177–179 (1991).
(1994). 54) Poole, K., Krebes, K., McNally, C., and Neshat, S.,
36) Walsh, C., Molecular mechanisms that confer anti- Multiple antibiotic resistance in Pseudomonas aerugi-
bacterial drug resistance. Nature, 406, 775–781 (2000). nosa: evidence for involvement of an efflux operon. J.
37) Livermore, D., Can better prescribing turn the tide of Bacteriol., 175, 7363–7372 (1993).
resistance? Nat. Rev. Microbiol., 2, 73–78 (2004). 55) Morshed, S. R. M., Lei, Y., Yoneyama, H., and Nakae,
38) Travis, J., Reviving the antibiotic miracle? Science, T., Expression of genes associated with antibiotic
264, 360–362 (1994). extrusion in Pseudomonas aeruginosa. Biochem. Bio-
39) Waksman, S. A., Reily, H. C., and Schatz, A., Strain phys. Res. Commun., 210, 356–362 (1995).
specificity and production of antibiotic substances. V. 56) Yoneyama, H., Ocaktan, A., Tsuda, M., and Nakae, T.,
Strain resistance of bacteria to antibiotic substances, The role of mex-gene products in antibiotic extrusion
especially to streptomycin. Proc. Natl. Acad. Sci. USA, in Pseudomonas aeruginosa. Biochem. Biophys. Res.
31, 157–164 (1945). Commun., 233, 611–618 (1997).
40) Murray, E., Vancomycin resistant enterococci. Am. J. 57) Paulsen, I. T., Multidrug efflux pumps and resistance:
Med., 102, 284–293 (1997). regulation and evolution. Curr. Opin. Microbiol., 6,
41) Neu, H. C., The crisis in antibiotic resistance. Science, 446–451 (2003).
257, 1064–1073 (1992). 58) Bolhuis, H., Molenaar, D., Poelarends, G., van Veen,
42) Coates, A., Hu, Y., Bax, R., and Page, C., The future H. W., Poolman, B., Driessen, A. J. M., and Konings,
challenges facing the development of new antimicro- W. N., Proton motive force-driven and ATP-dependent
bial drugs. Nat. Rev. Drug Discov., 1, 895–910 (2002). drug extrusion systems in multidrug-resistant Lacto-
43) Sievert, D. M., Boulton, M. L., Stoltman, G., Johnson, coccus lactis. J. Bacteriol., 176, 6957–6964 (1994).
D., Stobierske, M. G., Downes, F. P., Somsel, P. A., 59) Neyfakh, A. A., Mystery of multidrug transporters: the
Rudrik, J. T., Brown, W., Hafeez, W., Lundstrom, T., answer can be simple. Mol. Microbiol., 44, 1123–1130
Flanagan, E., Johnson, R., and Mitchell, J., Staph- (2002).
ylococcus aureus resistant to vancomycin: United 60) Murakami, S., Nakashima, R., Yamashita, E., and
States, 2002. Morbid. Mortal. Wkly Rep., 51, 565– Yamaguchi, A., Crystal structure of bacterial multidrug
567 (2002). efflux transporter AcrB. Nature, 419, 587–593 (2002).
44) Falkow, S., and Kennedy, D., Antibiotics, animals, and 61) Akama, H., Matsuura, T., Kashiwagi, S., Yoneyama,
people—again! Science, 291, 397 (2001). H., Narita, S., Tsukihara, T., Nakagawa, A., and
45) Witte, W., Medical consequences of antibiotic use in Nakae, T., Crystal structure of the membrane fusion
agriculture. Science, 279, 996–997 (1998). protein, MexA, of the multidrug transporter in Pseu-
46) Gustafson, R. H., and Bowen, R. E., Antibiotic use in domonas aeruginosa. J. Biol. Chem., 279, 25939–
animal agriculture. J. Appl. Microbiol., 83, 531–541 25942 (2004).
(1997). 62) Higgins, M. K., Bokma, E., Koronakis, E., Hughes, C.,
47) Wehener, H. C., Antibiotics in animal feed and their and Koronakis, V., Structure of the periplasmic
role in resistance development. Curr. Opin. Microbiol., component of a bacterial drug efflux pump. Proc.
6, 439–445 (2002). Natl. Acad. Sci. USA, 101, 9994–9999 (2004).
48) Casewell, M., Friis, C., Marco, E., McMullin, P., and 63) Koronakis, V., Sharff, A., Koronakis, E., Luisi, B., and
Phillips, I., The European ban on growth-promoting Hughes, C., Crystal structure of the bacterial mem-
antibiotics and emerging consequences for human and brane protein TolC central to multidrug efflux and
animal health. J. Antimicrob. Chemother., 52, 159–161 protein export. Nature, 405, 914–919 (2000).
(2003). 64) Akama, H., Kanemaki, M., Yoshimura, M., Tsukihara,
49) Tsiodras, S., Gold, H. S., Sakoulas, G., Eliopoulos, G. T., Kashiwagi, T., Yoneyama, H., Narita, S.,
M., Wennersten, C., Venkataraman, L., Moellering, R. Nakagawa, A., and Nakae, T., Crystal structure of the
C., and Ferraro, M. J., Linezolid resistance in a clinical drug discharge outer membrane protein, OprM, of
isolate of Staphylococcus aureus. Lancet, 358, 207– Pseudomonas aeruginosa: dual modes of membrane
208 (2001). anchoring and occluded cavity end. J. Biol. Chem.,
50) Gonzales, R. D., Schreckenberger, P. C., Graham, M. 279, 52816–52819 (2004).
B., Kelkar, S., DenBesten, K., and Quinn, J. P., 65) Fernandez-Recio, J., Walas, F., Federici, L., Pratap, J.
Infections due to vancomycin-resistant Enterococcus V., Bavro, V. N., Miguel, R. N., Mizuguchi, K., and
faecium resistant to linezolid. Lancet, 357, 1179 Luisi, B., A model of a transmembrane drug-efflux
(2001). pump from gram-negative bacteria. FEBS Lett., 578,
1072 H. YONEYAMA and R. KATSUMATA
5–9 (2004). resistance to erythromycin and other antibiotics. J.
66) Narita, S., Eda, S., Yoshihara, E., and Nakae, T., Biol. Chem., 258, 12702–12705 (1983).
Linkage of the efflux-pump expression level with 82) Cetinkaya, Y., Falk, P., and Mayhall, C. G., Vanco-
substrate extrusion rate in the MexAB-OprM efflux mycin-resistant enterococci. Clin. Microbiol. Rev., 13,
pump of Pseudomonas aeruginosa. Biochem. Biophys. 686–707 (2000).
Res. Commun., 308, 922–926 (2003). 83) Arias, C. A., Couvalin, P., and Reynolds, P. E., vanC
67) Murakami, S., and Yamaguchi, A., Multidrug-export- cluster of vancomycin-resistant Enterococcus gallina-
ing secondary transporters. Curr. Opin. Struct. Biol., rum BM4174. Antimicrob. Agents Chemother., 44,
13, 443–452 (2003). 1660–1666 (2000).
68) Yu, E. W., Aires, J. R., and Nikaido, H., AcrB 84) Bugg, T. D., Wright, G. D., Dutka-Malen, S., Arthur,
multidrug efflux pump of Escherichia coli: composite M., Couvalin, P., and Walsh, C. T., Molecular basis
substrate-binding cavity of exceptional flexibility gen- for vancomycin resistance in Enterococcus faecium
erates its extremely wide substrate specificity. J. BM4147: biosynthesis of a depsipeptide peptidoglycan
Bacteriol., 185, 5657–5664 (2003). precursor by vancomycin resistance proteins VanH and
69) Yu, E. W., McDermott, G., Zgurskaya, H. I., Nikaido, VanA. Biochemistry, 30, 10408–10415 (1991).
H., and Koshland, D. E., Jr., Structural basis of 85) Park, I. S., Lin, C. H., and Walsh, C. T., Bacterial
multiple drug-binding capacity of the AcrB multidrug resistance to vancomycin: overproduction, purification,
efflux pump. Science, 300, 976–980 (2003). and characterization of VanC2 from Enterococcus
70) Fisher, J., Belasco, J. G., Khosla, S., and Knowles, J. casseliflavus as a D-Ala-D-Ser ligase. Proc. Natl.
R., -lactamase proceeds via an acyl-enzyme inter- Acad. Sci. USA, 94, 10040–10044 (1997).
mediate: interaction of the Escherichia coli RTEM 86) Nagai, K., Davies, T. A., Jacobs, M. R., and
enzyme with cefoxitin. Biochemistry, 19, 2895–2901 Appelbaum, P. C., Effects of amino acid alterations
(1980). in penicillin-binding proteins (PBPs) 1a, 2b, and 2x
71) Knox, J. R., Moews, P. C., and Frere, J. M., Molecular on PBP affinities of penicillin, ampicillin, amoxicillin,
evolution of bacterial beta-lactam resistance. Chem. cefditoren, cefuroxime, cefprozil, and cefaclor in 18
Biol., 3, 937–947 (1996). clinical isolates of penicillin-susceptible, -intermediate,
72) Knox, J. R., Extended-spectrum and inhibitor-resistant and -resistant pneumococci. Antimicrob. Agents Che-
TEM-type beta-lactamases: mutations, specificity, and mother., 46, 1273–1280 (2002).
three-dimensional structure. Antimicrob. Agents Che- 87) Hakenbeck, R., Grebe, T., Zahner, D., and Stock, J. B.,
mother., 39, 2593–2601 (1995). Beta-lactam resistance in Streptococcus pneumoniae:
73) Kotra, L. P., Haddad, J., and Mobashery, S., Amino- penicillin-binding proteins and non-penicillin-binding
glycosides: perspectives on mechanisms of action and proteins. Mol. Microbiol., 33, 673–678 (1999).
resistance and strategies to counter resistance. Anti- 88) Zighelboim, S., and Tomasz, A., Penicillin-binding
microb. Agents Chemother., 44, 3249–3256 (2000). proteins of multiply antibiotic-resistant South African
74) Wright, G. E., Aminoglycoside-modifying enzymes. strains of Streptococcus pneumoniae. Antimicrob.
Curr. Opin. Microbiol., 2, 499–503 (1999). Agents Chemother., 17, 434–442 (1980).
75) Rodriguez-Fonseca, C., Amis, R., and Carrett, R. A., 89) Hakenbeck, R., Ellerbrock, H., Briese, T., Handwerger,
Fine structure of the peptidyl transferase center on 23S- S., and Tomasz, A., Penicillin-binding proteins of
like rRNAs deduced from chemical probing of anti- penicillin-susceptible and -resistant pneumococci: im-
biotic-ribosome complexes. J. Mol. Biol., 247, 224– munological relatedness of altered proteins and
235 (1995). changes in peptides carrying the beta-lactam binding
76) Moazed, D., and Noller, H. F., Chloramphenicol, site. Antimicrob. Agents Chemother., 30, 553–558
erythromycin, carbamycin and vernamycin B protect (1986).
overlapping sites in the peptidyl transferase region of 90) Hakenbeck, R., Mosaic genes and their role in
23S ribosomal RNA. Biochemie, 69, 879–884 (1987). penicillin-resistant Streptococcus pneumoniae. Elec-
77) Murray, I. A., and Shaw, W. V., O-Acetyltransferases trophoresis, 19, 597–601 (1998).
for chloramphenicol and other natural products. Anti- 91) Dowson, C. G., Coffey, T. J., Kell, C., and Whiley, R.
microb. Agents Chemother., 41, 1–6 (1997). A., Evolution of penicillin resistance in Streptococcus
78) Abraham, E. P., and Chain, E., An enzyme from pneumoniae: the role of Streptococcus mitis in the
bacteria able to destroy penicillin. Nature, 146, 837 formation of a low affinity PBP2B in S. pneumoniae.
(1940). Mol. Microbiol., 9, 635–643 (1993).
79) Bussiere, D., Muchmore, S. W., Dealwis, C. G., 92) Kuroda, M., Ohta, T., Uchiyama, I., Baba, T., Yuzawa,
Schluckebier, G., Nienaber, V. L., Edalji, R. P., H., Kobayashi, I., Cui, L., Oguchi, A., Aoki, K., Nagai,
Walter, K. A., Ladror, U. S., Holzman, T. F., and Y., Lian, J., Ito, T., Kanamori, M., Matsumura, H.,
Abad-Zapatero, C., Crystal structure of ErmC0 , an Maruyama, A., Murakami, H., Hosoyama, A.,
rRNA methyltransferase which mediates antibiotic Mizutani-Ui, Y., Takahashi, N. K., Sawano, T., Inoue,
resistance in bacteria. Biochemistry, 37, 7103–7112 R., Kaito, C., Sekimizu, K., Hirakawa, H., Kuhara, S.,
(1998). Goto, S., Yabuzaki, J., Kanehisa, M., Yamashita, A.,
80) Weisblum, B., Erythromycin resistance by ribosome Oshima, K., Furuya, K., Yoshino, C., Shiba, T.,
modification. Antimicrob. Agents Chemother., 39, 577– Hattori, M., Ogasawara, N., Hayashi, H., and
585 (1995). Hiramatsu, K., Whole genome sequencing of methi-
81) Skinner, R., Cundliffe, E., and Schmidt, F. J., Site of cillin-resistant Staphylococcus aureus. Lancet, 357,
action of a ribosomal RNA methylase responsible for 1225–1240 (2001).
Antibiotic Resistance in Bacteria 1073
93) Ryffel, C., Tesch, W., Birch-Machin, I., Reynolds, P. 110) Tao, J., and Schimmel, P., Inhibitors of amino acyl-
E., Barberis-Maino, L., Kayser, F. H., and Berger- tRNA synthetases as novel anti-infectives. Expert.
Bachi, B., Sequence comparison of mecA genes Opin. Invest. Drugs, 9, 1767–1775 (2000).
isolated from methicillin-resistant Staphylococcus aur- 111) Payne, D. J., Warren, P. V., Holmes, D. J., and
eus and Staphylococcus epidermidis. Gene, 94, 137– Lonsdale, J. Y., Bacterial fatty acid biosynthesis: a
138 (1990). genomics-driven target for antibacterial drug discov-
94) Ubukata, K., Nonoguchi, R., Song, M. D., Matsuhashi, ery. Drug Discovery Today, 6, 537–544 (2001).
M., and Konno, M., Homology of mecA gene in 112) Rohdich, F., Kis, K., Bacher, A., and Eisenreich, W.,
methicillin-resistant Staphylococcus haemolyticus and The non-mevalonate pathway of isoprenoids: genes,
Staphylococcus simulans to that of Staphylococcus enzymes and intermediates. Curr. Opin. Chem. Biol., 5,
aureus. Antimicrob. Agents Chemother., 34, 170–172 535–540 (2001).
(1990). 113) Clements, J. M., Coignard, F., Johnson, I., Chandler,
95) Hall, B. G., Predicting the evolution of antibiotic S., Palan, S., Waller, A., Wijkmans, J., and Hunter, M.
resistance genes. Nat. Rev. Microbiol., 2, 430–435 G., Antibacterial activities and characterization of
(2004). novel inhibitors of LpxC. Antimicrob. Agents Chemo-
96) McDevitt, D., and Rosenberg, M., Exploiting genomics ther., 46, 1793–1799 (2002).
to discover new antibiotics. Trends Microbiol., 9, 611– 114) Renau, T. E., Leger, R., Yen, R., She, M. W., Flamme,
617 (2001). E. M., Sangalang, J., Gannon, C. L., Chamberland, S.,
97) Rosamond, J., and Allsop, A., Harnessing the power of Lomovskaya, O., and Lee, V. J., Peptidomimetics of
the genome in the sesarch for new antibiotics. Science, efflux pump inhibitors potentiate the activity of levo-
287, 1973–1976 (2000). floxacin in Pseudomonas aeruginosa. Bioorg. Med.
98) Allsop, A., New antibiotic discovery, novel screens, Chem. Lett., 12, 763–766 (2002).
novel targets and impact of microbial genomics. Curr. 115) Flatman, R. H., Howells, A. J., Heide, L., Fiedler,
Opin. Microbiol., 1, 530–534 (1998). H.-P., and Maxwell, A., Simocyclinone D8, an inhib-
99) Alksne, L. E., and Projan, S. J., Bacterial virulence as a itor of DNA gyrase with a novel mode of action.
target for antimicrobial chemotherapy. Curr. Opin. Antimicrob. Agents Chemother., 49, 1093–1100
Biotechnol., 11, 625–636 (2000). (2005).
100) Mahan, M. J., Slauch, J. M., and Mekalanos, J. J., 116) Alksne, L. E., Burgio, P., Hu, W., Feld, B., Singh, M.
Selection of bacterial virulence genes that are specif- P., Tuckman, M., Petersen, P. J., Labthavikul, P.,
ically induced in host tissues. Science, 259, 686–688 McGlynn, M., Barbieri, L., McDonald, L., Bradford,
(1993). P., Dushin, R. G., Rothstein, D., and Projan, S. J.,
101) Chiang, S. L., Mekalanos, J. J., and Holden, D. W., In Identification and analysis of bacterial secretion inhib-
vivo genetic analysis of bacterial virulence. Annu. Rev. itors utilizing a SecA-LacZ reporter fusion system.
Microbiol., 53, 129–154 (1999). Antimicrob. Agents Chemother., 44, 1418–1427
102) Valdivia, R. H., and Falkow, S., Fluorescence-based (2000).
isolation of bacterial genes expressed within host cells. 117) Mazmanian, S. K., Liu, G., Jensen, E. R., Lenoy, E.,
Science, 277, 2007–2011 (1997). and Schneewind, O., Staphylococcus aureus sortase
103) Hensel, M., Shea, J. E., Gleeson, C., Jones, M. D., mutants defective in the display of surface proteins and
Dalton, E., and Holden, D. W., Simultaneous identi- in the pathogenesis of animal infections. Proc. Natl.
fication of bacterial virulence genes by negative Acad. Sci. USA, 97, 5510–5515 (2000).
selection. Science, 269, 400–403 (1995). 118) Darwin, K. H., Ehrt, S., Gutierrez-Ramos, J.-C.,
104) Rodriguez, E., and McDaniel, R., Combinatorial bio- Weich, N., and Nathan, C. F., The proteasome of
synthesis of antimicrobials and other natural products. Mycobacterium tuberculosis is required for resistance
Curr. Opin. Microbiol., 4, 526–534 (2001). to nitric oxide. Science, 302, 1963–1966 (2003).
105) McDaniel, R., Thamchaipenet, A., Gustafsson, C., Fu, 119) McKinney, T. K., Sharma, V. K., Craig, W. A., and
H., Betlach, M., and Ashley, G., Multiple genetic Archer, G. L., Persistence of Mycobacterium tuber-
modifications of the erythromycin polyketide synthase culosis in macrophages and mice requires the glyox-
to produce a library of novel ‘‘unnatural’’ natural ylate shunt enzyme isocitrate lyase. Nature, 406, 735–
products. Proc. Natl. Acad. Sci. USA, 96, 1846–1851 738 (2000).
(1999). 120) Barett, J. F., and Hoch, J. A., Two-component signal
106) Sutcliffe, J. A., The search for new antibiotics targeting transduction as a target for microbial anti-infective
the 50S ribozyme. ASM News, 70, 513–519 (2004). therapy. Antimicrob. Agents Chemother., 42, 1529–
107) Hansen, J. L., Ippolito, J. A., Ban, N., Nissen, P., 1536 (1998).
Moore, P. B., and Steitz, T. A., The structure of four 121) Mayville, P., Ji, G., Beavis, R., Yang, H., Goger, M.,
macrolide antibiotics bound to the large ribosomal Novick, R. P., and Muir, T. W., Structure-activity
subunit. Mol. Cell, 10, 117–128 (2002). analysis of synthetic autoinducing thiolactone peptides
108) Knowles, D. J. C., Foloppe, N. F., Matassova, N. B., from Stphylococcus aureus responsible for virulence.
and Murchie, A. I. H., The bacterial ribosome, a Proc. Natl. Acad. Sci. USA, 96, 1218–1223 (1999).
promising focus for structure-based drug design. Curr. 122) Mazel, D., Pochet, S., and Marliere, P., Genetic
Opin. Pharmacol., 2, 501–506 (2002). characterization of polypeptide deformylase, a distinc-
109) Yuan, Z., Trias, J., and White, R. J., Deformylase as a tive enzyme of eubacterial translation. EMBO J., 13,
novel antibacterial target. Drug Discovery Today, 6, 914–923 (1994).
954–961 (2001). 123) Mazel, D., Coic, E., Blanchard, S., Saurin, W., and
1074 H. YONEYAMA and R. KATSUMATA
Marliere, P., A survey of polypeptide deformylase domain I constituent activities. Proc. Natl. Acad. Sci.
function throughout the eubacterial lineage. J. Mol. USA, 94, 5588–5593 (1997).
Biol., 266, 939–949 (1997). 136) Heath, R. J., White, S. W., and Rock, C. O., Lipid
124) Adams, J. M., and Capecchi, M. R., N-formylmethio- biosynthesis as a target for antibacterial agents. Prog.
nyl-sRNA as the initiator of protein synthesis. Proc. Lipid Res., 40, 467–497 (2001).
Natl. Acad. Sci. USA, 55, 147–155 (1966). 137) McMurry, L. M., Oethinger, M., and Levy, S. B.,
125) Lucchini, G., and Bianchetti, R., Initiation of protein Triclosan targets lipid synthesis. Nature, 394, 531–532
synthesis in isolated mitochondria and chloroplasts. (1998).
Biochim. Biophys. Acta, 608, 54–61 (1980). 138) Rozwarski, D. A., Grant, G. A., Barton, D. H., Jacobs,
126) Guillon, J. M., Mechulam, Y., Schmitter, J. M., W. R., and Sacchettini, J. C., Modification of the
Blanquet, S., and Fayat, G., Disruption of the gene NADH of the isoniazid target (InhA) from Mycobac-
for Met-tRNA(fMet) formyltransferase severely im- terium tuberculosis. Science, 279, 98–102 (1998).
pairs growth of Escherichia coli. J. Bacteriol., 174, 139) Qiu, X., Janson, C. A., Court, R. I., Smyth, M. G.,
4294–4301 (1992). Payne, D. J., and Abdel-Meguid, S. S., Molecular basis
127) Meinnel, T., Mechulam, Y., and Blanquet, S., Methio- of triclosan activity involves a flipping loop in the
nine as translation start signal: a review of the enzymes active site. Protein Sci., 8, 2529–2532 (2001).
of the pathway in Escherichia coli. Biochimie, 75, 140) Moche, M., Schneider, G., Edwards, P., Dehesh, K.,
1061–1075 (1993). and Lindqvist, Y., Structure of the complex between
128) Apfel, C. M., Locher, H., Evers, S., Takacs, B., the antibiotic cerulenin and its target, beta-ketoacyl
Hubschwerlen, C., Pirson, W., Page, M. G. P., and carrier protein synthase. J. Biol. Chem., 274, 6031–
Keck, W., Peptide deformylase as an antibacterial drug 6034 (1999).
target: target validation and resistance development. 141) Qiu, X., Janson, C. A., Smith, W. W., Head, M.,
Antimicrob. Agents Chemother., 45, 1058–1064 Lonsdale, J., and Konstantinidis, A. K., Refined
(2001). structures of beta-ketoacyl carrier protein synthase
129) Clements, J. M., Beckett, R. P., Brown, A., Catlin, G., III. J. Mol. Biol., 307, 341–356 (2001).
Lobell, M., Palan, S., Thomas, W., Whittaker, M., 142) Strauss, E. J., and Falkow, S., Microbial pathogenesis:
Wood, S., Salama, S., Baker, P. J., Rodgers, H. F., genomics and beyond. Science, 276, 707–712 (1997).
Barynin, V., Rice, D. W., and Hunter, M. G., Antibiotic 143) Falkow, S., Molecular Koch’s postulates applied to
activity and characterization of BB-3497, a novel microbial pathogenicity. Rev. Infect. Dis., 10, S274–
peptide deformylase inhibitor. Antimicrob. Agents 276 (1988).
Chemother., 45, 563–570 (2001). 144) Cheung, A. L., Eberhardt, K. J., Chung, E., Yeaman,
130) Margolis, P. S., Hackbarth, C. J., Young, D. C., Chen, M. R., Sullam, P. M., Ramos, M., and Bayer, A. S.,
W. W. D., Yuan, Z., White, R., and Trias, J., Peptide Diminished virulence of a sar /agr mutant of Staph-
deformylase in Staphylococcus aureus: resistance to ylococcus aureus in the rabbit model of endocarditis.
inhibition is mediated by mutations in the formyl- J. Clin. Invest., 94, 1815–1822 (1994).
transferase gene. Antimicrob. Agents Chemother., 44, 145) Throup, J. P., Koretke, K. K., Bryant, A. P., Ingraham,
1825–1831 (2000). K. A., Chalker, A. F., Ge, Y., Marta, A., Wallis, N. G.,
131) Rohmer, M., Knani, M., Simonin, P., Sutter, B., and Brown, J. R., Holmes, D. J., Rosenberg, M., and
Sahm, H., Isoprenoid biosynthesis in bacteria: a novel Burnham, M. K., A genomic analysis of two-compo-
pathway for the early steps leading to isopentenyl nent signal transduction in Streptococcus pnermoniae.
diphosphate. Biochem. J., 295, 517–524 (1993). Mol. Microbiol., 35, 566–576 (2000).
132) Kazuyama, T., Shimizu, T., Takahashi, S., and Seto, 146) Throup, J. P., Zappacosta, F., Lunsford, R. D., Annan,
H., Fosmidomycin, a specific inhibitor of 1-deoxy-D- R. S., Carr, S. A., Lonsdale, J. T., Bryant, A. P.,
xylulose 5-phosphate reductoisomerase in the non- McDevitt, D., Rosenberg, M., and Burnham, M. K.,
mevalonate pathway for terpenoid biosynthesis. Tetra- The srhSR gene pair from Staphylococcus aureus:
hedron Lett., 39, 7913–7916 (1998). genomic and proteomic approaches to the identification
133) Hedl, M., Sutherlin, A., Wilding, E. I., Mazaulla, M., and characterization of gene function. Biochemistry,
McDevitt, D., Lane, P., Burgner, J. W., 2nd, 40, 10392–10401 (2001).
Lehnbeuter, K. R., Stauffacher, C. V., Gwynn, M. N., 147) Tang, H. B., DiMango, E., Bryan, R., Gambello, M.,
and Rodwell, V. W., Enteroccus faecalis acetoacetyl- Iglewski, B. H., Goldberg, J. B., and Prince, A.,
coenzyme A thiolase/3-hydroxy-3-methylglutaryl- Contribution of specific Pseudomonas aeruginosa
coenzyme A reductase, a dual-function protein of virulence factors to pathogenesis of pneumonia in a
isopentenyl diphosphate biosynthesis. J. Bacteriol., neonatal mouse model of infection. Infect. Immun., 64,
184, 2116–2122 (2002). 37–43 (1996).
134) Wildling, E. I., Brown, J. R., Bryant, A. P., Chalker, A. 148) Uchiya, K., Barbieri, M. A., Funato, K., Shah, A. H.,
F., Holmes, D. J., Ingraham, K. A., Iordanescu, S., So, Stahl, P. D., and Groisman, E. A., A Salmonella
S., Rosenberg, M., and Gwynn, M. N., Identification, virulence protein that inhibits cellular trafficking.
evolution, and essentiality of the mevalonate pathway EMBO J., 18, 3924–3933 (1999).
for isopentenyl diphosphate biosynthesis in gram- 149) Menard, R., Sansonetti, P. J., and Parsot, C., Nonpolar
positive cocci. J. Bacteriol., 182, 4319–4327 (2000). mutagenesis of the ipa genes defines IpaB, IpaC, and
135) Chirala, S. S., Huang, W. Y., Jayakumar, A., Sakai, K., IpaD as effectors of Shigella flexneri entry into
and Wakil, S. J., Animal fatty acid synthetase: func- epithelial cells. J. Bacteriol., 175, 5899–5906 (1993).
tional mapping and cloning and expression of the 150) Gaillot, O., Pellegrini, E., Bregenholt, S., and Berche,
Antibiotic Resistance in Bacteria 1075
P., The ClpP serine protease is essential for the 154) Ilangovan, U., Ton-That, H., Isahara, J., Schneewind,
intracellular parasitism and virulence of Listeria O., and Clubb, R. T., Structure of sortase, the trans-
monocytogenes. Mol. Microbiol., 35, 1286–1294 peptidase that anchors proteins to the cell wall of
(2000). Staphylococcus aureus. Proc. Natl. Acad. Sci. USA, 98,
151) Fischetti, V. A., Pancholi, V., and Schneewind, O., 6056–6061 (2001).
Conservation of a hexapeptide sequence in the anchor 155) Couvalin, P., and Davies, J., Antimicrobials: time to
region of surface proteins from gram-positive cocci. act! Editorial overview. Curr. Opin. Microbiol., 6,
Mol. Microbiol., 4, 1603–1605 (1990). 425–426 (2003).
152) Schneewind, O., Mihaylove-Petkov, D., and Model, P., 156) Schlaes, D. M., Gerding, D. N., John, J. F., Jr., Craig,
Cell wall sorting signals in surface proteins of gram- W. A., Bornstein, D. L., Duncan, R. A., Eckman, M.
positive bacteria. EMBO J., 12, 4803–4811 (1993). R., Farrer, W. E., Greene, W. H., Lorian, V., Levy, S.,
153) Mazmanian, S. K., Liu, G., Ton-That, H., and McGowan, J. E. Jr., Paul, S. M., Ruskin, J., Tenover, F.
Schneewind, O., Staphylococcus aureus sortase, an C., and Watanakunakorn, C., Guidelines for the
enzyme that anchors surface proteins to the cell wall. prevention of antimicrobial resistance in hospitals.
Science, 285, 760–763 (1999). Clin. Infect. Dis., 25, 583–599 (1997).

You might also like