Sunteți pe pagina 1din 7

Bioorganic & Medicinal Chemistry 22 (2014) 2409–2415

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


journal homepage: www.elsevier.com/locate/bmc

Synthesis, molecular docking and biological evaluation of


metronidazole derivatives containing piperazine skeleton as
potential antibacterial agents
She-Feng Wang  , Yong Yin  , Fang Qiao, Xun Wu, Shao Sha, Li Zhang ⇑, Hai-Liang Zhu ⇑
State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210093, People’s Republic of China

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

Article history: Metronidazole has a broad-spectrum antibacterial activity. Hereby a series of novel metronidazole deriv-
Received 22 January 2014 atives were designed and synthesized based on nitroimidazole scaffold in order to find some more potent
Revised 3 March 2014 antibacterial drugs. For these compounds which were reported for the first time, their antibacterial activ-
Accepted 4 March 2014
ities against Escherichia coli, Pseudomonas aeruginosa, Bacillus subtilis and Staphylococcus aureus were
Available online 13 March 2014
tested. These compounds showed good antibacterial activities against Gram-positive strains. Compound
4m represented the most potent antibacterial activity against S. aureus ATCC 25923 with MIC of 0.003 lg/
Keywords:
mL and it showed the most potent activity against S. aureus TyrRS with IC50 of 0.0024 lM. Molecular
Metronidazole derivatives
Antibacterial activities
docking of 4m into S. aureus tyrosyl-tRNA synthetase active site were also performed to determine the
S. aureus TyrRS probable binding mode.
Molecular docking Ó 2014 Elsevier Ltd. All rights reserved.

1. Introduction tRNA.15 Because aminoacyl-tRNA synthetases can recognize these


information including the coincident tRNA molecules and the ami-
Infections induced by antibiotic-resistant pathogens have al- no acids’ structures,16 they are essential to translate the coded
ready led to a crisis in people’s health care. With the advent of drug information into protein structures in nucleic acids.17 As a member
resistance, the microbes have developed several different kinds of of aminoacyl-tRNA synthetase family, tyrosyl-tRNA synthetase
resistance mechanisms. One of them is to reduce drug affinity by also plays an important role in protein synthesis. And it is a prom-
target-based mutation.1 Many problems still need to be solved even ising target for antibacterial agents as the topology of the ATP
though there are a lot of antimicrobial-drugs available such as keto- binding site and the interaction of bacteria aaRSs are different from
conazole, miconazole, metronidazole and so on (Scheme 1).2–9 For those of human.18–22
example, it has already caused serious menace because of the Nitroimidazole derivatives have showed broad varieties of bio-
appearance of multidrug resistant Gram-positive bacteria, in par- logical activities especially antimicrobial activity.23 5-Nitroimidaz-
ticular, methicillin-resistant Staphylococcus aureus and vancomy- ole based drugs have been applied to treat the infections induced
cin-resistant. In order to prevent this serious medical problem, it by bacteria and a range of pathogenic protozoan parasites for many
is urgent to elaborate new types of antibacterial agents.10 There- years because nitroimidazole derivatives can undergo bioreduction
fore, much of the research effort is put into the design of high effi- to produce electrophilic substances that can damage protein and
ciency antibacterial agents to combat resistant pathogens.11 nucleic acids.24 What’s important is the toxicology and metabolism
Aminoacyl-tRNA synthetases (aaRSs) have been proved to be of nitroimidazoles have been characterized, especially metronida-
antimicrobial targets,12 and they have been interesting targets in zole (Scheme 1).25,26 As one of the important nitroimidazole deriv-
antibacterial drug design.13,14 Aminoacyl-tRNAs can provide re- atives, metronidazole has been widely used as antimicrobial
quired substrates during protein synthesis. As in mRNA, the coded medicine. In recent years, much attention has been paid to modify
genetic information is translated into amino acid sequences which the pendant hydroxy group of metronidazole.27 Since piperazines
result proteins because the coded sequences complement with the and its derivatives are important pharmacophores, they are effec-
anticodon sequences which are built into a respective aminoacyl- tive ingredients in many marketed drugs like the merck HIV prote-
ase inhibitor Crixivan and Piperazinyl-linked ciprofloxacin dimers
⇑ Corresponding authors. Tel.: +86 25 83592572; fax: +86 25 83592672. which was reported as potent antibacterial agents against resistant
E-mail address: zhuhl@nju.edu.cn (H.-L. Zhu).
strains,28,29 and imidazole is found in many marketed drugs like
 
These authors contributed equally to this work. ketoconazole, miconazole and metronidazole and so on (Scheme 1),

http://dx.doi.org/10.1016/j.bmc.2014.03.004
0968-0896/Ó 2014 Elsevier Ltd. All rights reserved.
2410 S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415

O
CH 3 N N
Cl O O
Cl N a
N S Cl N S
OH O
O O
Cl O2 N O2 N
N N
N Cl N 1 2
N N
O O O N
OH R
N
Cl Cl O 2N N
O O b N
Ketoconazole Miconazole Metronidazole HN N R N N
N S
O
O NO2
O2N
Scheme 1. Chemical structure of metronidazole.
2 3a-3o 4a-4o
especially ketoconazole, it also contains piperazine skeleton. This
promoted us to synthesize new derivatives of metronidazole with Scheme 2. Reagents and conditions: (a) CH2Cl2, TEA, 0 °C, 5 h; (b) K2CO3, DMF,
80 °C, 22–24 h.
piperazine skeleton for the sake of finding new efficacy antibacterial
agents. Hence, we synthesized a series of novel metronidazole zole and piperazine derivatives. The key compound MET-OTs (2-
derivatives owing piperazine skeleton (4a–4o). In our preliminary (2-methyl-5-nitro-1H-imidazol-1-yl)ethyl 4-methylbenzenesulfonate,
work, the molecular docking was performed to validate which anti- compound 1) was synthesized by the method in our previous
bacterial target protein the designed compounds can work, the paper.35 The research showed when the ratio of MET-OTs and piper-
compounds were fitted into the ATP binding site of FabH (PDB code: azine derivatives was 1:1.2, the reaction temperature was 80 °C, and
1HNJ), bacterial DNA gyrase (PDB code: 3G75), bacterial thymidyl- the reaction time was 24 h, compounds 4a–4o were obtained with
ate kinase (PDB code: 4HEJ) and S. aureus tyrosyl-tRNA synthetase the higher yield. The chemical structures of these metronidazole
(PDB code: 1JIJ).30–34 The results have been plotted as a line-scatter derivatives were summarized in Table 1. All of the synthetic com-
graph and presented in Figure 1. It showed that the interaction en- pounds were characterized by 1H NMR, elemental analysis and mass
ergy between the designed compounds and protein 1JIJ is the low- spectrum, and they gave satisfactory analytical and spectroscopic
est. It means that the designed compounds are likely to display data, which were in full accordance with their depicted structures.
more potent inhibitory activity against S. aureus tyrosyl-tRNA syn-
thetase. So we studied their antibacterial activities against Bacillus 2.2. Biological activity
subtilis (B. subtilis) and Staphylococcus aureus (S. aureus) Escherichia
coli (E. coli), Pseudomonas aeruginosa (P. aeruginosa), and S. aureus 2.2.1. Antibacterial activity
tyrosyl-tRNA synthetase (PDB code: 1JIJ) inhibitory activities. The The synthesized compounds were tested for their antibacterial
results showed that the compounds showed good activity against activity against two Gram-negative bacterial strains: E. coli and P.
Gram-positive bacterial but no activity against the Gram-negative aeruginosa and two Gram-positive bacterial strains: B. subtilis and
bacterial. Docking simulation was performed using the X-ray crys- S. aureus by MTT method with MH medium (Mueller-Hinton med-
tallographic structure of the TyrRS of S. aureus in complex with ium: casein hydrolysate 17.5 g, soluble starch 1.5 g, beef extract
the most potent inhibitor to explore the binding mode of the com- 1000 mL). The MICs (minimum inhibitory concentrations) of the
pound at the active site. compounds against these bacteria were reported in Table 2, the
activity of reference drugs Penicillin G and Chloramphenicol was
2. Results and discussion also included. The results revealed that those compounds showed
good activity against Gram-positive bacterial but inactive against
2.1. Chemistry Gram-negative bacterial.
Most of the new compounds exhibited good inhibitory activities
Compounds 4a–4o were synthesized by the routes outlined in against both B. Subtilis and S. Aureus. As shown in Table 2. Compounds
Scheme 2. They were first designed and synthesized from metronida- 4l and 4m showed most potent activities with MIC values of 0.033,

Figure 1. The CDOCKER_INTERACTION_ENERGY (kcal/mol) obtained from the docking study of all synthesized compounds by the CDOCKER protocol (Discovery Studio 3.5,
Accelrys, Co. Ltd).
S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415 2411

Table 1 According to these data, we found that compound 4l which had


Structures of compounds 4a–4o a methoxy group on the 4-position of benzene ring showed higher
N
R antibacterial activity against B. Subtilis with MIC values of
N 0.033 lg/mL than compound 4h which had a methoxy group on
N
N the 2-position of benzene ring with MIC values of 0.272 lg/mL
NO2 and compound 4f which had a methoxy group on the 3-position
of benzene ring with MIC values of 0.805 lg/mL, the same trend
Compounds R Compounds R was also found in S. aureus. Compound 4g (MIC = 6.872 lg/mL)
F3 C with a chlorine on the 3-position of benzene ring showed worse
4a 4i inhibitory activity than compound 4d (MIC = 0.142 lg/mL) with a
chlorine on the 2-position of benzene ring. It indicated that com-
CH3
pounds with substitution at the para or ortho position exhibited
4b F 4j more potent antibacterial activities than compounds with substi-
H3 C tution at the meta position.
Cl Cl Among compounds 4a (MIC = 1.551 lg/mL), 4m (MIC = 0.011 lg/mL)
4c
Cl
4k and 4n (MIC = 0.185 lg/mL), the order of inhibitory activities
Cl
showed the potency of 4m > 4n > 4a, which indicated the activity of
Cl compounds with substitutes of benzhydryl were superior to that of
4d 4l phenyl. Compounds 4k and 4c, which introduced two chlorine atoms
H3 CO
at 2,3-position or 3,4-positon in benzene ring, displayed a dramatic
F loss in activity compared to the analogues bearing only one substitu-
4e 4m ent (4d,4g). In addition, compounds 4b (MIC > 50 lg/mL), 4e
(MIC > 50 lg/mL) and 4o (MIC > 50 lg/mL) indicated that com-
H3 CO pounds with substitution of fluorine exhibited almost no activities.
Varieties of substitutes of phenyl like trifluoromethyl, methyl and
4f 4n
halogen can lead to different antibacterial activities. For example,
Cl
the derivatives which have electron-withdrawing substituents (like
Cl
halogen and trifluoromethyl) displayed weaker activities against
4g 4o B. subtilis and S. aureus than those having electron-donating substit-
F F uents (such as methyl and methoxyl), which suggested that substi-
OCH3 tuent group can affect the inhibitory activities, and compounds
4h with electron-donating substituents exhibited better inhibitory
activities than those with electron-withdrawing substituents.

2.2.2. S. aureus TyrRS inhibitory activity


The S. aureus TyrRS inhibitory potency of the synthetic metroni-
dazole derivatives was examined and the results were summarized
Table 2
Antibacterial activities of synthetic compounds
in Table 3. Most of the tested compounds showed potent S. aureus
TyrRS inhibitory activities. Among them, compounds 4d, 4l and 4m
Compounds Minimum inhibitory concentrations (lg/mL) with better antibacterial activities also showed better inhibitory
Gram-positive Gram-negative activity toward S. aureus TyrRS. Compound 4m exhibited the most
B. subtilis S. aureus E. coli P. aeruginosa potent inhibitory activities against S. aureus TyrRS with IC50 of
4a 1.551 >50 >50 >50
0.0024 lM. Compounds 4l and 4d also displayed good inhibitory
4b >50 >50 >50 >50 with IC50 of 0.0067 lM and 0.0086 lM. Other tested compounds
4c 14.382 11.116 >50 >50 displayed moderate inhibitory activity with IC50 ranging from
4d 0.142 0.203 >50 >50 1.12 to 25.76 lM. Compound 4m with N-benzhydryl group substi-
4e >50 17.105 >50 >50
tuent showed more preferable inhibitory activity than compound
4f 0.805 0.656 >50 >50
4g 2.567 1.801 >50 >50 4a with N-phenyl group substituent. Compound 4l with p-substi-
4h 0.272 0.583 >50 >50
Table 3
4i 3.288 18.093 >50 >50
S. aureus TyrRS inhibitory activity of synthetic compounds
4j 4.312 1.609 >50 >50
4k 6.827 11.56 >50 >50 Compounds S. aureus TyrRS IC50 (lM) Hemolysis LC30a (mg/mL)
4l 0.033 0.123 >50 >50
4m 0.011 0.003 >50 >50 4a 20.47 >10
4n 0.185 0.593 >50 >50 4b 25.2 >10
4o >50 >50 >50 >50 4c 13.55 >10
Penicillin G 0.335 1.024 22.843 18.624 4d 0.0086 >10
Chloramphenicol 0.078 0.178 0.064 0.102 4e 23.37 >10
4f 7.65 >10
4g 10.52 >10
4h 1.12 >10
4i 15.28 >10
0.011 lg/mL and 0.123, 0.003 lg/mL against B. subtilis and S. aureus, 4j 12.56 >10
respectively, which were superior to the positive control Penicillin G 4k 16.2 >10
with MIC values of 0.335 and 1.024 lg/mL. 4d (MIC = 0.142 lg/mL), 4l 0.0067 >10
4n (MIC = 0.185 lg/mL) and 4h (MIC = 0.272 lg/mL) exhibited 4m 0.0024 >10
4n 1.72 >10
significant inhibitory against B. Subtilis, which were superior to the
4o 25.76 >10
positive control Penicillin G (MIC = 0.335 lg/mL) but worse than
a
Chloramphenicol (MIC = 0.078 lg/mL). Lytic concentration 30%.
2412 S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415

tuted chloro group displayed better inhibitory activity than com-


pounds 4h and 4f with o-substituted and m-substituted chloro
group. Compounds 4c and 4k with two chloro groups exhibited
worse inhibitory activity than compound 4d with single o-substi-
tuted chloro group.
The results of S. aureus TyrRS inhibitory activity of the test com-
pounds were corresponding to the structure–activity relationships
(SAR) of their antibacterial activities. This demonstrated that the
potent antibacterial activities of the synthetic compounds were
probably correlated to their TyrRS inhibitory activities.

2.2.3. Molecular docking


With the aim to better understanding of the structure–activity
relationships observed, molecular docking of the most potent
inhibitor 4m into the active site cavity of tyrosyl-tRNA synthetase
was performed on the binding model based on the tyrosyl-tRNA
synthetase complex structure (PDB ID: 1JIJ). The binding model
of compound 4m and TyrRs was described in Figure 2, and the
Figure 3. Binding mode of compound 4m with TyrRS. The enzyme is shown as
enzyme surface model was shown in Figure 3, which revealed that surface; while 4m docked structures are shown as sticks. This figure was made
the molecule is well filled in the active pocket. using PyMol.
As illustrated in Figure 2, 4m located in the substrate binding
site of tyrosyl-tRNA synthetase. The docking results revealed that
three amino acids Arg80, Gln196 and His50 located in the binding
pocket of protein played important roles in the combination with coordination bond with protons. As showed in Figure 2, a carboxy
compound 4m, which were stabilized by one p–p bond and two of Gln196 formed ammonium salt with N heterocyclic ring, then
hydrogen bonds. The p–p bond, of which their lengths was the oxygen of the carbonyl group formed a hydrogen bond with
4.45 Å, was formed between phene ring and His50. There is an the ammonium ion, and the length of H  O bond was 2.26 Å.
unoccupied orbital on the nitrogen atom, so it is easy to form a The other hydrogen bond was formed between the nitrogen of
amine of Arg80 and the ammonium ion with H  N bond length
of 2.49 Å. This molecular docking result, along with the biological
assay data, suggested that compound 4m was a potential inhibitor
of TyrRS.

3. Conclusion

A series of novel metronidazole derivatives have been designed


and synthesized, and their biological activities were also evaluated
as potent TryRs inhibitory. Compound 4m demonstrated the most
potent inhibitory activity (MIC = 0.011 lg/mL for B. subtilis and
MIC = 0.003 lg/mL for S. aureus), which was compared with the
positive control Penicillin G and Chloramphenicol. Docking simula-
tion was performed to position compound 4m into the S. aureus
TryRs active site to determine the potential binding model. Analy-
sis of the binding conformation of compound 4m displayed that
compound 4m was stabilized by hydrogen bonding interactions
with ASP80 and GLN196. Antibacterial assay results also showed
that these metronidazole derivatives had the potential to be devel-
oped for antibacterial agents against S. aureus. Particularly, com-
pound 4m has demonstrated significant S. aureus TryRs
inhibitory activity as a potential antibacterial agent.

4. Experiments

4.1. Materials and measurements

All chemicals and reagents used in current study were of analyt-


ical grade. TLC was performed on the glass-backed silica gel sheets
(silica GF254) and visualized in UV light (254 nm). Column chro-
matography was performed using silica gel (200–300 mesh) elut-
ing with ethyl acetate and petroleum ether. Melting points
(uncorrected) were determined on a XT4MP apparatus (Taike
Corp., Beijing, China). All the 1H NMR spectra were recorded on a
Bruker DPX 400 model Spectrometer in CDCl3 and chemical shifts
Figure 2. Binding mode of compound 4m with TyrRS from S. aureus. For clarity,
were reported in ppm (d). ESI-MS spectra were recorded on a Mar-
only interacting residues were labeled. Hydrogen bonding interactions are shown in iner System 5304 Mass spectrometer. Elemental analyses were
green dotted lines. This figure was made using PyMol. performed on a CHN-O-Rapid instrument.
S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415 2413

4.2. General method of synthesis of metronidazole derivatives 4.44–4.47 (t, J = 5.26 Hz, 2H, CH2), 3.51–3.63 (s, 4H, –CH2CH2–),
2.94–2.99 (s, 4H, –CH2CH2–), 2.50 (s, 3H, CH3). ESI-MS: 334.32
2-(2-Methyl-5-nitro-1H-imidazol-1-yl)ethyl 4-methylbenzene- (C16H21FN5O2, [M+H]+). Anal. Calcd for C16H20ClN5O2: C, 57.65; H,
sulfonate (2) was prepared according to our previous paper. Metro- 6.05; N, 20.01. Found: C, 57.60; H, 6.02; N, 20.02.
nidazole (3.14 g, 20 mmol) and Et3N (3.0 mL, 22 mmol) were
dissolved in CH2Cl2 (20 mL), and 4-methyl-benzenesufonyl chlo- 4.2.6. 1-(3-Methoxyphenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol-
ride (3.83 g, 20.1 mmol) in CH2Cl2 (10 mL) was added. The reaction 1-yl)ethyl)piperazine (4f)
mixture was stirred at 0 °C for 5 h followed by the addition of Brown, yield: 61%, mp: 45–48 °C. 1H NMR (400 MHz, CDCl3): d
30 mL ice water, and then the layer was separated and the aqueous 7.96 (s, 1H), 7.16–7.20 (t, J = 8.04 Hz, 1H, ArH), 6.50–6.53
layer was extracted with ethyl acetate (2  30 mL). The organic (t, J = 5 Hz, 3H, ArH), 4.61–4.63 (t, J = 5.26 Hz, 2H, CH2), 4.43–4.46
layer was combined and washed with saturated NaHCO3, and dried (t, J = 5.28 Hz, 2H, CH2), 3.78 (s, 3H, CH3O-), 3.47–3.60 (s, 4H, –CH2-
with anhydrous Na2SO4 for 0.5 h. Removal of the solvent gave a CH2–), 3.09–3.13 (s, 4H, –CH2CH2–), 2.49 (s, 3H, CH3). ESI-MS:
slight-yellow crystal of compound 2 (MET-OTs). Compounds 346.36 (C17H24N5O3, [M+H]+). Anal. Calcd for C17H23N5O3: C,
4a–4o were synthesized by the following procedure. MET-OTs 59.12; H, 6.71; N, 20.28. Found: C: 59.11; H: 6.71; N, 20.2.
(2 mmol) and K2CO3 (2 mmol) were dissolved in DMF (20 mL),
and then one of those substituted piperazines was added. The reac- 4.2.7. 1-(3-Chlorophenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol-
tion was stirred at 80 °C for 22–24 h. The reaction mixture was 1-yl)ethyl)piperazine (4g)
poured in water and extracted with ethyl acetate (3  50 mL). Brown, yield: 56%, mp: 66–68 °C. 1H NMR (400 MHz, CDCl3): d
The organic layer was combined and dried with anhydrous Na2SO4 7.96 (s, 1H), 7.15–7.19 (t, J = 8.12 Hz, 1H, ArH), 6.84–6.85 (d,
for 0.5 h. After the reaction mixture was vacuum distillated, the J = 6.48 Hz, 2H, ArH), 6.75–6.78 (m, 1H, ArH), 4.61–4.64 (t,
target product was separated by column chromatography. J = 5.28 Hz, 2H, CH2), 4.43–4.46 (t, J = 5.26 Hz, 2H, CH2), 3.47–3.59
(s, 4H, –CH2CH2–), 3.10–3.14 (s, 4H, –CH2CH2–), 2.49 (s, 3H, CH3).
4.2.1. 1-(2-(2-Methyl-5-nitro-1H-imidazol-1-yl)ethyl)-4-phenyl ESI-MS: 350.76 (C16H21ClN5O2, [M+H]+). Anal. Calcd for C16H20-
piperazine (4a) ClN5O2: C, 54.94; H, 5.76; N, 20.02. Found: C, 54.92; H,5.75; N,
Brown, yield: 47%, mp: 108–110 °C. 1H NMR (400 MHz, CDCl3): 20.02.
d 7.97 (s, 1H), 7.25–7.29 (m, 2H, ArH), 6.90–6.92 (d, J = 7.64 Hz, 3H,
ArH), 3.49–3.61 (s, 4H, –CH2CH2–), 3.09–3.13 (s, 4H, –CH2CH2–), 4.2.8. 1-(2-Methoxyphenyl)-4-(2-(2-methyl-5-nitro-1H-
2.49 (s, 3H, CH3), ESI-MS: 316.28 (C16H22N5O2, [M+H]+). Anal. Calcd imidazol-1-yl)ethyl)piperazine (4h)
for C16H21N5O2: C, 60.94; H, 6.71; N, 22.21. Found: C, 60.92; H, Yellow, yield: 52%, mp: 112–115 °C. 1H NMR (400 MHz, CDCl3):
6.72, N, 22.23. d 7.93 (s, 1H), 6.99–7.03 (m, 1H, ArH), 6.84–6.92 (m, 3H, ArH),
4.59–4.60 (d, J = 5.28 Hz, 2H, CH2), 4.41–4.44 (t, J = 5.22 Hz, 2H,
4.2.2. 1-(4-Fluorophenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol- CH2), 3.85 (s, 3H, CH3O-), 3.49–3.61 (s, 4H, –CH2CH2–), 2.93–2.99
1-yl)ethyl)piperazine (4b) (s, 4H, –CH2CH2–), 2.47 (s, 3H, CH3). ESI-MS: 346.36 (C17H24N5O3,
Brown, yield: 51%, mp: 65–68 °C. 1H NMR (400 MHz, CDCl3) : d [M+H]+). Anal. Calcd for C17H23N5O3: C, 59.12; H, 6.71; N, 20.28.
7.93 (s, 1H), 6.93–6.97 (t, J = 8.34 Hz, 2H, ArH), 6.83–6.85 (m, 2H, Found: C, 59.12; H, 6.70; N, 20.27.
ArH), 4.41–4.45 (t, J = 6.26 Hz, 2H, CH2), 3.04–3.07 (t, J = 4.86 Hz,
4H, –CH2CH2–), 2.71–2.72 (t, J = 6.26 Hz, 2H, CH2), 2.63–2.65 (t, 4.2.9. 1-(2-(2-Methyl-5-nitro-1H-imidazol-1-yl)ethyl)-4-(3-
J = 4.86 Hz, 4H, –CH2CH2–,), 2.52 (s, 3H, CH3). ESI-MS: 334.27 (C16- (trifluoromethyl)phenyl)piperazine (4i)
H21FN5O2, [M+H]+). Anal. Calcd for C16H20FN5O2: C, 57.65; H, 6.05; Yellow, yield: 45%, mp: 98–101 °C. 1H NMR (400 MHz, CDCl3)
N, 21.01. Found: C, 57.62, H, 6.04; N, 21.03. d:7.96 (s, 1H), 7.34–7.38 (t, J = 7.94 Hz, 1H, ArH), 7.03–7.06 (m,
3H, ArH), 4.62–4.65 (t, J = 5.26 Hz, 2H, CH2), 4.44–4.46 (t,
4.2.3. 1-(2,3-Dichlorophenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol- J = 5.28 Hz, 2H, CH2), 3.49–3.62 (s, 4H, –CH2CH2–), 3.14–3.18 (s,
1-yl)ethyl)piperazine (4c) 4H, –CH2CH2–), 2.50 (s, 3H, CH3). ESI-MS: 384.33 (C17H21F3N5O2,
Yellow, yield: 47%, mp: 90–93 °C. 1H NMR (400 MHz, CDCl3): d [M+H]+). Anal. Calcd for C17H20F3N5O2: C, 53.26; H, 5.26; N,
7.95 (s, 1H), 7.12–719 (m, 2H, ArH), 6.89–6.91 (m, 1H, ArH), 18.27. Found: C: 53.25; H, 5.27; N, 18.27.
4.61–4.64 (t, J = 5.26 Hz, 2H, CH2), 4.43–4.46 (t, J = 5.24 Hz, 2H,
CH2), 3.50–3.62 (s, 4H, –CH2CH2–), 2.92–2.97 (s, 4H, –CH2CH2–), 4.2.10. 1-(2,4-Dimethylphenyl)-4-(2-(2-methyl-5-nitro-1H-imid
2.49 (s, 3H, CH3). ESI-MS: 385.15 (C16H20Cl2N5O2, [M+H]+). Anal. azol-1-yl)ethyl)piperazine (4j)
Calcd for C16H19Cl2N5O2: C, 50.01; H, 4.98; N, 18.23. Found: C, Brown, yield: 55%, mp: 130–132 °C. 1H NMR (400 MHz, CDCl3)
49.98; H, 4.99; N, 18.22. d: 7.97 (s, 1H), 6.96–7.00 (t, J = 9.34 Hz, 2H, ArH), 6.86–6.88 (d,
J = 8.04 Hz, 1H, ArH), 4.61–4.64 (t, J = 5.24 Hz, 2H, CH2), 4.44–4.47
4.2.4. 1-(2-Chlorophenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol- (t, J = 5.24 Hz, 2H, CH2), 3.46–3.59 (s, 4H, –CH2CH2–), 2.77–2.82
1-yl) ethyl)piperazine(4d) (s, 4H, –CH2CH2–), 2.51 (s, 3H, CH3), 2.26 (s, 3H, CH3), 2.27 (s, 3H,
Yellow, yield: 59%, mp: 120–122 °C. 1H NMR (400 MHz, CDCl3): CH3). ESI-MS: 344.38 (C18H26N5O2, [M+H]+). Anal. Calcd for
d 7.96 (s, 1H), 7.34–7.37 (m, 1H, ArH), 7.20–7.26 (m, 1H, ArH), C18H25N5O2: C, 62.95; H, 7.34; N, 20.39. Found: C, 62.92; H, 7.36;
6.98–7.02 (m, 2H, ArH), 4.62–4.64 (t, J = 5.26 Hz, 2H, CH2), N, 20.38.
4.44–4.47 (t, J = 5.26 Hz, 2H, CH2), 3.51–3.63 (s, 4H, –CH2CH2–),
2.94–2.99 (s, 4H, –CH2CH2–), 2.50 (s, 3H, CH3). ESI-MS: 350.76 4.2.11. 1-(3,4-Dichlorophenyl)-4-(2-(2-methyl-5-nitro-1H-imid
(C16H21ClN5O2, [M+H]+). Anal. Calcd for C16H20ClN5O2: C, 54.94; azol-1-yl)ethyl)piperazine (4k)
H, 5.76; N, 20.02. Found: C, 54.88; H, 5.75, N, 20.01. Yellow, yield: 61%, mp: 106–109 °C. 1H NMR (400 MHz, CDCl3):
d 7.98 (s, 1H), 7.29–7.31 (d, J = 8.6 Hz, 1H, ArH), 6.96–6.97 (d,
4.2.5. 1-(2-Fluorophenyl)-4-(2-(2-methyl-5-nitro-1H-imidazol- J = 2.76 Hz, 1H, ArH), 6.73–6.76 (m, 1H, ArH), 4.64–4.67 (t,
1-yl)ethyl)piperazine (4e) J = 5.26 Hz, 2H, CH2), 4.46–4.48 (t, J = 5.26 Hz, 2H, CH2), 3.50–3.62
Yellow, yield: 48%, mp: 125–127 °C. 1H NMR (400 MHz, CDCl3): (s, 4H, –CH2CH2–), 3.11–3.14 (s, 4H, –CH2CH2–), 2.52 (s, 3H, CH3).
d 7.96 (s, 1H), 7.34–7.37 (m, 1H, ArH), 7.20–7.26 (m, 1H, ArH), ESI-MS: 385.15 (C16H20Cl2N5O2, [M+H]+). Anal. Calcd for C16H19Cl2-
6.98–7.02 (m, 2H, ArH), 4.62–4.64 (t, J = 5.26 Hz, 2H, CH2), N5O2: C, 50.01; H, 4.98; N, 18.23. Found: C, 50.03; H, 4.97; N, 18.24.
2414 S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415

4.2.12. 1-(4-Methoxyphenyl)-4-(2-(2-methyl-5-nitro-1H-imid performed at 37 °C in a mixture containing (final concentrations)


azol-1-yl)ethyl) piperazine (4l) 100 mM Tris/Cl pH 7.9, 50 mM KC1, 16 mM MgCl2, 5 mM ATP,
Yellow, yield: 46%, mp: 104–107 °C. 1H NMR (400 MHz, CDCl3): 3 mM DTT, 4 mg/mL E. coli MRE600 tRNA (Roche) and 10 lM
3
d 7.97 (s, 1H), 6.84–6.91 (t, J = 14.56 Hz, 4H, ArH), 4.62–4.65 (t, L-tyrosine (0.3 lM L-[ring-3,5- H] tyrosine (Perkin-Elmer, Specific
J = 5.26 Hz, 2H, CH2), 4.44–4.47 (t, J = 5.22 Hz, 2H, CH2), 3.77 (s, activity: 1.48–2.22 TBq/mmol), 10 lM carrier). TyrRS (0.2 nM)
3H, CH3O-), 3.51–3.63 (s, 4H, –CH2CH2–), 3.00–3.01 (s, 4H, CH2CH2- was pre-incubated with a range of inhibitor concentrations for
), 2.50 (s, 3H, CH3). ESI-MS: 346.36 (C17H24N5O3, [M+H]+). Anal. 10 min at room temperature followed by the addition of pre-
Calcd for C17H23N5O3: C, 59.12; H, 6.71; N, 20.28. Found: C, warmed mixture at 37 °C. After specific intervals, the reaction
59.13; H, 6.70; N, 20.27. was terminated by adding aliquots of the reaction mix into ice-cold
7% trichloroacetic acid and harvesting onto 0.45 lm hydrophilic
4.2.13. 1-Benzhydryl-4-(2-(2-methyl-5-nitro-1H-imidazol-1-yl) Durapore filters (Millipore Multiscreen 96-well plates) and
ethyl)piperazine (4m) counted by liquid scintillation. The rate of reaction in the experi-
Yellow, yield: 58%, mp: 40–43 °C. 1H NMR (400 MHz, CDCl3): d ments was linear with respect to protein and time with less than
7.93 (s, 1H), 7.37–7.39 (d, J = 7.36 Hz, 4H, ArH), 7.24–7.28 (m, 4H, 50% total tRNA acylation. IC50s corresponding to the concentration
ArH), 7.15–7.18 (t, J = 7.28 Hz, 2H, ArH), 4.54–4.57 (t, J = 5.18 Hz, at which half of the enzyme activity was inhibited by the com-
2H, CH2), 4.37–4.40 (t, J = 5.18 Hz, 2H, CH2), 4.20 (s, 1H, CH), 3.30– pound. The results were presented in Table 3.
3.43 (s, 4H, –CH2CH2–), 2.43 (s, 3H, CH3), 2.28–2.34 (s, 4H, –CH2CH2–
). ESI-MS: 406.46 (C23H28N5O2, [M+H]+). Anal. Calcd for C23H27N5O2:
4.5. Docking simulations
C, 72.33; H, 6.49; N, 14.54. Found: C, 72.34; H, 6.47; N, 14.55.

The crystal structures of TyrRS (PDB code: 1JIJ) was obtained


4.2.14. 1-((4-Chlorophenyl)(phenyl)methyl)-4-(2-(2-methyl-5-
from the Protein Data Bank (http://www.rcsb.org). Molecular
nitro-1H-imidazol-1-yl)ethyl)piperazine (4n)
docking of compound 4m into the three-dimensional X-ray struc-
Yellow, yield: 51%, mp: 50–52 °C. 1H NMR (400 MHz, CDCl3): d
ture of TyrRS was carried out using CDOCKER Dock protocol of Dis-
7.98 (s, 1H), 7.22–7.40 (m, 9H, ArH), 4.61–4.63 (t, J = 4 Hz, 2H,
covery Studio 3.5.
CH2), 4.43–4.45 (t, J = 5.16 Hz, 2H, CH2), 4.25 (s, 1H, CH),
3.36–3.49 (s, 4H, –CH2CH2–), 2.49 (s, 3H, CH3), 2.32–2.38 (s, 4H, –
Acknowledgment
CH2CH2–). ESI-MS:440.86 (C23H27ClN5O2, [M+H]+). Anal. Calcd for
C23H26ClN5O2: C, 67.50; H, 5.86; N, 13.57. Found: C, 67.51; H,
The work was financed by Natural Science Foundation of China
5.84; N, 13.55.
(No. J1103512).
4.2.15. 1-(Bis(4-fluorophenyl)methyl)-4-(2-(2-methyl-5-nitro-1H-
imidazol-1-yl)ethyl)piperazine (4o) References and notes
Brown, yield: 47%, mp: 47–50 °C. 1H NMR (400 MHz, CDCl3): d
1. Hooper, D. C. Emerging Infect. Dis. 2001, 7, 337.
7.90 (m, 4H, Ar), 6.97 (s, 4H, Ar), 4.35–4.38 (t, J = 6.3 Hz, 3H, 2. Appelbaum, P. C.; Hunter, P. A. Int. J. Antimicrob. Agents 2000, 16, 5.
CH2), 4.17 (s, 1H, CH), 2.61–2.66 (m, 2H, CH2), 2.49 (s, 4H, –CH2- 3. Mizuki, Y.; Fujawara, I.; Yamaguchi, T. J. Antimicrob. Chemother. 1996, 37, 41.
CH2–), 2.48 (s, 3H, CH3), 2.31–2.32 (s, 4H, –CH2CH2–). ESI-MS: 4. Ball, P. J. Antimicrob. Chemother. 2000, 46, 17.
5. Snaz-Nebot, V.; Valls, I.; Barbero, D.; Barbosa, J. Acta Chem. Scand. A 1997, 51,
442.33 (C23H26F2N5O2, [M+H]+). Anal. Calcd for C23H25F2N5O2: C, 896.
67.30; H, 5.65; N, 13.53. Found: C, 67.31; H, 5.64; N, 13.54. 6. Vazquez, J. A.; Sanchez, V.; Dmuchowski, C.; Dembry, L. M.; Sobel, J. D.; Zervos,
M. J. J. Infect. Dis. 1993, 168, 195.
7. Bodey, G. P. In Candidosis: Pathogenesis, Diagnosis and Treatment; Raven: New
4.3. Antibacterial activity
York, 1993. p 371.
8. Andriole, V. T.; Bodey, G. P. Pocket Guide to Systemic Antifungal Therapy;
The antibacterial activities of the synthetic compounds were Scientific Therapeutics: Springfield, NJ, 1994. p 371.
9. Current, W. L.; Tang, J.; Boylan, C.; Watson, P.; Zechkner, D.; Turner, W.;
tested against two Gram-negative bacterial strains: E. coli ATCC
Rodriguez, M.; Dixon, C.; Ma, D.; Radding, J. A. Glucan Biosynthesis as a Target
25922 and P. aeruginosa ATCC 27853, two Gram-positive bacterial for Antifungals: The Echinocandin Class of Antifungal Agents. In Antifungal
strains: B. subtilis ATCC 530 and S. aureus ATCC 25923, using meth- Agents: Discovery and Mode; Dixon, G. K., Copping, L. G., Hollomon, D. W., Eds.;
od recommended by National Committee for Clinical Laboratory BIOS: Oxford, 1995; p 143.
10. Leeb, M. Nature 2004, 431, 892.
Standards (NCCLS).36 11. Cui, Y.; Dang, Y.; Yang, Y.; Zhang, S. Eur. J. Med. Chem. 2005, 40, 209.
In vitro activities of the compounds were tested in Nutrient 12. Finn, J.; Tao, J. In Aminoacyl-tRNA Synthetase as Drug Targets in Aminoacyl-tRNA
broth (NB) for bacteria by the two-fold serial dilution method. Synthetases; Ibba, M., Ed.; Landes BioScience Press: Georgetown, TX, 2004; p
405; (b) Jarvest, R.; Berge, J.; Berry, V.; Boyd, H.; Brown, M.; Elder, J.; Forrest, A.;
Seeded broth (broth containing microbial spores) was prepared Fosberry, A.; Gentry, D.; Hibbs, M.; Jaworski, D.; O’Hanlon, P.; Pope, A.;
in NB from 24 h old bacterial cultures on nutrient agar (Hi-media) Rittenhouse, S.; Sheppard, R.; Slater-Radosti, C.; Worby, A. J. Med. Chem. 2002,
at 37 ± 1 °C. The bacterial suspension was adjusted with sterile sal- 45, 1959; (c) Finn, J.; Mattia, K.; Morytko, M.; Ram, S.; Yang, Y.; Wu, X.;
Silverman, J.; Mak, E.; Gallant, P.; Keith, D. Bioorg. Med. Chem. Lett. 2003, 13,
ine to a concentration of 1  104–105 CFU/mL. The tested com- 2231; (d) Finn, J.; Stidham, M.; Hilgers, M.; Kedar, G. C. Bioorg. Med. Chem. Lett.
pounds and reference drugs were prepared by two-fold serial 2008, 18, 3932; (e) Yu, X.; Finn, J.; Hill, J.; Wang, Z.; Keith, D.; Silverman, J.;
dilution to obtain the required concentrations of 100, 50, 25, Oliver, N. Bioorg. Med. Chem. Lett. 2004, 14, 1343; (f) Kim, S.; Lee, S. W.; Choi, E.
C.; Chio, S. Y. Appl. Microbiol. Biotechnol. 2003, 61, 278.
12.5, 6.25, 3.13 lg/mL and even lower concentration. The tubes
13. Ibba, M.; Soll, D. Annu. Rev. Biochem. 2000, 69, 617.
were incubated in BOD incubators at 37 ± 1 °C for bacteria. The 14. Ibba, M.; Thomann, H. U.; Hong, K. W.; Sherman, J. M.; Weygand-Durasevic, I.;
MICs were recorded by visual observations after 24 h (for bacteria) Sever, S.; Stange-Thomann, N.; Praetorius, M.; Soll, D. Nucleic Acids Symp. Ser.
1995, 33, 402.
of incubation. Penicillin G and chloramphenicol were used as stan-
15. Schimmel, P.; Tao, J.; Hill, J. FASEB J. 1998, 12, 1599.
dards for bacterial. The observed MICs are presented in Table 2. 16. Tao, J.; Schimmel, P. Expert Opin. Invest. Drugs 2000, 9, 1767.
17. Hurdle, J. G.; O’Neill, A. J.; Chopra, I. Antimicrob. Agents Chemother. 2005, 49,
4821.
4.4. S. aureus TyrRS inhibitory activity 18. Farhanullah, R.; Kang, T.; Yoon, E.-J.; Choi, E.-C.; Kim, S.; Lee, J. Eur. J. Med. Chem.
2009, 44, 239.
19. Van de Vijver, P.; Vondenhoff, G. H. M.; Kazakov, T. S.; Semenova, E.;
TyrRS activity was measured by aminoacylation using modifica- Kuznedelov, K.; Metlitskaya, A.; Van Aerschot, A.; Severinov, K. J. Bacteriol.
tions to previously described methods.37 The assays were 2009, 191, 6273.
S.-F. Wang et al. / Bioorg. Med. Chem. 22 (2014) 2409–2415 2415

20. Balg, C.; De Mieri, M.; Huot, J. L.; Blais, S. P.; Lapointe, J.; Chênevert, R. Bioorg. J. Med. Chem. 1994, 37, 3443; (c) Askin, D.; Eng, K. K.; Rossen, K.; Purick, R. M.;
Med. Chem. 2010, 18, 7868. Wells, K. M.; Volante, R. P.; Reider, P. J. Tetrahedron Lett. 1994, 35, 673; (d)
21. Wu, Y.; Yu, K.; Xu, B.; Chen, L.; Chen, X.; Mao, J.; Danchin, A.; Shen, X.; Qu, D.; Rossen, K.; Weissman, S. A.; Sagar, J.; Reamer, R. A.; Askin, D. A.; Volante, R. P.;
Jiang, H. J. Antimicrob. Chemother. 2007, 60, 502. Reider, P. J. Tetrahedron Lett. 1995, 36, 6419.
22. Hurdle, J. G.; O’Neill, A. J.; Chopra, I.; Antimicrob, J. Agents Chemother. 2005, 48, 29. Kerns, R. J.; Rybak, M. J.; Kaatz, G. W.; Vaka, F.; Cha, R.; Grucz, R. G.; Diwadkar,
4821. V. U. Bioorg. Med. Chem. Lett. 2003, 13, 2109.
23. Beena; Kumar, N.; Rohilla, R. K.; Roy, N.; Rawat, D. S. Bioorg. Med. Chem. Lett. 30. Luo, Y.; Li, H. Q.; Zhou, Y.; Li, Z. L.; Yan, T.; Zhu, H. L. ChemMedChem 2010, 5,
2009, 19, 1396. 1110.
24. Lord, E. M.; Harwell, L.; Koch, C. J. Cancer Res. 1993, 53, 5721. 31. Wang, Z.; Duan, Y.; Qin, Y.; Wang, P.; Luo, Y.; Wen, Q.; Yang, Q. Y.; Sun, J.; Hu,
25. Webster, L. T. Drugs Used in the Chemotherapy of Protozoal Infections. In The Y.; Sang, Y. Eur. J. Med. Chem. 2013, 65, 456.
Pharmacological Basis of Therapeutics; Gilman, A., Rall, T. W., Nies, A. S., Taylor, 32. Tanitame, A.; Oyamada, Y.; Ofuji, K.; Fujimoto, M.; Iwai, N.; Hiyama, Y.; Suzuki,
P., Eds.; Pergamon Press: New York, 1990; p 999. K.; Ito, H.; Terauchi, H.; Kawasaki, M. J. Med. Chem. 2004, 47, 3693.
26. Alvaro, R. F.; Wislocki, P. G.; Miwa, G. T.; Lu, A. Y. Chem. Biol. Interact. 1992, 82, 33. Sun, J.; Lv, P.; Yin, Y.; Yuan, R.; Ma, J.; Zhu, H. PLoS ONE 2013, 8, e69751.
21. 34. Qiu, X.; Janson, C. A.; Smith, W. W.; Green, S. M.; McDevitt, P.; Johanson, K.;
27. Mallia, B. M.; Mathur, A.; Subramanian, S.; Banerjee, S.; Sarma, H. D.; Carter, P.; Hibbs, M.; Lewis, C.; Chalker, A.; Fosberry, A.; Lalonde, J.; Berge, J.;
Venkatesh, M. Bioorg. Med. Chem. Lett. 2005, 15, 3398. Brown, P.; Houge-Frydrych, C. S.; Jarvest, R. L. Protein Sci. 2001, 10, 2008.
28. (a) Vacca, J. P.; Dorsey, B. D.; Schleif, W. A.; Levine, R. B.; McDaniel, S. L.; Darke, 35. Cao, P.; Fang, R. Q.; Li, H. Q.; Zhu, H. L. Chin. J. Struct. Chem. 2007, 26, 1320.
P. L.; Zugay, J.; Quintero, J. C.; Blahy, O. M.; Sardana, B. B.; Schlabach, A. J.; 36. National Committee for Clinical Laboratory Standards, Methods for Dilution
Graham, P. I.; Condra, J. H.; Gotlib, L.; Holloway, M. K.; Lin, J.; Chen, I. W.; Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically, 5th ed.,
Vastag, K.; Ostovic, D.; Anderson, P. S.; Emini, E. A.; Huff, J. R. Proc. Natl. Acad. National Committee for Clinical Laboratory Standards: Villanova, PA, 2000.
Sci. U.S.A. 1994, 91, 4096; (b) Vacca, J. P.; Dorsey, B. D.; Schleif, W. A.; Levine, R. Approved Standard. NCCLS Document M7–A5.
B.; McDaniel, S. L.; Darke, P. L.; Zugay, J.; Quintero, J. C.; Blahy, O. M.; Sardana, 37. Berge, J. M.; Broom, N. J. P.; Houge-Frydrych, C. S. V.; Jarvest, R. L.; Mensah, L.;
B. B.; Schlabach, A. J.; Graham, P. I.; Condra, J. H.; Gotlib, L.; Holloway, M. K.; Mcnair, D. J.; O’hanlon, P. J.; Pope, A. J.; Rittenhouse, S. J. Antibiot. 2000, 53,
Lin, J.; Chen, I. W.; Vastag, K.; Ostovic, D.; Anderson, P. S.; Emini, E. A.; Huff, J. R. 1282.

S-ar putea să vă placă și