You are on page 1of 12

Azucena and Mobashery

Aminoglycoside-modifying the ribosomal target of the drug; or (d) extrusion of the


aminoglycosides from the cell by efflux pumps. Of these
enzymes: mechanisms of resistance mechanisms, inactivation by aminoglycoside-
modifying enzymes is the most important both in terms of
catalytic processes and level and frequency of resistance conferred to the bac-
terium.
inhibition There are three types of aminoglycoside-modifying en-
zymes,each of which transfers a functional group to the amino-
glycoside structure thereby inactivating the antibiotic: (a)
Eduardo Azucena, Shahriar Mobashery aminoglycoside nucleotidyltransferases (ANTs) transfer a
nucleotide triphophates; (b) aminoglycoside acetyltransferases
Institute for Drug Design, Departments of Chemistry, Pharmacology and (AACs) transfer the acetyl group from acetyl-CoA; and (c)
Biochemistry and Molecular Biology,Wayne State University, Detroit, Michigan, aminoglycoside phosphotransferases (APHs) transfer the phos-
USA phoryl group from ATP.6,7 Each family of enzymes consists of
different isozymic forms that differ in substrate regiospecificity
Abstract The most prevalent mechanism for resistance to for their reactions. At least 50 different genes for aminoglyco-
aminoglycoside antibiotics is mediated through their enzymatic side-modifying enzymes are known in bacteria.7 A number of
modification in resistant organisms. Dozens of aminoglycoside- bacteria also harbor a unique bifunctional resistance enzyme
modifying enzymes are known at the gene sequence level, but that catalyzes both the AAC and APH reactions.6,8,9
few have been characterized in the details of their mechanism. Bacterial action of aminoglycosides in susceptible strains
This review summarizes the state of knowledge of the best involves an energy-requiring membrane transport into the
studied of these enzymes, focusing on their catalytic cell10–12 as well as interaction of the drug with the ribo-
mechanisms and inhibition. © 2001 Harcourt Publishers Ltd some.13–15 The rate-limiting step in the drug action is the slow
process of initial transport of the aminoglycoside across the
cytoplasmic membrane.11 Once inside the cell, aminoglyco-
side binds with the 16S rRNA of the 30S ribosomal frag-
INTRODUCTION
ment.16–18 This aminoglycoside-ribosome interaction causes
misreading of mRNA, leading to biosynthesis of non-

A
minoglycosides constitute a large family of water-
soluble, polycationic amino sugars of considerable functional, misfolded proteins.14,15 Some of the resulting non-
structural diversity (Fig. 1).The structural aspects of active proteins then interact with the cell membrane and
these antibiotics has been reviewed recently and will not be cause the creation of aberrant membrane structure and a
discussed here extensively.1,2 They are often broad-spectrum subsequent enhancement of cellular penetration by the
antibacterial agents that are products of bacterial or fungal aminoglycoside. This event augments the drug’s pleiotropic
metabolism. The structures of several of these natural prod- effects, which in turn collectively contribute to bacterial
ucts have been altered by chemists to generate semi- death.14,15 In resistant strains that possess aminoglycoside-
synthetic aminoglycosides with expanded activities, which inactivating enzymes, a different scenario occurs: drugs that
usually show aversion to modification by known important manage to enter the cell are modified by the resistance
resistance enzymes. enzyme, the modified drug is unable to interact with the
Shortly after the isolation of the first aminoglycoside, ribosome effectively, and the enhanced rate of aminoglyco-
streptomycin, from Streptomyces griseus in the 1940s,3 the side accumulation observed in susceptible strains does not
clinical importance of these mostly bactericidal agents was occur.19,20 Because drug uptake remains suppressed at low
realized in the treatment of tuberculosis.4,5 Over the decades rates, the cell survives and remains viable for as long as the
following this discovery, a series of aminoglycosides were rate of drug modification matches or surpasses the rate of
identified and characterized for clinical use in the treatment drug uptake.19 This requirement is often met readily: resis-
of bacterial infections caused by gram-positive and gram- tance enzymes are often expressed in high concentrations
negative organisms. Some aminoglycosides such as paro- and some of these enzymes have been shown to operate at
momycin have also been used in treatment of amoeboid or the diffusion limit.6
protozoan infections. In recent years, enthusiasm for the Structure-activity relationship studies on aminoglycosides
therapeutic use of aminoglycosides for purposes has been have established that their antibiotic activity depends little
waning because of toxicity issues, and there has been a trend on their hydroxyl groups. On the other hand, the number
towards the use of less toxic alternative drugs such as and positions of amino groups play a significant role in
β-lactams. Nevertheless, aminoglycosides remain important antibiotic activity in the order 2′, 6′-diamino > 6′-amino > 2′
weapons in the modern antimicrobial armamentarium, as a amino > absence of amine.21 Thus, according to this study, an
vast majority of gram-negative aerobes, including some mul- aminoglycoside with unsubstituted amino groups at posi-
tiresistant strains, remain vulnerable to some of these agents. tions 2′ and 6′ is generally found to possess greater antibac-
Bacterial resistance to aminoglycosides manifests itself terial activity than one with an unsubstituted amino group at
by one of the following mechanisms: (a) the presence of position 2′. Since the discovery of the antibiotic-inactivating
aminoglycoside-modifying enzymes that inactivate them; enzymes, there have been efforts to chemically modify the
(b) decreased cell membrane permeability towards amino- existing antibiotics to produce new drugs that are immune
glycoside uptake into the cell; (c) structural alteration in to inactivation, but at the same time retain their antibacterial

106 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
doi: 10.1054/drup.2001.0197, available online at http://www.idealibrary.com on
Aminoglycoside-modifying enzymes

R4O 6" R3 R4
4" R2 4" CH2OH
5" O 4' "O'
HO 2' 3' HO
H 3N 3" 2"
1" R1O OH HO
1"
OH
6' 3" 2" O
+ HO 1' O NH3 O HO
O 6 4 5' NH3+ IV +
+ 2'
3' 4'
III 5 O H3N OH
R3NH NH3+ O 1' O
1
2
3
I HO 6 4 5' 6'
R1
5
II III HO
R2NH 2 NH3+ I
1 3
aminoglycoside R1 R2 R3 R4 II

kanamycin A OH OH H H aminoglycoside R1 R2 R3 R4
kanamycin B NH3 + OH H H
neomycin NH3+ H H CH2NH3+
amikacin OH OH L-AHBA H
butirosin NH3+ L-AHBA (no ring IV)
tobramycin NH3+ OH R3 CONH2
lividomysin OH H H CH2NH3+
dibekacin NH3+ H H H
paromomycin OH H CH2NH3 + H
r`ibostamycin NH3 + H (no ring IV)

OH
4" 5" 2'
R2
R7 O 3' 4'
1"
R1 6' R3
H3N 2" O
+ 3" HO 1' 5'
OH O 6 5 4
O R5
III R4
R6NH 2 NH3+
1 3

II I

aminoglycoside R1 R2 R3 R4 R5 R6 R7

gentamicin A NH3+ OH OH H OH H H
gentamicin B OH OH OH H NH3+ H CH3
gentamicin B1 OH OH OH CH3 NH3+ H CH3
gentamicin C1 NH3 + H H CH3 NH2CH3 H CH3
gentamicin C1a NH3+ H H H NH3+ H CH3
gentamicin C2 NH3 + H H CH3 NH3 + H CH3
sisomicin NH3 + H 4' H NH3 + H CH3
netilmicin NH3+ H 4' H NH3+ CH2CH3 CH3

AHBA = 4-amino-1-hydroxybutyryl.
Fig. 1 Structure of representative aminoglycosides.

activity. This strategy has led to considerable success in the been forthcoming. First, resistance to aminoglycosides has
fight against penicillin resistance. For instance, the use of emerged gradually and not explosively, as documented for
methicillin, a derivative of the early penicillins, overcame the case of β-lactam antibiotics.22 This is in part due to the
enzyme-mediated inactivation in most penicillin-resistant, fact that β-lactams have been used very extensively clinically,
gram-positive organisms.22 Such strategy has been relatively in contrast to aminoglycosides. On the other hand, the
modest for aminoglycosides, since many of the structural enzymes of resistance to aminoglycosides have not been as
requirements for their antimicrobial activity run parallel to numerous as β-lactamases. They have also been difficult to
those for their enzymatic inactivation.23,24 Nonetheless, semi- study and few laboratories have been willing to undertake
synthetic aminoglycosides have enjoyed clinical success in such efforts. The cornerstone of success in development of
the past several years. Combinations of inhibitors of modify- inhibitors for these enzymes hinges on our ability to under-
ing enzymes and enzyme-susceptible aminoglycosides have stand the basic properties of these resistance enzymes.
not entered clinical use. This contrasts to β-lactam antibi- This report presents a discussion of the best studied of
otics, where clinical use of combinations of β-lactamase these enzymes and presents the results for inhibitors that
inhibitors and penicillins trace back to the mid-1980s.22 have been reported for members of each class of amino-
There are a number of reasons that such a strategies have not glycoside-modifying enzymes. Most of these inhibitors were

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 107
Azucena and Mobashery
designed based on the information obtained from kinetics could enhance the electrophilicity of the latter and thus
and mechanisms of the particular groups of enzymes. Hence, would facilitate the nucleophilic attack by the antibiotic.
only enzymes on which mechanistic studies were per- These authors also proposed the possibility of rotation for
formed, and against which inhibitors have been designed, are ring II of kanamycins such that the 4′′-hydroxyl of ring III
discussed here. could also be adenylated by ATP.This, however, is not possi-
ble for the neomycins, as they lack the internal symmetry in
AMINOGLYCOSIDE NUCLEOTIDYL TRANSFERASES their structure that exist within kanamycins.
The order of binding of the substrates to ANT(4′)
Mechanism was determined by kinetic studies using spectinomycin, a
Most of the mechanistic studies on aminoglycoside kanamycin A analog.28 Inhibition by spectinomycin was found
nucleotidyltransferases have been carried out on two to be competitive versus kanamycin and noncompetitive
enzymes: ANT(4′) and ANT(2′′)-I. The crystal structure of versus ATP, indicating that spectinomycin binds to the
ANT(4′) isolated from Staphylococcus aureus has been elu- free enzyme and not to the E.ATP complex. On the other
cidated.25,26 Several members of the kanamycin and hand, the nonhydrolyzable ATP analogs AMP and AMPCP
neomycin family of aminoglycosides, as well as five (β,γ-methyleneadenosine 5′-triphosphate) are competitive
nucleotide triphosphates (ATP, GTP, CTP, TTP, and UTP) are against ATP but uncompetitive against kanamycin A.Therefore,
substrates for this enzyme.27 Kanamycins and neomycins are these inhibitors bind not to the free enzyme but rather to the
similar in structure for rings I and II, but differ in ring III. E. kanamycin A complex. Substrate binding to the active
While kanamycins have a six-membered ring III, neomycins site is an ordered process, with antibiotic binding taking
have a five-membered ring III (some members have a six- place prior to nucleotide binding. Determination of the
membered ring IV) (Fig. 1).The X-ray crystal structure of the order of product release, which adds further support for
enzyme reveals that several active-site residues interact via the proposed order of substrate binding, was provided by
hydrogen bonds with rings I and II of aminoglycosides, but product-inhibition experiments using the turnover product
relatively fewer residues interact with the rest of the antibi- AMP-kanamycin A. AMP-kanamycin was found to be a com-
otic structure (Fig. 2).The lack of interaction between active- petitive inhibitor of kanamycin A and noncompetitive
site residues and ring III (and IV) accounts for both the inhibitor of ATP.This finding implicates the nucleotidylated-
broad substrate specificity of the enzyme and the compara- aminoglycoside (not pyrophosphate) as the last product
ble rates of turnover for kanamycins and neomycins. released from the enzyme, since it supposes that the amino-
Similarly, the small differences in V and V/Km values among glycoside portion of the product inhibitor binds only to the
the five nucleotide substrates are due to the relatively few free enzyme.The kinetic mechanism that is most consistent
interactions between active-site residues and nucleotide with these inhibition data is an ordered Bi-Bi mechanism. In
base. A catalytic mechanism proposed by Chen-Goodspeed this mechanism, aminoglycoside binds first, followed by the
et al. for ANT(4′) based on the crystal structure and on pH nucleotide.After bond-making and bond-breaking, pyrophos-
dependence of the kinetic parameters postulates that Glu- phate leaves the enzyme first, before the nucleotidylated
145 (Fig. 2) acts as a general base for the activation of the 4′- aminoglycoside does (Fig. 3A). Substrate inhibition is ob-
hydroxyl of the aminoglycoside, and in so doing, prepares served with all aminoglycoside substrates tested for ANT(4′).
the antibiotic for its subsequent nucleophilic attack at the This observation predicates that aminoglycoside, the sub-
α-phosphoryl group of the nucleotide triphosphate.27 strate that binds first to the enzyme, may bind to the antibi-
Moreover, the close proximity between the positively otic-binding site of the E-AMP-aminoglycoside (E.Q) before
charged Lys-149 and the nucleotide α-phosphoryl group the product is completely released, thereby forming a

Glu-141
Ser-94
O
O H
O−

NH3+ NH3+
II
O Ser-39 Thr-187 Lys-149
O O
HO O NH3+
III OH
I O O
HO OH OH H H
HO Glu-145 NH2
NH3+ OH +H3N
−O
O O O H N N
O− O O O
NH3+ −O −O P O P O P O N N
O O
Glu-67 Glu-145
−O O− O
Mg2+
Lys-74
HO OH
Glu-76

Fig. 2 Representation of the interactions between active site residues of ANT(4′) with the substrates,ATP and kanamycin A.27

108 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
Aminoglycoside-modifying enzymes
NMP-aminoglycoside) in a dead-end fashion (Fig. 3B).
Analogous to the mechanism of ANT(4′), binding of B to E.Q
involves the release of the aminoglycoside portion of NMP-
aminoglycoside, while the nucleotide portion remains bound
to the enzyme.The release of the aminoglycoside moiety of
the product is believed to be driven by the exothermic
hydrolysis of a phosphate anhydride bond of NTP, together
with the stereochemical inversion of the α-phosphate, which
is consistent with a mechanism of direct attack by the amino-
glycoside.31 This initial step also exposes the specific aminogly-
coside binding site in the E.Q complex and allows a second
aminoglycoside molecule to bind to the enzyme, a process
that reaches rapid equilibrium, as confirmed by pH- and vis-
cosity-effects data.32 A step in which only a portion of the
product is detached from the enzyme occurs. This event is
subsequent to a slow conformational change, which delays
the release of the product from the enzyme, thereby allow-
Fig. 3 (A) Scheme for the ordered Bi-Bi kinetic mechanism of ing the E.Q.B to accumulate and the inhibition to occur at
ANT(4′).The horizontal line represents the reaction aminoglycoside levels only slightly above the Km values.
proceeding from left to right. E = enzyme; Secondary binding of a substrate inhibitor to the E.Q
A = aminoglycoside; B = nucleotide triphosphate; complex can be tested by the addition of Q to reaction mix-
P = pyrophosphate; Q = nucleotidylated aminoglycoside.
tures to increase the steady-state levels of E.Q, and thus
(B) Scheme for the Theorell-Chance kinetic mechanism of
enhance substrate inhibition.33,34 However, increased Q con-
ANT(2′′)-I.The horizontal line represents the reaction
proceeding from left to right. E = enzyme;A = nucleotide centration does not increase, but rather decreases, substrate
triphosphate; B = aminoglycoside; P = pyrophosphate; inhibition for the ANT(2′′)-I enzyme.This is due to the unique
Q = nucleotidylated aminoglycoside. feature of the Theorell-Chance mechanism, wherein virtually
all of the enzyme already exists as E.Q during steady-state
dead-end complex, E.Q.A, that prevents the full release of the turnover. Therefore, the observed decrease in substrate in-
product. In this case, the aminoglycoside portion of Q is hibition with increase in concentration of Q indicates the
released first from its binding site, while the AMP portion formation of an E.Q.Q complex. Product inhibition by AMP-
remains enzyme-bound. tobramycin (product Q, Fig. 3) was found to be noncompeti-
Another well-studied nucleotidyltransferase is ANT(2′′)-I. tive against ATP.30 Normally, for enzymes that follow the
This enzyme can turn over several aminoglycosides from Theorell-Chance mechanism, product inhibition should be
both the gentamicin and the kanamycin families of antibi- competitive with ATP since Q and ATP both compete for free
otics (Fig. 1).29 The significant differences in the kcat values enzyme.Therefore, for the Theorell-Chance mechanism, it is
for turnover of gentamicins and kanamycins might be attrib- necessary to presuppose that the tobramycin portion of Q
utable to structural differences in ring III, the site of binds to the exposed aminoglycoside-binding site of the E.Q
nucleotidylation.Although a variety of aminoglycosides with and forms E.Q.Q rather than to the free enzyme. The
considerable diversity of structures can be modified by nucleotidylated tobramycin resembles what may be termed a
ANT(2′′)-I, these aminoglycosides have an equatorial 5- bisubstrate analog inhibitor that normally would have a dis-
hydroxyl group in common—presumably a requisite for sociation constant much less than those of either tobramycin
optimal binding to the enzyme.Apart from this, there seems or ATP. However,the dissociation constant for AMP-tobramycin
to be no other requirement for a hydroxyl group at any other was instead found to be an order of magnitude greater than
site except for the site of nucleotidylation. It was suggested that for tobramycin. An explanation for this comes from pH-
that the relative positions of rings I and III may switch in and viscosity-dependence kinetic studies32 that suggest that
binding to the active site, reminiscent of the situation for the aminoglycoside portion of AMP-aminoglycoside is released
ANT(4′).29 Thus, the site occupied by the 6′-amino group of prior to the release of pyrophosphate and that pyrophosphate
kanamycins can be occupied by the 3′′-amino group of the blocks binding of AMP-aminoglycoside to the enzyme.
gentamicins. The ‘natural’ aminoglycoside substrates of ANT(2′′)-I are
Inhibition kinetics of ANT(2′′)-I supports a Theorell- turned over to products with such high-affinity enzyme bind-
Chance kinetic mechanism,30 with nucleotide binding to the ing properties that they practically behave as suicide
enzyme occurring prior to aminoglycoside binding, and the inhibitors.30 Hence, the mechanism by which these enzymes
release of pyrophosphate preceding that of the nucleotidy- confer resistance to bacteria during the initial encounter with
lated aminoglycoside (Fig. 3B). Release of the adenylated the aminoglycoside is an open question. One potential expla-
product is the rate-limiting step. Substrate inhibition is nation to this paradox might be that antibiotic resistance is
observed with nearly all aminoglycosides at concentrations the result of the fast rate of capture of aminoglycosides by
only slightly above their Km values. Substrate inhibition the enzyme upon their entry into the cell, rather than their
by aminoglycosides of ATP binding was shown to be subsequent rate of inactivation by catalytic turnover.30
partial uncompetitive and it arises as a result of binding of For this strategy to be practical, either the uptake of the
aminoglycoside (B) to the E.Q complex (enzyme. antibiotic into bacteria must be relatively slow, or the levels of

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 109
Azucena and Mobashery
expression of the enzymes must be high. Unfortunately, the
literature does not shed light on these issues. O
OR1
R4 7 2
1
Inhibition 3

As discussed above for the mechanism of ANT(2′′), there 6 R2


5
appears to be a requirement for an equatorial hydroxyl group at 4

the 5 position of the 2-deoxystreptamine ring for effective bind- R3


ing of the aminoglycoside to this enzyme. Since the 5-hydroxyl
group is not critical for the antibiotic activity, this position is a
logical candidate for modification as a strategy for development aminoglycoside R1 R2 R3 R4
of effective antimicrobial aminoglycoside.21,35 Thus, semisyn- tropolone H H H H
thetic drugs such as 5-epi-sisomicin36,37,38 and 5-epi-gentamicin39 7-hydroxytropolone (7-HT) H H H OH
(Fig. 4), both of which have the 5-hydroxyl group at the axial 3,5,7-Tri(hydroxymethyl)- H CH2OH CH2OH CH2OH
position, are found to be effective against a broad spectrum of tropolone
7-hydroxytropolone COMe H H COMe
bacteria that harbor resistance enzymes, including ANT(2′′), diacetate
AAC(3),AAC(2′).ANT(2′′) was shown to have about 9- and 55-
fold higher Km for 5-epi-sisomicin and 5-epi-gentamicin B, Fig. 5 Structures of tropolone and tropolone derivatives.
respectively, compared to the corresponding parent com-
pounds, sisomicin and gentamicin.39
The results of these in vitro inhibition studies were found
7-Hydroxytropolone (7-HT, Fig. 5), a fermentation product
to parallel those of in vivo experiments, demonstrating
of certain Pseudomonas and Streptomyces species, and some
potentiation of tobramycin activity by 7-HT against
of its derivatives were found to be potent inhibitors specific for
tobramycin-resistant strains. For example, growth of
ANT(2′′) and ANT(4′).40,41,42 Kinetic studies with 7-HT showed
Escherichia coli strains that harbor ANT(2′′) was not affected
that in combination with aminoglycosides tobramycin, gen-
by tobramycin, gentamicin, amikacin, or dibekacin alone.
tamicin, kanamycin A or dibekacin, this compound can inhibit
However, each of these antibiotics was shown to inhibit
ANT(2′′).40 Certain derivatives of 7-HT, such as 7-hydrox-
growth of the same E. coli strains when combined with 7-HT.
ymethyltropolone, 3,5,7-tri(hydroxymethyl)-tropolone, and 7-
The synergistic effect between 7-HT and various aminogly-
HT diacetate can cause complete inhibition of adenylation of
cosides was not observed in E. coli strains that harbor APHs
certain aminoglycosides at inhibitor concentrations of 50
or AACs, which is an indication of the specificity of the inhi-
µg/mL.42 Replacement of the hydroxyl group at the 7-position
bition towards aminoglycoside nucleotidylation. It is interest-
with bromo, nitro, carboxyl, or amino group gave inactive
ing to note, however, that 7-HT did not inhibit the APH
derivatives,40 emphasizing the importance of the 7-hydroxyl
enzymes, despite the requirement by these enzymes for
group for the inhibitory activity. Inhibition due to 7-HT is com-
ATP.40
petitive with respect to ATP (Ki = 10 µM) and not competitive
The effects of a combination of tobramycin plus 7-HT
with respect to tobramycin. Whether inhibition against
and of tobramycin alone on [3H]dihydrostreptomycin
tobramycin was uncompetitive or noncompetitive has not
([3H]DHS) uptake was examined in an E. coli strain that har-
been clearly determined.40
bors tobramycin-adenylating and streptomycin-phosphory-
lating enzymes and in a susceptible E. coli strain.40 In the
susceptible strain, accumulation of [3H]DHS occurs after a
short lag period in the absence of tobramycin. However, the
uptake of [3H]DHS occurred at an increased rate and with-
OH
out a lag when susceptible cells were pre-exposed to
+
H3C O H 3N tobramycin, indicating that preincubation with tobramycin
H3CHN O
facilitates the further entry of aminoglycosides. When the
OH 5 susceptible cells were preincubated with 7-HT or 7-HT plus
O O
tobramycin prior to incubation with [3H]DHS, the cellular
H 3N NH3+ NH3+
+ OH uptake of [3H]DHS was unaffected. In contrast, uptake of
[3H]DHS was inadequate in the ANT/APH-bearing strain
5-epi-sisomicin
without pre-exposure to tobramycin and was only slightly
OH enhanced with pre-exposure to tobramycin. Preincubation
HO
with a combination of tobramycin and 7-HT, however,
O OH resulted in a dramatic increase in [3H]DHS uptake. These
H3 C HO
H3CHN O results may be due to the observed reduced antibiotic
NH3+
OH 5 uptake that accompanies aminoglycoside modification in
O O
H3N NH3+ resistant strains.10,19,20 The effectiveness of the combination
+ OH of 7-HT and aminoglycosides in inhibiting bacterial cell
5-epi-gentamicin B growth is therefore credited to the aminoglycoside-ribo-
some interaction, which results in increased aminoglyco-
Fig. 4 Structures of 5-epi-sisomicin and 5-epi-gentamicin B. side uptake.

110 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
Aminoglycoside-modifying enzymes
AMINOGLYCOSIDE ACETYLTRANSFERASES of the other substrate, while in an ordered sequential mecha-
nism only one set of data should show this dependence.
Mechanism
Additional data from kinetics studies established a rapid equi-
The first aminoglycoside-modifying enzyme ever reported in
librium feature for substrate binding in this mechanism.45
bacteria (second only to penicillinase as an antibiotic-
Another aminoglycoside acetyltransferase enzyme,
modifying enzyme) was aminoglycoside 6′-N-acetyltrans-
AAC(3)-I, has been subjected to detailed kinetic analysis.47,48
ferase type-IV [AAC(6′)-IV, also known as AAC(6′)-Ib].43
Sixteen aminoglycosides and derivatives were found to be
Radika and Northrop established the kinetic mechanism for
substrates for this enzyme, including members of the gen-
this enzyme as rapid equilibrium sequential random Bi-Bi
tamicin and kanamycin families. Since the site of aminogly-
(Fig. 6A)44,45 using a kinetic diagnostic that they developed
coside modification is the 2-deoxystreptamine ring,
for distinguishing the ping-pong (Fig. 6B) from the sequential
derivatives containing only two rings, one of which contains
mechanisms, as well as for distinguishing among sequential
the site of modification, are among the substrates for this
mechanisms.46 In this diagnostic analysis, concentration of
enzyme.These include neamine, sisomine, gentamine Cla, and
substrate A is varied against a fixed saturating concentration
tobramine—two-ring derivatives of neomycin, sisomicin,
of the alternative substrates B, B′, B′′, etc., and B is similarly
gentamicin Cla, and tobramycin, respectively.47 Initial-velocity
varied against a series of the alternative substrates A. This
kinetic experiments with tobramycin, sisomicin, and gentam-
method was amenable to the study of AAC(6′)-IV, since it can
icin Cla revealed that AAC(3)-I exhibits a sequential mecha-
transfer a variety of acyl groups (acetyl, n-propionyl, n-
nism.48 Gentamicin Cla was found to be a substrate inhibitor
butyryl, n-valeryl, isovaleryl, and isohexyl).45 The initial-
of the enzyme, and this inhibition is partially uncompetitive
velocity kinetics for discriminating the ping-pong
with respect to acetyl-CoA.This is evidence for a sequential
mechanism from the sequential mechanisms requires the
mechanism, arising from binding of the second gentamicin
use of substrate concentrations at the vicinity of the value for
Cla to the enzyme after catalysis, forming E.CoA.gentamcin
Km.This may prove difficult when the Km value is very low, as
Cla complex and preventing the release of the CoA.This par-
is the case for substrates of AAC(6′)-IV (Km for acetyl-CoA =
tially uncompetitive substrate inhibition suggests an incom-
0.8 µM).45 In addition, initial-velocity patterns show differ-
plete trapping of CoA by gentamicin Cla. It was therefore
ences in intercepts and slopes for the disparate kinetic
postulated that a degree of randomness in substrate binding
mechanisms of only a factor of 2 or less, which in turn may
exists that is coupled to partial rate limitation of turnover by
produce imprecise results.The Radika-Northrop method cir-
the rate of product release. Based on these results, a random
cumvents the requirement for near-Km substrate concentra-
Bi-Bi mechanism (Fig. 6A) was proposed for this enzyme.This
tions and shows greater variance in V/Km among kinetic
proposed mechanism was further validated by product
mechanisms.
inhibition experiments in which CoA was shown to be
The data obtained for AAC(6′)-IV using this method indi-
competitive against acetyl-CoA and noncompetitive against
cated that V/Km for the second substrate was dependent on
tobramycin, while acetyl-tobramycin was competitive and
the identity of the antibiotic, and the V/Km for an aminoglyco-
noncompetitive with reference to tobramycin and acetyl-
side was dependent on the nature of the nucleotide.44 This
CoA, respectively.48 An important characteristic of both prod-
mutual dependence of V/Km for one substrate on the identity
uct and substrate inhibitions by gentamicin Cla is the
of the other substrate is consistent only with a random
synergism that exists between CoA binding and gentamicin
sequential mechanism. In the ping-pong mechanism, the V/Km
Cla binding, i.e. the presence of CoA on the enzyme enhances
for a substrate should not exhibit dependence on the identity
binding of gentamicin Cla and vice versa.Additional support
for the proposed mechanism came from dead-end inhibition
and multiple inhibition patterns.48
The initial-velocity pattern for sisomicin showed an
upward curve at low concentrations. This indicates a non-
rapid equilibrium for enzyme-substrate binding in which the
reaction Vmax is greater than the rate constant for the release
of the substrate from the enzyme.49 In fact, the rate constant
for binding of sisomicin to enzyme during catalytic turnovers
is found to approach the normal diffusion-controlled limits,
making the product release rate limiting. On the other hand,
the initial-velocity pattern for tobramycin is linear, with its
binding rate constant two orders of magnitude less than that
for sisomicin.48 In this case, substrate binding takes place as a
rapid equilibrium, and catalytic steps of bond-making and
bond-breaking become primarily rate limiting. Thus, an
antibiotic-dependent shift from rapid to nonrapid equili-
brium random mechanism occurs as one shifts from to-
bramycin (poor substrate) to sisomicin (good substrate).48
Fig. 6 Kinetic scheme for (A) random Bi-Bi and (B) ping-pong Crystal structures of two aminoglycoside acetyltrans-
mechanisms.A and B = substrates; P and Q = products; ferases have recently been determined. These are AAC(3)-Ia
E = enzyme; F = modified enzyme. from Serratia marcescens50 and AAC(6′)-li from Enterococcus

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 111
Azucena and Mobashery
faecium.51 AAC(3)-Ia was observed to be similar in folding to in modifying the structures of existing aminoglycosides to
the yeast histone acetyltransferase HATI and N-myristoyltrans- produce drugs that are refractory to enzyme inhibition with-
ferase—both protein-acylating enzymes and members of the out impairing their antibacterial activity. Unfortunately, from
GCN5-related N-acetyltransferase superfamily. the correlation of the antimicrobial activity with the amino-
Comparison of the crystal structure of AAC(6′)-Ii in com- glycoside structures, many of the structural requirements of
plex with acetyl-CoA with S. marcescens AAC(3)-la and Hat1 aminoglycosides for enzymatic activity run parallel to those
revealed that it belongs to this superfamily of enzymes as for antimicrobial activity.Thus, there are very few chemically
well. Although the sequence identity among these enzymes modified aminoglycoside derivatives that have been found to
was negligible, functional studies show that this aminoglyco- retain activity against bacterial strains harboring these
side-modifying enzyme possesses protein-acetylation activity. enzymes. Derivatives that possess antimicrobial properties
include 5-epi-sisomicin36,38 and netilmicin55,56 which are
Inhibition shown to be resistant to modification by AAC(3)-I.
A tight-binding, bisubstrate analog has been synthesized as
an inhibitor of AAC(3)-I.52 The design of this inhibitor was
based on the proposed kinetic mechanism and aminoglyco- AMINOGLYCOSIDE PHOSPHOTRANSFERASES
side structure-function studies for this enzyme, as described
above.48 (1) Substrate binding is sequential, that is both Mechanism
antibiotic and CoA must be present on the enzyme before Among the known APHs, the best studied is APH(3′)-IIIa.This
any product is released and no covalent bonds would be enzyme confers resistance to a broad spectrum of aminogly-
formed between the enzyme and either of the two sub- cosides, including members of the kanamycin and neomycin
strates; (2) substrate binding is synergistic; (3) interactions of groups of aminoglycosides, most of which were also found to
gentamicin C1a with active site residues occur primarily on be relatively poor substrate inhibitors for this enzyme.57 The
the purpurosamine and garosamine rings (rings I and III), structural basis for substrate inhibition is not well elucidated.
with few interactions with 2-deoxystraptamine; (4) acyl One possible explanation is that the drug may bind improp-
transfer by the enzyme accommodates acetyl-CoA and propi- erly to the active site of the enzyme.The substrate specificity
onyl-CoA, but not butyryl-CoA or malonyl-CoA.The inhibitor of this enzyme indicates that there may be little stringency for
(Fig. 7) was enzymatically prepared using chloroacetyl-CoA 4,6-disubstituted aminoglycosides (e.g. kanamycins) and 4,5-
in place of acetyl-CoA. During the enzymatic reaction with disubstituted aminoglycoside (e.g. neomycins), and that the
gentamicin C1a, the chloroacetyl moiety was transferred to aminocyclitol ring is significant for binding. Interestingly,
the 3 position of gentamicin C1a, and the sulfur of the CoA lividomycin A, which lacks the 3′-hydroxyl group, was found
product displaced chlorine on the transferred acyl group via to be a substrate for APH(3′)-IIIA.57 The site of modification of
nucleophilic attack. this aminoglycoside was found to be the 5′′-hydroxyl group.58
The bisubstrate inhibitor gave a Ki of 0.5 to 20 nM against Butirosin and neomycin, both of which possess 3′ and 5′′
AAC(3)-I.53 When tested against a resistant E. coli strain that hydroxyl groups, were found to be phosphorylated at either
harbored AAC(3)-I, the inhibitor did not show any effect on sites.59 NMR analysis of butirosin bound to a ternary
the antibiotic activity of gentamicin presumably due to its Cr.ATP.APH(3′)-IIIa complex shows that both 3′-and 5′′-
inability to cross the cell membrane. In addition to this hydroxyl groups are in close proximity to the γ-phosphoryl
inhibitor, several aminoglycosides that were tested as non- group of ATP.60 It is speculated that the first phosphorylation
substrates for AAC(3)-I exhibited competitive inhibition occurs at random at either position, followed by a conforma-
against substrate aminoglycosides, neomycin B being the tional change that precedes the second phosphorylation.This
best inhibitor having a submicromolar iK.47 Also, two semi- conformational change draws the negatively charged phos-
synthetic kanamycin derivatives, 6′-N-methylkanamycin A phoryl group on the modified aminoglycoside away from the
and 3′, 4′-dideoxy-6-′-N-methylkanamycin B have been shown incoming second ATP molecule.
to possess strong antibacterial activity against strains having It was proposed that the chemical mechanism of the
6′-N-acetylating enzymes, as they are resistant to modifica- phosphoryl transfer reaction between the aminoglycoside
tion at the 6′-position.54 and ATP among APH(3′)s involves a conserved histidine
Detailed structure-function relationships for acylation of residue that would relay the phosphoryl group from the γ-
several aminoglycyosides with AAC(6′)-IV and AAC(3)-I have position of ATP to the aminoglycoside 3′-hydroxyl group.61
been reported.45,47 These data provide insights for strategies This notion was disproved, at least in the case of APH(3′)-IIIa,
when conversion of the equivalent His residue (His-188) in
APH(3′)-IIIa failed to abolish the enzyme activity, thereby
OH
+ supporting the suggestion that phosphoryl transfer occurs
H3C O H3N
H3N HO O through direct attack of the 3′-hydroxyl group of the amino-
NH2 + NH3+
OH O
H3N
ON
glycoside on the γ-phosphoryl group of ATP.62 The crystal
N N + O
−OO−O O O O structure of this enzyme predicted that residue Asp-190,
N N O P P S which is in close proximity to the aminoglycoside 3′-
O O O N N
OH H H hydroxyl group, acts as the catalytic base that abstracts the
HO P O− 3′-hydroxyl proton to facilitate attack at ATP.63 This predic-
−O
O
tion was validated by site-directed mutagenesis, which
Fig. 7 Structure of acetylgentamicin-CoA inhibitor. demonstrated that replacement of this Asp-190 with alanine

112 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
Aminoglycoside-modifying enzymes
resulted in at least a 650-fold decrease in kcat.63 Site-directed rapid equilibrium random mechanism for this enzyme.68
mutagenesis also established the importance of interaction Thus, similarly to the case of APH(3′)-IIIa, this enzyme
of Lys-44 with the α-and β-phosphates of ATP, as this residue exhibits a direct displacement mechanism for the γ-phospho-
may impart greater electrophilicity to the γ-phosphoryl ryl of ATP to the aminoglycoside. In the absence of aminogly-
group for enhanced nucleophilic attack by the aminogly- cosides, this enzyme was also shown to hydrolyze ATP with a
coside by its electrostatic interaction.63 Further structural Km.ATP value in the same range as that for the aminoglycoside
analyses of APH(3′)-IIIa-bound aminoglycosides by NMR phosphotransferase activity of the enzyme (Km = 49 ± 1 µM).
demonstrated that amikacin and butirosin can assume sev- This indicates that aminoglycoside binding does not influ-
eral different arrangements at the active site of the ence the enzyme affinity for ATP.68 Insofar as the physiologic
enzyme.64,65 It is presumed that not all of the bound confor- ATP concentration in bacteria is well above this Km value, it
mations lead to productive phosphoryl transfer. This may would appear that this enzyme is constantly hydrolyzing ATP
provide explanation for the observed low kcat/Km for these in vivo. However, the kcat for the ATP hydrolysis is signifi-
aminoglycosides.57 cantly lower than that for the aminoglycoside phosphoryla-
APH(3′)-IIIa was subjected to kinetic studies to determine tion, such that the ATPase activity does not compete with
its catalytic mechanism.66 Initial velocity patterns ruled out phosphorylation of aminoglycosides when these drugs are
the ping-pong mechanism and supports a sequential mecha- present.
nism.ATP would bind first, followed by the aminoglycoside, APH(3′)-IIa differs from APH(3′)-Ia in that it modifies both
as established by dead-end inhibition patterns.Tobramycin is butirosin and lividomycin.This enzyme also possesses ATPase
not a substrate for this enzyme, but was found to be a strong activity in the absence of aminoglycosides (Km.ATP = 52 ± 19
competitive inhibitor with reference to kanamycin A, but µM), similarly to what is observed for the type Ia counter-
uncompetitive inhibitor relative to ATP. Both AMP and AMP- part.69 It is believed that electrostatic interactions (ion pair-
PNP (a nonhydrolizable ATP isostere) exhibited competitive ing and hydrogen binding) are significant for both substrate
inhibition versus ATP, and noncompetitive versus amikacin. binding and catalysis among aminoglycoside-modifying en-
The uncompetitive substrate inhibition observed with zymes.To assess the significance of these interactions, seven
kanamycin against ATP is indicative of dead-end binding of distinct deaminated analogs of neamine and kanamycin A
kanamycin to the E.ADP complex, suggesting a partial rate- were synthesized by Roestamadji et al., and were tested with
limiting release of ADP. Noncompetitive product inhibition APH(3′) types Ia, IIa, and IIIa.70,71 All seven molecules were
with kanamycin phosphate versus ATP can be circumvented shown to be exceedingly poor substrates for both APH(3′)-Ia
by increasing kanamycin A concentration. This establishes and APH(3′)-IIa, arguing for the critical role of electrostatic
the order of product release as follows: rapid release of interactions between the aminoglycoside and the active sites
kanamycin phosphate, followed by a slow release of ADP. of these enzymes.Although the Km values for these deamino
These results collectively argue for a Theorell-Chance mecha- derivatives increased only modestly relative to those for the
nism for this enzyme in which ATP binds first, followed by parent aminoglycosides, the kcat values were dramatically
aminoglycoside, and the rapid release of kanamycin phos- decreased by several orders of magnitutde.70,71 Surprisingly,
phate taking place before the slow release of ADP. These APH(3′)-IIIa seemed to interact with the deaminated amino-
results are corroborated further by solvent-isotope effect glycoside derivatives in quite a different fashion than
measurements and thio-and viscosity-effect experiments.67 A APH(3′)-Ia and -IIa, as it showed no clear kinetic pattern
small solvent isotope effect measured for reactions carried among most of these deaminated aminoglycosides and the
out in deuterium oxide ruled out the possibility that the parent compounds.
enzyme-mediated deprotonation of the aminogycoside 3′-
hydroxyl group is the rate-limiting step. Likewise, the phos- Inhibition
phate-transfer step (from ATP to the aminoglycoside) was The deaminated analogs of neamine and kanamycin A,
ruled out as the predominant contributor to the overall rate described above, were shown to possess biological activity
of reaction, since it produced only a small thio-effect when against strains that harbor aminoglycoside 3′-phoaphotrans-
ATP was replaced by ATPγS.Alteration of the viscosity of the ferases types Ia and IIa, indicating that these compounds
assay solution did not affect the V/Km for aminoglycosides, were not modified by these enzymes in vivo.70 Thus, the syn-
but had a slope effect on the V/Km for ATP.This identified ATP thesis of such antibiotic derivatives that have reduced elec-
binding and ADP release as diffusion-controlled processes. trostatic interactions with their corresponding enzymes may
However, since under physiologic conditions, aminoglyco- prove to be valuable in the circumvention of resistance to
side (not ATP) is limiting, ADP release was identified as the antibiotics. Roestamadji et al. also synthesized mechanism-
primary contributor to the enzymatic reaction rate.This pro- based inactivators (Fig. 8) whose design was based on mech-
posal is consistent with the Theorell-Chance mechanism (see anistic studies with APH(3′)s implicating an active-site
Figure 3B). catalytic base in activation of the 3′-hydroxyl proton of
Two additional APHs,APH(3′)-Ia and APH(3′)-IIa, have also the substrate aminoglycoside.72 These compounds are 2′-
been studied.APH(3′)-I is the most common APH is gram-neg- deamino-2′-nitro derivatives of neamine and kanamycin B.
ative bacteria. Kinetic analyses by the alternative-substrate Upon phosphorylation of 2′-deamino-2′-nitro-neamine at its
method46 (using amikacin, kanamycin A, and neomycin as 3′ position, the phosphoryl group is spontaneously elimi-
aminoglycoside substrates, and ATP, GTP, and UTP as nated.As a result, an electrophilic inactivating species is gen-
nucleotide substrates), together with product inhibition erated, which then captures an active-site nucleophilic
(with kanamycin A 3′-phosphate and ADP) determined a residue.A covalent bond is thus formed between the enzyme

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 113
Azucena and Mobashery

HO
2' 4'
3'
O2N OH 6" OH
HO 1' O 6' 4" HO
5' NH3+ 5" O 2' 4'
6 4 3'
5 O HO O2N OH
HO 1" 6'
NH3+ +H3N 3" 2" HO 1' O
2 4 NH3+
+H3N 1 3 OH
6 5 5'
O O
+H3N 2 NH3+
1 3

Fig. 8 Structures of mechanism-based inactivators for APH(3′)s.The structure on the left is 2′-deamino-2′-nitro-neamine; the
structure on the right is 2′-deamino-2′-nitro-kanamycin B.

HO 4'
2'
H2N
3'
OH 2' HO 4'
3'
1' O H2N OH
HO 6'
4 5' NH2 1' O
6
O HO 6'
HO 5
6 4
5' NH2
5 O
NH2 HO
HN 2
1 3 NH
H2N 1
2
3
Br Br
O O

O
Br HO 4' 2' HO 4'
2' 3' 3'
OH O
N OH H2N
H O HO 1' O 6' Br
HO 5'
6'
4
5' NH
6 4 NH2 6
5 O
5 O HO
HO
NH2 H2N 2 NH2
H2N 2 1 3
1 3

Fig. 9 Structures of bromoacretylated analogs of neamine.

and the inactivator and an irreversible inactivation of the γ-CH2ATP and/or kanamycin A, the observed rate of inactiva-
enzyme ensues.This mechanism is validated by the detection tion was attenuated. Therefore, inactivation is directed
of the released inorganic phosphate during the inactivation towards the active site.To see whether the inactivators react
process, which is rapid and time-dependent, among other with any of the five thiols of cysteine residues69—potential
experiments.72 sites of preferential modification by the inactivators—the
The 2′-deamino-2′-nitro derivative of kanamycin B was cyanylated enzyme, which has all its cysteine thiols pro-
also found to inactivate APH(3′) via a mechanism similar to tected with the nitrile group, was prepared. The activity of
that proposed for 2′-deamino-2′-nitro-neamine, except that the enzyme upon cyanylation was found to be unaffected,
the enzyme inactivated by 2′-deamino-2′-nitro-kanamycin B and the kinetic parameters for the enzyme inactivation with
regained activity over several hours.This observed difference the cyanylated and uncyanylated enzymes were virtually the
in the inactivation profile could be attributed to interaction same.
of the two compounds with different active-site residues. The close similarity in the chemical mechanism and overall
This is the only account of a mechanism-based inhibition for three dimensional structure between APH(3′)-IIIa and protein
aminoglycoside resistance enzymes.72 kinases led to the investigation of possible sensitivity of APHs
Four bromoacetylated analogs of neamine were synthe- to known inhibitors of protein kinases. Of the three classes of
sized by Roestamadji et al. (Fig. 9) as affinity inactivators for inhibitors tested, the isoquinolinesulfonamides were found to
APH(3′)-IIa.73 In the presence of ATP, the enzyme could phos- be good inhibitors of APH(3′)-IIIa and the bifunctional
phorylate these compounds, but only at higher Km values, AAC(6′)-APH(2′′).74 Unfortunately, these compounds did not
ranging from 3-to 86-fold greater than that for the unmodi- reverse antibiotic resistance in enterococcal strains that har-
fied neamine. Since Km may approximate Ks here, the effect bor either the aph(3′)-IIIa gene or aac(6′)-aph(2′′) gene.
of structural modification of neamine in these compounds is In a recent publication, Wong et al. Synthesized various
largely on binding, and not turnover (i.e. kcat). On the other dimers of neamine linked via either amides or 1,2-hydrox-
hand, in the absence of ATP, all four compounds inactivated yamine moieties.75 These dimers were designed to identify
APH(3′)-IIa in a time-dependent and saturable fashion. The bivalent aminoglycosides that would interact with biotiny-
inactivated protein did not recover activity despite extensive lated E. coli 16S rRNA A-site—a model for the target site of
dialysis to remove the inactivator molecules, suggestive of aminoglycosides in bacteria.Three of these compounds were
covalent bond formation between the enzyme and the in- shown to be potent inhibitors of the APH(2′′) activity of the
activators. When the enzyme was preincubated with β, bifunctional enzyme AAC(6′)-APH(2′′), with a Kis as low as 0.1

114 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
Aminoglycoside-modifying enzymes
µM. Furthermore, these dimers show antimicrobial activity, 9. Azucena E, Grapsas I, Mobashery S. Properties of a bifunctional
with the most active dimer exhibiting MIC of 6.25 µM bacterial antibiotic resistance enzyme that catalyzes ATP-
against E. coli, and a Kd of 40 nM for the biotinylated 16s dependent 2′′-phosphorylation and acetyl-CoA-dependent
rRNA A-site. 6′-acetylation of aminoglycosides. 1997; 119: 2317–2318.
10. Bryan LE,Van Den Elzen HM. Streptomycin accumulation in
susceptible and resistant strains of Escherichia coli and Pseudomonas
CONCLUDING REMARKS
aeruginosa.Antimicrob Agents Chemother 1976; 9: 928–938.
In this review we have attempted to provide biochemical 11. Bryan LE,Van Den Elzen HM. Effects of membrane- energy
information for each of the three classes of bacterial resis- mutations and cations on streptomycin and gentamicin
tance enzymes for aminoglycosides.The emphasis was placed accumulation by bacteria: a model for entry of streptomycin and
on surveying the published body of information on the gentamicin in susceptible and resistant bacteria.Antimicrob Agents
respective mechanisms of these enzymes, along with infor- Chemother 197; 12: 163–177.
mation on their inhibition.There are scores of such inactivat- 12. Taber HW, Mueller JP,Arrow AS. Bacterial uptake of
ing enzymes for these antibiotics, yet mechanistic knowledge aminoglycoside antibiotics. Microbiol Rev 1987; 51: 439–457.
exists only for a handful. To date, there are no examples of 13. Hurwitz CC, Braun B, Rosano CL. Role of ribosome recycling in
combination drugs of aminoglycoside-modifying enzyme uptake of dihydrostreptomycin by sensitive and resistant
inhibitors and aminoglycosides. However, in order to counter Escherichia coli. Biochim Biophys Acta 1981; 652: 168–176.
the resistance produced by these enzymes, detailed mecha- 14. Davis BD, Chen LL,Tai PC. Misread protein creates membrane
nistic understanding of them is a critical first step. channels: an essential step in the bacterial action of
aminoglycosides. Proc Natl Acd Sci USA 1986; 83: 6164–6168.
15. Davis BD. Mechanisms of action of aminoglycosides. Microbiol Rev
1987; 51: 341–350.
Received 23 February 2001;Accepted 6 April 2001
16. Moazed D, Noller HF. Interaction of antibiotics with functional
sites in 16S ribosomal RNA. Nature 1987; 27: 389–394.
Correspondence to: Shahriar Mobashery, Institute for Drug Design,
17. Woodcock J, Moazed D, Cannon M, Davies J, Noller HF. Interaction
Departments of Chemistry, Pharmacology and Biochemistry and Molecular
of antibiotics with A- and P-site-specific bases in 16S ribosomal
Biology, Wayne State University, 5101 Cass Avenue, Detroit, MI 48202, USA.
RNA. EMBO J 1991; 10: 3099–3103.
Tel.: +313 577 3924; Fax: +1 313 577 8822;
18. Fourmy D, Recht MI, Blanchard SC, Puglisi JD. Structure of the A
E-mail: som@gordon.chem.wayne.edu
site of Escherichia coli 16S ribosomal RNA complexed with an
aminoglycoside antibiotic. Science 1996; 274: 1367–1371.
19. Dickie PL, Bryan LE, Pickard MA. Effect of enzymatic adenylation
on dihydrostreptomycin accumulation in Escherichia coli carrying an
References R-factor: model explaining aminoglycoside resistance by
1. Haddad J, Kotra LP, Mobashery S.Aminoglycoside antibiotics: inactivating mechanisms. Antimicrob Agents Chemother 1978; 14:
structures and mechanism of action. In:Wang G, Bertozzi C, eds. 569–580.
Glycochemistry: principles, synthesis, and applications. New York, 20. Holtje, JW. Induction of streptomycin uptake in resistant strains of
Marcel Dekker, 2001, 307–351. Escherichia coli. Biochim Biophys Acta 1979; 15: 177–181.
2. Haddad J, Liu MZ, Mobashery S. Methodologies in syntheses of 21. Benveniste R, Davies J. Structure-activity relationships among the
aminoglycoside antibiotics. In:Wang G, Bertozzi C, eds. aminoglycoside antibiotics: role of hydroxyl and amino groups.
Glycochemistry: principles, synthesis, and applications. New York, Antimicrob Agents Chemother 1973; 4: 402–409.
Marcel Dekker, 2001, 353–424. 22. Bush, K, Mobashery S. How beta-lactamases have driven
3. Achatz A, Bugie E,Waksman SA. Streptomycin, a substance pharmaceutical drug discovery – From mechanistic knowledge to
exhibiting antibiotic activity against Gram-positive and Gram- clinical circumvention Adv Exp Med Biol 1998; 456: 71–98.
negative bacteria. Proc Soc Exp Biol Med 1944; 55: 66–69. 23. Kotra LP, Mobashery S. A renaissance of interest in
4. Waksman SA, Lechevalier HA. Neomycin, a new antibiotic active aminoglycoside antibiotics, Curr Org Chem 2000; 5: 89–111.
against streptomycin-resistant bacteria, including tuberculosis 24. Kotra LP, Haddad J, Mobashery S. aminoglycoside antibiotics:
organisms. Science 1949; 109: 305–307. perspectives on mechanisms of action, resistance and strategies to
5. Hinshaw HC, Feldman WH. Streptomycin in treatment of clinical counter resistance, Antimicrob Agents Chemother 2000; 44:
tuberculosis: preliminary report. Mayo Clin 1945; 35: 1147–1152. 3249–3256.
6. Wright GD, Berghuis AM, Mobashery S. [AU:Title?] In: Rosen BP, 25. Joshua S, Liao HH, Kanikula AM, Benning MW, Rayment M, Holden
Mobashery S, eds. Resolving the antibiotic paradox. News York, HM. Molecular structure of kanamycin nucleotidyltransferase
Plenum Press, 1998, 33–34. determined to 3.0 Å resolution. Biochemistry 1993; 32:
7. Shaw KJ, Rather PN, Hare RS, Miller GH. Molecular genetics of 11977–11984.
aminoglycoside resistance genes and familial relationships of the 26. Pedersen LC, Benning MW, Holden HM. Structural investigation of
aminoglycoside-modifying enzymes. Microbiol Rev 1993; 57: 138–163. the antibiotics and ATP-binding sites in kanamycin
8. Ferretti JJ, Gilmore KS, Courvallin P. Nucleotide sequence analysis nucleotidyltransferase. Biochemistry 1995; 34: 13305–13311.
of the gene specifying the bifunctional 6′aminoglycoside 27. Chen-Goodspeed M,Vanhooke JL, Holden HM, Raushel FM.
acetyltransferase 2′′-aminoglycoside phosphotransferase enzyme in Kinetic mechanism of kanamycin nucleotidyltransferase
Streptococcus faecalis and identification and cloning of gene regions from Staphylococcus aureus. Bioorg Chem 1999; 27:
specifying the two activities. J Bacteriol 1986; 167: 631–638. 395–408.

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 115
Azucena and Mobashery
28. Fromm HJ. Use of competitive inhibitors to study substrate 47. Williams JW, Northrop DB. Substrate specificity and structure-
binding order. Methods Enzymol 1979; 63: 467–485. activity relationships of gentamicin acetyltransferase I. J Biol Chem
29. Gates CA, Northrop DB. Substrate specificities and structure- 1978; 17: 5908–5914.
activity relationships for the nucleotidylation of antibiotics 48. Williams JW, Northrop DB. Kinetic mechanism of gentamicin
catalyzed by aminoglycoside nucleotidyltransferase 2′′-I. acetyltransferase I. J Biol Chem 1978; 253: 5902–5907.
Biochemistry 1988; 27: 3820–3825. 49. Bar-Tana J, Cleland WW. Rabbit muscle phosphofructokinase II:
30. Gates CA, Northrop DB. Alternative substrate and inhibition product and dead-end inhibition. J Biol Chem 1974; 249:
kinetics of aminoglycoside nucleotidyltransferase 2′′-I in support f 1271–1276.
a Theorell-Chance kinetic mechanism. Biochemistry 1988; 27: 50. Wolf E,Vassiley A, Makino Y, Sali A, Nakatani Y, Burley SK. Crystal
3826–3833. structure of a GCN5-related N-acetyltransferase: Serratia
31. Van Pelt JE, Iyengar R, Frey PA. Gentamycin nucleotidyltransferase: marcescens aminoglycoside 3-N-acetyltransferase. Cell 1998; 94:
stereochemical inversion at phosphorus in enzymatic 2′- 439–449.
deoxyadenyltransfer to tobramycin. J Biol Chem 1986; 261: 51. Wybenga-Groot LE, Draker K,Wright GD, berghuis AM. Crystal
15995–15999. structure of an aminoglycoside 6′-N-acetyltransferase: defining the
32. Gates CA, Northrop DB. Determination of the rate- limiting GCN5-related N-acetyltransferase superfamily fold. Struct Fold
segment of aminoglycoside nucleotidyltransferase 2′′-I by pH- and Des 1999; 7: 497–507.
viscosity-dependent kinetics. Biochemistry 1988; 27: 3820–3825. 52. Williams JW, Northrop DB. Synthesis of a tight- binding,
33. Northrop BD, Cleland WW.The kinetics of pig heart multisubstrate analog inhibitor of gentamicin acetyltransferase I.
triphosphorydine nucleoride-isocitrate dehydrogenase II: dead-end J Antibiot 1979; 32: 1147–1154.
and multiple inhibition studies. J Biol Chem 1974; 249: 2928–2931. 53. Jencks WP. Binding energy, specificity, and enzyme catalysis: the
34. Rife JE, Cleland WW. Kinetic mechanism of glutamate Circe effect.Adv Enz 1975; 43: 219–410.
dehydrogenase. Biochem 1980; 19: 2321–2328. 54. Umezawa H, Nishimura Y. Synthesis of 6′-N-methylkanamycin and
35. Moellering RC Jr. In vitro antibacterial activity of the 3′, 4′-dideoxy- 6′-N-methylkanamycin B active against resistant
aminoglycoside antibiotics. Rev Infect Dis 1983; 5: S212–S232. strains having 6′-N-acetylating enzymes. J Antibiot 1972; 25:
36. Waitz JA, Miller GH, Moss Ejr, Chiu PJS. Chemotherapeutic 743–745.
evaluation of 5-episisomicin (Sch 225910), a new semisynthetic 55. Kabins SA, Nathan C, Cohen S. In vitro comparison of netilmicin, a
aminoglycoside.Antimicrob Agents Chemother 1978; 13: 41–48. semisyhtetic derivative of sisomicin, and four other aminoglycoside
37. Fu KP, Neu HC. Activity of 5-episisomicin compared with that of antibiotics.Antimicrob Agents Chemother 1976; 10: 139–145.
other aminoglycosides.Antimicrob Agents Chemother 1978; 14: 56. Miller GH,Arcieri G,Weinstein MJ,Waitz JA. Biological activity of
194–200. netilmicin, a broad-spectrum semisynthetic aminoglycoside
38. Kabins SA, Nathan C. In vitro activity of 5-episisomicin in bacteria antibiotic.Antimicrob Agents Chemother 1976; 10: 827–836.
resistant to other aminoglycosdie antibiotics.Antimicrob Agents 57. McKay GA,Thompson PR,Wright GD. Broad spectrum
Chemother 1978; 14: 391–397. aminoglycoside phosphotransferase type III from Enterococcus:
39. Vastola AP,Altschaefl J, Harford S. 5-epi-sisomicin and 5-epi- overexpression, purification, and substrate specificity. Biochemistry
gentamicin B: substrates for aminoglycoside-modifying enzymes 1994; 33: 6936–6944.
that retain activity against aminoglycoside-resistant bacteria. 58. Kondo S,Yamamoto H, Naganawa H, Umezawa H. (title) J
Antimicrob Agents Chemother 1980; 17: 798–802. Antibiotics 1972; 25: 483–484.
40. Allen NE, Alborn WE Jr, Hobbs JN Jr, Kirst HA. 7- 59. Thompson PR, Hughes DW,Wright GD. Regiospecificity of
Hydroxytropolone: an inhibitor of aminoglycoside-2′′-O- aminoglycoside phosphotransferase from Enterococci and
adenyltransferase.Antimicrob Agents Chemother 1982; 22: Staphylococci (APH(3′)-IIIa). Biochemistry 1996; 35: 8686–8695.
824–831. 60. Cox JR, Mckay GA,Wright GD, Serpersu EH.Arrangement of
41. Kirst HA, Marconi GG, Counter FT, Ensminger PW, Jones ND, substrates at the active site of an aminoglycoside antibiotic 3′-
Chaney MO,Toth JE, and Allen NE. Synthesis and characterization phosphotransferase as determined by NMR. J Am Chem Soc 1996;
of a novel inhibitor of an aminoglycoside- inactivating enzyme. 118: 1295–1301.
J Antibiotics 1982; 12: 1651–1657. 61. Martin P, Jullien E, Courvallin P. Nucleotide sequence of
42. Saleh NA, Zwiefak A, Peczynska-Czoch W, Mordarski M, Pulverer Acinetobacter braumannii aphA-6 gene: evolutionary and functional
G. New inhibitors for aminoglycoside-adenyltransferase. Zbl Bakt implications of sequence homologies with nucleotide-binding
Hyg A 1988; 270: 66–75. proteins, kinases and other aminoglycoside-modifying enzymes.
43. Okamoto S, Suzuki Y. Chloramphenicol-, dihydrostreptomycin-, and Mol Microbiol 1988; 2: 615–625.
kanamycin-inactivating enzymes from multiple drug-resistant 62. Thompson PR, Hughes DW,Wright GD. Mechanism opf
Escherichia coli carrying episome ‘R’. Nature 1965; 208: 1301–1303. aminoglycoside 3′-phosphotransferase type IIIa: His 188 is not a
44. Radika K, Northrop DB.The kinetic mechanism of kanamycin phosphate-accepting residue. Chem Biol 1996; 3: 747–755.
acetyltransferase derived from the use of alternative antibiotics 63. Hon W-C, McKay GA,Thompson PR, Sweet RM,Yang DSC,Wright
and coenzymes. J Biol Chem 1984; 258: 12543–12546. GA, Berghuis AM. Structure of an enzyme required for
45. Radika K, Northrop DB. Substrate specificity and structure activity aminoglycoside antibiotic resistance reveals homology to
relationships for acetylation of antibiotics catalyzed by kanamycin eukaryotic protein kinase. Cell 1997; 89: 887–895.
acetyltransferase. Biochemistry 1984; 23: 5118–5122. 64. Cox JR, McKay GA,Wright GD, Serpersu EH. Arrangement of
46. Radika K, Northrop DB. A new kinetic diagnostic for enzymatic substrates at the active site of an aminoglycoside antibiotic 3′-
mechanisms using alternative substrates. Anal Biochem 1984; 141: phosphotransferase as determined by NMR. J Am Chem Soc 1996;
413–417. 118: 1295–1301.

116 Drug Resistance Updates (2001) 4, 106–117  2001 Harcourt Publishers Ltd
Aminoglycoside-modifying enzymes
65. Cox JR, Serpersu EH. Biologically important conformations of and resistance enzymes as an effective means to overcoming
aminoglycoside antibiotics bound to an aminoglycoside 3′- bacterial drug resistance. J Am Chem Soc 1995; 117:
phosphotransferase as determined by transferred nuclear 11060–11069.
Overhauser effect spectroscopy. Biochemistry 1997; 36: 71. McKay GA, Roestamadji J, Mobashery S,Wright GD. Recognition of
2353–2359. aminoglycoside antibiotics by enterococcal-staphylococcal
66. McKay GA,Wright GD. Kinetic mechanism of aminoglycoside aminoglycoside 3′- phosphotreansferase type IIa: role of substrate
phosphotransferase type IIIa: evidence for a Theorell-Chance amino groups. Antimicrob Agents Chemother 1996; 40:
mechanism. J Biol Chem 1995; 270: 24686–24692. 2648–2650.
67. McKay GA,Wright GD. Catalytic mechanism of enterococcal 72. Roestamadji J, Grapsas I, Mobashery S. Mechanism-based
kanamycin kinase (APH(3′)-IIIa): viscosity, thio, and solvent isotope inactivation of bacterial aminoglycoside 3′-phosphotransferases. J
effects support a Theorell-Chance mechanism. Biochemistry 1996; Am Chem Soc 1995; 117: 80–84.
35: 8680–8685. 73. Roestamadji J., Mobashery, S.The use of neamine as a molecular
68. Siregar JJ, Miroshnimov K, Mobashery S. Purification, template: inactivation of bacterial antibiotic resistance enzyme
characterization, and investigation of the mechanism of aminoglycoside 3′- phosphotransferase type IIa, Bioorg Med Chem
aminoglycoside 3′- phospotransferase type Ia. Biochemistry 1995; Lett 1998; 8: 3483–3486.
34: 12681–12688. 74. Daigle, DM, McKay, GA,Wright, GD. Inhibition of aminoglycoside
69. Siregar JJ, Lerner SA, Mobashery S. Purification and antibiotic resistance enzymes by protein kinase inhibitors. J Biol
characterization of aminoglycoside 3′-phosphotransferase type IIa Chem 1997; 272: 24755–24758.
and kinetic comparison with a new mutant enzyme. Antimicrob 75. Sucheck SJ,Wong AL, Koeller KM, Boehr DD, Draker K, Sears P,
Agens Chemother 1994; 38: 641–647. Wright GD,Wong C-H. Design of bifunctional antibiotics that
70. Roestamadji J, Grapsas I, Mobashery S. Loss of individual target bacterial rRNA and inhibit resistance-causing enzymes. J Am
interactions between aminoglycoside antibiotics Chem Soc 2000; 122: 5230–5231.

 2001 Harcourt Publishers Ltd Drug Resistance Updates (2001) 4, 106–117 117

You might also like