You are on page 1of 7

European Journal of Medicinal Chemistry 60 (2013) 333e339

Contents lists available at SciVerse ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Original article

Identification of novel antimycobacterial chemical agents through the


in silico multi-conformational structure-based drug screening of
a large-scale chemical library
Yuji Koseki a, Tomohiro Kinjo a, Maiko Kobayashi a, Shunsuke Aoki a, b, *
a
Department of Bioscience and Bioinformatics, Graduate School of Computer Science and Systems Engineering, Kyushu Institute of Technology, 680-4 Kawazu, Iizuka-shi,
Fukuoka 820-8502, Japan
b
Biomedical Informatics Research and Development Center (BMIRC), Kyushu Institute of Technology, 680-4 Kawazu, Iizuka-shi, Fukuoka 820-8502, Japan

a r t i c l e i n f o a b s t r a c t

Article history: The increasing prevalence of drug-resistant tuberculosis, which is resistant to effective multiple
Received 1 October 2012 antibiotic, presents a major global health threat. The thymidine monophosphate kinase (TMPK) of
Received in revised form Mycobacterium tuberculosis (M. tuberculosis), which is an essential enzyme for the maintenance of the
3 December 2012
thymidine triphosphate pools, is considered an attractive target for the development of effective anti-
Accepted 7 December 2012
Available online 20 December 2012
biotics against tuberculosis. In this study, we attempted to identify novel chemical compounds that
specifically target the M. tuberculosis TMPK (mtTMPK). We performed in silico structure-based drug
screening using the crystal structure data of mtTMPK and a large-scale virtual compound library, which
Keywords:
Antibiotics
is composed of 6,192,930 chemicals. Through a three-step screening method using the DOCK and GOLD,
Mycobacterium we identified ten chemical compounds that were predicted to have high binding affinity to the active site
In silico structure-based drug screening cleft of the mtTMPK. We then evaluated the antibiotic effects of these chemical compounds on model
Thymidine monophosphate kinase (TMPK) mycobacteria strains. As a result, we found that a chemical compound, K10, completely inhibited the
growth of Mycobacterium vanbaalenii (M. vanbaalenii) and Mycobacterium smegmatis (M. smegmatis).
Moreover, K10 does not exhibit any toxic effects on the growth of enterobacteria and mammalian cells.
The structural and experimental information regarding this novel chemical compound, K10, is likely to
be useful for the hit-to-lead optimization of new antibiotics for the treatment of tuberculosis.
Ó 2012 Elsevier Masson SAS. All rights reserved.

1. Introduction is often incomplete, which has resulted in increasing prevalences of


multidrug-resistant TB (MDR-TB), extensively drug-resistant TB
Mycobacterium tuberculosis is the causative agent of human (XDR-TB) and totally drug-resistant TB (TDR-TB) in many areas
tuberculosis (TB), which is one of the major infectious diseases and around the world [9e11]. Therefore, the discovery of new anti-TB
causes approximately 1.8 million deaths every year worldwide. drugs is important for the treatment against the increasing cases
These deaths also include a large number of human immunodefi- of MDR-, XDR- and TDR-TB.
ciency virus (HIV) co-infected patients [1], which makes sense The anti-TB drugs in current development target various biolog-
because HIV infection is known to be a deadly synergistic factor for ical pathways, such as mycobacterial cell wall synthesis, DNA/RNA
TB [2]. The Bacille Calmette-Guérin (BCG) vaccine shows protective synthesis and protein synthesis [12,13]. The M. tuberculosis thymi-
effects against childhood TB. However, the protective efficacy of the dine monophosphate kinase (mtTMPK, EC 2.7.4.9) is proposed as an
BCG vaccine is limited and incomplete against adult TB [3e5]. In attractive target for the treatment of TB [14]. mtTMPK catalyses the
most cases, TB is currently treated with a cocktail of four first-line reversible phosphorylation of deoxythymidine 50 -monophosphate
drugs, which include isoniazid, rifampin, ethambutol and strepto- (dTMP) to deoxythymidine 50 -diphosphate in the presence of Mg2þ
mycin, for at least six months [6e8]. However, the drug compliance and adenosine triphosphate (ATP). It is a crucial enzyme in both the
de novo and the salvage pathways of DNA synthesis for bacterial
growth [15e17]. Previously, several research groups have reported
* Corresponding author. Department of Bioscience and Bioinformatics, Graduate the successful development of nucleotide analogue inhibitors of
School of Computer Science and Systems Engineering, Kyushu Institute of Tech-
nology, 680-4 Kawazu, Iizuka-shi, Fukuoka 820-8502, Japan. Tel.: þ81 948 29 7819;
mtTMPK [18,19]. Therefore, mtTMPK is considered one of the
fax: þ81 948 29 7801. significant targets for the development of novel TB therapeutic
E-mail address: aokis@bio.kyutech.ac.jp (S. Aoki). chemical compounds.

0223-5234/$ e see front matter Ó 2012 Elsevier Masson SAS. All rights reserved.
http://dx.doi.org/10.1016/j.ejmech.2012.12.012
334 Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339

mammalian cells. The structural and experimental information of


the active compound K10 will contribute to the development of
anti-TB medical drugs.

2. Results

2.1. Molecular docking and scoring of putative binding affinities

We performed a three-step screening process using DOCK (first


screening) and GOLD (single- and multi-conformation screening)
to identify chemicals that potentially bind to the active site cleft of
mtTMPK. The three-dimensional structure of mtTMPK is charac-
terized by nine a-helices (H1eH9) and five b-sheets (S1eS5), which
is also called the Rossmann fold [20,21]. We focused on the
following 14 amino acid residues: Asp 9, Arg 14, Phe 36, Pro 37, Tyr
39, His 53, Phe 70, Asp 94, Arg 95, Asn 100, Tyr 103, Arg 153, Ala 161
and Asp 163 (Fig. 1) [17,18,21]. All of these residues are located in
the active site cleft of mtTMPK and comprise the phosphate binding
loop (P-loop) and phosphate donor binding site (LID region) [21].
From a constructed structural database of 6,192,930 chemicals, the
DOCK screening identified 43,932 chemicals that are potentially
capable of binding to the active site cleft with an estimated binding
free energy of less than 45 kcal/mol. This cut-off was chosen
because nucleotide analogue inhibitors of mtTMPK [18,22] have
estimated binding free energies of approximately 45 kcal/mol.
Following the DOCK screening, we performed a GOLD screening
using a single conformation per chemical and identified the top
3100 chemical compounds. During the first GOLD screening
(single-conformation GOLD screening), we initially identified the
Fig. 1. The three-dimensional structure of M. tuberculosis TMPK (mtTMPK). The top 1000 ranked chemical compounds with GOLD scores of more
secondary structures are indicated by ribbon representations (a-helix: orange, b-sheet:
than 70 because our previous research demonstrated that
green). The amino acid residues of the active site and Mg2þ ion are shown as a stick
model and a sphere model, respectively. (For interpretation of the references to colour compounds with GOLD scores of over 60 can affect enzyme activity
in this figure legend, the reader is referred to the web version of this article.) at concentrations lower than several hundred mM [23,24]. A
previous study has suggested that the accuracy of docking simu-
lations with multiple conformers is improved relative to the
In this study, we used a three-step hierarchical in silico structure- accuracy of simulations with single conformers [25]. We then
based drug screening (SBDS) to screen for chemical compounds that performed a second GOLD screening using multiple conformations
are potentially able to bind to the active site of mtTMPK. After per chemical structure (five structural conformations for each
making careful predictions of proteineligand interactions using compound). To remove the already evaluated chemicals, we elim-
multi-conformational data of the chemical compounds, we selected inated several chemical compounds from the previously identified
ten compounds that exhibited average GOLD scores over 70 from the 1000 compounds according to the bioactivity assay results in the
large-scale virtual chemical compounds library (6,192,938 chem- PubChem database [26]. The elimination of chemical compounds
icals) that was analysed. Moreover, we experimentally demonstrated was based on the following three criteria: (1) the compound was
that four compounds have antibiotic effects against Mycobacterium not identified through high-throughput screening, (2) the
vanbaalenii that are similar to those of isoniazid. Among them, three compound did not have potentially toxic effects (i.e., cytotoxicity),
chemical compounds had strong inhibitory effects on the growth of and (3) the compound had one or more direct hydrogen bond
the model mycobacteria strains. In addition, two of the active (H-bond) interactions with the active site. Among the remaining
compounds did not exhibit any significant toxic effects on the chemical compounds with average GOLD scores of over 70, we
growth of Escherichia coli (E.coli) and the viability of cultured selected ten chemical compounds, K1eK10 (Table 1), that form

Table 1
List of the ten selected chemical compounds that were identified by the in silico screen.

No. Compound name GOLD score

Single conf. Multiple conf.a


K1 N-{2-[4-(aminosulfonyl)phenyl]ethyl}-2-(3-isopropoxyphenyl)-4-quinolinecarboxamide 86.33 88.17  0.95
K2 N,N0 -bis(4-methylphenyl)-6-[(5-methyl-1,3,4-thiadiazol-2-yl)thio]-1,3,5-triazine-2,4-diamine 78.63 85.98  0.21
K3 N-[4-(benzyloxy)-phenyl]-1-(4-nitrobenzoyl)-4-piperidinecarboxamide 75.58 84.56  0.28
K4 N-[2({2-[(4-ethoxyphenyl)amino]-2-oxoethyl}thio)-1,3-benzothiazol-6-yl]-2-methylbenzamide 76.28 84.23  0.11
K5 benzyl [(6-chloro-2-oxo-4-propyl-2H-chromen-7-yl)oxy]acetate 80.23 79.85  0.16
K6 2-(4-chloro-2-methylphenoxy)-N-[2-methoxy-5-(2-oxo-2H-chromen-3-yl)phenyl]acetamide 73.00 79.14  0.49
K7 benzyl [(6-chloro-4-ethyl-2-oxo-2H-chromen-7-yl)oxy]acetate 70.91 76.00  0.33
K8 methyl {4-[(4-tert-butylbenzoyl)amino]phenoxy}acetate 74.31 74.79  0.18
K9 ethyl [(4-butyl-6-chloro-2-oxo-2H-chromen-7-yl)-oxy]acetate 72.67 74.53  0.45
K10 8-quinolinyl 4-[(4-chlorophenyl)sulfonyl]benzoate 74.00 71.92  0.47
a
Each value represents the mean  SEM.
Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339 335

Fig. 2. General flowchart for the three-step in silico SBDS used to identify novel
antimycobacterial compounds targeting mtTMPK.

H-bond interactions with at least one of the target amino acids


(Fig. 2). Furthermore, the relationship between the kinetic param-
eter of mtTMPK inhibitors (Ki values ¼ 7.2e69 mM) [18,22] and the
GOLD score was verified by the in silico docking study. As a result,
the correlation coefficients (r2) for the correlations between the Ki
values and the single- and multi-conformational GOLD scores were Fig. 3. Inhibitory effects of the candidate chemicals (K1eK10) on the growth of the
0.76 and 0.80, respectively (Table 2). model mycobacteria: (A) M. vanbaalenii and (B) M. smegmatis. The concentrations of all
of the chemicals were (A) 50 mM or (B) 100 mM. DMSO (0.3%) and isoniazid (50/
100 mM) were used as the negative and positive controls, respectively. Each value
2.2. In vitro antimycobacterial activity assay represents the mean  SEM of four independent experiments. Dunnett’s multiple
comparison test was performed. The statistical significance is indicated in all of the
figures (**p < 0.01, n.s. not significant).
We then performed antimycobacterial activity assays to confirm
that the ten chemicals (K1eK10) are actually able to exhibit
inhibitory effects on the growth of two model mycobacteria:
M. vanbaalenii and Mycobacterium smegmatis. These model bacteria We also examined the effects of the four chemicals (K1, K2, K7 and
were utilized because of their amino acid similarities to K10) on M. smegmatis growth. After 24 h, K1 and K2 (100 mM)
M. tuberculosis [TMPK of M. vanbaalenii (96%) and M. smegmatis moderately inhibited the growth of M. smegmatis compared with
(97%)] according to the NCBI protein blast search [27] and because the positive control (isoniazid). Similarly to isoniazid, K10 (100 mM)
these strains are harmless (biosafety level 1) [28]. The inhibitory completely inhibited the growth of M. smegmatis, whereas K7
effects of the ten candidate chemicals that were identified through (100 mM) has no inhibitory effect on the growth of M. smegmatis
the hierarchical in silico SBDS on the growth of M. vanbaalenii were (Fig. 3B). The chemical structure and physicochemical properties of
tested. K1, K2, K7 and K10 (all at concentrations of 50 mM) signifi- the active hits are indicated in Fig. 4A and Table 3, respectively.
cantly inhibited the growth of M. vanbaalenii compared with the Furthermore, we investigated the dose-dependent effects of the
negative control (DMSO). In addition, after 18 h, these effects were active hits on the growth of M. vanbaalenii to determine the IC50
similar to or higher than those obtained with the positive control values. The experimentally determined IC50 values of K1, K2 and
(isoniazid). In contrast, the six other compounds (K3eK6, K8 and K10 for M. vanbaalenii were 15 mM, 22 mM and 20 mM, respectively
K9) had no significant or weak inhibitory effects on M. vanbaalenii (Fig. 4BeD). However, these values are higher than the IC50 value of
growth compared with the negative and positive controls (Fig. 3A). isoniazid for M. vanbaalenii (2.6 mM; data not shown).

Table 2
Evaluation of the relationship between the GOLD score and the kinetic parameter (Ki) for nucleotide analogue inhibitors of mtTMPK and isoniazid-NAD.

Compound name Ki (mM)a GOLD score

Single conf. Multiple conf.b


N-[(30 -deoxythymidine-30 -yl)methyl]-N0 -phenylthiourea 69.0 73.81 78.78  0.33
N-[(30 -deoxythymidine-30 -yl)methyl]-N0 -(4-methylphenyl)thiourea 36.0 75.57 82.16  0.36
N-(4-chlorophenyl)-N0 -[(30 -deoxythymidine-30 -yl)methyl]thiourea 21.0 76.89 82.63  0.77
1-[3-(azidomethyl)-2,3-dideoxy-5-O-phosphoryl-b-D-erythro-pentofuranosyl]thymine 12.0 78.41 83.94  0.35
1-[3-(aminomethyl)-2,3-dideoxy-5-O-phosphoryl-b-D-erythro-pentofuranosyl]thymine 10.5 76.67 85.30  0.17
30 -azide-30 -deoxythymidine monophosphate 10.0 80.05 84.62  0.52
N-(3,4-dichlorophenyl)-N0 -[(30 -deoxythymidine-30 -yl)methyl]thiourea 7.2 78.77 84.72  0.53
Isoniazid-NAD (INH-NAD)c 7.5  104 107.43 127.10  2.99
a
The Ki values were based on previously published data [18,22,49].
b
Each value represents the mean  SEM.
c
INH-NAD inhibits enoyleacyl carrier protein reductase (mtInhA) [6,42].
336 Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339

Fig. 4. Chemical structural formula of the active hits (A) and dose-dependent effects of the active hits (K1, K2 and K10) on the growth of M. vanbaalenii: (B) K1, (C) K2 and (D) K10.
The bacterial growth rate (%) was determined through a 24 h cultivation of M. vanbaalenii. Each plotted value represents the mean  SD of four independent experiments.

2.3. Enterobacteria and mammalian cell toxicity assay H-bond interactions with Asp 9, Arg 14, Asp 94 and Arg 95. Moreover,
the 4-methylphenyl ring in K2 forms an areneecation interaction
We performed chemical compound toxicity assays to confirm with Arg 153 (Fig. 6B). The sulfonyl and benzoate in K10 form H-bond
that the chemical compounds do not have toxic effects on both interactions with His 53 and Arg 95. The 4-chlorophenyl and
a model enterobacteria (E. coli) and mammalian, MadineDarby 8-quinolinyl rings in K10 additionally form areneearene interactions
Canine Kidney (MDCK) and human neuroblastoma (SH-SY5Y) with Tyr 39 and Phe 70, respectively (Fig. 6C).
cells. The active hit chemicals identified by the antimycobacterial
assays were investigated using E. coli growth and mammalian cell 3. Discussion
cytotoxicity assays. All of the compounds showed no toxic effect on
the growth of E. coli BL21 (data not shown) and JM 109 (Fig. 5A). K1 Using a three-step in silico SBDS (DOCK, single-conformation
and K10 showed no significant toxic effects on cultured MDCK and GOLD and multi-conformation GOLD) of 6,192,930 chemical
SH-SY5Y cells, whereas compound K2 had significant toxic effects compounds, we identified ten candidate compounds that could
on these mammalian cells (Fig. 5B and C). potentially inhibit mtTMPK activity. We then evaluated the actual
inhibitory effects of the compounds on the growth of model
2.4. Predicted binding modes of active hit chemical compounds mycobacteria and identified four active compounds (compound K1,
K2, K7 and K10) that were able to inhibit M. vanbaalenii growth.
A proteineligand interaction fingerprint (PLIF) analysis was per- Two chemicals (K1 and K2) also exhibited moderate inhibitory
formed to better understand the putative interaction between effects on the growth of M. smegmatis. Similarly to isoniazid, K10
mtTMPK and the active chemicals. The predicted best binding modes completely inhibited the growth of both M. vanbaalenii and
of the three active chemicals (K1, K2 and K10) are presented in Fig. 6. M. smegmatis and exhibited no significant toxic effects on E. coli,
All of the chemicals are predicted to be located near the active site MDCK and SH-SY5Y cells. A PLIF analysis of these active chemicals
cleft of mtTMPK. The 3-isopropoxyphenyl ring in K1 forms an arenee showed that all of the chemicals highlighted a key H-bond inter-
cation interaction with Arg 153 and a H-bond interaction with the action with Arg 95, which is found in the active site of mtTMPK [21].
main chain of Ala 161. Moreover, the 4-quinolinecarboxamide in K1 Recently, a study that used live-cell imaging with a microfluidic
forms H-bond interactions with Arg 95 and Arg 153. In addition, the device to culture M. smegmatis demonstrated that the cell division
4-aminosulfonyl ring in K1 forms a hydrophobic interaction with Phe of M. smegmatis is similar to that of M. tuberculosis, which suggests
70 and a H-bond interaction with Asn 100 (Fig. 6A). The 1,3,5-triazine that M. smegmatis is an appropriate model mycobacteria and will
and the (5-methyl-1,3,4-thiadiazol-2-yl)thio rings in K2 form contribute to the development of antibiotics for TB [29]. These
results suggested that the structural information of K10 is useful in
the research and development of novel TB therapeutic chemicals.
Table 3 Several nucleotide analogue inhibitors for mtTMPK have been
Physicochemical properties of the active hit compounds. developed [18,19]. However, these chemicals have low permeability
Chemical Molecular log P H-bond H-bond tPSAa to the mycobacterial cell wall and are cleaved by phosphatase or
name weight acceptor donor phosphorylase [19,30]. In contrast, the chemical K10 is an artificially
K1 490 4.71 5 2 111.4 synthesised chemical that is composed of hydrophobic chemical
K2 422 5.68 5 2 88.5 groups, 8-quinolinyl, (4-chlorophenly)sulfonyl and benzoate. Thus,
K10 424 5.12 5 0 73.3 K10 is expected to have a higher permeability and stability than
a
Topological molecular polar surface area. the nucleotide analogues. According to the structureeactivity
Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339 337

Fig. 5. Toxic effects of the chemicals (K1, K2 and K10) on the model enterobacteria and mammalian cells: (A) E. coli JM 109, (B) MDCK cells and (C) SHeSY5Y cells. The concentration
of all of the chemicals was 50 mM (IC90 value for mycobacteria growth). DMSO (0.3%) and isoniazid (50 mM) were used as the negative and positive controls, respectively. Each value
represents the mean  SEM of four independent experiments. Dunnett’s multiple comparison test was performed (**p < 0.01, n.s. not significant).

Fig. 6. Putative intermolecular interactions between mtTMPK and the hit chemicals by PLIF analysis: (A) K1, (B) K2 and (C) K10. The H-bond, areneearene and areneecation
interactions are indicated by green, blue dotted and blue dashed lines, respectively. (For interpretation of the references to colour in this figure legend, the reader is referred to the
web version of this article.)

relationships (SAR), it is hypothesized that an areneearene biological experiments with ten candidate chemicals that were
interaction with Tyr 39, a H-bond interaction with His 53 and an predicted by the three-step in silico SBDS and successfully identi-
areneearene/hydrophobic interaction with Phe 70 are important for fied three novel potential anti-mycobacteria chemicals (30% hit
the antibiotic effects (Fig. 6). The targeted amino acid residues are rate). Thus, our screening methods could contribute to the further
consistent with a previous structural analysis of the complexes of identification of hit compounds for the development of therapeutic
inhibitors with mtTMPK [17]. Thus, the structural information of the chemicals against not only TB but also other diseases.
chemical compounds identified in this study is likely to be useful in
the design of new antibiotics against TB. 4. Conclusion
Many medical drugs have been developed through the typical
processes, such as the identification of active chemicals using a HTS In the present study, we used a three-step in silico SBDS
method and the optimization of their physicochemical properties approach with a large-scale chemical library to identify ten
through the synthesis of related analogues. However, these chemicals that were predicted to have a high binding affinity to
processes are costly and require much time [31]. In contrast, mtTMPK. The results of the in vitro biological assays indicated
computer-aided drug design, which is based on molecular docking, that one of these chemical compounds, K10, exhibits a potent
pharmacophore modelling, three-dimensional quantitative antimycobacterial effect and no toxicity. Thus, the structural and
structure-activity (3D-QSAR) [32] and molecular dynamics (MD) predicted proteineligand interaction information for this novel
[33], is a shortcut technique that enables a more rapid hit identi- chemical compound, K10, will be helpful for further hit-to-lead
fication than HTS [34]. Several research groups have suggested that optimization of new antibiotics against drug-resistant TB.
a multi-step in silico SBDS is very effective and exhibits reasonable
accuracy [35,36]. In addition, the docking of multiple conforma- 5. Experimental protocols
tions of chemicals into an active site demonstrated that the docking
accuracy was improved compared to the docking of a single 5.1. Chemical compounds library
conformation per chemical [25]. Previously, two novel anti-TB
agents were identified through in silico SBDS using a single For the in silico SBDS, we used a chemical compounds library
conformation of the chemicals (2.5% hit rate) [37]. In this study, we (6,192,930 commercially available compounds) that was derived
developed a novel hierarchical multistep SBDS method, performed from the web-based database ZINC [38]. This compounds library is
338 Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339

ADME/Tox-Drug-Like filtered (Absorption, Distribution, Metabo- K3: N-[4-(benzyloxy)phenyl]-1-(4-nitrobenzoyl)-4-piperidine


lism, Excretion and Toxicity) [39]. The three-dimensional structures carboxamide (16267193);
of mtTMPK inhibitors [18,22] were generated using the Builder K4: N-[2-({2-[(4-ethoxyphenyl)amino]-2-oxoethyl}thio)-1,3-
module in the Molecular Operating Environment (MOE) version benzothiazol-6-yl] -2-methylbenzamide (00680597);
2010.10 [40]. The multi-conformation structural data of the K5: benzyl [(6-chloro-2-oxo-4-propyl-2H-chromen-7-yl)oxy]
compounds was generated using the LowMode MD module in the acetate (01210682);
MOE 2010.10 with the default search parameters: rejection K6: 2-(4-chloro-2-methylphenoxy)-N-[2-methoxy-5-(2-oxo-
limit ¼ 100, iteration limit ¼ 10,000, RMS gradient ¼ 0.005, MM 2H-chromen-3-yl)phenyl] acetamide (01065313);
iteration limit ¼ 500, RMSD limit ¼ 0.25 and conformation K7: benzyl [(6-chloro-4-ethyl-2-oxo-2H-chromen-7-yl)oxy]
limit ¼ 10,000 [40,41]. acetate (01142847);
K8: methyl {4-[(4-tert-butylbenzoyl)amino]phenoxy}acetate
5.2. Preparation of protein structure (00619622);
K9: ethyl [(4-butyl-6-chroro-2-oxo-2H-chromen-7-yl)oxy]
The crystal structure data for mtTMPK (PDB ID: 1MRN) [17] acetate (02479771); and.
and mtInhA (PDB ID: 2IDZ) [42] were obtained from the Research K10: 8-quinolinyl 4-[(4-chlorophenyl)sulfonyl]benzoate
Collaboratory for Structural Bioinformatics Protein Data Bank [43]. (01138659).
These two structure files were based on mtTMPK and mtInhA
complexed with inhibitors, P1-(adenosine-50 )-P5-(thymidine-50 )- 5.5. Bacterial strains
pentaphosphate (Ap5T) and INH-NAD, respectively. Before using
the mtTMPK and mtInhA crystal structures in the in silico SBDS, all M. vanbaalenii and M. smegmatis were obtained from the RIKEN
of the crystal waters and the inhibitor (Ap5T, INH-NAD) were BioResource Center (Saitama, Japan) [28]. E. coli JM 109 and BL 21
removed. In addition, hydrogen atoms were added to mtTMPK and were kindly gifted from Dr. S. Sueda (Kyushu Institute of Technology).
mtInhA using the MMFF94x force field, and the energy was mini-
mized using the Protonate 3D and Energy Minimize modules of 5.6. Bacterial growth assay
MOE 2010.10. As part this process, all of the heavy atoms, except the
hydrogen atoms, were tethered to relieve any short contacts [40]. The bacteria were cultured using modified methods that were
previously described [48]. Briefly, M. vanbaalenii and M. smegmatis
5.3. Hierarchical in silico structure-based drug screening were grown overnight or for 2 days at 300 rpm and 37  C in 3 ml of
culture medium [3.7% Brain heart infusion broth (Difco)]. E. coli was
The hierarchical in silico SBDS was performed using UCSF DOCK grown overnight at 300 rpm and 37  C in 3 ml of liquid culture [1%
version 6.4 [44] and CCDC GOLD suite version 5.0.1 [45]. The three- Tryptone (BD), 0.5% Yeast extract (BD) and 0.5% NaCl (Wako)]. We
step in silico SBDS was conducted using the energy-minimized used a 6 x dilution of the M. vanbaalenii culture, an 8 x dilution of
structure. The proteineligand interaction space was restricted to the M. smegmatis culture and a 10 x dilution of the E. coli culture for
the active site cleft of mtTMPK. For the first screening with DOCK, the respective growth assays. In the assays, the diluted bacteria
we constructed the molecular surface and generated the interac- were seeded in 96-well assay plates (NUNC) and incubated in
tion cleft using the DMS and SPHGEN programs, respectively [44]. culture medium containing one of the chemicals. In the
In the rigid ligand-based DOCK screening, the grid-based energy M. vanbaalenii and M. smegmatis growth assays, 0.3% DMSO was
scoring function, which is composed of van der Waals and elec- used as the negative control and isoniazid (LKT) was used as the
trostatic interaction energies, was applied to estimate the potential positive control. Ampicillin (SIGMA) was used as the positive
binding affinities [44]. Following the screening with rigid condi- control in the E. coli growth assay. After 4, 8, 18 and 24 h, we
tions in DOCK, the protein-chemical binding affinities were pre- measured the absorbance of the culture media at 595 nm with
dicted with the GOLD program, which uses a genetic algorithm a micro plate reader (BioRad).
(GA) method for the docking and conformation search [45,46]. The
docking studies were performed using the default evolutionary 5.7. Mammalian cell toxicity assay
parameters for GOLD (single- and multi-conformation screening):
operations ¼ 100,000, island ¼ 5, migration ¼ 15, mutation ¼ 95 The cytotoxic effect of the chemical compounds on mammalian
and crossover ¼ 95. The ten docking poses for each chemical cells was assayed using the Cell Counting Kit-8 (DOJIN), which was
compound were saved for use in the proteineligand interaction used to measure the number of living cells. The MDCK and SH-SY5Y
analyses. The analysis of the in silico SBDS results was performed cells were seeded in 100-mm dishes and maintained in DMEM
using the proteineligand interaction fingerprint (PLIF) and Ligand medium (Wako) supplemented with 10% FBS (GIBCO), 100 units/ml
Interactions module in MOE 2010.10 [40]. All of the calculations penicillin, 100 mg/ml streptomycin (GIBCO) and 2 mM L-Glutamine
were performed on clusters of personal computers running the (GIBCO). The MDCK and SH-SY5Y cells were seeded in 96-well
Linux operating system. assay plates (CORNING) at 5.0  103 and 1.5  104 cells/well,
respectively, and were incubated 6 h or overnight at 37  C in 5%
5.4. Chemical compounds CO2. For cell starvation, the culture medium of the MDCK and
SH-SY5Y cells was replaced by fresh DMEM medium containing
All of the chemical compounds were purchased from the 0.25% FBS, and the cells were then incubated overnight and for 2
ChemBridge Corporation (San Diego, CA) [47] and dissolved in days, respectively. After cell starvation, the culture medium of the
dimethyl sulfoxide (DMSO, SIGMA) for the in vitro assays. The MDCK and SH-SY5Y cells was replaced by fresh medium (0.25%
chemical compounds (ZINC IDs given in parentheses) used were FBS) containing one of the chemical compounds or isoniazid
the following: (50 mM) as the negative control. The MDCK and SH-SY5Y cells were
K1: N-{2-[4-(aminosulfonyl)phenyl]ethyl}-2-(3-isopropoxyphenyl)- then incubated for 1 day and 2 days, respectively. We then added
4-quinolinecarboxamide (02867600); 10 ml/well of Cell Counting Kit-8, and, after 3 h incubation, we
K2: N,N0 -bis(4-methylphenyl)-6-[(5-methyl-1,3,4-thiadiazol-2-yl) measured the absorbance of WST-8 formazan at 450 nm with
thio]-1,3,5-triazine-2,4-diamine (00708281); a micro plate reader (BioRad).
Y. Koseki et al. / European Journal of Medicinal Chemistry 60 (2013) 333e339 339

5.8. Statistical analysis [21] I.L. de la Sierra, H. Munier-Lehmann, A.M. Gilles, O. Barzu, M. Delarue, X-ray
structure of TMP kinase from Mycobacterium tuberculosis complexed with
TMP at 1.95 angstrom resolution, J. Mol. Biol. 311 (2001) 87e100.
All of the statistical analyses were performed using R version [22] V. Vanheusden, H. Munier-Lehmann, M. Froeyen, L. Dugue, A. Heyerick, D. De
2.15.0 (The R Foundation for Statistical Computing, Vienna, Keukeleire, S. Pochet, R. Busson, P. Herdewijn, S. Van Calenbergh, 3 ’-C-
Austria) and GraphPad Prism version 4 (GraphPad Software, Inc., branched-chain-substituted nucleosides and nucleotides as potent inhibitors
of Mycobacterium tuberculosis thymidine monophosphate kinase, J. Med.
San Diego, CA). Chem. 46 (2003) 3811e3821.
[23] K. Hirayama, S. Aoki, K. Nishikawa, T. Matsumoto, K. Wada, Identification of
novel chemical inhibitors for ubiquitin C-terminal hydrolase-L3 by virtual
Acknowledgements screening, Bioorg. Med. Chem. 15 (2007) 6810e6818.
[24] T. Mitsui, K. Hirayama, S. Aoki, K. Nishikawa, K. Uchida, T. Matsumoto, T. Kabuta,
The authors are very thankful to Dr. S. Fujii (Kyushu Institute of K. Wada, Identification of a novel chemical potentiator and inhibitors of UCH-L1
by in silico drug screening, Neurochem. Int. 56 (2010) 679e686.
Technology) for the valuable comments and Mr. K. Tsuruta (Kyushu [25] N. Sauton, D. Lagorce, B.O. Villoutreix, M.A. Miteva, MS-DOCK: accurate
Institute of Technology) for the excellent technical support. multiple conformation generator and rigid docking protocol for multi-step
virtual ligand screening, BMC Bioinf. 9 (2008) 184.
[26] Q. Li, T. Chen, Y. Wang, S.H. Bryant, PubChem as a public resource for drug
References discovery, Drug Discovery Today 15 (2010) 1052e1057.
[27] M. Johnson, I. Zaretskaya, Y. Raytselis, Y. Merezhuk, S. McGinnis, T.L. Madden,
[1] World Health Organization, Global Tuberculosis Control: WHO Report 2011, NCBIBLAST: a better web interface, Nucleic Acids Res. 36 (2008) W5eW9.
ISBN 978 92 4 156438 0. [28] RIKEN, RIKEN BioResource Center, Japan, <http://www.jcm.riken.go.jp>.
[2] K.H. Mayer, C.D. Hamilton, Synergistic pandemics: confronting the global HIV [29] B.B. Aldridge, M. Fernandez-Suarez, D. Heller, V. Ambravaneswaran, D. Irimia,
and tuberculosis epidemics, Clin. Infect. Dis. 50 (2010) S67eS70. M. Toner, S.M. Fortune, Asymmetry and aging of mycobacterial cells lead to
[3] G.A. Colditz, T.F. Brewer, C.S. Berkey, M.E. Wilson, E. Burdick, H.V. Fineberg, variable growth and antibiotic susceptibility, Science 335 (2012) 100e104.
F. Mosteller, Efficacy of BCG vaccine in the prevention of tuberculosis meta- [30] M.J. Perez-Perez, E.M. Priego, A.I. Hernandez, M.J. Camarasa, J. Balzarini,
analysis of the published literature, JAMA 271 (1994) 698e702. S. Liekens, Thymidine phosphorylase inhibitors: recent developments and
[4] S.H.E. Kaufmann, A.J. McMichael, Annulling a dangerous liaison: vaccination potential therapeutic applications, Mini Rev. Med. Chem. 5 (2005) 1113e1123.
strategies against AIDS and tuberculosis, Nat. Med. 11 (2005) S33eS44. [31] G.M. Keseru, G.M. Makara, Hit discovery and hit-to-lead approaches, Drug
[5] T.H.M. Ottenhoff, S.H.E. Kaufmann, Vaccines against tuberculosis: where are Discovery Today 11 (2006) 831e839.
we and where do we need to go? PLoS Pathog. 8 (2012) 1e12. [32] X.-Y. Lu, Y.-D. Chen, Y.-J. Jiang, Q.-D. You, Discovery of potential new InhA
[6] B.R. Bloom, C.J.L. Murray, Tuberculosis: commentary on a reemergent killer, direct inhibitors based on pharmacophore and 3D-QSAR analysis followed by
Science 257 (1992) 1055e1064. in silico screening, Eur. J. Med. Chem. 44 (2009) 3718e3730.
[7] A.E. Belanger, G.S. Besra, M.E. Ford, K. Mikusova, J.T. Belisle, P.J. Brennan, [33] Y. Koseki, T. Kinjo, M. Kuroki, S. Aoki, Aberrant structures of Parkinson’s
J.M. Inamine, The embAB genes of Mycobacterium avium encode an arabinosyl disease-associated ubiquitin C-terminal hydrolase L1 predicted by molecular
transferase involved in cell wall arabinan biosynthesis that is the target for the dynamics, Chem. Phys. Lett. 535 (2012) 163e168.
antimycobacterial drug ethambutol, Proc. Natl. Acad. Sci.U. S. A. 93 (1996) [34] M. Miteva, Hierarchical structure-based virtual screening for drug design,
11919e11924. Biotechnol. Biotechnol. Equip. 22 (2008) 634e638.
[8] A.P. Carter, W.M. Clemons, D.E. Brodersen, R.J. Morgan-Warren, B.T. Wimberly, [35] T. Pencheva, O.S. Soumana, I. Pajeva, M.A. Miteval, Post-docking virtual
V. Ramakrishnan, Functional insights from the structure of the 30S ribosomal screening of diverse binding pockets: comparative study using DOCK,
subunit and its interactions with antibiotics, Nature 407 (2000) 340e348. AMMOS, X-Score and FRED scoring functions, Eur. J. Med. Chem. 45 (2010)
[9] C. Dye, Global epidemiology of tuberculosis, Lancet 367 (2006) 938e940. 2622e2628.
[10] A.A. Velayati, M.R. Masjedi, P. Farnia, P. Tabarsi, J. Ghanavi, A.H. ZiaZarifi, [36] S. Kalyaanamoorthy, Y.-P.P. Chen, Structure-based drug design to augment hit
S.E. Hoffner, Emergence of new forms of totally drug-resistant tuberculosis discovery, Drug Discovery Today 16 (2011) 741e748.
Bacilli Super extensively drug-resistant tuberculosis or totally drug-resistant [37] M. Muddassar, J.W. Jang, H.S. Gon, Y.S. Cho, E.E. Kim, K.C. Keum, T. Oh,
strains in Iran, Chest 136 (2009) 420e425. S.-N. Cho, A.N. Pae, Identification of novel antitubercular compounds through
[11] N.R. Gandhi, P. Nunn, K. Dheda, H.S. Schaaf, M. Zignol, D. van Soolingen, hybrid virtual screening approach, Bioorg. Med. Chem. 18 (2010) 6914e6921.
P. Jensen, J. Bayona, Multidrug-resistant and extensively drug-resistant [38] J.J. Irwin, B.K. Shoichet, ZINC e a free database of commercially available
tuberculosis: a threat to global control of tuberculosis, Lancet 375 (2010) compounds for virtual screening, J. Chem. Inf. Model. 45 (2005) 177e182.
1830e1843. [39] C.A. Lipinski, Drug-like properties and the causes of poor solubility and poor
[12] Z. Ying, The magic bullets and tuberculosis drug targets, Annu. Rev. Phar- permeability, J. Pharmacol. Toxicological Methods 44 (2000) 235e249.
macol. Toxicol 45 (2005) 529e564. [40] Molecular Operating Environment (MOE), 2010. 10, Chemical Computing
[13] B. Villemagne, C. Crauste, M. Flipo, A.R. Baulard, B. Deprez, N. Willand, Group Inc, 1010 Sherbooke St. West, Suite #910, Montreal, QC, Canada, H3A
Tuberculosis: the drug development pipeline at a glance, Eur. J. Med. Chem. 51 2R7, 2010.
(2012) 1e16. [41] P. Labute, LowModeMD-implicit low-mode velocity filtering applied to
[14] S. Pochet, L. Dugue, G. Labesse, M. Delepierre, H. Munier-Lehmann, Compar- conformational search of macrocycles and protein loops, J. Chem. Inf. Model.
ative study of purine and pyrimidine nucleoside analogues acting on the 50 (2010) 792e800.
thymidylate kinases of Mycobacterium tuberculosis and of humans, Chem- [42] M.V.B. Dias, I.B. Vasconcelos, A.M.X. Prado, V. Fadel, L.A. Basso, W.F. De
biochem 4 (2003) 742e747. Azevedo Jr., D.S. Santos, Crystallographic studies on the binding of isonicotinyl-
[15] M.A. Moors, D.A. Portnoy, Identification of bacterial genes that contribute to NAD adduct to wild-type and isoniazid resistant 2-trans-enoyl-ACP (CoA)
survival and growth in an intracellular environment, Trends Microbiol. (1995) reductase from Mycobacterium tuberculosis, J. Struct. Biol. 159 (2007) 369e380.
83e85. [43] H.M. Berman, J. Westbrook, Z. Feng, G. Gilliland, T.N. Bhat, H. Weissig,
[16] H. Munier-Lehmann, A. Chaffotte, S. Pochet, G. Labesse, Thymidylate kinase of I.N. Shindyalov, P.E. Bourne, The protein data bank, Nucl. Acids Res. 28 (2000)
Mycobacterium tuberculosis: a chimera sharing properties common to 235e242.
eukaryotic and bacterial enzymes, Protein Sci. 10 (2001) 1195e1205. [44] P.T. Lang, S.R. Brozell, S. Mukherjee, E.F. Pettersen, E.C. Meng, V. Thomas,
[17] A. Haouz, V. Vanheusden, H. Munier-Lehmann, M. Froeyen, P. Herdewijn, S. Van R.C. Rizzo, D.A. Case, T.L. James, I.D. Kuntz, DOCK 6: combining techniques to
Calenbergh, M. Delarue, Enzymatic and structural analysis of inhibitors designed model RNA-small molecule complexes, RNA 15 (2009) 1219e1230.
against Mycobacterium tuberculosis thymidylate kinase - new insights into the [45] G. Jones, P. Willett, R.C. Glen, A.R. Leach, R. Taylor, Development and validation
phosphoryl transfer mechanism, J. Biol. Chem. 278 (2003) 4963e4971. of a genetic algorithm for flexible docking, J. Mol. Biol. 267 (1997) 727e748.
[18] I. Van Daele, H. Munier-Lehmann, M. Froeyen, J. Balzarini, S. Van Calenbergh, [46] G. Jones, P. Willett, R.C. Glen, Molecular recognition of receptor site using
Rational design of 5’-thiourea-substituted alpha-thymidine analogues as a genetic algorithm with description of desolvation, J. Mol. Biol. 245 (1995)
thymidine monophosphate kinase inhibitors capable of inhibiting mycobac- 43e53.
terial growth, J. Med. Chem. 50 (2007) 5281e5292. [47] ChemBridge, San Deigo, CA, <http://www.chembridge.com>.
[19] O. Familiar, H. Munier-Lehmann, J. Antonio Ainsa, M.-J. Camarasa, M.-J. Perez- [48] Y. Izumizono, S. Arevalo, Y. Koseki, M. Kuroki, S. Aoki, Identification of novel
Perez, Design, synthesis and inhibitory activity against Mycobacterium tuber- potential antibiotics for tuberculosis by in silico structure-based drug
culosis thymidine monophosphate kinase of acyclic nucleoside analogues with screening, Eur. J. Med. Chem. 46 (2011) 1849e1856.
a distal imidazoquinolinone, Eur. J. Med. Chem. 45 (2010) 5910e5918. [49] R. Rawat, A. Whitty, P.J. Tonge, The isoniazid-NAD adduct is a slow, tight-
[20] A. Lavie, I.R. Vetter, M. Konrad, R.S. Goody, J. Reinstein, I. Schlichting, Structure binding inhibitor of InhA, the Mycobacterium tuberculosis enoyl reductase:
of thymidylate kinase reveals the cause behind the limiting step in AZT adduct affinity and drug resistance, Proc. Natl. Acad. Sci.U. S. A. 100 (2003)
activation, Nat. Struct. Mol. Biol. 4 (1997) 601e604. 13881e13886.

You might also like