You are on page 1of 13

doi:10.1016/j.jmb.2004.10.030 J. Mol. Biol.

(2005) 345, 115–127

The Crystal Structure of E. coli 1-Deoxy-D-xylulose-5-


phosphate Reductoisomerase in a Ternary Complex
with the Antimalarial Compound Fosmidomycin and
NADPH Reveals a Tight-binding Closed Enzyme
Conformation
Aengus Mac Sweeney1†, Roland Lange1†, Roberta P. M. Fernandes2
Henk Schulz1, Glenn E. Dale1, Alice Douangamath1, Philip J. Proteau2
and Christian Oefner1*
1
Morphochem AG The key enzyme in the non-mevalonate pathway of isoprenoid biosyn-
WRO-1055/338 thesis, 1-deoxy-D-xylulose 5-phosphate reductoisomerase (DXR) has been
Schwarzwaldallee 215 shown to be the target enzyme of fosmidomycin, an antimalarial,
CH-4058, Basel, Switzerland antibacterial and herbicidal compound. Here we report the crystal
2 structure of selenomethionine-labelled Escherichia coli DXR in a ternary
College of Pharmacy, Oregon complex with NADPH and fosmidomycin at 2.2 Å resolution. The
State University, Corvallis
structure reveals a considerable conformational rearrangement upon
OR 97331-3507, USA
fosmidomycin binding and provides insights into the slow, tight binding
inhibition mode of the inhibitor. Although the inhibitor displays an
unusual non-metal mediated mode of inhibition, which is an artefact most
likely due to the low metal affinity of DXR at the pH used for
crystallization, the structural data add valuable information for the rational
design of novel DXR inhibitors. Using this structure together with the
published structural data and the 1.9 Å crystal structure of DXR in a
ternary complex with NADPH and the substrate 1-deoxy-D-xylulose 5-
phosphate, a model for the physiologically relevant tight-binding mode of
inhibition is proposed. The structure of the substrate complex must be
interpreted with caution due to the presence of a second diastereomer in
the active site.
q 2004 Elsevier Ltd. All rights reserved.
*Corresponding author Keywords: DXR; IspC; crystal structure; fosmidomycin; malaria

Introduction
Malaria remains a significant cause of mortality
† A.M.S. & R.L. contributed equally. in many areas of the world, with over 300 million
Present addresses: R. Lange, Actelion-Percurex AG, cases and one million deaths per year.1 The
Schwarzwaldallee 215, 4058 Basel, Switzerland; A.
escalating incidence of malaria and the develop-
Douangamath, Evotec OAI, 151 Milton Park, Abingdon,
Oxon OX14 4SD, UK. ment of drug resistance highlights the urgent need
Abbreviations used: DTT, dithiothreitol; DXP, 1-D- to identify novel antimalarial target enzymes and
deoxyxylulose-5-phosphate; DXR, DXP drugs. Recently two phosphonic acid antibiotics,
reductoisomerase; fosmidomycin/FR-31564, 3-(N- fosmidomycin (FR-31564) and its derivative
hydroxy)aminopropylphosphonic acid; NADPH, FR-900098 have been identified as promising agents
nicotinamide adenine dinucleotide phosphate; NCS, non- for the development of a new antimalarial chemo-
crystallographic symmetry; NTA, nitrilotriacetic acid therapy.2,3 Originally, these compounds were
agarose; SeMet, selenomethionine; TCEP, Tris(2-
carboxyethyl)-phosphine; TIM, triose phosphate identified as spheroplast-inducing antibacterials
isomerase. inhibiting most Gram-negative and some Gram-
E-mail address of the corresponding author: positive bacteria.4 Initial studies undertaken to
christian.oefner@morphochem.ch illuminate the mode of action indicated that

0022-2836/$ - see front matter q 2004 Elsevier Ltd. All rights reserved.
116 Crystal Structure of DXR

fosmidomycin blocks bacterial isoprenoid bio- inhibition is eliminated when the enzyme DXR is
synthesis. 5 Subsequently, it was shown that preincubated with fosmidomycin and NADPH but
fosmidomycin and FR-900098 (Figure 1) are specific not when NADPH is omitted from the preincu-
inhibitors of 1-deoxyxylulose-5-phosphate reducto- bation mix it has been concluded that the tight
isomerase (DXR; EC 1.1.1.267) also known as 2-C- binding of the inhibitor requires prior formation of
methyl-D-erythritol synthase (MEP synthase) and a DXR–NADPH complex. A slow onset of inhi-
IspC.2,6,7 DXR is the second enzyme of the bition, as displayed by fosmidomycin with DXR, is
mevalonate-independent pathway for the bio- typically associated with a conformational change
synthesis of isopentenyl diphosphate (IPP) and of the enzyme.
dimethylallyl diphosphate (DMAPP), the two Four crystal structures of DXR from E. coli have
basic precursors for isoprenoids. 6 Genomic been determined to high and medium resolution as
analyses of the phylogenetic distribution indicate the enzyme alone (1K5H),15 a binary complex with
that the genes coding for this mevalonate-indepen- NADPH (1JVS),16 a complex with fosmidomycin
dent pathway are present in most Gram-negative and manganese (1ONP)17 and a complex with
and some Gram-positive bacteria, in higher plants bisphosphonate inhibitors and manganese.18 The
and Plasmodium falciparum but not in mammals, crystal structure of DXR from Zymomonas mobilis, a
fungi and archaebacteria.8,9 Accordingly, the phylo- bacterium widely used for ethanol production, has
genetic distribution of dxr (yaeM) is consistent with recently been reported as an apoenzyme (1ROK)
the activity spectrum of fosmidomycin as an and a binary complex with NADPH (1ROL).19 The
antibacterial, antimalarial and herbicidal agent. V-shaped protein displays an intrinsic flexibility15,16
The enzymatic properties of DXR and its mode of and it has been proposed that binding of mag-
inhibition by fosmidomycin have been analyzed in nesium, substrate or inhibitor could induce a
detail. DXR catalyzes the conversion of 1-deoxy-D- conformational change. Conformational differences
xylulose-5-phosphate (DXP) to 2-C-methyl-D-ery- between the three molecules of the asymmetric unit
thritol phosphate (MEP), requiring NADPH and a of the apoenzyme, resulting from different crystal
divalent metal such as Mn2C, Mg2C or Co2C (Figure contacts, revealed that the connective domain
2).6,10,11 Based on the stereochemical features of the (residues 160–270) can function as a hinge. High
NADPH-dependent reduction, DXR is classified as temperature factors (approx. 100 Å2) in the pub-
a class B dehydrogenase.12,13 Recent mechanistic lished structures, with the exception of molecule C
investigations suggest a two-step reaction involving of the apoenzyme (PDB code 1K5H)15 which is
an isomerization of DXP to 2-C-methyl-D-erythrose- stabilized by crystal contacts, reveal disorder in
4-phosphate followed by NADPH-dependent loop 206–216. However, the structure of DXR
reduction to form 2-C-methyl-D-erythritol-4-phos- complexed with fosmidomycin in the presence of
phate. Two possible mechanisms have been pro- manganese through soaking experiments17 reveals
posed for isomerization by DXR, an alpha-ketol no structural rearrangement and little order in loop
rearrangement and a retroaldolization/aldolization 206–216, suggesting that the observed complex
(Figure 2).14 The divalent ion is required for the might represent the initial, lower affinity (210 nM)
reduction step but whether it is required for the binding state of fosmidomycin. Published data have
initial isomerization step has not been determined. shown that slow, tight binding inhibition requires a
The catalytic role of His209 has also not been conformational change, which occurs only upon
determined but it is likely that it plays a role in prior binding of NADPH.11
binding the substrate in the correct orientation and Here we report the structure of both wild-type
conformation. It has been shown that mutation and SeMet-labelled DXR determined from crystals
of His209 to Gln causes a 5200-fold decrease in cocrystallized in the presence of the inhibitor
kcat/Km.10 fosmidomycin, magnesium and the required cosub-
An analysis of the progress curves for MEP strate NADPH. The enzyme undergoes the
synthesis in the presence of inhibitor revealed a expected rearrangement upon complex formation
slow, tight-binding mode of inhibition by fosmido- and the structure reveals a binding mode of
mycin and two kinetically distinguishable Ki values fosmidomycin that is not mediated by a metal ion.
have been reported for the Escherichia coli enzyme The ternary complex of the DXR, NADPH and the
(Ki w215 nM, Ki w21 nM).11 As the slow onset of substrate DXP is also presented and is used to create
a model of metal-mediated fosmidomycin binding.

Results

Overall structure

The overall structure of the complexed enzyme is


similar to that described,15–19 with a symmetry-
related molecule forming the physiologically active
Figure 1. The DXR inhibitors fosmidomycin (A) and homodimer in the case of the fosmidomycin
FR-900098 (B). complexes. The structure is composed of three
Crystal Structure of DXR 117

Figure 2. The conversion of DXP


to MEP via the intermediate 2-C-
methyl- D -erythrose-4-phosphate.
The carbon numbering scheme for
DXP is shown.

domains: a larger N-terminal NADPH binding observed which revealed almost identical confor-
domain comprising 150 residues, a connective or mations despite differences in crystal packing.
linker domain (residues 160–270) and a smaller The most notable difference between the pub-
C-terminal alpha-helical domain comprising resi- lished structures of DXR and the fosmidomycin and
dues 312 to 398 (Figure 3). The N-terminal domain substrate ternary complexes observed in the pre-
exhibits an a/b topology with a seven-stranded sence of NADPH is an induced fit adaptation of the
parallel b-sheet and seven a-helices. The connective connective and C-terminal domains upon small
domain comprises five helices and a four-stranded molecule binding, resulting in a closed enzyme
b-sheet with one parallel and two antiparallel conformation. Superimposing the NADPH binding
alignments. The open side of the b-sheet, i.e. the domain of the fosmidomycin complex structure
side that is not covered by loops or helices, forms with that of the binary complex reveals a 12.58
part of the protein surface. The C-terminal domain domain rotation of the remaining domains (Figure
is a four-helix bundle and is joined to the connective 3). Residues 206–216 form a flexible “lid”, which
domain by an extended loop (residues 186–216), closes upon substrate or fosmidomycin binding to
which spans the entire central domain and contains shield the active site from the solvent. The ternary
eight conserved residues. As is the fold, the dimer complex structures reveal for the first time a fully
interface is identical to that observed in the non- ordered conformation of loop 206–216 that is not
crystallographic symmetry (NCS)-related dimers of dependent on crystal contacts. Gly185 appears to
the known structures. With the exception of the act as a hinge, with Ser186 forming a hydrogen
N-terminal six-histidine tag, all residues of the bond with His209 in the closed enzyme confor-
protein are well ordered. The structure of DXR in mation. The dihedral angle phi of G185 is altered by
complex with NADPH (PDB accession code 1JVS, approximately 358 between the open (binary
molecule A) was used for comparison. In the NADPH complex) and closed conformations of
structure of the substrate complex, two crystal- DXR. A comparison of the open and closed enzyme
lographically independent monomers were conformations using the conformational analysis
118 Crystal Structure of DXR

disordered. The entire NADPH molecule is clearly


visible in the substrate structure. In this NADPH
conformation, two additional hydrogen bonds are
formed with Gly215(N) compared to the NADPH
binary complex. The favorable hydrogen bonding
of NADPH in the closed conformation may account
for its mandatory binding prior to the formation of
the high affinity DXR–fosmidomycin complex.
Interestingly, the published DXR–NADPH complex
exhibits a partially closed conformation that is
between the most open structure of DXR alone
and the closed structure of the DXR-NADPH–
fosmidomycin complex described here (Figure 4).
In order to investigate the driving force for
“closure” of the enzyme, the solvent-accessible
surface areas of the ternary complexes and the
published NADPH binary complex16 were com-
pared. Upon adopting the closed conformation, the
hydrophobic surface area of DXR is decreased by
4.1% (710 Å2) while the polar surface area is
increased by 2.9% (400 Å2). Both molecules of the
dimer were used in all surface area calculations,
and similar values were obtained for the substrate
and fosmidomycin ternary complexes. The solvent-
exposed surfaces of the hydrophobic residues
Trp212, Ile218 and Met276 in particular are signifi-
cantly reduced upon inhibitor binding.

Active site

Figure 3. The physiological homodimer of DXR. The Within the active site region, the inhibitor and
lower monomer is colored yellow (N-terminal domain), substrate bind to both the connective and C-term-
green (connective domain), blue (C-terminal domain) and inal domains and appear to induce the observed
grey (flexible “lid” loop). The upper monomer is colored conformational change by tethering the domains in
dark blue with the “lid” loop shown in red. The upper a closed conformation. In addition, the flexible loop
monomer has been superimposed with the binary connecting both domains (residues 206–216) under-
complex of NADPH with DXR by fitting the N-terminal
domains (residues 1–153) and the regions of binary goes an induced fit adaptation and adopts a
complex showing the greatest structural difference conformation that allows it to function as a “lid”
(residues 154–230 and 298–398, average Ca deviation over the active site. This significant rearrangement
3.2 Å each domain) are shown in magenta. The average points to its important role in substrate binding. The
Ca deviation of the omitted regions is 0.8 Å. highly conserved residues Met276, Met214 and

software DynDom20 indicates a domain movement


upon inhibitor binding, involving the region of
Gly185 as a hinge for the structural alteration.
The binding of the adenine and pentose phos-
phate moieties of NADPH is identical to that
observed in the structure of the binary complex.16
The nicotinamide ring of NADPH is disordered in
the fosmidomycin complexes but not in the sub-
strate complex. The NADPH molecule in the SeMet-
labelled DXR–fosmidomycin complex structure
turns away from the active site and is disordered
from the nicotinamide ring to the nearest phosphate
link. This appears to be caused by the conformation
of SeMet214, which occludes the binding pocket of
the nicotinamide ring. In the wild-type DXR–
fosmidomycin and DXR–substrate complexes
NADPH can be clearly seen to extend to the active Figure 4. This orientation shows DXR with nothing
site, although the nicotinamide moiety of the bound (green), NADPH bound (yellow) and NADPH and
fosmidomycin complex remains partially fosmidomycin bound (blue).
Crystal Structure of DXR 119

Trp212 form a barrier between the active site and Fosmidomycin binding
the solvent and the b-indole of Trp212 provides the
key interaction with the substrate or fosmidomycin Within the solvent-shielded cavity that is formed
backbone. This arrangement results in the for- upon closure of the “lid” residues 206–216, the
mation of an intramolecular interface of 160 Å2, of inhibitor backbone lies parallel to the b-indole of
which about 80% is of hydrophobic character. The Trp212 at a distance of approximately 4 Å. The
most notable effect of the conformational change is hydroxamic acid moiety of the inhibitor binds to the
the shielding of the active site region from the side-chains of Glu152 and Glu231 and to the
solvent. The electron density for NADPH and loop backbone nitrogen of Ser151 (Figure 6). A change
206–216 of the substrate complex are shown in of 1208 in the chi-1 torsion angles of both Asp150
Figure 5. and Glu234 residues results in the exchange of the
The substrate-binding cavity can be considered to hydrogen bond between Asp150 and Glu152 (6.0 Å
consist of three regions: a positively charged pocket, versus 2.8 Å in the published binary complex with
which binds the phosphonate moiety of fosmido- NADPH) with a bond between Asp150 and Glu234
mycin, a hydrophobic region around the carbon (3.0 Å versus 5.0 Å in the binary complex). Remark-
backbone and an amphipathic region, which binds ably, no magnesium ion was observed in either this
the hydroxamic acid function. The active site structure or those of the apoenzyme and binary
contains the highly conserved residues Asp150, complex, despite the fact that it is essential for
Glu152, Glu231, Glu234, His209 and His257. The activity and was included in the crystallization
experiments. Furthermore, the clustered acidic
clustered acidic Asp and Glu residues are thought
residues of the active site do not give rise to
to bind the catalytically essential divalent cation, by
octahedral geometry, which is required for mag-
analogy with the structure of acetohydroxy acid
nesium binding. This is most likely due to the low
isomeroreductase.21 DXR and acetohydroxy acid
pH of the crystallization buffer (pH 5.0), which
isomeroreductase catalyze similar reactions reduces the metal-binding affinity of DXR. A
although the two enzymes are unrelated by schematic diagram of fosmidomycin binding to
sequence or structure. The acidic residues at the SeMet-labelled DXR is shown in Figure 6(C).
active site of the NADPH binary complex interact Upon superimposing the hydroxamate-binding
with the highly conserved basic residues Lys125, region of the fosmidomycin complex with the
His153 and Lys228 and the formation of a complex corresponding region of the NADPH binary com-
with the substrate or fosmidomycin causes sub- plex, the phosphonate moiety of the inhibitor is
stantial changes in the hydrogen bonding network observed to lie 1.9 Å from the position occupied by
of these residues. The proximity of the carboxylates the sulphate ion in the binary complex. The
of Asp150, Glu152 and Glu231 suggests that they phosphonate-binding region of DXR is also dis-
interact via hydrogen bonds and raises uncertainty placed by 1.9 Å toward the hydroxamate binding
concerning their protonation states.15 In the closed region, in a movement that matches that of the
conformation of the enzyme, the newly created overall domain. Fosmidomycin forms no hydrogen
hydrophobic nature of the active site may alter the bond with His209, which appears to plays an
pKa of these residues further, as is observed for the important role in binding the substrate in the
catalytic base Glu165 of triose phosphate isomerase correct orientation for catalysis (Figure 7).10 This
(TIM).22 may be due to the presence of a monoanionic

Figure 5. An FoKFc omit electron


density map of NADPH and loop
206–216 of the substrate–NADPH–
DXR ternary complex, contoured at
3s.
120 Crystal Structure of DXR

Figure 6. (A) An FoKFc omit electron density map of fosmidomycin in the active site of DXR, contoured at 3s. Met276
is shown in orange on the left and Met214 is shown in two conformations: that of SeMet214 in magenta (right) and that of
wild-type Met214 in orange (left). (B) Hydrogen bonding interactions of the inhibitor fosmidomycin in the active site of
DXR. (C) A schematic diagram of fosmidomycin binding to SeMet-labelled DXR.

phosphonate species of fosmidomycin resulting fosmidomycin. The more open inhibitor-binding


from crystallization at pH 5.0, instead of the pocket of the wild-type DXR–fosmidomycin com-
dianionic species that exists at a physiological pH. plex remains largely shielded from the solvent but
Interestingly, Met214 of the selenomethionine- merges with the binding pocket of the nicotinamide
labelled fosmidomycin complex occupies a differ- ring of NADPH. Overall, the two protein structures
ent position to that observed in the wild type are almost identical in all other respects.
complex structure, with atoms Se and CE lying
closer (3.0 and 6.1 Å) to the corresponding atoms of Substrate binding
Met276 (Figure 6(A)). In this conformation, Met214
forms a hydrophobic interaction with Met276 rather The phosphate moiety of the substrate forms
than the inhibitor backbone. This conformational hydrogen bonds with Ser186, Asn227, Lys228 and
change can be explained by the fact that seleno- with the catalytically essential His209 (Figure 7).
methionine is more hydrophobic than methionine, These residues are hydrogen bonded to a sulphate
with a difference of 0.17 kcal/mol in their free ion in the apoenzyme and NADPH binary complex
energies of transfer from octanol to water.23 As can structures. The substrate backbone interacts with
be seen from the omit map (Figure 6(A)), SeMet214 the b-indole of Trp212 and the carbonyl group of C2
partially occupies both possible conformations is hydrogen bonded to Glu152 and the backbone
although the conformation shown in Figure 6(B) is nitrogen of Ser151. The hydroxyl group of C3 is
favored. Whether the conformational difference involved in hydrogen bonds with Lys125 and
between Met214 and SeMet214 reflects a flexibility Glu231, while the C4 hydroxyl group is hydrogen
of Met214 that is required for substrate binding bonded to Glu152, Asn227 and Lys228. The C3
remains unclear. In the SeMet-labelled enzyme a hydroxyl group of the alternative diastereomer is
closed cavity of 713 Å3 is formed upon inhibitor not involved in any hydrogen bonding interactions.
binding, of which 464 Å3 is occupied by The nicotinamide ring of NADPH adopts an
Crystal Structure of DXR 121

Figure 7. (A) An FoKFc omit electron density map of the substrate DXP in the active site of DXR, contoured at 3s. The
position of the hydroxyl group in the L-configuration at C4 is depicted in green. (B) Hydrogen bonding interactions of
the substrate DXP in the active site of DXR. The position of the hydroxyl group in the L-configuration at C4 is depicted in
green. (C) A schematic diagram of the substrate DXP binding to DXR. For clarity, the bond between the carboxylate of
E231 and the hydroxyl group of C4 of DXP has been omitted.

orientation that would allow the transfer of a Model of tight-binding mode of fosmidomycin
hydride from the pro-S hydrogen of C4 of the
nicotinamide ring to C2 of the proposed intermedi- A model of metal-mediated fosmidomycin inhi-
ate 2-C-methylerythrose 4-phosphate, which is in bition of the closed form of DXR (shown in Figure 8)
keeping with the classification of DXR as a class B
dehydrogenase.12 A schematic diagram of DXP
binding to DXR is shown in Figure 7(C).
The electron density indicates that two diaster-
eomers are bound to the enzyme, although NMR
analysis of the substrate indicated the presence of a
single diastereomer. For this reason, it was not
possible to unambiguously interpret this crystal
structure with regard to the enzyme mechanism.
However, because the structure was determined at
a physiologically relevant pH, it has been possible
to use both available structures to create a model
for the tight-binding inhibitor of fosmidomycin.
The reason for the presence of a second dia-
stereomer is unclear and may be due to epimeriza-
tion under the crystallization conditions or the
selective binding of a small amount of the non- Figure 8. A model of metal-mediated fosmidomycin
physiological diastereomer by metal-free DXR. binding to DXR in its tight-binding conformation.
122 Crystal Structure of DXR

was prepared based on the structure on the ternary attempt was made to determine the Kms at pH 5.0,
complexes and the published structure of fosmido- but the enzyme activity was negligible in either
mycin bound to DXR in its open conformation.17 citrate or acetate buffer at this pH. The Kms
The phosphonate moiety of fosmidomycin is bound determined at pH 7.8 for the ions Co2C and Mn2C
in a similar fashion to the phosphate group of the are 2 mM and 17 mM, respectively, similar to the Kms
substrate. The hydroxamic acid moiety coordinates found for these ions with the DXR from Synecho-
a divalent cation that is bound by residues D150, cystis (Co2C 10 mM; Mn2C 15 mM)23 and Mycobacter-
E152 and E231, the same residues that coordinate ium tuberculosis (Co2C 1.2 mM; Mn2C 21 mM).24 The
the metal in the published structure of the binary Km value observed for Mg2C was 210 mM. Although
complex of DXR and fosmidomycin.17 The metal this value is higher than those determined for Mn2C
binding site was modelled using the octahedral and Co2C, it is lower than the values found with the
binding site that was observed in the metal-free Synechocystis (2400 mM; 11-fold) and Mycobacterium
substrate complex as well as the observed matching tuberculosis (1200 mM; sixfold) DXRs.24,25 Mg2C has
position of the metal in the published fosmidomy- been proposed to be the physiologically relevant
cin binary complex. The inhibitor was adjusted metal ion activator for DXR.11
slightly to accommodate the metal ion. The inter- An increase in the Km values for Mn2C and Mg2C
actions of the metal with the inhibitor and the was observed when the assays were carried out at
enzyme are similar to those observed in the pH 6.0 (Table 1). The Km for Mn2C increased 31-fold
published structure of fosmidomycin bound to and for Mg2C 81-fold. A similar pH effect was
DXR. As in the published structure a single water observed using the M. tuberculosis DXR, where the
molecule completes the octahedral coordination of Km for Mn2C increased 243-fold from 7 mM at pH
the metal, however in the model this water 8.75 to 1.7 mM at pH 6.0.24 The increase in Km is
molecule is also hydrogen bonded to K125. The most likely due to a change in protonation state of
favorable hydrophobic interactions of the inhibitor the carboxylate ligand(s) for the divalent metal
with Trp212, Met214 and Met276 that were ion.24 The carboxylic acid functional group is a
observed in the structure of the ternary complex poorer ligand than a carboxylate. Due to the low pH
of fosmidomycin, NADPH and DXR are preserved of the crystallization buffer and a concentration of
in this model. By contrast, these interactions are not 10 mM Mg2C, it appears as though the absence of a
observed in the published structure of the binary Mg2C in the crystal structure results from a
complex of fosmidomycin and DXR. Met276 and decreased affinity for the divalent ion at low pH.
the nicotinamide ring of NADPH also contribute to
the formation of a hydrophobic binding pocket. The
tight-binding conformation of the enzyme results in
a small and totally enclosed inhibitor-binding Discussion
pocket. This model is consistent with the fact that The structures of both ternary complexes reveal a
no inhibitors of DXR that are significantly larger dramatic conformational change of DXR that is
than fosmidomycin have been reported to date. It induced by substrate or inhibitor binding in the
also reveals that the only possibility to extend the presence of NADPH. This remarkable flexibility of
molecule will involve either displacement of the DXR plays an important role in substrate binding
nicotinamide ring of NADPH or targeting the open and catalysis and is similar to that of other
conformation of DXR. We propose that this model isomerases in which a domain or loop movement
represents the physiologically relevant tight-bind- is required to shield a reactive intermediate from
ing conformation of DXR in complex with the surrounding solvent.22,26 Based on the slow
fosmidomycin. timescale of DXR domain movement compared to
the rate of inhibitor diffusion, it is possible to
Kinetics explain the slow onset of inhibition that has been
observed for fosmidomycin. The formation of
The Michaelis constants (Km) for the divalent additional hydrogen bonds between the enzyme
cations Mg2C, Mn2C and Co2C with E. coli DXR and NADPH in the closed conformation correlates
were determined at two different pHs, pH 7.8 and with the observation that NADPH is essential for
pH 6.0. The results are summarized in Table 1. An tight binding of the inhibitor.11 In addition, the
favorable enthalpic interactions due to the
increased buried hydrophobic surface in the closed
Table 1. Michaelis constants for divalent cations using conformation may compensate for the entropic cost
E. coli DXR of a more rigid closed structure. The increase in
buried hydrophobic surface area in the closed
Km (mM)
conformation, together with its occurrence under
Cations pH 7.8 pH 6.0 different crystal packing conditions obtained by
Mn 2C
17G2 520G60 cocrystallization experiments indicates that the
Mg2C 210G20 17,000G2000 observed conformation most likely represents that
Co2C 2.0G0.7 nd of the complex in solution.
nd, not determined.
The structure of the ternary complex of DXR with
NADPH and fosmidomycin differs markedly from
Crystal Structure of DXR 123

that published by Steinbacher et al.,17 in which the of a second diastereomer (at C-3), it is impossible to
hydroxamic acid of fosmidomycin co-ordinates a draw any firm conclusion about the enzyme
manganese ion which is bound to Asp150, Glu152 mechanism based on this structure. Two features
and Glu234. In the previously published structure, of the substrate-binding mode are noteworthy.
the octahedral geometry of the manganese-binding Firstly, the catalytic residue His209 is hydrogen
site is completed by three water molecules and the bonded to the phosphate moiety of the substrate,
active site lies on the enzyme surface due to the which is separated by four atoms (a distance of
open conformation of the enzyme. No hydrophobic 5.1 Å) from the scissile carbon-carbon bond. Thus, it
interactions were observed between the methylene is tempting to speculate that the role of the histidine
linker of fosmidomycin and the flexible “lid” loop of residue might involve “pre-orientation” of the
DXR (residues 206–216) is disordered. By contrast, substrate in an orientation and conformation that
in the tight-binding conformation fosmidomycin is favors retroaldolization or a-ketol rearrangement.
buried within a solvent-shielded binding site. The The role of phosphate binding in stabilizing
active sites of both structures are shown in Figure 9, conformations that favor productive turnover has
in which both fosmidomycin molecules have been been reported for the enzyme ribulose-bisphos-
superimposed. The greatest difference between the phate carboxylase (RuBisCo).27 However, further
two active sites is observed in loop 206–216, which biochemical and structural studies will be required
is largely disordered in the previously published to determine the exact role of the histidine residue.
structure of fosmidomycin bound to DXR. In Secondly, a distorted octahedral coordination site
addition, the region containing Met276 is closer to involving Glu152, Glu231, Asp150, Lys125 and the
fosmidomycin in the ternary complex than in the carbonyl group of C2 and the hydroxyl group of C3
previously published structure. The absence of the of the substrate appears to be the most likely
expected metal ion from the structure can be magnesium-binding site. This contrasts with the
explained by the low metal affinity of DXR in the fosmidomycin complex in which no metal-binding
crystallization buffer. The fact that the same enzyme site was observed.
conformation was observed in both ternary com- In summary, our findings reveal the closed
plexes, despite different crystallization conditions conformation of DXR and allow us to propose a
and crystal packing, allows us to conclude that the model for the tight-binding mode of fosmidomycin,
structures reveal the physiologically relevant sub- which will be of use for the development of novel
strate/inhibitor binding conformation of DXR. The DXR inhibitors.
previously solved structure of DXR in an open
conformation in complex with fosmidomycin17 may
represent the initial, lower affinity enzyme–inhibitor Materials and Methods
complex.
The ternary complex of DXR, NADPH and Cloning, overexpression and purification
substrate reveals significant differences between
the binding modes of the inhibitor and substrate, The E. coli dxr (ispC) gene (yaeM, accession number
even in their phosph(on)ate moieties. However, AE000126) encoding 1-deoxy-D-xylulose-5-phosphate
because the electron density indicates the presence reductoisomerase (EC 1.1.1.267) was amplified by

Figure 9. The superimposed


active sites of the published DXR-
fosmidomycin complex and the
ternary complex of fosmidomycin,
NADPH and DXR. The previously
published complex is shown in
yellow and the ternary complex is
shown in grey.
124 Crystal Structure of DXR

polymerase chain reaction (PCR) from genomic E. coli MnCl2, 2 mM DTT, 1 mg/ml BSA buffer at pH 8 contain-
K12 (W3110) DNA using the oligonucleotide primers ing varying concentrations of NADPH and DXP. The
Ecdxrfor (5 0 -GGATGTCATATGAAGCAACTCACCATTC) enzymatic reaction rate was measured by monitoring the
and Ecdxrrev (5 0 -GACTATATCACTGGATCCCTACGC changes at A340 nm corresponding to the oxidation of
AAC). The respective PCR product was purified, digested NADPH (30340 Z 6:22 cmK1 mMK1). The substrate (DXP)
with NdeI and BamHI and inserted into the NdeI-BamHI was synthesized by the method described by Blagg &
digested vector pDS-6hisNdeI (27). Insertion into pDS- Poulter.30
NdeI6his resulted in a recombinant dxr gene with a 5 0 -end
coding for an N-terminal tag of additional 14 residues Kinetic studies
including six consecutive histidine residues (H6-DXR).
The dxr nucleotide sequence in pDS-NdeI-6hisEcdxr was The typical assay system for kinetic studies contained
verified by applying the dye terminator cycle sequencing 100 mM Tris–HCl (pH 7.8), 1 mM MnCl2, 2 mM DXP,
technology on an ABI PRISM 310 Genetic Analyzer. For 0.2 mM NADPH, and 1 mg/ml of BSA in a final volume
routine expression of H6-DXR the E. coli host strain BL21 of 1 ml. Assays were conducted at pH 6.0 used 100 mM
was first transformed with pREP4 (lacIq, Qiagen) and Mes buffer. The reaction was initiated by adding the
then with pDS-NdeI-6hisEcdxr. Expression of H6-DXR enzyme (5 nM at pH 7.8; 50 nM at pH 6.0) to the total
was induced by the addition of 0.25 mM isopropyl-b-D- assay mixture. The oxidation of NADPH was monitored
thiogalactopyranoside to mid-log cells growing in nutri- at 37 8C in a Hitachi U-2000 Spectrophotometer. The
ent rich medium (NZ-amine 1%, Yeast extract, NaCl) kinetic parameters for three metal ions, Mn2C, Mg2C and
supplemented with 0.2% (w/v) fructose, 100 mg/ml Co2C, were determined at pH 7.8 and 6.0. Kinetic
ampicillin and 25 mg/ml kanamycin at 30 8C. Cells were parameters were determined in triplicate using at least
grown for an additional 2.5 hours and then harvested by seven varying concentrations of these ions. The concen-
centrifugation. tration ranges for Mn2C, Mg2C and Co2C were 2–
In vivo incorporation of SeMet into H6-DXR was 2,000 mM, 50–60,000 mM and 0.4–500 mM, respectively.
achieved by applying the methionine pathway inhibition Kinetic parameters were calculated by fitting the initial
method.28,29 BL21 pREP4 pDS-NdeI-his6-Ecdxr was velocity versus substrate concentration to the Michaelis
grown overnight in minimal medium M9 (30) sup- equation, vZ ðVmax ½SÞ=ðKm C ½SÞ, using the non-linear
plemented with 0.2% fructose, 1 mg/ml thiamin, 25 mg/ml regression analysis option of GraphPad Prism software.
kanamycin and 100 mg/ml ampicillin at 30 8C. Two liters
of the same medium were inoculated with cells from the Crystallization
overnight culture to reach an absorbance of A600 nmZ
0.12). Cells were harvested after 4.5 hours growth, Crystallization screening was carried out by the
centrifuged and resuspended in two liters of fresh, modified microbatch method,31 adding 1ul of protein to
prewarmed (30 8C) minimal medium containing 1ul of crystallization solution from the INDEX screen
additional amino acids (L-lysine, L-phenylalanine, L- (Hampton Research catalogue No. HR2-134). A Douglas
threonine at 100 mg/l; L-isoleucine, L-leucine, L-valine at Instruments Impax robot was used for pipetting. Crystals
50 mg/l). After 15 minutes growth in the presence of of the DXR-substrate complex were grown using DXR at a
these amino acids expression of H6-DXR was induced by concentration of 10 mg/ml containing 3 mM NADPH
adding 0.5 mM IPTG. After two hours cells were and 10 mM substrate. The reservoir buffer contained 25%
harvested (6000 g, 15 minutes) and stored at K80 8C. (w/v) PEG 3350, 200 mM Li2SO4, 100 mM Bis-Tris (pH
H6-DXR protein has been purified by a combination of 6.5). Crystals appeared overnight as thin plates of
Ni-NTA affinity chromatography and size exclusion maximum dimensions 0.1 mm!0.1 mm!0.03 mm and
chromatography. Cells were resuspended in ice-cold were mounted directly from the microbatch plate.
lysis buffer (50 mM NaH2PO4, 300 mM NaCl, 10 mM For DXR-fosmidomycin complex crystals, wild-type
imidazole 5 mM TCEP pH 8) containing Protease Inhibi- (non-SeMet-labelled) and SeMet-labelled E. coli DXR,
tor Cocktail (Roche). After incubation in the presence of crystals were optimized using the hanging-drop vapor-
1 mg/ml lysozyme cells were broken by ultrasonic diffusion method at a protein concentration of 10 mg/ml,
treatment. Cell debris were removed by centrifugation containing 3 mM NADPH and 10 mM fosmidomycin.
(20,000 g, 30 minutes, 4 8C). The bacterial lysate was The protein solution contained 100 mM NaCl, 50 mM
mixed with 50% Ni2C-NTA (Qiagen) and incubated for Tris–HCl (pH 8.0), 5% glycerol, 5 mM TCEP and 10 mM
one hour at 4 8C. The lysate-Ni2C-NTA mixture was MgCl2. The reservoir buffer contained 2.6 M NaCl,
loaded onto an Econo Pac column (Biorad) and washed 100 mM acetate (pH 5.0). Crystals grew to a maximum
with 12 volumes of wash buffer (NaH2PO4, 300 mM NaCl, size of 0.1 mm!0.1 mm!0.1 mm over three days. 1.6 M
20 mM imidazole, pH 8). H6-DXR was eluted with buffer sodium citrate was used as a cryoprotectant for low
(NaH2PO4, 300 mM NaCl, 250 mM imidazole). H6-DXR temperature measurements (with a soak time of 1–2
containing fractions were identified by SDS-PAGE, seconds).
pooled and concentrated by ultrafiltration using ULTRA-
FREE-15 filter devices (Millipore, MWCO 30,000). An Data collection and processing
aliquot of 10 ml of the H6-DXR containing solution was
purified on a Superdex 75 gel filtration column equili- The substrate complex crystals belong to the orthor-
brated with 50 mM Tris, 100 mM NaCl, 2 mM MgCl2, hombic space group C2221 with cell dimensions aZ
5 mM TCEP, 5% (v/v) glycerol. H6-DXR eluted as a single 107.57, bZ122.85, cZ128.55 Å. They contain two mol-
peak. For the preparation of metal-free H6-DXR, the metal ecules in the asymmetric unit and diffract to a resolution
(MgCl2) was omitted from all buffers and 20 mM EDTA of 1.9 Å. The data set was collected at the SLS beamline
was added to the buffer used for size exclusion X06SA at PSI, Villigen, Switzerland and diffraction
chromatography. Protein concentrations of pure fractions intensities were recorded on a MAR CCD detector at a
were determined by absorbance at 280 nm. Enzymatic wavelength of 0.97818 Å.
activity of H6-DXR was routinely determined at 30 8C in The fosmidomycin complex crystals belong to the
40 mM Tris–HCl buffer containing 2 mM MgCl2 or 2 mM trigonal space group P3121 with cell dimensions aZbZ
Crystal Structure of DXR 125

Table 2. Data collection and refinement statistics

SeMet, NADPH, Wild-type, NADPH, Wild-type, NADPH,


fosmidomycin fosmidomycin substrate

Data collection
Space group P3(1)21 P3(1)21 C222(1)
Cell dimensions aZbZ109.17, cZ90.63 aZbZ109.14, cZ91.29 aZ107.06, bZcZ128.55
Limiting resolution (Å) (last shell) 2.20 (2.34–2.20) 2.65 (2.82–2.65) 1.90 (2.02–1.90)
Reflections 31,221 18,451 62,616
I/sI (overall/last shell) 7.64 (1.97) 12.1 (2.98) 12.6 (2.73)
Rmerge (%) (overall/last shell) 12.9 (46.5) 14.3 (44.4) 9.6 (35.9)
Redundancy 3.8 6.8 4.8
Completeness (%) (overall/last shell) 97.5 (94.5) 99.2 (95.5) 93 (95.3)
Refinement
Resolution 20–2.2 20–2.65 20–1.90
Rcryst (%) 18.7 21.1 17.4
Rfree (%) 21.2 25.8 21.2
Non-hydrogen protein atoms 3030 3030 6060
Solvent molecules 265 111 639
B-factor (Å2)
Protein 23.8 25.5 19.8
Solvent 32.0 25.4 32.0
Fosmidomycin 24.9 25.4 18.5
NADPH 49.5 33.5 14.6
Rmsd
Bond length (Å) 0.010 0.007 0.007
Bond angle (8) 1.175 1.063 1.136

109.17, cZ90.63 Å. They contain one molecule in the structures with excellent stereochemistry. All residues
asymmetric unit and diffract to a resolution of 2.65 Å for fall within the allowed regions of the Ramachandran plot
the wild-type enzyme and 2.2 Å for the SeMet-labelled and meet all stereochemical requirements of the program
protein. Data collection and refinement statistics are PROCHECK.38 The refined structure of the fosmido-
summarized in Table 2. Both data sets were measured mycin-complexed E. coli enzyme has an R-factor of 18.7%
with Cu Ka radiation provided from a NONIUS FR591 (RfreeZ20.2%) for the SeMet-labelled protein and 21.1%
rotating anode generator equipped with an OSMIC (RfreeZ25.8%) for wild-type DXR (Table 2). The refined
mirror system. Diffraction intensities were recorded on structure of the substrate-complexed enzyme has an R-
a MAR-Research image plate area detector. All diffraction factor of 17.4% (RfreeZ21.2%).
data were further processed and scaled with DENZO and
SCALEPACK.32 Protein Data Bank accession numbers

Molecular replacement and refinement The structures have been deposited with the RCSB
Protein Data Bank under the accession codes 1Q0H, 1Q0L
The ternary complex of E. coli DXR with NADPH and and 1Q0Q.
fosmidomycin was solved by molecular replacement,
where phases were obtained with the AMoRe software33,34,
using the refined coordinates of the apoenzyme (PDB
accession code 1K5H, molecule A).15 The Eulerian angles
and translation parameters are aZ22.538, bZ38.298, gZ Acknowledgements
128.618, xZK31.12 Å, yZ73.09 Å, zZK46.83 Å for the
single molecule in the asymmetric unit. The ternary We thank Allan D’Arcy for excellent assistance in
complex of DXR with NADPH and DXP was solved using protein crystallization. We also thank the staff of the
the coordinates of the fosmidomycin complex described Swiss Light Source, Paul Scherrer Institute, Villigen,
here. The Eulerian angles and translation parameters are Switzerland for assistance with data collection.
aZ88.28, bZ0.08, gZ0.08, xZ52.91 Å, yZK32.86 Å, zZ
R.P.M.F. has a PhD scholarship from CNPq (Brazil).
2.80 Å for molecule A and aZ151.198, bZ0.008, gZ0.008,
xZ52.82 Å, yZ32.34 Å, zZ34.95 Å for molecule B of the
asymmetric unit.
Maximum likelihood refinement was performed with References
REFMAC5,35 using parameters for ideal stereochemistry
as described.36 Manual rebuilding was accomplished 1. Frankish, H. (2002). Scientists reach milestone in
using the graphics program MOLOC.37 The initial malaria research. Lancet, 360, 1075.
difference Fourier map of the fosmidomycin complex 2. Jomaa, H., Wiesner, J., Sanderbrand, S., Altincicek, B.,
clearly revealed a residual electron density corresponding Weidemeyer, C., Hintz, M. et al. (1999). Inhibitors of
to fosmidomycin and a partially bound molecule of the nonmevalonate pathway of isoprenoid biosyn-
NADPH, while that of the substrate-complexed enzyme thesis as antimalarial drugs. Science, 285, 1573–1576.
revealed a residual electron density corresponding to the 3. Missinou, M. A., Borrmann, S., Schindler, A., Issifou,
partially racemized substrate and NADPH. S., Adegnika, A. A., Matsiegui, P. B. et al. (2002).
Progressive introduction of ligand and solvent mol- Fosmidomycin for malaria. Lancet, 360, 1941–1942.
ecules with good geometry led to the complexed 4. Mine, Y., Kamimura, T., Nonoyama, S., Nishida, M.,
126 Crystal Structure of DXR

Goto, S. & Kuwahara, S. (1980). In vitro and in vivo two bisphosphonate:1-deoxyxylulose-5-phosphate


antibacterial activities of FR-31564, a new phosphonic reductoisomerase complexes. J. Am. Chem. Soc. 126,
acid antibiotic. J. Antibiot. (Tokyo), 33, 36–43. 10824–10825.
5. Shigi, Y. (1989). Inhibition of bacterial isoprenoid 19. Ricagno, S., Grolle, S., Bringer-Meyer, S., Sahm, H.,
synthesis by fosmidomycin, a phosphonic acid-con- Lindqvist, Y. & Schneider, G. (2004). Crystal structure
taining antibiotic. J. Antimicrob. Chemother. 24, 131– of 1-deoxy-D-xylulose-5-phosphate reductoisomerase
145. from Zymomonas mobilis at 1.9-A resolution. Biochim.
6. Takahashi, S., Kuzuyama, T., Watanabe, H. & Seto, H. Biophys. Acta, 1698, 37–44.
(1998). A 1-deoxy-D-xylulose 5-phosphate reductoi- 20. Hayward, S., Kitao, A. & Berendsen, H. J. (1997).
somerase catalyzing the formation of 2-C-methyl-D- Model-free methods of analyzing domain motions in
erythritol 4-phosphate in an alternative nonmevalo- proteins from simulation: a comparison of normal
nate pathway for terpenoid biosynthesis. Proc. Natl mode analysis and molecular dynamics simulation of
Acad. Sci. USA, 95, 9879–9884. lysozyme. Proteins: Struct. Funct. Genet. 27, 425–437.
7. Lichtenthaler, H. K. (2000). Non-mevalonate isopre- 21. Biou, V., Dumas, R., Cohen-Addad, C., Douce, R., Job,
noid biosynthesis: enzymes, genes and inhibitors. D. & Pebay-Peyroula, E. (1997). The crystal structure
Biochem. Soc. Trans. 28, 785–789.
of plant acetohydroxy acid isomeroreductase com-
8. Boucher, Y. & Doolittle, W. F. (2000). The role of lateral
plexed with NADPH, two magnesium ions and a
gene transfer in the evolution of isoprenoid biosyn-
herbicidal transition state analog determined at 1.65 A
thesis pathways. Mol. Microbiol. 37, 703–716.
9. Lange, B. M. & Croteau, R. (1999). Isoprenoid resolution. EMBO J. 16, 3405–3415.
biosynthesis via a mevalonate-independent pathway 22. Harris, T. K., Abeygunawardana, C. & Mildvan, A. S.
in plants: cloning and heterologous expression of 1- (1997). NMR studies of the role of hydrogen bonding
deoxy-D-xylulose-5-phosphate reductoisomerase in the mechanism of triosephosphate isomerase.
from peppermint. Arch. Biochem. Biophys. 365, 170– Biochemistry, 36, 14661–14675.
174. 23. Gassner, N. C., Baase, W. A., Hausrath, A. C. &
10. Kuzuyama, T., Takahashi, S., Takagi, M. & Seto, H. Matthews, B. W. (1999). Substitution with seleno-
(2000). Characterization of 1-deoxy-D-xylulose 5- methionine can enhance the stability of methionine-
phosphate reductoisomerase, an enzyme involved in rich proteins. J. Mol. Biol. 294, 17–20.
isopentenyl diphosphate biosynthesis, and identifi- 24. Argyrou, A. & Blanchard, J. S. (2004). Kinetic and
cation of its catalytic amino acid residues. J. Biol. chemical mechanism of Mycobacterium tuberculosis 1-
Chem. 275, 19928–19932. deoxy-D-xylulose-5-phosphate isomeroreductase. Bio-
11. Koppisch, A. T., Fox, D. T., Blagg, B. S. & Poulter, C. D. chemistry, 43, 4375–4384.
(2002). E. coli MEP synthase: steady-state kinetic 25. Yin, X. & Proteau, P. J. (2003). Characterization of
analysis and substrate binding. Biochemistry, 41, 236– native and histidine-tagged deoxyxylulose 5-phos-
243. phate reductoisomerase from the cyanobacterium
12. Proteau, P. J., Woo, Y. H., Williamson, R. T. & Phaosiri, Synechocystis sp. PCC6803. Biochim. Biophys. Acta,
C. (1999). Stereochemistry of the reduction step 1652, 75–81.
mediated by recombinant 1-deoxy-D-xylulose 5-phos- 26. Marks, G. T., Harris, T. K., Massiah, M. A., Mildvan,
phate isomeroreductase. Org. Letters, 1, 921–923. A. S. & Harrison, D. H. (2001). Mechanistic impli-
13. Radykewicz, T., Rohdich, F., Wungsintaweekul, J., cations of methylglyoxal synthase complexed with
Herz, S., Kis, K., Eisenreich, W. et al. (2000). phosphoglycolohydroxamic acid as observed by X-
Biosynthesis of terpenoids: 1-deoxy-D-xylulose-5- ray crystallography and NMR spectroscopy. Biochem-
phosphate reductoisomerase from Escherichia coli is istry, 40, 6805–6818.
a class B dehydrogenase. FEBS Letters, 465, 157–160. 27. Larimer, F. W., Harpel, M. R. & Hartman, F. C. (1994).
14. Hoeffler, J. F., Tritsch, D., Grosdemange-Billiard, C. & Beta-elimination of phosphate from reaction inter-
Rohmer, M. (2002). Isoprenoid biosynthesis via the mediates by site-directed mutants of ribulose-bispho-
methylerythritol phosphate pathway. Mechanistic sphate carboxylase/oxygenase. J. Biol. Chem. 269,
investigations of the 1-deoxy-D-xylulose 5-phosphate 11114–11120.
reductoisomerase. Eur. J. Biochem. 269, 4446–4457. 28. Guerrero, S. A., Hecht, H. J., Hofmann, B., Biebl, H. &
15. Reuter, K., Sanderbrand, S., Jomaa, H., Wiesner, J.,
Singh, M. (2001). Production of selenomethionine-
Steinbrecher, I., Beck, E. et al. (2002). Crystal structure
labelled proteins using simplified culture conditions
of 1-deoxy-D-xylulose-5-phosphate reductoisomerase,
and generally applicable host/vector systems. Appl.
a crucial enzyme in the non-mevalonate pathway of
isoprenoid biosynthesis. J. Biol. Chem. 277, 5378–5384. Microbiol. Biotechnol. 56, 718–723.
16. Yajima, S., Nonaka, T., Kuzuyama, T., Seto, H. & 29. Doublie, S. & Carter, C. W. J. (1992). Preparation of
Ohsawa, K. (2002). Crystal structure of 1-deoxy-D- selenomethionyl protein crystals. In Crystallization of
xylulose 5-phosphate reductoisomerase complexed Nucleic Acids and Proteins. A Practical Approach
with cofactors: implications of a flexible loop move- (Ducruix, A. & Giege, R., eds), Oxford University
ment upon substrate binding. J. Biochem. (Tokyo), 131, Press.
313–317. 30. Blagg, B. S. & Poulter, C. D. (1999). Synthesis of 1-
17. Steinbacher, S., Kaiser, J., Eisenreich, W., Huber, R., Deoxy-D-xylulose and 1-Deoxy-D-xylulose-5-phos-
Bacher, A. & Rohdich, F. (2003). Structural basis of phate. J. Org. Chem. 64, 1508–1511.
fosmidomycin action revealed by the complex with 31. D’Arcy, A., Mac Sweeney, A., Stihle, M. & Haber, A.
2-C-methyl-D-erythritol 4-phosphate synthase (IspC). (2003). The advantages of using a modified micro-
Implications for the catalytic mechanism and anti- batch method for rapid screening of protein crystal-
malaria drug development. J. Biol. Chem. 278, 18401– lization conditions. Acta Crystallog. sect. D, Biol.
18407. Crystallog. 59, 396–399.
18. Yajima, S., Hara, K., Sanders, J. M., Yin, F., Ohsawa, K., 32. Otwinowski, Z. (1993). Oscillation data reduction
Wiesner, J. et al. (2004). Crystallographic structures of program. In Proceedings of the CCP4 Study Weekend:
Crystal Structure of DXR 127

Data Collection and Processing (Waywey, L., Isaacs, N. 36. Engh, R. & Huber, R. (1991). Accurate bond and angle
& Bailey, S., eds), pp. 55–62, SERC Daresbury parameters for X-ray protein refinement. Acta Crystal-
Laboratory, Daresbury, UK. log. sect. A, 47, 961–963.
33. Collaborative Computational Project, N4. (1994). The 37. Gerber, P. (1992). Peptide mechanics: a force field for
CCP4 Suite: programs for protein crystallography. peptides and protens working with entire residues as
Acta Crystallog. sect. D, 50, 760–763. small units. Biopolymers, 32, 1003–1017.
34. Navaza, J. (2001). Implementation of molecular 38. Laskowski, R. A., MacArthur, M. W., Moss, D. S. &
replacement in AMoRe. Acta Crystallog. sect. D, 57, Thornton, J. M. (1993). PROCHECK: a program to
1367–1372. check the stereochemical quality of protein structures.
35. Murshudov, G. N., Vagin, A. A. & Dodson, E. J. (1997). J. Appl. Crystallog. 26, 283–291.
Refinement of macromolecular structures by the
maximum-likelihood method. Acta Crystallog. sect.
D, 53, 240–255.

Edited by R. Huber

(Received 28 July 2004; received in revised form 1 October 2004; accepted 11 October 2004)

You might also like