You are on page 1of 21

Biochem. J. (2014) 461, 87–98 (Printed in Great Britain) doi:10.

1042/BJ20140209 87

Alternative substrates reveal catalytic cycle and key binding events in the
reaction catalysed by anthranilate phosphoribosyltransferase from
Mycobacterium tuberculosis

www.biochemj.org
Tammie V. M. COOKSON*1 , Alina CASTELL†1,2 , Esther M. M. BULLOCH†, Genevieve L. EVANS†, Francesca L. SHORT†3 ,
Edward N. BAKER†, J. Shaun LOTT†4 and Emily J. PARKER*4
*Maurice Wilkins Centre for Molecular Biodiscovery, Biomolecular Interaction Centre and Department of Chemistry, University of Canterbury, 20 Kirkwood Avenue, Christchurch 8140,
New Zealand
†Maurice Wilkins Centre for Molecular Biodiscovery and School of Biological Sciences, University of Auckland, 3 Symonds Street, Auckland 1142, New Zealand

AnPRT (anthranilate phosphoribosyltransferase), required for PRPP, in a catalytically relevant position. Soaking the analogues
the biosynthesis of tryptophan, is essential for the virulence for variable periods of time provides evidence for anthranilate
of Mycobacterium tuberculosis (Mtb). AnPRT catalyses the located at transient positions during transfer from the outer site to
Mg2 + -dependent transfer of a phosphoribosyl group from the inner catalytic site. PRPP and Mg2 + binding have been shown
PRPP (5 -phosphoribosyl-1 -pyrophosphate) to anthranilate to to be associated with the rearrangement of two flexible loops,
form PRA (5 -phosphoribosyl anthranilate). Mtb-AnPRT was which is required to complete the inner anthranilate-binding site. It
shown to catalyse a sequential reaction and significant substrate is proposed that anthranilate first binds to the outer site, providing
inhibition by anthranilate was observed. Antimycobacterial an unusual mechanism for substrate capture and efficient transfer
fluoroanthranilates and methyl-substituted analogues were shown to the catalytic site following the binding of PRPP.
to act as alternative substrates for Mtb-AnPRT, producing
the corresponding substituted PRA products. Structures of the
enzyme complexed with anthranilate analogues reveal two distinct
binding sites for anthranilate. One site is located over 8 Å (1 Å = Key words: anthranilate phosphoribosyltransferase (AnPRT),
0.1 nm) from PRPP at the entrance to a tunnel leading to the active fluoroanthranilate, Mycobacterium tuberculosis, trpD, tryptophan
biosynthesis, tuberculosis.

Biochemical Journal
site, whereas in the second, inner, site anthranilate is adjacent to

INTRODUCTION of the gene encoding the enzyme that catalyses the second
committed step in tryptophan biosynthesis, AnPRT (anthranilate
The worldwide emergence of extensively drug-resistant phosphoribosyltransferase) (also known as trpD; EC 2.4.2.18),
Mycobacterium tuberculosis (Mtb), the causative agent of TB has a decreased ability to multiply in murine macrophages
(tuberculosis) [1], has been suggested to be one of the most and is essentially avirulent in both immunocompetent and
profound challenges facing global health [2]. Treatment of TB immunocompromised mice [6]. AnPRT, like the other enzymes
requires a multidrug approach [3] and thus there is a desperate involved in tryptophan biosynthesis, is absent from mammals, so
need for new and effective anti-TB drugs that function via novel inhibitors of this enzyme may have limited mammalian toxicity,
modes of action [4]. strengthening it further as an attractive target for anti-TB drug
Recent studies have shown that host CD4 + T-cells employ design.
tryptophan starvation in an attempt to eradicate M. tuberculosis AnPRT is a member of the PRT (phosphoribosyltransferase)
infection [5]. This innate immune response is effective against family of enzymes characterized by their common substrate
organisms that are natural tryptophan auxotrophs, but as PRPP (5 -phosphoribosyl-1 -pyrophosphate) [7], which acts as
the tryptophan biosynthetic pathway can normally overcome the phosphoribosyl donor in a substitution reaction with a
this starvation in M. tuberculosis, the host immune response nitrogenous, and generally aromatic, base [8]. At least four distinct
is rendered ineffective. Drugs that inhibit the tryptophan structural types of PRT have been identified, with AnPRT the sole
biosynthetic pathway could therefore work synergistically with representative of a type III PRT [9]. Type I PRTs are the most
the host immune response to starve M. tuberculosis of this common, and enzymes in this class act on purines and pyrimidines
essential amino acid. A tryptophan auxotrophic mutant of as part of salvage pathways (e.g. adenine PRT, hypoxanthine PRT
the virulent M. tuberculosis strain H37Rv, with a deletion or uracil PRT) or in the case of orotate PRT as part of the de

Abbreviations: AnPRT, anthranilate phosphoribosyltransferase; Eco , Escherichia coli ; FA, fluoroanthranilate; F-PRA, fluoro-PRA; InGP, indole glycerol
phosphate; InGPS, indole glycerol phosphate synthase; KIE, kinetic isotope effect; MA, methylanthranilate; MESG, 2-amino-6-mercapto-7-methylpurine
ribonucleoside; M-PRA, methyl-PRA; Mtb , Mycobacterium tuberculosis ; NP, nucleoside phosphorylase; PNP, purine nucleoside phosphorylase; PR, 5 -
phosphoribosyl; PRA, 5 -phosphoribosyl anthranilate; PRAI, 5 -phosphoribosyl anthranilate isomerase; PRPP, 5 -phosphoribosyl-1 -pyrophosphate; PRT,
phosphoribosyltransferase; Sso , Sulfolobus solfataricus ; TB, tuberculosis.
1
These authors contributed equally to this work.
2
Present address: Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, 171 77 Stockholm, Sweden.
3
Present address: Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K.
4
Correspondence may be addressed to either of these authors (email s.lott@auckland.ac.nz or emily.parker@canterbury.ac.nz).
The structural co-ordinates reported for Mycobacterium tuberculosis anthranilate phosphoribosyltransferase complexed with anthranilate or anthranilate
analogues have been deposited in the PDB under codes 4N5V, 4N8Q, 4N93, 4OWM, 4OWN, 4OWO, 4OWQ, 4OWS, 4OWU and 4OWV.


c The Authors Journal compilation 
c 2014 Biochemical Society
88 T. V. M. Cookson and others

novo pyrimidine biosynthetic pathway [9]. Quinolate PRT and inhibitors show binding across both S1 and S2 and S2 and S3
nicotinate PRT are type II PRTs, and ATP PRT, which catalyses [23,26].
the first step of histidine biosynthesis, is the only type IV PRT In the present study, we investigated the Mtb-AnPRT catalytic
identified to date. mechanism using compounds that have been reported recently to
The displacement of PPi from PRPP by a nitrogenous base have antimycobacterial activity both in vitro and in a mouse model
that is catalysed by PRTs takes place with inversion of the of disease [5]. The present study reveals that Mtb-AnPRT exhibits
stereochemistry at C1 of the ribose ring. Reactions of this a broad substrate tolerance, and analysis of the crystal structures
type can be either SN 1-like, in which PPi release precedes of the enzyme in complex with these alternative substrates has
nucleophilic attack on PRPP by the base, generating a short- illuminated some details of the reaction mechanism.
lived oxocarbenium ion intermediate, or SN 2-like, where the
nucleophilic attack and PPi loss occur in the same reaction step.
KIE (kinetic isotope effect) experiments with type I orotate PRT EXPERIMENTAL
are consistent with a transition state that has extensive ribo-
Materials
oxocarbenium ion character [10–13]. Structures of type I PRT
enzymes in complex with substrates and transition state analogues Unless otherwise stated, all chemicals were obtained from Sigma–
show that C1 of the ribosyl ring adopts various positions away Aldrich. Anthranilate was obtained from Merck Schuchardt. 4-
from PPi and towards the nitrogenous base, as would be consistent Fluoroanthranilate was obtained from Sigma–Aldrich, whereas
with an SN 1 mechanism [9,14,15]. It has been proposed that 3-, 5- and 6-fluoroanthranilate and 3- and 4-methylanthranilate
type I PRTs guide catalysis by binding PRPP in an orientation were obtained from Fluorochem.
that favours its breakdown into PR (5 -phosphoribosyl) and PPi
[16,17].
AnPRT catalyses the transfer of PR from PRPP to anthranilate. Protein preparation
AnPRT is structurally distinct from the other PRT types, and in- The Mtb-AnPRT protein was expressed and purified as described
stead has a similar structural fold, quaternary structure and active- previously [23]. The Escherichia coli (Eco) trpFC gene had been
site location to that of type II NPs (nucleoside phosphorylases) cloned previously into the expression vector pProEX-HTb, which
[18,19]. Type II NP enzymes catalyse glycosyl transfer reactions adds an N-terminal His6 tag and rTEV (recombinant tobacco
that result in the production of ribose 1-phosphate, which is the etch virus) site. This plasmid was transformed into E. coli
near-reverse reaction of that catalysed by AnPRT [18,19]. To date, BL21(DE3) cells containing plasmids (pBB528 and pBB541) for
most of the NPs characterized using KIEs are of type I and have co-expression of chaperonins (GroES and GroEL) [27].
been shown to have SN 1-like transition states. The only type II The E. coli fusion protein of PRAI (5 -phosphoribosyl
NP characterized by KIEs, human thymidine phosphorylase, has anthranilate isomerase) and InGPS (indole glycerol phosphate
been proposed to have an SN 2 transition state [20,21]. Thus it is synthase) was purified as described for the Mtb-AnPRT protein
not clear whether AnPRT catalyses an SN 1 or an SN 2 reaction with the omission of the size-exclusion chromatography step. For
(Figure 1). Further evidence about the catalytic mechanism of the NMR analyses, the proteins were dialysed against 150 mM
AnPRT will aid in inhibitor design. NH4 HCO3 (pH 8.0).
We have previously determined crystal structures of Mtb- Protein concentrations were determined using a Nanodrop ND-
AnPRT with and without PRPP (PDB codes 2BPQ, 1ZVW 1000 spectrophotometer (Thermo Scientific) and molar absorp-
and 3QR9) [22,23]. The enzyme crystallizes as a homodimer tion coefficient values of 34 615 and 40 715 litres · mol − 1 · cm − 1
with one active site in each subunit. Each monomer comprises for Mtb-AnPRT and Eco-PRAI–InGPS respectively.
two domains: a smaller α-helical N-terminal domain, and a
larger C-terminal domain containing both α-helices and β-sheets.
A hinge region, consisting of the α4–β1, β3–α8 and α9–β4 Kinetics
connecting loops, links the two domains. Dimer formation occurs
through interactions between the helical N-terminal domains, as A coupled assay where PRA (5 -phosphoribosyl anthranilate)
is also observed for the AnPRT enzymes from Acinetobacter formation is detected by the formation of InGP (indole glycerol
baylyi (PDB code 4GTN), Sulfolobus solfataricus (Sso) [24], phosphate) was used to determine the enzyme kinetic parameters
Pectobacterium carotovorum (PDB codes 1KGZ and 1KHD) of Mtb-AnPRT as described previously [23].
[19], Thermus thermophilus (PDB codes 1V8G and 2ELC) The final concentrations of the following components were
and Xanthomonas campestris (PDB code 4HKM). The PRPP included in the coupled assay: PRPP (0–65 μM), MgCl2 (1 mM),
substrate has been shown to bind in the C-terminal domain Mtb-AnPRT (0.1 μM), Eco-PRAI–InGPS (1.7 μM), anthranilate
of the enzyme, at the bottom of a deep cleft, with the PPi or anthranilate analogue (0–10 μM) and Tris buffer (50 mM
group buried the most deeply. When compared with the ligand- Tris/HCl and 150 mM NaCl, pH 8.0). Reactions were initiated
free protein structure, two loops (the β1–α5 and β2–α6 loops) with the addition of Mtb-AnPRT, with initial rates of reaction
can be identified in the PRPP- and Mg2 + -bound structure that determined by least-squares fit of the initial rate data. Raw data
move out of the PRPP-binding site in order to accommodate were fitted to the ternary mechanism equation:
the PRPP substrate. Lee et al. [22] used in silico docking
to predict the binding site of the enzyme’s second substrate, Vmax [A] [B]
v=
anthranilate, and surprisingly predicted two sites despite a K A K B + K B [A] + K A [B] + [A] [B]
1:1 reaction stoichiometry with PRPP (Figure 1). Two similar
anthranilate-binding sites (S1 and S2) were observed directly using GraFit 5 (Erithacus Software).
in Sso-AnPRT (PDB codes 1ZYK and 2GVQ) [25], and a The commercially available EnzChek® Pyrophosphate Assay
third anthranilate-binding site most distal to the PRPP (S3) has Kit (Invitrogen) was used to determine the kinetic parameters
been proposed to be part of a substrate capture mechanism that pertaining to alternative substrate turnover, as not all compounds
was also utilized in the binding of Mtb-AnPRT inhibitors [23]. were turned over by Eco-PRAI–InGPS. Along with PRA,
Structures of Mtb-AnPRT in complex with bi-anthranilate-like Mtb-AnPRT produces PPi in a 1:1 stoichiometric ratio with


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 89

Figure 1 The biosynthesis of tryptophan from anthranilate


Two possible mechanisms are shown for the reaction catalysed by AnPRT (shown in grey): an SN 1-like reaction passing through an oxocarbenium ion intermediate is depicted as the top route, and
an SN 2-like reaction where dissociation of PPi and addition of anthranilate take place in the same reaction step as the bottom route.

anthranilate. In this assay, the PPi produced by the Mtb-AnPRT anthranilate/anthranilate analogue (0–1 mM), MESG (200 μM),
reaction is converted first into Pi by inorganic pyrophosphatase PNP (1 unit), inorganic pyrophosphatase (0.03 unit) and reaction
(1:2), which is then reacted with the substrate MESG (2- buffer (50 mM Tris/HCl and 1 mM MgCl2 , pH 7.5, containing
amino-6-mercapto-7-methylpurine ribonucleoside), which has an 100 μM sodium azide) made up to 1 ml. Reactions were initiated
absorbance maximum of 330 nm, by PNP (purine nucleoside with the addition of Mtb-AnPRT, with initial rates of reaction
phosphorylase) in a 1:1 ratio. The products of this reaction determined by least-squares fit of the initial rate data. Apparent
are ribose 1-phosphate and 2-amino-6-mercapto-7-methylpurine; K m values were obtained by fitting the data to the Michaelis–
the latter can be monitored spectrophotometrically at 360 nm. Menten equation:
Inorganic pyrophosphatase, PNP and Eco-PRAI–InGPS were all
added in excess to ensure that the AnPRT-catalysed reaction was Vmax [S]
V =
the rate-limiting step. The molar absorption coefficient for 2- K m + [S]
amino-6-mercapto-7-methylpurine production from anthranilate
and PRPP was determined to be 26 200 litres · mol − 1 · cm − 1 under whereas substrate inhibition data were fitted to the substrate
assay conditions using known concentrations of anthranilate. inhibition equation:
All assays were measured with a Varian Cary 100 UV-VIS
Vmax [S]
spectrophotometer using quartz cuvettes thermally equilibrated V =  
to 22 ◦ C. The final concentrations of the following components K m + [S] 1 + [S]
Ki
were included in the coupled assay: PRPP (120 μM), MgCl2
(1 mM), Mtb-AnPRT (0.1 μM), Eco-PRAI–InGPS (6 μM), using GraFit 5.


c The Authors Journal compilation 
c 2014 Biochemical Society
90 T. V. M. Cookson and others

Mass spectroscopy of programs [32]. Chain A of the original Mtb-AnPRT–PRPP


Solutions for determination of the mode of epimerization of PRA complex structure (PDB code 1ZVW) [22], without ligands, was
via MS contained 2 mM PRPP, 2 mM anthranilate, 1 mM MgCl2 used as the search model for structure determination by molecular
and 20 mM NaCNBH3 made up to 2 ml with Tris buffer. Solutions replacement using MOLREP [33], and as the initial model
were mixed by inversion and 0.2 μM Mtb-AnPRT was added. for refinement of the AnPRT–anthranilate analogue complex
After a 5 min reaction time, solutions were spun through Vivaspin structures. Refinement and model building was performed with
10 000 Da molecular-mass cut-off concentrators to remove the Coot [34] and Refmac5 [35], or autoBUSTER [36], with restraints
enzyme present, flash-frozen with liquid nitrogen and left to freeze for the anthranilate analogues generated by using the PRODRG
dry (FreeZone 2.5, Labconco) for 4–5 days. Dried products were server [37]. Restraints on bond lengths and angles were based
then resuspended in ultrapure water and analysed via UHR-TOF on the ideal values of Engh and Huber [38] and model quality
(ultra-high-resolution TOF) MS/MS (Bruker maXis 3G). was assessed using MolProbity [39]. Full data collection and
Solutions for analysis of Mtb-AnPRT/Eco-PRAI–InGPS refinement statistics are given in Supplementary Table S1.
reaction products contained 1.2 mM PRPP, 400 μM an- Figures illustrating structural details were prepared using PyMOL
thranilate/fluoroanthranilate or 2 mM methylanthranilate, 2 mM (Schrödinger; http://www.pymol.org), with maps generated using
MgCl2 , 0.6 μM Mtb-AnPRT, 6 μM Eco-PRAI–InGPS made up the FFT program [40] in the CCP4 suite [41].
to 500 μl with ultrapure water for anthranilate/fluoroanthranilate The crystallographic co-ordinates and structure factors have
or 5 mM NH4 HCO3 for methylanthranilate. Reactions with been deposited in the RCSB PDB (Supplementary Table S1).
anthranilate or fluoroanthranilate were initiated with Mtb-AnPRT
immediately before analysis via MS, whereas reactions for
methylanthranilate were incubated with all substituents present RESULTS
for 1 h before analysis. Mtb -AnPRT catalyses a sequential reaction and is inhibited by high
levels of its substrate anthranilate
NMR experiments To examine the kinetic mechanisms of the reaction, initial rate
measurements were taken at various concentrations of both
Assay solutions for analysis of Mtb-AnPRT-catalysed reactions substrates. The kinetic profile is entirely consistent with a
by 1 H-NMR spectroscopy contained 2–2.5 mM PRPP, 0.5–1 mM sequential mechanism for this enzyme (Figure 2A). In addition,
anthranilate or anthranilate analogue, 0.2–0.8 μM Mtb-AnPRT, for assays performed at higher concentrations of the substrate
2 mM MgCl2 , 0.25 mM trimethylsilyl propanoic acid, 80–90 % anthranilate, a significant fall in the enzyme activity was observed,
(v/v) 2 H2 O and 10 mM NH4 HCO3 (pH 8). The 1D 1 H-NMR indicating that the enzyme is subject to significant substrate
experiments were acquired at 25 ◦ C on a DRX-400 spectrometer inhibition (Figure 2B and Table 1).
(Bruker), or an AV-600 spectrometer (Bruker) fitted with a
cryoprobe, and using presaturation of the water resonance [28]. A
1D 1 H-NMR spectrum of each assay solution was collected before
addition of Mtb-AnPRT, and then after addition of the enzyme Fluorine- and methyl-substituted analogues of anthranilate act as
at 100–300 s intervals, with averaging over 16–64 transients. In alternative substrates for Mtb -AnPRT
experiments with Eco-PRAI–InGPS, this enzyme was added at In the process of screening a targeted library of 165 compounds
a concentration of 2.5 μM to an NMR sample in which PRA or for inhibitors of Mtb-AnPRT, we discovered several compounds,
PRA analogue had been formed in situ using the assay conditions including 5MA (5-methylanthranilate) and orthanilate, that are
detailed above, without removal of Mtb-AnPRT. alternative substrates for the enzyme [23]. The fluoroanthranilates
inhibit mycobacterial growth in a tryptophan-dependent manner
[5] and we established that 4FA (4-fluoroanthranilate) was also
Crystallization an alternative substrate of Mtb-AnPRT. These results prompted
us to investigate a series of anthranilate analogues, with fluorine-
Crystallization of native Mtb-AnPRT has been described
or methyl-substitutions on the aromatic ring, as substrates
previously [23]. For soaking experiments and data collection,
for Mtb-AnPRT (Table 1, and Supplementary Figures S1 and
native crystals were removed from the crystallization drop with
S2 at http://www.biochemj.org/bj/461/bj4610087add.htm). The
a nylon loop. The native Mtb-AnPRT crystals were soaked
anthranilate analogue with C6 fluorine substitution has been
in a mixture of the mother liquor with 5 mM PRPP, 2.5 mM
identified previously as inhibiting Pseudomonas aeruginosa
MgCl2 and the alternative substrate to a final concentration
pathogenesis in mice [42], and both C5 and C6 fluoroanthranilate
of 1 mM or anthranilate to a final concentration of 5 nM
analogues have shown promising bactericidal activity against
for various periods of time (Supplementary Table S1 at
M. tuberculosis in the absence of tryptophan [5]. Orthanilate,
http://www.biochemj.org/bj/461/bj4610087add.htm). The soaked
which carries a sulfonyl group in place of the carboxy group of
crystals were quickly pulled through a drop of cryoprotectant
anthranilate, was also tested in the present study. The ability of
containing the mother liquor and 26 % (v/v) glycerol before being
Mtb-AnPRT to process these alternative substrates was probed
flash-cooled in liquid nitrogen.
using a combination of UV-based assays, MS and 1 H-NMR
spectroscopy.
In order to assess the ability of Mtb-AnPRT to process
Data collection, structure solution and refinement each anthranilate analogue, a coupled UV-based assay for
Data were collected at the Australian Synchrotron, Beamline PPi production was used. PPi production by Mtb-AnPRT was
MX2, within 72 h. The crystals of the Mtb-AnPRT–anthranilate observed in the presence of all alternative substrates and PRPP,
analogue complexes diffracted to a maximum resolution that and kinetic parameters were determined for these anthranilate
varied between 1.89 and 2.43 Å (1 Å = 0.1 nm). X-ray diffraction analogues (Table 1).
data were indexed, integrated and scaled with the autoPROC suite Fluorine substitution around the aromatic ring of anthranilate
[29], XDS [30] and SCALA [31] or with the HKL2000 suite had a relatively minor effect on the rate of catalysis, with a


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 91

decrease in kcat values of 25–40 %. The apparent K m for the


fluorinated analogues, however, is highly dependent on the site
of substitution, with fluorine substitution on C3 or C5 resulting
in an 18- and 6-fold increase in K m respectively, whereas fluorine
substitution on either C4 or C6 produces similar K m values to
that for anthranilate. The biggest decrease in kcat /K m is observed
for the alternative substrate 3FA, where the electron-withdrawing
fluorine substituent is found adjacent to the nucleophilic amino
group. K i values for substrate inhibition by these analogues are
again highly dependent on the site of substitution, but show a
different trend, with fluorine substitution on C3 and C4 resulting
in a 24- and 8-fold increase in K i respectively compared with
anthranilate, but substitution on either C5 or C6 having relatively
little effect. Of the anthranilate analogues tested in the present
study, 6FA has kinetic parameters that are most similar to those
of anthranilate.
Methylation of the anthranilate ring has a more significant
effect on the rate of catalysis, with this substitution at C3, C5
or C6 resulting in a ∼3-fold reduction in kcat , and substitution at
C4 yielding a 10-fold reduction. All of the methylated analogues
have much poorer affinity for the enzyme, with apparent K m values
that are increased by at least two orders of magnitude relative
to that of anthranilate. Substrate inhibition was not observed
with these analogues, although this analysis was limited by the
concentrations of methylated analogue required.

1
H-NMR spectroscopy confirms the production of substituted PRA
products from anthranilate analogues

To confirm that the hydrolysis of PPi in the presence


of the anthranilate analogues and PRPP indeed results in
product formation, the enzymatic production of substituted
PRA analogues by Mtb-AnPRT was followed using 1 H-NMR
spectroscopy. Anthranilate, and its substituted analogues, were
each incubated with an excess of PRPP and Mtb-AnPRT, and
1
H-NMR spectra were recorded at 100–300 s intervals as the
reactions proceeded. In the reaction with the native substrate
anthranilate, there was a clear consumption of PRPP over time,
as demonstrated by the loss of signal at 5.80 p.p.m. due to its
anomeric proton, and a corresponding gain of signals between 6.8
and 7.8 p.p.m. and between 5.3 and 5.7 p.p.m., which are due to
PRA (Figure 3, and Supplementary Figure S3 and Supplementary
Table S2 at http://www.biochemj.org/bj/461/bj4610087add.htm).
As observed previously [43], PRA is unstable and was observed to
break down to anthranilate and ribose, with a peak concentration
of PRA observed after approximately 12 min. The anthranilate
released again forms a substrate for Mtb-AnPRT, allowing the
PRA formation reaction to proceed until all of the PRPP is
consumed (Figure 3A).
Unexpectedly, two anomeric protons (at 5.70 and 5.31 p.p.m.)
and two sets of aromatic proton peaks were identified
Figure 2 Plots for determination of the catalytic mechanism and corresponding to the product PRA. This indicates that both
anthranilate kinetic parameters α- and β-epimers at the C1 position of the ribose moiety
(A) Mtb -AnPRT kinetic data obtained using the enzyme-coupled UV assay, fitted to the ternary of PRA are present in the product mixture. The 5.70 and
mechanism equation and presented as a double-reciprocal plot. PRPP was used at 12.5 μM 5.31 p.p.m. resonances were assigned to the α- and β-epimers
(䊐), 25 μM () or 62.5 μM (䊊). The K m values for anthranilate and PRPP were determined to of PRA respectively on the basis of chemical shifts and coupling
be 0.9 + + + −1
− 0.6 μM and 12 − 4 μM respectively, with a k cat value of 1.1 − 0.1 s . (B) Substrate constants reported for related compounds [44,45]. From the
inhibition is observed at high concentrations of anthranilate, with a K i value of 45 +
− 6 μM.
Results were obtained using the pyrophosphate UV assay in the presence of 120 μM PRPP,
relative integrals of the two anomeric proton peaks, the ratio of
and fitted to the substrate inhibition equation. (C) Michaelis–Menten plot for determination the α- to the β-epimer is approximately 7:3 at equilibrium under
of the apparent K m of anthranilate, as measured by the pyrophosphate assay in the presence the conditions of the assay.
of 120 μM PRPP. At lower concentrations of anthranilate, no inhibition is observed, with an As with other PRT enzymes [46,47], it is reasonable to
apparent K m value for anthranilate of 1.7 + + −1
− 0.1 μM, and a k cat value of 2.0 − 0.1 s . expect that the Mtb-AnPRT reaction proceeds with inversion of
stereochemistry at the C1 position on the ribose ring of PRPP.
Crystal structures of several different AnPRT enzymes [19,22,25]


c The Authors Journal compilation 
c 2014 Biochemical Society
92 T. V. M. Cookson and others

Table 1 Apparent K m values, turnover number, specificity constant and substrate inhibition K i values for substituted anthranilate substrates of Mtb -AnPRT
N/A indicates that the value was not measured, as no substrate inhibition was observable under assay conditions. The Michaelis–Menten plots are shown in Supplementary Figures S1 and S2 at
http://www.biochemj.org/bj/461/bj4610087add.htm.

Substrate Structure K m value (μM) k cat value (s − 1 ) k cat /K m (s − 1 · μM − 1 ) K i value (μM)

Anthranilate 1.7 +
− 0.1 2.0 +
− 0.1 1.2 +
− 0.1 45 +
−6

3FA 31 +
−3 1.4 +
− 0.03 0.05 +
− 0.006 1090 +
− 330

4FA 2.1 +
− 0.2 1.2 +
− 0.1 0.6 +
− 0.1 380 +
− 100

5FA 10 +
−1 1.3 +
− 0.1 0.13 +
− 0.01 60 +
− 11

6FA 1.4 +
− 0.1 1.3 +
− 0.1 0.9 +
− 0.1 34 +
−5

3MA 290 +
− 20 0.63 +
− 0.03 0.002 +
− 0.0002 N/A

4MA 490 +
− 130 0.15 +
− 0.02 0.0003 +
− 0.0001 N/A

5MA 570 +
− 120 0.62 +
− 0.07 0.0011 +
− 0.0004 N/A

6MA 800 +
− 120 0.71 +
− 0.07 0.0009 +
− 0.0002 N/A

Orthanilate 130 +
− 10 0.1 +
− 0.004 0.0008 +
− 0.0001 N/A

provide supporting evidence for this claim, as it can be seen that analysed using MS. Evidence for the reduced imine was indeed
anthranilate can only approach from the β-position, thereby only observed, with m/z peaks present at 352.0782 and 374.0603 Da,
creating β-PRA. It is therefore likely that the α-PRA observed corresponding to the masses of the reduced imine and its sodium
is formed after the reaction due to rapid epimerization of β- salt respectively.
PRA in solution, a phenomenon that has been identified in other The Mtb-AnPRT-catalysed turnover of PRPP in the presence
PRA-like compounds such as phosphoribosyl amine [44,48]. of all of the anthranilate analogues was followed using 1 H-NMR
Epimerization is likely to occur via intramolecular rearrange- spectroscopy. For all of these alternative substrates, excluding
ment through an imine intermediate (Supplementary Figure 3MA, the concurrent formation of the corresponding PRA ana-
S4 at http://www.biochemj.org/bj/461/bj4610087add.htm). The logue was detected (Supplementary Figure S5 and Supplementary
products of the Mtb-AnPRT reaction, formed in the presence of Table S2 at http://www.biochemj.org/bj/461/bj4610087add.htm).
NaCNBH3 to trap the proposed iminium ion intermediate, were As observed for native PRA, the fluorine- and methyl-substituted


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 93

Figure 3 Progress curves for Mtb -AnPRT-catalysed reactions followed by 1 H-NMR spectroscopy
PRPP (2.5 mM) and anthranilate (A) or 4FA (B) (1 mM) were incubated with Mtb -AnPRT (0.8 μM) at 25 ◦ C and pH 8, and 1 H-NMR spectra were collected at 105-s intervals. Approximate
concentrations during the reaction were estimated on the basis of peak integrals for H1 of PRPP, H8 of anthranilate and H8 of PRA (α-PRA and β-PRA combined), relative to an internal TSP
(trimethylsilyl propionate) standard. Full details of the 1 H-NMR assay are given in the Experimental section.

PRAs accumulate as a mixture of α- and β-epimers in an Structures of Mtb -AnPRT in complex with fluorine- and
approximate 7:3 ratio in the assay solution. methyl-substituted anthranilates define two distinct
Fluorinated and sulfonylated analogues of PRA were observed substrate-binding sites
to be significantly more stable than PRA itself. As illustrated
for 4FA in Figure 3(B), the stability of the product, 4F-PRA Structures of Mtb-AnPRT soaked with the anthranilate analogues
(4-fluoro-PRA), results in a more distinct biphasic progress for various time periods prior to crystal harvesting and freezing
curve during monitoring of the Mtb-AnPRT-catalysed reaction were determined to gain structural insight into the binding of these
by NMR spectroscopy. Initially, the reaction progresses quickly alternative substrates. The lower turnover numbers of the enzyme
until 4FA is consumed, and then it continues at a lower with these analogues has allowed substrate-bound complexes to be
rate governed by the breakdown of 4F-PRA to release 4FA, visualized, which are difficult to access with the natural substrate
until the excess PRPP is consumed. Conversely, the inability anthranilate. The structures of these complexes reveal a consistent
to detect the PRA analogue resulting from turnover of 3MA pattern where S3, the substrate-capture site, is occupied first, after
may reflect a shorter half-life of 3M-PRA (3-methyl-PRA) which the substrate moves through the tunnel to a position adja-
relative to PRA itself. NMR spectroscopy was also used to cent to PRPP, the catalytic site (S1). The existence of more than
monitor directly the degradation of PRA and two substituted one binding site for anthranilate has previously been inferred from
PRA analogues in solution (Supplementary Figure S6 at modelling [22] and inhibition [23,26] studies and by analogy with
http://www.biochemj.org/bj/461/bj4610087add.htm). results for the Sso-AnPRT enzyme [25]. Notably, however, no dir-
The substituted PRA analogues were also shown to be ect structural information for Mtb-AnPRT is yet available showing
substrates for the next two enzymes of the tryptophan biosynthesis the substrate anthranilate bound in a catalytically relevant mode.
pathway from E. coli PRAI and InGPS, which are expressed The structure of Mtb-AnPRT soaked with PRPP, Mg2 + and
as a single protein with two catalytic domains [49]. The 4FA for 6 min most clearly defines the two major sites S1 and S3
formation of fluorinated and methylated InGP analogues was (Figure 4), and is used here as the reference structure to describe
observed by MS, UV spectroscopy and 1 H-NMR spectroscopy the active site and understand the reaction mechanism. In this
(Supplementary Figures S7–S9 and Supplementary Table S3 complex, both sites are fully occupied by the 4FA substrate and
at http://www.biochemj.org/bj/461/bj4610087add.htm) in the the same interactions are made between enzyme and substrate in
presence of Eco-PRAI–InGPS. In the 1 H-NMR experiments, both molecules of the Mtb-AnPRT dimer. The enzyme structure
it was observed that both epimers of PRA or 4F-PRA were is unchanged from its ligand-free state by substrate binding, and
consumed in line with the predicted rapid interconversion no domain movements are necessary to bring the anthranilate
of these isomers. Unlike the other PRA analogues studied, substrate into contact with the PRPP as was proposed for Sso-
sulfonyl-PRA could theoretically undergo PRA isomerization AnPRT [25]. Superposition of the 4FA complex on to the
catalysed by PRAI to yield CdRP [1-(o-carboxyphenylamino)- substrate-free and liganded (with PRPP, Mg2 + and inhibitors)
1 -deoxyribulose-5 -phosphate], but not the decarboxylation Mtb-AnPRT structures previously described [23] gave RMSDs of
catalysed by InGPS to form the indole InGP. Consistent with 0.22–0.88 Å for all Cα atoms.
this, the formation of InGP was not observed on incubation of The pocket within which the substrate binds in its catalytically
sulfonyl-PRA with Eco-PRAI–InGPS. However, the formation competent S1 position is bounded by the PRPP ribose ring and
of a small amount of an uncharacterized compound or residues from the β1–α5 and β2–α6 loops that close over PRPP
compounds was indicated by the appearance of a new set (Val106 , Gly107 , Thr108 , His136 and Asn138 ), together with Ala179 ,
of 1 H-NMR peaks (Supplementary Figure S8 at http://www. Arg193 , Gly206 and Pro207 . The binding mode of the 4FA molecule
biochemj.org/bj/461/bj4610087add.htm). at S1 is shown in Figure 4(C). It has several interactions with


c The Authors Journal compilation 
c 2014 Biochemical Society
94 T. V. M. Cookson and others

Figure 4 Structure of Mtb -AnPRT soaked with PRPP, Mg2 + and 4FA for 6 min
4FA is shown bound in both the inner catalytic S1 (orange) and the outer S3 (yellow) of chain A. PRPP is shown in magenta, with Mg2 + as black spheres. Polar contacts are shown as yellow
broken lines, and water molecules as red spheres. Analogous binding modes and interactions were observed for chain B. (A) Monomer of Mtb -AnPRT with PRPP, Mg2 + and 4FA bound, and with
key structural features indicated. (B) Surface representation showing 4FA bound at S1 and S3. (C) 4FA bound in the inner catalytic site (S1). (D) 4FA bound in the outer site (S3). (E) Omit map
calculated before the addition of ligands to the model, where the F o − F c map is contoured at 3σ and coloured green and red for positive and negative density respectively. (F) The 2F o − F c map
for the ligands after final refinement is shown in blue, contoured at 1σ .


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 95

the surrounding structure including hydrogen bonds between its parameters seen between substrates with substituent groups at
amino group and the carbonyl oxygen of Gly107 and the C2 different positions.
hydroxy group of PRPP, and hydrogen bonds made between The movement of anthranilate-like substrates from S3 into the
its carboxy group, the side-chain amide group of Asn138 , the η- catalytic S1 is more complex than it appears at first sight. At
nitrogen of Arg193 and several water molecules. The other notable S3, the amino group for a number of the alternative substrates
interaction involves the aromatic ring, which is sandwiched is oriented towards His183 , hydrogen-bonding to the backbone
between the Cα of Gly206 and the Cβ of Asn138 . In this orientation, carbonyl oxygen. The ring must be flipped 180◦ , however, to
the amino nitrogen is 4.3 Å from the C1 atom of the PRPP orient the amino group towards the PRPP molecule in the substrate
ribose ring, in position to attack this atom from the β position. pocket. This could happen by normal ‘breathing’ of the protein
The 4-fluoro-substituent of 4FA is well accommodated, making structure. However, the structure of the 6MA complex after
hydrophobic contacts with Val106 Cγ 2 (3.2 Å) and with the side a 30 min soak shows positive electron density at a third site,
chain of His136 (3.2–3.4 Å). The same binding mode is seen for the equivalent to the previously described S2 [26], which is located
5FA structure, where the fluorine moiety is situated ∼3.5 Å from between S1 and S3 (Figure 5E). S2 is formed in a mostly
the centre of the His136 imidazole ring. Structures of Mtb-AnPRT hydrophobic pocket between the side chains of Met86 , Ala179 ,
with 6FA or 6MA bound also show binding at S1, where the C6- His183 , Tyr186 and His136 . The electron density at this site is more
substituent projects unimpeded along the substrate tunnel. The poorly defined, and cannot be accurately modelled, suggesting
accommodation of 4FA, 5FA and 6FA at S1 is consistent with the that it is a transient site that lacks anchor points for the substrate
similarity of their kinetic parameters to that of the native substrate, molecule.
anthranilate. Crystal structures of complexes with 3MA, 4MA and Mtb-AnPRT soaked with PRPP, Mg2 + and anthranilate for
5MA did not show binding at S1, and may be indicative of steric 4 s shows electron density present in the substrate-capture site
clashes amplified by the larger methyl substitutions. Likewise, alone. The observation that anthranilate can bind in this outer
3FA accommodated in this site would result in unfavourable site without PRPP or Mg2 + present provides some insight into
electrostatic interactions between the fluorine substituent and the the order of substrate-binding events, and is consistent with
Gly107 backbone carbonyl. the recent report by Evans et al. [26] that a bi-anthranilate-like
The second binding site of 4FA, which we designate the inhibitor can bind in both S2 and S3 in the absence of PRPP
substrate-capture site, is located ∼8 Å from S1 (distance measured or Mg2 + . Previous comparisons of the Mtb-AnPRT PRPP-bound
between the centres of the two rings), in a position at the and ligand-free structures [22] show clear rearrangement of the
entrance to the tunnel that extends into the active site. This site is flexible β1–α5 and β2–α6 loops upon PRPP/Mg2 + binding, and
different from a more buried site (S2) that was predicted by Mtb- these rearrangements are likely to be necessary to construct the
AnPRT modelling [22] and which is occupied by anthranilate inner catalytic binding site for anthranilate. These flexible loops
in Sso-AnPRT structures [25], but is coincident with the outer remain disordered in the 4-s anthranilate soak structure, indicating
inhibitor-binding site (S3) seen for many inhibitor complexes of that binding to the outer site S3 does not require prior PRPP/Mg2 +
Mtb-AnPRT [23,26]. That it differs in detail from the second binding or lead to the rearrangement of the loops. Binding of
anthranilate site found for Sso-AnPRT [25] is not surprising anthranilate at S3 may precede PRPP/Mg2 + binding, with the
given that the residues involved are mostly non-conserved. In S3 enzyme effectively sequestering this substrate until it can proceed
(Figure 4D), the substrate is held in position by stacking between to the inner catalytic site S1 to enable reaction with PRPP.
Pro180 on one side and the side chains of Tyr186 and Ala190 on the
other, and by hydrogen bonds between its carboxy group, Arg194
DISCUSSION
and two waters, and between its amino group, a water molecule
and the carbonyl oxygen of His183 . PRTs are present in the biosynthetic pathways of tryptophan
and histidine, as well as playing a role in nucleotide-salvage
and -synthesis pathways, and as such are important enzymes
for bacterial metabolism. Their potential as antibacterial drug
Early binding events as deduced from the substrate complex targets makes the understanding of their mechanisms of significant
structures
interest. The present study provides insight into the reaction
All the crystal structures of Mtb-AnPRT complexed with the mechanism of Mtb-AnPRT, and includes the first crystal structures
fluorine- and methyl-substituted anthranilates show binding at of Mtb-AnPRT in complex with both PRPP and anthranilate
S3, even for the very shortest soaking times. This clearly points analogues. These structures provide a clearer picture of how
to binding at this site as the earliest event in substrate binding. We anthranilate is transported from the bulk solvent to the catalytic
illustrate this for the 4FA substrate after a 6 s soak (Figure 5). For pocket for a productive reaction with PRPP.
this complex, the binding mode in S3 is already well defined for Mtb-AnPRT catalyses a sequential reaction and catalytic
both Mtb-AnPRT molecules of the dimer, with the substrate in the activity is inhibited by high concentrations of the substrate
same orientation as seen in the longer 6-min 4FA-soaked complex. anthranilate. Mtb-AnPRT is remarkably tolerant of substitution on
When the crystal structures of the other substrate complexes are the phenyl ring of anthranilate. Fluorine- and methyl-substituted
compared, the substrate is always bound in S3 with its anthranilate analogues of anthranilate were accepted as alternative substrates,
ring stacked between Pro180 and Tyr186 as for the 4FA complex, and and substituted PRA products were identified by NMR and
with its carboxy group hydrogen-bonded to Arg194 . The substrate’s MS studies. Unsurprisingly, with its lower steric demands,
aromatic ring can, however, be flipped to give two different modes fluorine substitution was more efficiently accommodated than the
of interaction by the amino group, hydrogen-bonded either to the introduction of a methyl group. The bactericidal activity of 5FA
backbone oxygen of His183 (4FA, 5FA, 4MA, 5MA and 6MA) and 6FA against M. tuberculosis demonstrated by Zhang et al. [5]
or to Oδ1 of Asn138 (3FA, 6FA and 3MA). It is possible that the is only observed in the absence of tryptophan, consistent with
fluorine or methyl substituents force the substrate to adopt one of these compounds acting by targeting tryptophan biosynthesis.
two orientations in S3 to minimize unfavourable interactions with This activity could plausibly originate from the toxicity of
the surrounding residues. These interactions and orientations of fluorinated intermediates generated by the turnover of these
the substrates at S3 could contribute to the variation in kinetic compounds by Mtb-AnPRT and subsequent pathway enzymes.


c The Authors Journal compilation 
c 2014 Biochemical Society
96 T. V. M. Cookson and others

Figure 5 Structures of Mtb -AnPRT soaked with PRPP, Mg2 + and either 6MA for 30 min or 4FA for 6 s
6MA is shown bound at both S1 (cyan) and S3 (purple) of chain A, and 4FA (green) is shown bound at S3 of chain B. PRPP is shown in yellow, with Mg2 + as green spheres. Polar contacts are
shown as yellow broken lines, and water molecules as red spheres. Analogous binding modes and interactions were observed for chain B of the 6MA structure, and in chain A of the 4FA structure. (A
and B) Binding of ligands in the active site for the 6MA and 4FA structures respectively. (C and D) 2F o − F c maps for the ligands after final refinement for the 6MA and 4FA structures respectively.
The 2F o − F c maps are shown in blue and are contoured at 1σ . (E and F) Omit maps for the 6MA and 4FA structures respectively. The omit maps displayed were calculated before the addition of
ligands to the models, and show the F o − F c map contoured at 3σ and coloured green and red for positive and negative density respectively.

In crystal structures of Mtb-AnPRT reported previously [22,23], only productively bind at the inner site S1 after PRPP has bound.
it has been observed that residues from the flexible loops β1– The crystal structures solved in the present study strengthen
α5 and β2–α6 that close over the PRPP molecule after binding this claim further, as 4FA can clearly be seen to bind to the
into the substrate pocket form the contacts for anthranilate residues from the flexible loops at S1, namely Asn138 and Gly107 .
binding in S1. This observation, coupled with our findings of It can also be observed that the above-mentioned flexible PRPP-
a sequential mechanism, strongly indicates that anthranilate can binding loops are in the ‘open’ position in the ligand-free structure


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 97

and closed in the 4FA/PRPP/Mg2 + -bound and PRPP/Mg2 + - Evans, Tammie Cookson, Esther Bulloch, Emily Parker, Edward Baker and Shaun Lott
bound structures, indicating that the enzyme is only primed for revised the paper before acceptance.
efficient anthranilate binding at the catalytic S1 once PRPP is
bound. In contrast, the AnPRT enzymes from S. solfataricus
and Saccharomyces cerevisiae have both been reported to exhibit ACKNOWLEDGEMENTS
random sequential Bi Bi mechanisms [25,50]. For Sso-AnPRT, We gratefully acknowledge Dr Joseph Mire, Dr Inna Kriger and Professor Jim Sacchettini
this discrepancy is surprising given the similarity of the substrate- (Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX,
binding modes between the two structures. U.S.A.) for the phenotypic high-throughput screens that identified 4FA, initial tryptophan
The closure of flexible loops over a substrate-binding pocket biosynthesis inhibition experiments involving 4FA and for providing us with a small
in Mtb-AnPRT is not unique among the PRT family. Substrate amount of 4FA compound for initial experiments. The Mtb -AnPRT expression plasmid was
originally supplied by Dr Farah Javid-Majd (Department of Biochemistry and Biophysics,
binding instigates conformational change in both type I and Texas A&M University). We also acknowledge Dr Jackie Kendall, Dr Julie Spicer and
type II PRTs, either by flexible loop rearrangement (type I) or by Professor Bill Denny (Auckland Cancer Society Research Centre, Auckland, New Zealand)
relative domain rotation (type II). In both cases, the importance for providing the compounds for the initial targeted screen against Mtb -AnPRT, and for the
of enclosing the active site has been correlated with protecting the 5MA, 6MA and orthanilate compounds tested in the present study. This work was performed
hypothesized highly reactive carbocation intermediate. Previous as part of the M. tuberculosis Structural Genomics Consortium (http://www.webtb.org).
experiments on orotate PRT, ATP-PRT and hypoxanthine PRT
using KIEs strongly support the idea of an SN 1-like reaction
[12,13]. The observation that none of the fluoroanthranilate FUNDING
analogues in the present study significantly adversely affects The research was funded by grants from the Health Research Council of New Zealand,
the overall kcat value of Mtb-AnPRT could indicate that the the Foundation for Research, Science and Technology of New Zealand and the Tertiary
nucleophilicity of the amino group is not of great significance Education Commission of New Zealand, through the Maurice Wilkins Centre.
to the overall reaction rate, consistent with either a dissociative
SN 1-like mechanism for AnPRT or rate-limiting substrate-binding
or release steps. REFERENCES
The substituted analogues allow direct observation of two
distinct binding sites for anthranilate in Mtb-AnPRT: an inner 1 Shah, N. S., Wright, A., Bai, G. H., Barrera, L., Boulahbal, F., Martin-Casabona, N.,
catalytic site (S1) and an outer substrate-capture site (S3). Drobniewski, F., Gilpin, C., Havelkova, M., Lepe, R. et al. (2007) Worldwide emergence of
extensively drug-resistant tuberculosis. Emerg. Infect. Dis. 13, 380–387 CrossRef
Additionally, some evidence for passage of anthranilate between
2 Upshur, R., Singh, J. and Ford, N. (2009) Apocalypse or redemption: responding to
the outer S3 and the catalytic S1 is seen via an intermediate
extensively drug-resistant tuberculosis. Bull. World Health Organ. 87, 481–483
binding site (S2), in a manner consistent with previously observed CrossRef
inhibitor-binding studies [26]. The role of S3 in trapping 3 Goldberg, D. E., Siliciano, R. F. and Jacobs, Jr, W. R. (2012) Outwitting evolution: fighting
anthranilate and the flexibility of the loops associated with the drug-resistant TB, malaria, and HIV. Cell 148, 1271–1283 CrossRef
active site may have a direct role in the productive use of 4 Gandhi, N. R., Nunn, P., Dheda, K., Schaaf, H. S., Zignol, M., van Soolingen, D., Jensen,
highly reactive PRPP in the transferase reaction. If anthranilate P. and Bayona, J. (2010) Multidrug-resistant and extensively drug-resistant tuberculosis:
is first captured in the outer S3 and PRPP then binds, leading a threat to global control of tuberculosis. Lancet 375, 1830–1843 CrossRef
to concomitant formation of the higher-affinity inner catalytic 5 Zhang, Y. J., Reddy, M. C., Ioerger, T. R., Rothchild, A. C., Dartois, V., Schuster, B. M.,
site by loop rearrangements, efficient shuttling of anthranilate Trauner, A., Wallis, D., Galaviz, S., Huttenhower, C. et al. (2013) Tryptophan biosynthesis
to the catalytically relevant S1 is achieved. This sequence of protects mycobacteria from CD4 T-cell-mediated killing. Cell 155, 1296–1308
events allows anthranilate to efficiently react with the developing CrossRef
oxocarbenium ion intermediate derived from PRPP and 6 Smith, D. A., Parish, T., Stoker, N. G. and Bancroft, G. J. (2001) Characterization of
minimizes exposure of this highly reactive intermediate to water. auxotrophic mutants of Mycobacterium tuberculosis and their potential as vaccine
candidates. Infect. Immun. 69, 1142–1150 CrossRef
Although potentially facilitating efficient formation of PRA
7 Sinha, S. C. and Smith, J. L. (2001) The PRT protein family. Curr. Opin. Struct. Biol. 11,
by AnPRT, the outer S3 may also be somewhat of an Achilles
733–739 CrossRef
heel for the enzyme. These studies reveal that the substrate 8 Musick, W. D. (1981) Structural features of the phosphoribosyltransferases and their
anthranilate significantly inhibits this enzyme, and it is tempting relationship to the human deficiency disorders of purine and pyrimidine metabolism. CRC
to speculate that this inhibition may arise from non-productive Crit. Rev. Biochem. 11, 1–34 CrossRef
binding of anthranilate to S3 as anthranilate is transferred to the 9 Schramm, V. L. and Grubmeyer, C. (2004) Phosphoribosyltransferase mechanisms and
productive catalytic S1. It is noteworthy that bactericidal 5FA and roles in nucleic acid metabolism. Prog. Nucleic Acid Res. Mol. Biol. 78, 261–304
6FA differ from the other alternative substrates in their ability to CrossRef
mimic the anthranilate substrate inhibition, and this contrasting 10 Zhang, Y. and Schramm, V. L. (2010) Pyrophosphate interactions at the transition states of
kinetic behaviour is suggestive of different affinities of the two Plasmodium falciparum and human orotate phosphoribosyltransferases. J. Am. Chem.
anthranilate sites for these analogues. Moreover, several inhibitors Soc. 132, 8787–8794 CrossRef
of this enzyme have been shown to utilize S2 and S3 [23,26], 11 Zhang, Y., Luo, M. and Schramm, V. L. (2009) Transition states of Plasmodium falciparum
suggesting that this unique allosteric feature of this enzyme may and human orotate phosphoribosyltransferases. J. Am. Chem. Soc. 131, 4685–4694
be fruitfully targeted for inhibition, and may be the basis for new CrossRef
12 Tao, W., Grubmeyer, C. and Blanchard, J. S. (1996) Transition state structure of Salmonella
anti-TB therapeutics.
typhimurium orotate phosphoribosyltransferase. Biochemistry 35, 14–21 CrossRef
13 Goitein, R. K., Chelsky, D. and Parsons, S. M. (1978) Primary 14 C and α secondary 3 H
substrate kinetic isotope effects for some phosphoribosyltransferases. J. Biol. Chem.
253, 2963–2971
AUTHOR CONTRIBUTION 14 Tao, W., Grubmeyer, C. and Blanchard, J. S. (1996) Transition state structure of Salmonella
Shaun Lott, Tammie Cookson, Alina Castell, Esther Bulloch and Emily Parker conceived and typhimurium orotate phosphoribosyltransferase. Biochemistry 35, 14–21 CrossRef
designed the experiments. Tammie Cookson, Alina Castell, Esther Bulloch and Francesca 15 Fedorov, A., Shi, W., Kicska, G., Fedorov, E., Tyler, P. C., Furneaux, R. H., Hanson, J. C.,
Short performed the experiments. Tammie Cookson, Alina Castell, Esther Bulloch and Gainsford, G. J., Larese, J. Z., Schramm, V. L. and Almo, S. C. (2001) Transition state
Emily Parker analysed the data. Emily Parker, Tammie Cookson, Alina Castell, Esther structure of purine nucleoside phosphorylase and principles of atomic motion in
Bulloch, Genevieve Evans and Edward Baker wrote the paper. Alina Castell, Genevieve enzymatic catalysis. Biochemistry 40, 853–860 CrossRef


c The Authors Journal compilation 
c 2014 Biochemical Society
98 T. V. M. Cookson and others

16 Krahn, J. M., Kim, J. H., Burns, M. R., Parry, R. J., Zalkin, H. and Smith, J. L. (1997) 34 Emsley, P. and Cowtan, K. (2004) Coot: model-building tools for molecular graphics. Acta
Coupled formation of an amidotransferase interdomain ammonia channel and a Crystallogr. D Biol. Crystallogr. 60, 2126–2132 CrossRef
phosphoribosyltransferase active site. Biochemistry 36, 11061–11068 CrossRef 35 Murshudov, G. N., Vagin, A. A. and Dodson, E. J. (1997) Refinement of macromolecular
17 Smith, J. L. (1998) Glutamine PRPP amidotransferase: snapshots of an enzyme in action. structures by the maximum-likelihood method. Acta Crystallogr. D Biol. Crystallogr. 53,
Curr. Opin. Struct. Biol. 8, 686–694 CrossRef 240–255 CrossRef
18 Mayans, O., Ivens, A., Nissen, L. J., Kirschner, K. and Wilmanns, M. (2002) Structural 36 Blanc, E., Roversi, P., Vonrhein, C., Flensburg, C., Lea, S. M. and Bricogne, G. (2004)
analysis of two enzymes catalysing reverse metabolic reactions implies common ancestry. Refinement of severely incomplete structures with maximum likelihood in BUSTER-TNT.
EMBO J. 21, 3245–3254 CrossRef Acta Crystallogr. D Biol. Crystallogr. 60, 2210–2221 CrossRef
19 Kim, C., Xuong, N. H., Edwards, S., Yee, M. C., Spraggon, G. and E Mills, S. (2002) The 37 Schuttelkopf, A. W. and van Aalten, D. M. F. (2004) PRODRG: a tool for high-throughput
crystal structure of anthranilate phosphoribosyltransferase from the enterobacterium crystallography of protein–ligand complexes. Acta Crystallogr. D Biol. Crystallogr. 60,
Pectobacterium carotovorum . FEBS Lett. 523, 239–246 CrossRef 1355–1363 CrossRef
20 Birck, M. R. and Schramm, V. L. (2004) Binding causes the remote [5 -3 H]thymidine 38 Engh, R. A. and Huber, R. (1991) Accurate bond and angle parameters for X-ray
kinetic isotope effect in human thymidine phosphorylase. J. Am. Chem. Soc. 126, protein-structure refinement. Acta Crystallogr. A 47, 392–400 CrossRef
6882–6883 CrossRef 39 Chen, V. B., Arendall, W. B., III, Headd, J. J., Keedy, D. A., Immormino, R. M., Kapral, G.
21 Birck, M. R. and Schramm, V. L. (2004) Nucleophilic participation in the transition state J., Murray, L. W., Richardson, J. S. and Richardson, D. C. (2010) MolProbity: all-atom
for human thymidine phosphorylase. J. Am. Chem. Soc. 126, 2447–2453 CrossRef structure validation for macromolecular crystallography. Acta Crystallogr. D Biol.
22 Lee, C. E., Goodfellow, C., Javid-Majd, F., Baker, E. N. and Lott, J. S. (2006) The crystal Crystallogr. 66, 12–21 CrossRef
structure of TrpD, a metabolic enzyme essential for lung colonization by Mycobacterium 40 Ten Eyck, L. F. (1985) Fast Fourier-transform calculation of electron-density maps.
tuberculosis , in complex with its substrate phosphoribosylpyrophosphate. J. Mol. Biol. Methods Enzymol. 115, 324–337 CrossRef
355, 784–797 CrossRef 41 Winn, M. D., Ballard, C. C., Cowtan, K. D., Dodson, E. J., Emsley, P., Evans, P. R., Keegan,
23 Castell, A., Short, F. L., Evans, G. L., Cookson, T. V. M., Bulloch, E. M. M., Joseph, D. D. R. M., Krissinel, E. B., Leslie, A. G. W., McCoy, A. et al. (2011) Overview of the CCP4 suite
A., Lee, C. E., Parker, E. J., Baker, E. N. and Lott, J. S. (2013) Substrate capture and current developments. Acta Crystallogr. D Biol. Crystallogr. 67, 235–242 CrossRef
mechanism of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase 42 Lesic, B., Lépine, F., Déziel, E., Zhang, J., Zhang, Q., Padfield, K., Castonguay, M.-H.,
provides a mode for inhibition. Biochemistry 52, 1776–1787 CrossRef Milot, S., Stachel, S., Tzika, A. A. et al. (2007) Inhibitors of pathogen intercellular signals
24 Ivens, A., Mayans, O., Szadkowski, H., Wilmanns, M. and Kirschner, K. (2001) as selective anti-infective compounds. PLoS Pathog. 3, 1229–1239 CrossRef
Purification, characterization and crystallization of thermostable anthranilate 43 Sterner, R., Kleemann, G. R., Szadkowski, H., Lustig, A., Hennig, M. and Kirschner, K.
phosphoribosyltransferase from Sulfolobus solfataricus . Eur. J. Biochem. 268, (1996) Phosphoribosyl anthranilate isomerase from Thermotoga maritima is an extremely
2246–2252 CrossRef stable and active homodimer. Protein Sci. 5, 2000–2008 CrossRef
25 Marino, M., Deuss, M., Svergun, D. I., Konarev, P. V., Sterner, R. and Mayans, O. (2006) 44 Cheng, Y. S., Murray, M., Schendel, F., Otvos, J., Wehrli, S. and Stubbe, J. (1987)
Structural and mutational analysis of substrate complexation by anthranilate Chemical characterization of phosphoribosylamine, a substrate for newly discovered
phosphoribosyltransferase from Sulfolobus solfataricus . J. Biol. Chem. 281, trifunctional protein containing glycineamide ribonucleotide synthetase activity. Adv.
21410–21421 CrossRef Enzyme Regul. 26, 319–333 CrossRef
26 Evans, G. L., Gamage, S. A., Bulloch, E. M. M., Baker, E. N., Denny, W. A. and Lott, J. S. 45 Trandinh, S., Neumann, J. M., Thiery, J. M., Huynhdinh, T., Igolen, J. and Guschlbauer, W.
(2014) Re-purposing the chemical scaffold of the anti-arthritic lobenzarit to target (1977) Configuration and conformation of α-anomers and β-anomers of C-nucleosides
tryptophan biosynthesis in Mycobacterium tuberculosis . ChemBioChem 15, by proton magnetic-resonance spectroscopy: new criterion for determination of
852–864 CrossRef α-anomers and β-anomers. J. Am. Chem. Soc. 99, 3267–3273 CrossRef
27 de Marco, A. (2011) Molecular and chemical chaperones for improving the yields of 46 González-Segura, L., Witte, J. F., McClard, R. W. and Hurley, T. D. (2007) Ternary complex
soluble recombinant proteins. Methods Mol. Biol. 705, 31–51 CrossRef formation and induced asymmetry in orotate phosphoribosyltransferase. Biochemistry
28 Bax, A. (1985) A spatially selective composite 90◦ radiofrequency pulse. J. Magn. Reson. 46, 14075–14086 CrossRef
65, 142–145 47 Sharma, V., Grubmeyer, C. and Sacchettini, J. C. (1998) Crystal structure of quinolinic
29 Vonrhein, C., Flensburg, C., Keller, P., Sharff, A., Smart, O., Paciorek, W., Womack, T. and acid phosphoribosyltransferase from Mycobacterium tuberculosis : a potential TB drug
Bricogne, G. (2011) Data processing and analysis with the autoPROC toolbox. Acta target. Structure 6, 1587–1599 CrossRef
Crystallogr. D Biol. Crystallogr. 67, 293–302 CrossRef 48 Schendel, F. J., Cheng, Y. S., Otvos, J. D. and Wehrli, S. (1988) Characterization and
30 Kabsch, W. (2010) XDS. Acta Crystallogr. D Biol. Crystallogr. 66, 125–132 CrossRef chemical properties of phosphoribosylamine, an unstable intermediate in the de novo
31 Evans, P. (2006) Scaling and assessment of data quality. Acta Crystallogr. D Biol. purine biosynthetic pathway. Biochemistry 27, 2614–2623 CrossRef
Crystallogr. 62, 72–82 CrossRef 49 Yanofsky, C., Platt, T., Crawford, I. P., Nichols, B. P., Christie, G. E., Horowitz, H.,
32 Otwinowski, Z. and Minor, W. (1997) Processing of X-ray diffraction data collected in VanCleemput, M. and Wu, A. M. (1981) The complete nucleotide sequence of the
oscillation mode. Methods Enzymol. 276, 307–326 CrossRef tryptophan operon of Escherichia coli . Nucleic Acids Res. 9, 6647–6668 CrossRef
33 Vagin, A. and Teplyakov, A. (2010) Molecular replacement with MOLREP. Acta Crystallogr. 50 Hommel, U., Lustig, A. and Kirschner, K. (1989) Purification and characterization of
D Biol. Crystallogr. 66, 22–25 CrossRef yeast anthranilate phosphoribosyltransferase. Eur. J. Biochem. 180, 33–40 CrossRef

Received 13 February 2014/7 April 2014; accepted 9 April 2014


Published as BJ Immediate Publication 9 April 2014, doi:10.1042/BJ20140209


c The Authors Journal compilation 
c 2014 Biochemical Society
Biochem. J. (2014) 461, 87–98 (Printed in Great Britain) doi:10.1042/BJ20140209

SUPPLEMENTARY ONLINE DATA


Alternative substrates reveal catalytic cycle and key binding events in the
reaction catalysed by anthranilate phosphoribosyltransferase from
Mycobacterium tuberculosis
Tammie V. M. COOKSON*1 , Alina CASTELL†1, 2 , Esther M. M. BULLOCH†, Genevieve L. EVANS†, Francesca L. SHORT†3 ,
Edward N. BAKER†, J. Shaun LOTT†4 and Emily J. PARKER*4
*Maurice Wilkins Centre for Molecular Biodiscovery, Biomolecular Interaction Centre and Department of Chemistry, University of Canterbury, 20 Kirkwood Avenue, Christchurch 8140,
New Zealand
†Maurice Wilkins Centre for Molecular Biodiscovery and School of Biological Sciences, University of Auckland, 3 Symonds Street, Auckland 1142, New Zealand

Figures S1–S9 and Tables S1–S3 appear on the following pages.

1
These authors contributed equally to this work.
2
Present address: Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, 171 77 Stockholm, Sweden.
3
Present address: Division of Molecular Microbiology, College of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, U.K.
4
Correspondence may be addressed to either of these authors (email s.lott@auckland.ac.nz or emily.parker@canterbury.ac.nz).
The structural co-ordinates reported for Mycobacterium tuberculosis anthranilate phosphoribosyltransferase complexed with anthranilate or anthranilate
analogues have been deposited in the PDB under codes 4N5V, 4N8Q, 4N93, 4OWM, 4OWN, 4OWO, 4OWQ, 4OWS, 4OWU and 4OWV.


c The Authors Journal compilation 
c 2014 Biochemical Society
T. V. M. Cookson and others

Figure S1 Michaelis–Menten graphs for calculation of K m values for alternative substrates of Mtb -AnPRT
Results were obtained using the EnzChek® Pyrophosphate Assay Kit with PRPP held at a saturating concentration of 120 μM. Reactions were initiated with the addition of anthranilate/alternative
substrate. Results were plotted using GraFit 5.


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase

Figure S2 Substrate inhibition graphs for calculation of K i values for alternative substrates of Mtb -AnPRT
Results were obtained using the EnzChek® Pyrophosphate Assay Kit with PRPP held at a saturating concentration of 120 μM. Reactions were initiated with the addition of anthranilate/alternative
substrate. Results were plotted using GraFit 5.


c The Authors Journal compilation 
c 2014 Biochemical Society
T. V. M. Cookson and others

Figure S3 Monitoring reaction catalysed by Mtb -AnPRT using 1 H-NMR spectroscopy

(A) Spectrum of assay solution containing α-PRPP (2.5 mM) and anthranilate (1 mM) at pH 8 before incubation with Mtb -AnPRT. Note that the commercially obtained α-PRPP also contains β-PRPP
as an impurity, which is not a substrate for Mtb -AnPRT. (B) Spectrum of assay solution following incubation with Mtb -AnPRT (0.8 μM) at 25 ◦ C for 10 min. The substrates PRPP and anthranilate
have been consumed, and both α-PRA and β-PRA are present at an approximate 7:3 molar ratio. Chemical shifts are given relative to TSP (δ 0 p.p.m.). Full details of the NMR assay composition
and collection of 1 H-NMR spectra are given in the Experimental section of the main text.

Figure S4 Epimerization of the Mtb -AnPRT product PRA via an imine intermediate and its reduction to the open-chain form


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase

1
Figure S5 H NMR spectra for the production of substituted PRAs from anthranilate analogues
The upper spectrum of each panel shows the anthranilate analogue (1 mM) and PRPP (2 mM) before the addition of Mtb -AnPRT. The lower 1 H-NMR spectrum is following incubation of this sample
with 0.2 μM Mtb -AnPRT for 20 min at 25 ◦ C. Full details of the NMR assays are given in the Experimental section of the main text.

c The Authors Journal compilation 
c 2014 Biochemical Society
T. V. M. Cookson and others

Figure S6 Decomposition of PRA and PRA analogues as followed by 1 H-


NMR spectroscopy
PRPP (2.5 mM) and anthranilate or analogue (1 mM) was incubated with 0.8 μM Mtb -AnPRT to
form PRA or PRA analogue. Following consumption of the substrates, the rate of decomposition
of PRA or PRA analogue was monitored at 25 ◦ C and at pH 8 by collecting 1 H-NMR spectra at
1.5–5 min intervals. Rates of degradation were measured on the basis of peak integrals for H8 of
PRA (α-PRA and β-PRA combined), or analogue, relative to an internal TSP standard. Results
were fitted to a first-order exponential decay curve to determine the half-life of each compound.
It has been noted previously that PRA undergoes hydrolysis to yield ribose 5-phosphate and
anthranilate in a first-order exponential decay process with a half-life of 450 s at 25 ◦ C and
pH 7.5 [3]. By following the rate of decrease in the 1 H-NMR resonance of H8 of PRA, after
the consumption of all the PRPP substrate, a half-life of 580 + − 10 s for PRA was determined
under the conditions of our 1 H-NMR spectroscopy assay at 25 ◦ C and pH 8. This analysis was
repeated for 4F-PRA and sulfonyl-PRA, which are formed from the action of Mtb -AnPRT on 4FA
and orthanilate respectively. Both of these analogues are substantially stabilized relative to native
PRA, with half-lives of 2960 + +
− 50 and 6610 − 30 s determined for 4F-PRA and sulfonyl-PRA
respectively.

Figure S7 Wavelength scans of the Mtb -AnPRT/Eco -PRAI–InGPS reaction


for fluoro-/methyl-anthranilate with PRPP
The absorbance can be seen to increase in the range 260–290 nm for each substrate. Solutions
for analysis of FA contained 1 mM MgCl2 , 0.1 μM Mtb -AnPRT, 1.7 μM Eco -PRAI–InGPS,
0.6 mM PRPP and 100 μM FA (3FA, 4FA, 5FA or 6FA). Solutions for analysis of MA contained
1 mM MgCl2 , 30 μM Mtb -AnPRT, 150 μM Eco -PRAI–InGPS, 0.6 mM PRPP and 0.5 mM MA
(5MA or 6MA). Reactions were scanned from 360 to 220 nm with a cycle length time of 2.5 min.
Solutions were thermally equilibrated for 5 min before initiation with alternative substrate and
either allowed to go to completion or to a maximum of 50 cycles.


c The Authors Journal compilation 
c 2014 Biochemical Society
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase

Figure S8 Turnover of PRA analogues by Eco-PRAI–InGPS monitored by 1 H-NMR spectroscopy


(A) 4F-PRA was formed by the incubation of 4FA (1 mM) and PRPP (2.5 mM) with Mtb -AnPRT (0.8 μM). (B) Addition of Eco -PRAI–InGPS (2.5 μM) to this 4FA-PRA sample resulted in
complete conversion into 4-fluoro-InGP. δ 7.76 (dd, 5.5, 8.8 Hz, H6), 7.42 (s, H1 ), 7.25 (dd, 10.1, 2.2 Hz, H3), 6.98 (ddd, 2.2, 9.4 Hz, H5), 5.06 (d, 8.2 Hz, H3 ). (C) Sulfonyl-PRA was
formed by the incubation of orthanilate (0.5 mM) and PRPP (2.5 mM) with Mtb -AnPRT (0.8 μM). (D) Addition of Eco -PRAI–InGPS (2.5 μM) to this sulfonyl-PRA sample resulted in the
production of a new set of minor resonances as indicates by the broken arrows (δ 7.70, 7.42, 6.84 and 6.74 p.p.m.). We speculate that these new resonances may correspond to the CdRP
[1-(o -carboxyphenylamino)-1 -deoxyribulose-5 -phosphate] analogue 1-(o -sulfonylphenylamino)-1-deoxyribulose-5-phosphate, formed from isomerization of sulfonyl-PRA by PRAI. Samples were
incubated at 25 ◦ C for approximately 10 min following the addition of each enzyme. Peaks marked by an asterisk result from an imidazole impurity present in the PRAI–InGPS preparation used for
these experiments. Full details of NMR assay composition and collection of 1 H-NMR spectra are given in the Experimental section of the main text.

1
Figure S9 H-NMR spectrum of InGP
PRA was formed by the incubation of anthranilate (1 mM) and PRPP (2.5 mM) with Mtb -AnPRT (0.8 μM). Addition of Eco -PRAI–InGPS (2.5 μM) and incubation at 25 ◦ C for approximately 10 min
resulted in complete conversion into InGP. Full details of NMR assay composition and collection of 1 H-NMR spectra are given in the Experimental section of the main text. δ 7.83 (d, 8 Hz, H6), 7.54
(d, 8.2 Hz, H3), 7.46 (s, H1 ), 7.27 (dd, 7.6 Hz, H4), 7.19 (dd, 7.5 Hz, H5), 5.10 (d, 8.2 Hz, H3 ).


c The Authors Journal compilation 
c 2014 Biochemical Society

c The Authors Journal compilation 

T. V. M. Cookson and others


Table S1 Data and refinement statistics for all complexes
Values for the outer resolution shell are shown in parentheses. R merge =  hklI (hkl ) − <I (hkl )>/ hklI (hkl ), where <I (hkl )> is the mean of the symmetry equivalent reflections of I (hkl ). The B factor is the average atomic temperature factor. R work =
|F o | − |F c |/|F o |. R free =  T |F o | − |F c |/ T |F o | (where T is a test dataset of 5 % of the total reflections randomly chosen and set aside before refinement). Ramachandran outliers were calculated using MolProbity [1]. Ideal RMSD values from Engh and Huber
[2]. NA, not applicable.
c 2014 Biochemical Society

4FA 4FA 6MA 3FA 5FA 6FA 3MA 4MA 5MA Anthranilate

Soak time 6 min 6s 30 min 2 min 5 min 5 min 2 min 2 min 4 min 4s
Data collection
PDB code 4N5V 4N8Q 4N93 4OWM 4OWN 4OWO 4OWQ 4OWS 4OWU 4OWV
Space group P 21 2 1 2 1 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21 P 21 21 21
Cell dimensions
a , b , c (Å) 79.6, 92.5, 121.1 79.6, 91.0, 120.0 79.6, 92.2, 121.4 79.3, 92.0, 120.9 79.6, 92.3, 120.8 79.4, 92.1, 121.3 79.4, 92.3, 121.7 79.7, 91.8, 120.6 79.5, 91.8, 120.8 79.6, 91.7,
120.4
α, β, γ (◦ ) 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90 90, 90, 90
Number of 71077 52754 58211 52873 52044 61270 71647 34257 71474 69457
unique
reflections
Resolution 73.52–1.90 120.0–2.08 121.4–2.03 120.9–1.99 120.8–2.11 121.3–1.99 121.7–1.89 120.6–2.43 120.8–1.89 120.4–1.90
range (Å) (1.95– 1.90) (2.20–2.08) (2.14–2.03) (2.10–1.99) (2.22–2.11) (2.10–1.99) (2.00–1.89) (2.56–2.43) (1.99–1.89) (2.00–1.90)
R merge 0.041 (0.483) 0.031 (0.203) 0.039 (0.233) 0.085 (0.485) 0.084 (0.496) 0.102 (0.494) 0.072 (0.480) 0.087 (0.495) 0.065 (0.485) 0.153 (1.047)
I /σ (I ) 14.7 (1.8) 17.2 (3.9) 17.2 (3.7) 12.2 (2.8) 17.9 (3.9) 14.0 (3.5) 17.8 (3.5) 17.6 (3.9) 20.6 (3.8) 8.2 (1.8)
Completeness 99.9 (100) 100 (99.9) 100 (100) 86.9 (90.0) 100 (100) 99.8 (99.4) 100 (100) 100 (100) 100 (100) 99.3 (99.7)
(%)
Multiplicity 2.0 (1.00) 2.0 (2.0) 2.0 (2.0) 4.6 (4.5) 7.2 (7.4) 7.3 (7.4) 7.3 (7.1) 7.2 (7.4) 7.2 (6.9) 5.0 (4.8)
Refinement
Number of
atoms, B
factor (Å2 )
Protein 5093, 25.7 5045, 33.4 5028, 22.7 5020, 21.8 4960, 25.7 5014, 19.8 4993, 21.9 4892, 33.4 4989, 21.4 4963, 19.9
Solvent 514, 35.1 294, 36.3 545, 32.9 453, 33.4 404, 35.8 513, 33.3 510, 34.5 204, 38.1 516, 35.1 485, 31.4
PRPP 44, 26.7 NA 44, 28.5 44, 35.4 22, 40.1 44, 27.0 44, 29.7 NA 44, 28.1 NA
Mg2 + 4, 29.3 NA 4, 27.2 4, 27.6 4, 28.2 4, 22.8 3, 26.4 NA 4, 22.9 NA
Analogue 44, 23.1 22, 47.0 44, 42.0 22, 43.1 44, 43.1 44, 27.2 22, 34.9 22, 34.7 22, 35.3 20, 46.4
Pyrophosphate NA NA NA NA 9, 27.8 NA NA 18, 64.0 NA NA
R work /R free 18.4/21.8 19.9/23.4 18.1/22.1 17.2/21.3 17.0/19.4 16.3/19.3 16.9/19.2 18.1/22.7 16.5/19.5 19.5/22.1
(%/%) (30.7/30.5) (23.3/28.7) (21.7/27.8) (24.2/28.1) (21.8/23.6) (22.5/26.4) (24.5/26.8) (23.8/32.4) (23.3/26.6) (40.6/43.7)
Ramachandran 0.29 0.59 0.29 0.29 0.29 0.29 0.29 0.30 0.29 0.29
outliers (%)
RMSD
Bond lengths 0.014 0.015 0.014 0.010 0.009 0.009 0.009 0.011 0.009 0.010
(Å)
Bond angles (◦ ) 1.417 1.476 1.462 1.454 1.441 1.441 1.479 1.469 1.475 1.436
Alternative substrates of Mycobacterium tuberculosis anthranilate phosphoribosyltransferase

Table S2 Chemical shift assignment of 1 H-NMR spectra of PRA and PRA analogues produced by Mtb -AnPRT
Chemical shifts are given relative to TSP (δ 0 p.p.m.). Full details of NMR assay composition and collection of 1 H NMR spectra are given in the Experimental section of the main text. ND, not detected;
NA, not applicable.

Assignment of chemical shifts (δ, p.p.m.) PRA 3M-PRA 4M-PRA 5M-PRA 6M-PRA 3F-PRA 4F-PRA 5F-PRA 6F-PRA Sulfonyl-PRA

H1 a 5.70 ND 5.67 5.66 5.60 5.71 5.62 5.65 5.62 5.69
H1 b 5.31 ND 5.29 5.28 5.23 5.39 5.24 5.25 5.25 5.32
H3a 7.05* ND 6.89 6.95* 6.85 NA 6.76* 7.02* 6.82 7.12*
H3b 6.88 6.84 6.81
H4a 7.41* ND NA 7.25* 7.16* 7.22* NA 7.17* 7.27* 7.47*
H4b
H5a 6.86* ND 6.70* NA 6.75 6.93* 6.55* NA 6.61* 6.93*
H5b 6.77
H6a 7.80* ND 7.71* 7.63 NA 7.55 7.83* 7.51 NA 7.73*
H6b 7.61 7.60
α-H1 a coupling constant (Hz) 3.7 ND 4.0 3.8 3.6 3.8 3.9 3.6 4.1 3.6
β-H1 coupling constant (Hz) 6.6 ND 6.4 6.4 6.9 5.4 6.7 6.8 6.9 6.7
Approximate ratio α/β 7:3 ND 6:4 7:3 7:3 7:3 7:3 7:3 7:3 8:2
*Peaks from the two epimers of PRA closely overlap preventing unique assignment. For PRA and the majority of PRA analogues, there was significant overlap in the aromatic peaks of the two
epimers, preventing unique assignment of these resonances to an epimer and complicating analysis of peak types, integrals and coupling constants in this region.

Table S3 Mass of substituted InGP products arising from turnover of anthranilate analogues by Mtb -AnPRT and Eco -PRAI–InGPS
Solutions contained 1.2 mM PRPP, 0.6 μM Mtb -AnPRT, 6 μM Eco -PRAI–InGPS, 2 mM MgCl2 and either 400 μM fluoro-anthranilate or 2 mM methyl-anthranilate. − indicates that the expected
product could not be observed.

Substrate Formula of product Expected mass of product (g/mol) Observed mass of product (g/mol)

Anthranilate C11 H14 NO6 PNa 310.0451 310.0441


6FA C11 H13 FNO6 PNa 328.0357 328.0348
5FA C11 H13 FNO6 PNa 328.0357 328.0349
4FA C11 H13 FNO6 PNa 328.0357 328.0347
3FA C11 H13 FNO6 PNa 328.0357 328.0350
6MA C12 H16 NO6 PNa 324.0607 324.0597
5MA C12 H16 NO6 PNa 324.0607 324.0600
4MA C12 H16 NO6 PNa 324.0607 324.0605
3MA C12 H16 NO6 PNa 324.0607 –

REFERENCES
1 Chen, V. B., Arendall, W. B., Headd, J. J., Keedy, D. A., Immormino, R. M., Kapral, G. J.,
Murray, L. W., Richardson, J. S. and Richardson, D. C. (2010) MolProbity: all-atom
structure validation for macromolecular crystallography. Acta Crystallogr. D Biol.
Crystallogr. 66, 12–21 CrossRef PubMed
2 Engh, R. A. and Huber, R. (1991) Accurate bond and angle parameters for X-ray
protein-structure refinement. Acta Crystallogr. A 47, 392–400 CrossRef
3 Sterner, R., Kleemann, G. R., Szadkowski, H., Lustig, A., Hennig, M. and Kirschner, K.
(1996) Phosphoribosyl anthranilate isomerase from Thermotoga maritima is an
extremely stable and active homodimer. Protein Sci. 5, 2000–2008 CrossRef PubMed

Received 13 February 2014/7 April 2014; accepted 9 April 2014


Published as BJ Immediate Publication 9 April 2014, doi:10.1042/BJ20140209


c The Authors Journal compilation 
c 2014 Biochemical Society

You might also like