Next Article in Journal
First Total Synthesis of Varioxiranol A
Next Article in Special Issue
Consequences of IDH1/2 Mutations in Gliomas and an Assessment of Inhibitors Targeting Mutated IDH Proteins
Previous Article in Journal
Drug-1,3,4-Thiadiazole Conjugates as Novel Mixed-Type Inhibitors of Acetylcholinesterase: Synthesis, Molecular Docking, Pharmacokinetics, and ADMET Evaluation
Previous Article in Special Issue
Aminobenzosuberone Scaffold as a Modular Chemical Tool for the Inhibition of Therapeutically Relevant M1 Aminopeptidases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Synthesis and AChE-Inhibitory Activity of New Benzimidazole Derivatives

by
Ulviye Acar Cevik
1,2,
Begüm Nurpelin Saglik
1,2,
Serkan Levent
1,2,
Derya Osmaniye
1,2,
Betul Kaya Cavuşoglu
1,2,
Yusuf Ozkay
1,2,* and
Zafer Asim Kaplancikli
1
1
Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, 26210 Eskişehir, Turkey
2
Doping and Narcotic Compounds Analysis Laboratory, Faculty of Pharmacy, Anadolu University, 26210 Eskişehir, Turkey
*
Author to whom correspondence should be addressed.
Molecules 2019, 24(5), 861; https://doi.org/10.3390/molecules24050861
Submission received: 31 January 2019 / Revised: 22 February 2019 / Accepted: 26 February 2019 / Published: 28 February 2019
(This article belongs to the Special Issue Medicinal Chemistry in Europe)

Abstract

:
Alzheimer’s disease (AD), one of the main causes of aged dementia, is a progressive and degenerative neurological disorder characterized by loss of cognition and memory. Although the symptomatic treatment of AD, particularly acetylcholinesterase inhibitors (AChEIs) based on the ‘cholinergic hypothesis’, has been successful in clinic, at present there is no cure for this disease. In this study, we designed compounds carrying benzimidazole and triazole rings on the same chemical skeleton so as to investigate their potential acetylcholinesterase and butyrylcholinesterase activity. Furthermore, molecular modeling study was performed to determine the binding mode of the best inhibitor to the AChE. Among them, compounds 3d and 3h, which featured 3,4-dihydroxy substitution at the phenyl ring and 5(6)-chloro substitution at the benzimidazole ring were found to be potent inhibitors of AChE. The inhibition kinetics of the two most active derivatives 3d and 3h were further studied. The kinetic displayed increasing slope and increasing intercept, which is consistent with a mixed inhibition. The IC50 and Ki values of 3d are 31.9 ± 0.1 nM and 26.2 nM, respectively. Compound 3h exhibited IC50 of 29.5 ± 1.2 nM and Ki of 24.8 nM. The above data compared favorably with data for donepezil (21.8 ± 0.9 nM) the reference compound in our study.

Graphical Abstract

1. Introduction

Neurodegeneration is defined as a progressive and often untreatable sequence that causes loss of specific neuron types, disregulation of neurons, distribution and central nervous system (CNS) dysfunction. Alzheimer’s disease (AD), which is one of the most important central nervous system disorders among various neurodegenerative diseases, is the fourth leading cause of death among the elderly. According to the World Alzheimer’s Report 2016, 46.8 million people worldwide were predicted by dementia in 2015 and the number of patients was predicted to triple by 2050. AD clinically includes the progressive degeneration of brain tissue that is influenced by the absence in acetylcholine (ACh) and has multifactorial pathology [1,2,3,4,5]. Accumulation of extracellular amyloid-beta (Aβ) in senile plaques, loss of cholinergic activity in certain parts of brain and intracellular neurofibrillary tangles including the hyperphosphorylated tau protein as well as neuroinflammation, responsible for neurodegenerative processes observed in AD [6,7,8].
The cholinergic hypothesis is one of the oldest, most robust and clinically confirmed among cases of mild severe dementia and Alzheimer’s hypotheses that suggest a relationship between the onset and progression of the disease. One of the major ways to increase the level of acetylcholine (ACh), which plays an important role in attention, learning, memory and motivation, is the inhibition of cholinesterase enzymes (ChEs). Cholinesterases belonging to the class of hydrolase enzyme are a widely distributed enzyme classified as acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). In patients with AD, an imbalance occurs between AChE and BChE. AChE inhibitors are preferred in the treatment of AD to protect the AChE norms because AChE exhibits more hydrolytic activity than BChE. Crystal structural studies with both AChE enzyme have revealed that they have different binding sites, containing the aromatic patch (AP), oxyanionic hole, peripheral anionic site (PAS), catalyist active site (CAS), acyl site and anionic site (AS). To improve effective acetylcholinesterase inhibitors (AChEI), acetylcholinesterase activity must be inhibited in the oxidation hole region (AS) and in the peripheral anionic region (PAS), because it causes accumulation of bound AChE amyloid peptide plaques in PAS [9,10,11,12,13,14,15,16,17]. Although there are many ongoing studies for the treatment of AD, only some drugs have been accepted by the FDA, such as donepezil, rivastigmin and tacrine [18].
Bioisosterism is a special molecular modification process that is applied over a lead compound (LC) and is a medicinal chemistry strategy for the rational design of new drugs. The capability of a group of bioisosteres to prompt similar biological activity has been recognized in compounds with common physicochemical properties such as benzazoles. They provide a single point for chemical variation, with a heteroatom (N, O, S) at the 1-position and with a nitrogen at the 3-position. This property facilitated a systematic investigation [19,20,21]. When the literature studies are evaluated, it can be learned that benzazole derivatives have inhibitory activity on acetylcholinesterase enzyme [17,22,23,24,25,26,27,28,29,30]. In addition, some benzimidazole derivatives have been shown to be useful as amyloid imaging probes because of their high binding affinity to Aβ aggregates and high uptake into the brain [31]. Also, benzimidazole scaffold is the ring isoster of indanone pharmacophore of donepezil which is one of the most important AChEI [32]. Benzimidazoles with different biological activities were thought to be an appropriate starting point for AD.
Considering that there is a limited choice of medicines available for the treatment of AD, we focused on using benzimidazole as a lead compound to develop new drug candidates to combat AD. In this study, 16 new benzimidazole derivatives were synthesized and cholinesterase inhibition activity was evaluated.

2. Results and Discussion

2.1. Chemistry

The syntheses of the target compounds 3ap were realized as summarized in Scheme 1. Known triazoles 1ad were synthesized according to our already published method [33,34]. The S-alkylation of the triazole ring of compounds 1ad with an appropriate 2-bromoacetophenone moiety 2ad using potassium carbonate as base generated the target derivatives 3ap.

2.2. Anticholinesterase Activity Assay

New benzimidazole-triazole derivatives were designed, synthesized, and evaluated for their potential to inhibit AChE and BChE. First, all the synthesized compounds were tested at 10−3 M and 10−4 M concentrations. Table 1 presents the acetylcholinesterase and butyrylcholinesterase inhibition of synthesized compounds and donepezil and tacrine at initial concentrations. According to activity results, the series generally displayed better inhibitor activity against AChE than BChE activity. At the 10−4 M concentration, compounds 3d, 3h, 3l and 3p indicated more than 50% activity. Then, the IC50 values of the active compounds were determined using 10−3–10−9 M concentrations against AChE along with the reference drug donepezil (Table 2). The IC50 value was calculated as 21.8 ± 0.9 nM for donepezil. The IC50 values of the compounds (5(6)chlorobenzimidazole) (3d and 3h) and (5(6)fluorobenzimidazole) (3l and 3p) were calculated as 31.9 ± 0.1 nM, 29.5 ± 1.2 nM and 169.4 ± 7.2 nM, 139.9 ± 5.1 nM, respectively for AChE. The compounds 3d, 3h, 3l and 3p among the series were those having the 3,4-dihydroxy substituent on the phenyl attached to benzimidazole. The strongest inhibitions against AChE were observed with 3d and 3h having a chloro substituent on the benzimidazole ring. The results show that replacing the methyl group attached to the triazole with an ethyl group does not create a significant activity change. The results demonstrated that the compounds 3d and 3h showed IC50 values similar activities compared to the reference drug against AChE.

2.3. Enzyme Kinetic Studies

Enzyme kinetic studies were carried out in order to determine the mechanism of AChE inhibition. The type of inhibition was forecast using linear Lineweaver-Burk plots. The substrate rate curves in the absence and presence of the most potent compounds 3d and 3h were recorded in the enzyme kinetics analysis. These compounds were prepared at concentrations of 2 × IC50, IC50 and IC50/2. The initial velocity measurements were recorded by using different substrate concentrations ranging from 600 µM to 18.75 µM for AChE enzyme. The secondary plots of the Km/Vmax (slope) versus varying concentrations were used to calculate Ki (intercept on the x-axis) values of compounds [35]. The graphical analyses of the steady-state inhibition data for AChE enzyme are shown in Figure 1 and Figure 2, respectively.
Reversible inhibition consists of the following subtypes: mixed-type, competitive, uncompetitive, or noncompetitive. These types of inhibition can be determined with the help of Lineweaver-Burk plots. As known from Lineweaver-Burk graphics, mixed-type inhibition can be refereed if the lines do not cross x- or y-axis at the same point. The Lineweaver-Burk plots of compounds 3d and 3h are presented in Figure 1 and Figure 2. As can be understand from these figures, compounds 3d and 3h display mixed type inhibition for AChE enzyme. Namely, both compounds can bind to the free enzyme or the enzyme-substrate complex. Also, this means that tested compounds 3d and 3h are reversible inhibitors, so these compounds could bind to the enzyme by noncovalent interactions such as hydrophobic interactions, ionic bonds, and hydrogen bonds without forming any chemical bonds or reacting with the enzyme. It is known that these interactions occur quickly and can be easily reversed. Therefore, the enzyme and inhibitor complex is rapidly degraded immediately prior to irreversible inhibition. This feature of tested compounds allows that such these reversible inhibitors display a lower risk of side effects than irreversible inhibitors. As a result, enzyme kinetics studies have shown the biological importance of compounds 3d and 3h due to their mixed type forces, unlike irreversible enzyme inhibitors.

2.4. Molecular Docking Studies

Docking studies were performed in order to gain more insight into the binding mode of compounds 3d, 3h, 3l and 3p to AChE enzyme. Studies were carried out by using the X-ray crystal structure of Homo sapiens AChE (hAChE PDB ID:4EY7) [36] obtained from Protein Data Bank server (www.pdb.org).
The docking poses of the compounds 3d and 3h are presented in Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7, whereas the poses of donepezil, compounds 3l and 3p are presented in Supporting Information Figures S1–S5. As seen from Figure S1 it is known that donepezil could bind strongly to the enzyme active region owing to its dual binding sites. The 5,6-dimethoxyindanone moiety binds to the peripheral anionic site (PAS) of the enzyme active region by interacting with the Trp286 and Phe295 amino acid residues. The rest of the structure, the benzylpiperidine group, binds to the catalytic anionic site (CAS) by interacting with Trp86. Especially as known that the interaction with Trp286 is essential for binding to PAS, whereas interaction with Trp86 is a key point of locating into CAS. When analyzing the docking results of compounds 3d, 3h, 3l and 3p (Supporting Information Figures S2–S5, Figure 3, Figure 4, Figure 5, Figure 6 and Figure 7), it can be seen that these compounds settle into the CAS and PAS regions of enzyme active site in a similar position as donepezil. Lipophilic parts of the compounds consist of fluoro/chloro-benzimidazole ring, while 1-(3,4-dihydroxyphenyl)-2-((4-methyl/ethyl-5-substitued-4H-1,2,4-triazol-3-yl)thio)ethan-1-ones are constituted by a polar basic center. The docking poses indicate that the PAS region of AChE interacts with lipophilic groups in the structure by interacting with Trp286, whereas the polar and basic groups bind to the CAS region of the enzyme by interacting with the Trp86 amino acid residue.
Two and three-dimensional binding modes of compounds 3d and 3h are given in Figure 4, Figure 5, Figure 6 and Figure 7. According to the docking poses, these compounds have six common interactions. The 5(6)-chloro-benzimidazole ring interacts with the phenyl of the Trp286 indole by a π–π interaction. The nitrogen atom of triazole forms a hydrogen bond with the hydroxy of Tyr124. The 3,4-dihydroxyphenyl ring creates a π–π interaction with the indole phenyl of Trp86. This dihydroxy substituent is also essential for polar interactions. The hydroxy group at the 4 position of the phenyl ring forms two hydrogen bonds with the carbonyl of Gly120 by acting as a hydrogen donor and the hydroxy of Tyr133 by acting as a hydrogen acceptor. The other hydroxy moiety, at the 3 position of the phenyl, creates another hydrogen bond with the carbonyl of Glu202.
The main difference between compounds 3d and 3h that they carry methyl and ethyl moieties, respectively, at the 4 position of the triazole ring. This suggests that the additional interaction of these compounds is caused by small differences in the conformational direction. Compound 3d has a π-π interaction between the triazole ring and the phenyl of Phe338. On the other hand, the hydroxy group at the 3 position of the phenyl ring forms an additional hydrogen bond with the carbonyl of Gly120 for compound 3h. Also, it is seen that the change of methyl at the 4 position of the triazole ring to an ethyl group does not cause a significant difference in the enzyme inhibition profile of these compounds. Furthermore, the substituents on the phenyl ring at the end of the structure are responsible for binding to the enzyme active site strongly and thus producing differences in terms of enzyme inhibition potency. It can be suggested that the presence of groups capable of hydrogen bonding at the 3 and 4 positions of the phenyl ring allow the compounds to be placed in stronger and more convenient positions. For compounds 3d and 3h, the hydroxy moiety at the 3 and 4 positions provide hydrogen bonds and therefore this situation enables them to show a more compatible binding potential than other compounds in the series. All these interactions show that compounds 3d and 3h could bind to AChE enzyme active region in a similar and proper conformation by similar interactions. Besides, it can be explained with the help of docking studies that these compounds display similar inhibition profiles on AChE enzyme with IC50 values of 31.9 ± 1.0 nM and 29.5 ± 1.2 nM, respectively.
The structurally fundamental difference of compounds 3l and 3p from compounds 3d and 3h is that the chlorine substituent on the benzimidazole ring is replaced by a fluorine atom. Thus, it may be expected that compounds 3l and 3p exhibit similar binding properties such as compounds 3d and 3h and demonstrate similar activity on AChE enzyme. However, according to the enzyme inhibition results, compounds 3l and 3p are the most potent derivatives after compounds 3d and 3h in the series. Compound 3l shows an AChE enzyme inhibition profile with an IC50 value of 169.4 ± 7.2 nM, however, compound 3p has an IC50 value of 139.9 ± 5.1 nM on AChE enzyme. The docking results help explain this difference. The poses of compounds 3l and 3p are presented in Supporting Information Figures S2–S5.
By comparing Figure 4 and Figure 5 and Supporting Information Figures S2 and S3, it can be seen that compound 3l shows the same five interactions as compound 3d, but compound 3l does not have the interaction between the triazole ring and the phenyl of Phe338. Also, the hydrogen bond between the hydroxy moiety at the 4 position of the phenyl ring anthe d hydroxy of Tyr133 could not be seen in the compound 3l.
The interaction differences observed between compounds 3d and 3l are also observed between compounds 3h and 3p (Figure 6 and Figure 7 and Supporting Information Figures S4 and S5). For compound 3p, the nitrogen atom of the triazole ring cannot form a hydrogen bond with the Tyr124 amino acid residue. Moreover, the hydroxy group at the 4 position of the phenyl ring does not form a hydrogen bond with hydroxy of Tyr133. All these losses of interactions mentioned above may explain why compounds 3l and 3p exhibit a lower inhibition profile than compounds 3d and 3f. Besides, it can be suggested that the presence of a fluorine atom instead of a chlorine on the benzimidazole ring cause distinctness in terms of conformational directions because the chlorine atom has a larger atomic volume in comparison with a fluorine atom. These conformational differences caused the interaction losses observed in compounds 3l and 3p.
It can be understood by looking a the enzyme inhibition assay that none of the synthesized compounds display significant activity against BChE enzyme. In order to illuminate this situation in silico, docking studies were performed for compound 3h by using the X-ray crystal structure of Homo sapiens BChE (hBChE PDB ID:4BDS) obtained from the Protein Data Bank server (www.pdb.org). The docking pose of compound 3h on BChE enzyme is presented in Supporting Information Figure S6. As can be seen from this figure, compound 3h could not bind to the BChE enzyme active region properly. This compound shows two interactions related only to the phenyl ring at the end of the structure. Namely, it is unable to display a general interaction profile with its whole chemical structure. The phenyl ring interacts with the imidazole of Hid438. Also, a hydrogen bond is observed between the hydroxy group at the 4 position of the phenyl ring and the carbonyl of Glu197. Consequently, it can be seen that compound 3h, selected as the example for the docking study on BChE enzyme, could not settle in the BChE enzyme active site in a strong and proper position as in AChE enzyme. The reason for the low BChe activity of the synthesized compounds can be explained as the inability of the general chemical skeleton to locate into the active site.

3. Materials and Methods

3.1. Chemistry

All chemicals used in the study were purchased either from Sigma-Aldrich (St. Louis, MO, USA) or Merck KGaA (Darmstadt, Germany), and used without further chemical or biological purification. Microwave syntheses were realized by using a Monowave 300 high-performance microwave reactor (Anton-Paar, Graz, Austria). Melting points of synthesized compounds were determined by using an automatic melting point determination system (MP90 series, Mettler-Toledo, Ohio, OH, USA) and were uncorrected. 1H-NMR and 13C-NMR spectra were recorded in DMSO-d6 by a Bruker digital FT-NMR spectrometer (Bruker Bioscience, MA, USA) at 300 MHz and 75 MHz, respectively. High resolution mass spectrometric studies were performed using an LCMS-IT-TOF system (Shimadzu, Kyoto, Japan). Chemical purities of the compounds were checked by classical TLC applications performed on silica gel 60 F254 (Merck KGaA); LCMS-IT-TOF chromatograms were also used for the same purpose.

3.1.1. 5(6)-Chloro/fluoro-2-((4-methylcarboxylate)phenyl)-1H-benzimidazole

In a microwave synthesis reactor vial (Anton-Paar, Graz, Austria, 30 mL), equipped with a magnetic stirrer, methyl-4-formyl benzoate (2.4 g, 0.015 mol) in DMF (10 mL) and sodium bisulfite (2.85 g, 0.015 mol) were heated under conditions of 240 °C and 10 bar for 5 min. The mixture was cooled, 5-chloro/fluoro-1,2-phenylenediamine (0.015 mol) was added into the vial and then reaction mixture was kept under same reaction conditions in the microwave reactor for 5 min. The mixture was cooled and poured into iced-water. The obtained solid was filtered, washed with water, dried and recrystallized from ethanol [33,34].

3.1.2. 4-(5(6)-Chloro/fluoro-1H-benzimidazol-2-yl)benzohydrazide

5(6)-Chloro/fluoro-2-((4-methylcarboxy)phenyl)-1H-benzimidazole (0.01 mol) and excess of hydrazine hydrate (5 mL) in ethanol (15 mL) was heated under the conditions of 150 °C and 10 bar for 10 min in the microwave synthesis reactor (Anton-Paar, Graz, Austria). The mixture was cooled and poured into iced-water. The obtained solid was filtered, washed with water, dried and recrystallized from ethanol [33,34].

3.1.3. N-methyl/ethyl-2-[4-(5(6)-chloro/fluoro-1H-benzimidazol-2-yl)benzoyl]hydrazine-1-carbothioamide

4-(5(6)-Chloro/fluoro-1H-benzimidazol-2-yl)benzohydrazide (0.01 mol) and methyl isothiocyanate or ethyl isothiocyanate (0.012 mol) in ethanol were refluxed for 2 h. The precipitated product was filtered, washed with ethanol and dried [34].

3.1.4. 4-Methyl/Ethyl-5[4-5(6)-chloro/fluoro-1H-benzimidazol-2-yl)phenyl)-4H-1,2,4-triazole-3-thiol (1ad)

N-methyl/ethyl-2[4-(5(6)-chloro/fluoro-1H-benzimidazol-2-yl)benzoyl]hydrazine-1-carbothio-amide (0.001 mol) in ethanol was refluxed under stirring for 2 h in the presence of NaOH (0.012 mol). After completion of reaction, the solution was acidified with HCl 37%, the precipitate was filtered, washed with water, dried and then recrystallized from ethanol [34].

3.1.5. 2-(4-(4-Methyl/ethyl-5-(2-(substitutedphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro/fluoro-1H-benzimidazoles 3ap

A solution of 1a or 1b (0.001 mol) in acetone (10 mL), an appropriate substituted 2-bromoacetophenone derivative (0.001 mol) and potassium carbonate (0.138 g, 0.001 mol) were refluxed at 40 °C for 12 h. The solvent was evaporated, residue was washed with water, dried and recrystallized from ethanol. The mass spectra of the compounds (3ap) are available in the Supplementary Materials.
2-(4-(4-Methyl-5-(2-(4-cyanophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3a). Yield: 84%. M.p. 269.5–271.8 °C. 1H-NMR: δ = 3.72 (3H, s, –CH3), 4.98 (2H, s, –CH2–), 7.25 (1H, t, J = 8.5 Hz, benzimidazole C–H), 7.57 (1H, d, J = 8.5 Hz, benzimidazole C-H), 7.72 (1H, br.s., benzimidazole C–H), 7.91 (2H, d, J = 8.5 Hz, 4-cyanophenyl C–H), 8.05 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 8.18 (2H, d, J = 8.5 Hz, 4-cyanophenyl C–H), 8.33 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 13.29 (1H, s, benzimidazole –NH). 13C-NMR: δ (ppm): 32.50, 41.19, 111.67, 113.30, 116.02, 118.55, 118.90, 120.78, 122.78, 123.45, 127.41, 128.77, 129.25, 129.53, 131.33, 133.29, 139.01, 145.19, 150.88, 155.29, 193.45. [M + H]+ calcd for C25H17ClN6OS: 485.0930; found: 485.0946.
2-(4-(4-Methyl-5-(2-(4-bromophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3b). Yield: 82%. M.p. 279.1–281.4 °C. 1H-NMR: δ = 3.72 (3H, s, –CH3), 4.93 (2H, s, –CH2–), 7.25 (1H, d, J = 8.1 Hz, benzimidazole C–H), 7.61–7.75 (2H, m, benzimidazole C–H), 7.77 (2H, d, J = 8.5 Hz, 4-bromophenyl C–H), 7.91 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H), 7.97 (2H, d, J = 8.6 Hz, 4-bromophenyl C–H), 8.33 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H), 13.27 (1H, s, benzimidazole –NH). 13C-NMR: δ = 32.51, 41.11, 111.64, 113.30, 118.90, 120.80, 122.92, 123.14, 126.81, 127.41, 128.39, 128.78, 129, 130.91, 131.33, 132.35, 134.76, 151.01, 155.25, 193.20. [M + H]+ calcd for C24H17BrClN5OS: 538.0060; found: 538.0098.
2-(4-(4-Methyl-5-(2-(4-methylphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3c). Yield: 79%. M.p. 254.9–256.3 °C. 1H-NMR: δ = 2.38 (3H, s, CH3), 3.71 (3H, s, –CH3), 4.91 (2H, s, –CH2–), 7.25 (1H, dd, J = 8.6–2.0 Hz, benzimidazole C–H), 7.34-7.37 (2H, m, Ar–C–H), 7.62-7.70 (2H, m, Ar–C–H), 7.89–7.94 (4H, m, Ar–C–H), 8.33 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 13.32 (1H, s, benzimidazole –NH). 13C-NMR: δ = 21.67, 32.48, 41.27, 106.76, 117.24, 123.14, 127.41, 127.81, 128.81, 129.03, 129.25, 129.82, 130.80, 131.30, 133.21, 133.70 144.77, 151.16, 152.23, 155.21, 193.35. [M + H]+ calcd for C25H20ClN5OS: 474.1148; found: 474.1150.
2-(4-(4-Methyl-5-(2-(3,4-dihydroxyphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benzimidazole (3d). Yield: 76%. M.p. 261.2–262.8 °C. 1H-NMR: δ = 3.71 (3H, s, –CH3), 4.80 (2H, s, –CH2), 6.81 (1H, d, J = 8.0 Hz, dihydroxyphenyl C–H), 7.25 (1H, dd, J = 8.6–2.0 Hz, benzimidazole C–H), 7.38-7.45 (2H, m, Ar–C–H), 7.6–7.73 (2H, m, Ar–CH), 7.91 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 8.33 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 13.26 (1H, s, benzimidazole -NH). 13C–NMR: δ = 32.47, 41.09, 114.67, 115.26, 115.62, 122.70, 123.13, 127.16, 127.41, 128.47, 128.85, 129.26, 130.77, 131.30, 138.96, 146.15, 151.41, 152.2, 152.79, 155.18, 191.65. [M + H]+ calcd for C24H18ClN5O3S: 492.0877; found: 492.0892.
2-(4-(4-Ethyl-5-(2-(4-cyanophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3e). Yield: 81%. M.p. 258.7–259.9 °C. 1H-NMR: δ = 1.28 (3H, t, J = 7.2, –CH3), 4.12 (2H, q, J = 7.2 Hz, –CH2), 5.03 (2H, s, –CH2), 7.24 (1H, dd, J = 8.6–1.9 Hz, benzimidazole C–H),7.62-7.68 (2H, m, benzimidazole C–H), 7.85 (2H, d, J = 8.4 Hz, 4-cyanophenyl C–H), 8.04 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H), 8.19 (2H, d, J = 8.4 Hz, 4-cyanophenyl C–H), 8.33 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H), 13.27 (1H, s, benzimidazole –NH). 13C-NMR: δ = 15.51, 36.23, 41.15, 116.04, 118.55, 119.28, 123.15, 127.55, 128.90, 129.29, 129.51, 129.83, 131.46, 132.93, 133.29, 139.03, 144.94, 150.39, 152.19, 154.78, 193.31. [M + H]+ calcd for C26H19ClN6OS: 499.1092; found: 499.1102.
2-(4-(4-Ethyl-5-(2-(4-bromophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3f). Yield: 80%. M.p. 249.3–251.4 °C. 1H-NMR: δ = 1.28 (3H, t, J = 7.20, –CH3), 4.12 (2H, q, J = 7.2 Hz, –CH2), 4.99 (2H, s, –CH2–), 7.26 (1H, dd, J = 8.6–2.0 Hz, benzimidazole C-H), 7.63–7.69 (2H, m, benzimidazole C–H), 7.79 (2H, d, J = 8.6 Hz, 4-bromophenyl), 7.85 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H), 7.98 (2H, d, J = 8.6 Hz, 4-bromophenyl), 8.33 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H). 13C-NMR: δ = 15.52, 34.01, 41.06, 116.52, 120.40, 123.20, 127.23, 127.56, 128.39, 128.98, 129.33, 130.92, 131.40, 131.89, 132.38, 134.82, 139.48, 150.50, 152.19, 154.73, 193.10. [M + H]+ calcd for C25H19BrClN5OS: 552.0249; found: 552.0255.
2-(4-(4-Ethyl-5-(2-(4-methylphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benz-imidazole (3g). Yield: 74%. M.p. 247.9–249.6 °C. 1H-NMR: δ = 1.28 (3H, t, J = 7.2, –CH3), 2.43 (3H, s, –CH3), 4.11 (2H, q, J = 7.2 Hz, –CH2), 4.98 (2H, s, –CH2–), 7.26 (1H, d, J = 8.4 Hz, benzimidazole C–H), 7.37 (2H, d, J = 8.2, 4-methylphenyl C–H), 7.60–7.75 (2H, m, benzimidazole C–H), 7.85 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 7.95 (2H, d, J = 8.2 Hz, 4-methylphenyl C–H), 8.33 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 13.28 (1H, s, benzimidazole –NH). 13C-NMR: δ = 15.53, 21.68, 33.58, 41.22, 111.68, 113.31, 118.98, 123.48, 127.54, 128.43, 129.03, 129.32, 129.85, 130.59, 131.43, 132.59, 133.27, 144.79, 150.42, 150.66, 154.69, 193.21. [M + H]+ calcd for C26H22ClN5OS: 488.1300; found: 488.1306.
2-(4-(4-Ethyl-5-(2-(3,4-dihydroxyphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-chloro-1H-benzimidazole (3h). Yield: 83%. M.p. 262.3–264.1 °C. 1H-NMR: δ = 1.27 (3H, t, J = 7.2 Hz, –CH3), 4.11 (2H, q, J = 7.2 Hz, –CH2), 4.88 (2H, s, –CH2–), 6.86 (1H, d, J = 8.3 Hz, dihydroxyphenyl C–H), 7.25 (1H, dd, J = 8.5–1.9 Hz, benzimidazole C–H), 7.40–7.47 (2H, m, dihydroxyphenyl C–H), 7.63–7.69 (2H, m, benzimidazole C–H), 7.85 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H), 8.34 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H). 13C-NMR: δ = 15.53, 36.24, 41.08, 111.36, 115.66, 118.96, 120.97, 122.48, 123.17, 125.42, 127.55, 128.29, 129.03, 129.31, 131.43, 133.31, 133.57, 145.86, 150.85, 151.93, 152.23, 154.66, 191.71. [M + H]+ calcd for C25H20ClN5O3S: 506.1038; found: 506.1048.
2-(4-(4-Methyl-5-(2-(4-cyanophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3i). Yield: 76%. M.p. 260.9–262.7 °C. 1H-NMR: δ = 3.72 (3H, s, –CH3), 4.98 (2H, s, –CH2), 7.08–7.11 (1H, m, benzimidazole C–H), 7.34–7.73 (2H, m, benzimidazole C–H), 7.90 (2H, d, J = 8.5 Hz, 4-cyanophenyl C–H), 8.05 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H), 8.19 (2H, d, J = 8.5 Hz, 4-cyanophenyl C–H), 8.32 (2H, d, J = 8.5 Hz, 1,4-disubstituted benzene C–H), 13.21 (1H, s, benzimidazole –NH). 13C-NMR: δ = 32.51, 41.17, 98.31 (d, J = 27.3 Hz), 104.95 (d, J = 23.3 Hz), 112.66 (d, J = 10.9 Hz), 120.44, 127.25 (d, J = 8.1 Hz), 128.51, 129.04, 129.25, 131.49, 132.25, 133.31, 135.74, 140.97, 144.75, 150.85, 152.82 (d, J = 2.8 Hz), 155.31, 157.95 (d, J = 233.5 Hz), 193.47. HRMS (m/z): [M + H]+ calcd for C25H17FN6OS: 469.1227; found: 469.1241.
2-(4-(4-Methyl-5-(2-(4-bromophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3j). Yield: 73%. M.p. 295.8–298.1 °C. 1H-NMR: δ = 3.71 (3H, s, –CH3), 4.93 (2H, s, –CH2–), 7.10 (1H, s, benzimidazole C–H), 7.37–7.56 (2H, m, benzimidazole C–H), 7.79 (2H, d, J = 8.2 Hz, 4-bromo-phenyl C–H), 7.90 (2H, d, J = 8.0 Hz, 1,4-disubstituted benzene C–H), 7.97 (2H, d, J = 8.2 Hz, 4-bromophenyl C–H), 8.32 (2H, d, J = 8.0 Hz, 1,4-disubstituted benzene C–H), 13.21 (1H, s, benzimidazole –NH). 13C-NMR: δ = 32.51, 41.09, 98.45 (d, J = 26.8 Hz), 104.82 (d, J = 24.3 Hz), 113.42 (d, J = 11.4 Hz), 121.52, 127.23 (d, J = 8.9 Hz), 128.93, 129.27, 130.94, 132.37, 134.5, 137.4, 140.39, 150.25, 152.73 (d, J = 2.6 Hz), 153.21, 155.70, 161.03 (d, J = 228.6 Hz), 193.56. HRMS (m/z): [M + H]+ calcd for C24H17BrFN5OS: 522.0374; found: 522.0394.
2-(4-(4-Methyl-5-(2-(4-methylphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3k). Yield: 71%. M.p. 284.3–286.9 °C. 1H-NMR: δ = 2.39 (3H, s, CH3), 3.71 (3H, s, –CH3), 4.92 (2H, s, –CH2–), 7.07–7.14 (1H, m, benzimidazole C–H), 7.36 (2H, d, J = 8.1 Hz, 4-methylphenyl C–H), 7.42-7.46 (1H, m, Ar–C–H), 7.62–7.66 (1H, m, Ar–CH C–H), 7.89–7.95 (4H, Ar–C–H), 8.33 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H). 13C-NMR: δ = 21.67, 32.48, 41.25, 99.46 (d, J = 25.3 Hz), 101.78, 104.85 (d, J = 21.3 Hz), 111.29 (d, J= 10.2 Hz), 116.37, 120.44, 127.52 (d, J = 9.6 Hz), 128.78, 129.05, 129.28, 129.83, 131.23, 133.22, 144.78, 151.18 (d, J = 2.9 Hz), 155.20, 159.37 (d, J = 236.8 Hz), 193.36. HRMS (m/z): [M + H]+ calcd for C25H20FN5OS: 458.1437; found: 458.1445.
2-(4-(4-Methyl-5-(2-(3,4-dihydroxyphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benzimidazole (3l). Yield: 77%. M.p. 277.8–279.2 °C. 1H-NMR: δ = 3.71 (3H, s, –CH3), 4.82 (2H, s, –CH2–), 6.85 (1H, d, J = 8.2 Hz, dihydroxyphenyl C–H), 7.09 (1H, br.s., Ar–C–H), 7.39–7.56 (4H, m, Ar–C–H), 7.90 (2H, d, J = 8.2 Hz, 1,4-disubstituted benzene C–H), 8.32 (2H, d, J = 8.2 Hz, 1,4-disubstituted benzene C–H), 13.22 (1H, s, denzimidazole –NH). 13C-NMR: δ = 32.46, 41.07, 98.33 (d, J = 28.9 Hz), 104.99 (d, J = 30.7 Hz), 110.81, 111.49 (d, J = 11.4 Hz), 112.75, 115.64, 120.41, 122.51, 127.38 (d, J = 9.4 Hz), 128.72, 129.26, 131.48, 132.21, 145.83, 150.75, 151.59 (d, J = 2.6 Hz), 153.62, 155.21, 162.78 (d, J = 236.8 Hz), 191.88. HRMS (m/z): [M + H]+ calcd for C24H18FN5O3S: 476.1176; found: 476.1187.
2-(4-(4-Ethyl-5-(2-(4-cyanophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3m). Yield: 70%. M.p. 225.4–227.1 °C. 1H-NMR: δ = 1.27 (3H, t, J = 7.2 Hz,–CH3), 4.11 (2H, q, J = 7.2 Hz, –CH2), 5.03 (2H, s, –CH2–), 7.10 (1H, t, J = 8.5 Hz, benzimidazole C–H), 7.43 (1H, br. s, benzimidazole C–H), 7.62 (1H, br.s., benzimidazole C–H), 7.83 (2H, d, J = 8.4 Hz, 4-cyanophenyl C–H), 8.05 (2H, d, J = 8.4, 1,4-disubstituted benzene C–H), 8.19 (2H, d, J = 8.4, 4-cyanophenyl C–H), 8.31 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 13.24 (1H, s, benzimidazole –NH). 13C-NMR: δ = 15.49, 40.30, 41.08, 98.19 (d, J = 25.2 Hz), 104.57 (d, J = 31.1 Hz), 111.55 (d, J = 11.3 Hz), 116.02, 118.56, 127.40, 128.71, 129.42 (d, J = 14.9 Hz), 131.60, 133.31, 134.81, 139.03, 142.18, 150.39, 152.18, 152.75 (d, J= 2.8 Hz), 154.81, 162.24 (d, J = 228.4 Hz), 193.33. HRMS (m/z): [M + H]+ calcd for C26H19N5FN6OS: 483.1386; found: 483.1398.
2-(4-(4-Ethyl-5-(2-(4-bromophenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3n). Yield: 73%. M.p. 265.8–267.9 °C. 1H-NMR: δ = 1.28 (3H, t, J = 7.2 Hz, –CH3), 4.11 (2H, q, J = 7.2 Hz, –CH2), 4.99 (2H, s, –CH2–), 7.06–7.13 (1H, m, benzimidazole C–H), 7.43 (1H, d, J = 8.5 Hz, benzimidazole C–H), 7.61-7.66 (1H, m, benzimidazole C–H), 7.79 (2H, d, J = 8.5 Hz, 4-bromophenyl C–H), 7.84 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H), 7.98 (2H, d, J = 8.5 Hz, 4-bromophenyl C–H), 8.32 (2H, d, J = 8.3 Hz, 1,4-disubstituted benzene C–H), 13.20 (1H, s, benzimidazole –NH). 13C-NMR: δ = 15.53, 40.80, 48.13, 99.45 (d, J = 24.8 Hz), 105.64 (d, J = 22.4 Hz), 111.25 (d, J = 11.2 Hz), 124.32, 127.40 (d, J = 9.2 Hz), 128.39, 128.80, 129.31, 130.92, 132.38, 134.81, 139.13, 150.47, 151.60 (d, J = 2.4 Hz), 152.15, 154.76, 159.28 (d, J = 234.1 Hz), 193.09. HRMS (m/z): [M + H]+ calcd for C25H19BrFN5OS: 536.0537; found: 536.0550.
2-(4-(4-Ethyl-5-(2-(4-methylphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benz-imidazole (3o). Yield: 82%. M.p. 260.6–262.2 °C. 1H-NMR: δ = 1.28 (3H, t, J = 7.1 Hz,–CH3), 2.39 (3H, s, CH3), 4.10 (2H, q, J = 7.1 Hz,–CH2), 4.98 (2H, s, –CH2–), 7.10 (1H, t, J = 8.5 Hz, benzimidazole C–H), 7.37 (2H, d, J = 7.7 Hz, 4-methylphenyl C–H), 7.42 (1H, br.s, benzimidazole C–H), 7.63 (1H, br.s, benzimidazole C–H), 7.84 (2H, d, J = 8.1 Hz, 1,4-disubstituted benzene C–H), 7.95 (2H, d, J = 7.8 Hz, 4-methylphenyl C–H), 8.32 (2H, d, J = 8.1 Hz, 1,4-disubstituted benzene C–H), 13.23 (1H, s, benzimidazole –NH). 13C-NMR: δ = 15.53, 21.68, 36.28, 41.21, 99.63 (d, J = 27.6 Hz), 104.32 (d, J = 24.8 Hz), 111.52 (d, J = 11.6 Hz), 123.46, 127.39 (d, J = 10.2 Hz), 128.82, 129.03, 129.30, 129.84, 131.62, 133.26, 144.79, 150.64 (d, J = 2.4 Hz), 152.72, 154.72, 159.24 (d, J = 234.7 Hz), 193.21. HRMS (m/z): [M + H]+ calcd for C26H22FN5OS: 472.1602; found: 472.1604.
2-(4-(4-Ethyl-5-(2-(3,4-dihydroxyphenyl)-2-oxo-ethylthio)-4H-1,2,4-triazol-3yl)-phenyl)-5(6)-fluoro-1H-benzimidazole (3p). Yield: 81%. M.p. 243.9–245.4 °C. 1H-NMR: δ = 1.26 (3H, t, J = 7.2 Hz,–CH3), 4.10 (2H, q, J = 7.2 Hz,–CH2), 4.77 (2H, s, –CH2), 6.48 (1H, d, J = 8.4 Hz, dihydroxyphenyl C–H), 7.04–7.11 (1H, m., benzimidazole C–H), 7.20 (1H, d, J = 2.3 Hz, dihydroxyphenyl C–H), 7.34 (1H, dd, J = 8.4–2.3 Hz, dihydroxyphenyl C–H), 7.42 (1H, dd, J = 9.5–2.4 Hz, benzimidazole C–H), 7.60–7.64 (1H, m, Ar–C–H), 7.83 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H), 8.35 (2H, d, J = 8.4 Hz, 1,4-disubstituted benzene C–H). 13C-NMR: δ = 15.56, 36.04, 41.16, 101.79 (d, J = 30.8 Hz), 110.90 (d, J = 24.8 Hz), 111.84, 116.50 (d, J = 12.3 Hz), 121.39, 122.55, 124.21, 128.86, 129.35 (d, J = 8.9 Hz), 130.61, 131.75, 134.81, 139.16, 147.72, 151.16 (d, J = 3.9 Hz), 152.40, 154.65, 159.21 (d, J = 241.6 Hz), 160.87, 189.78. HRMS (m/z): [M + H]+ calcd for C25H20FN5O3S: 490.1328; found: 490.1344.

3.2. Anticholinesterase Activity Assay

The inhibitory activities of the compounds against AChE and BChE were determined in 96-well plates by modified Ellman’s method as defined in our previous studies [17,37,38,39]. Donepezil and tacrine were used as reference drugs. A robotic system, Biotek Precision XS (Winooski, VT, USA), was used for all pipetting processes in the enzyme inhibition assay. Measurements of the percentage inhibition were carried out at 412 nm by using a BioTek-Synergy H1 microplate reader (Winooski, VT, USA). Inhibition potencies of synthesized compounds and IC50 of selected derivatives were calculated as reported previously [17,37,38,39].

3.3. Kinetic Studies of Enzyme Inhibition

Enzyme kinetic studies were performed for compounds 3d and 3h in order to determine the inhibition type on AChE. Enzyme kinetic assay was carried out using Ellman’s method according to the previous studies reported by our research group [17,37,38,39].

3.4. Molecular Docking Studies

In order to determine the binding modes of compounds 3d and 3h on AChE enzyme active site, docking studies were performed by using in silico procedure. X-ray crystal structure of AChE (PDB ID: 4EY7) [34] was retrieved from the Protein Data Bank server (www.pdb.org). The docking procedure was applied as described in our previous works [17,37,38,39].

4. Conclusions

AChE and BuChE are enzymes which play an important role in memory and cognition. They catalyse the hydrolysis of acetylcholine causing a loss of communication between nerve cells. This leads to a loss of brain function and causes AD. Pharmaceutical research has thus been focusing on cholinesterase inhibitors as treatments for cognitive disorders. This study aimed to design and synthesized benzimidazole-triazole derivatives and tested their activity against AChE and BuChE. Compounds 3d and 3h showed good AChE inhibition, but poor selectivity for BChE. In order to clarify the type of inhibition and their in silico properties, enzyme kinetics and binding studies were carried out for these compounds. These compounds were assessed as reversible and mixed-type AChE enzyme inhibitors. Also, it was shown that compounds 3d and 3h could bind to the AChE enzyme active site in a similar manner and proper positions. Consequently, this study presents important results for developing new agents for neurodegenerative diseases such as Alzheimer’s diseases.

Supplementary Materials

The following are available online at https://www.mdpi.com/1420-3049/24/5/861/s1.

Author Contributions

Y.O., Z.A.K., conceived and designed the experiments; U.A.C. performed the synthesis; S.L. performed analysis studies; B.N.S. performed activity tests; B.N.S. performed docking studies; D.O., B.K.C., B.N.S., S.L., U.A.C., Y.O. and Z.A.K. wrote the paper.

Funding

This study was financially supported by Anadolu University Scientific Projects Fund, Project No: 1805S189.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Madav, Y.; Wairkar, S.; Prabhakar, B. Recent therapeutic strategies targeting beta amyloid and tauopathies in Alzheimer’s disease. Brain Res. Bull. 2019, 146, 171–184. [Google Scholar] [CrossRef] [PubMed]
  2. Tan, S.H.; Karri, V.; Tay, N.W.R.; Chang, K.H.; Ah, H.Y.; Ng, P.Q.; Ho, H.S.; Keh, H.W.; Candasamy, M. Emerging pathways to neurodegeneration: Dissecting the critical molecular mechanisms in Alzheimer’s disease, Parkinson’s disease. Biomed. Pharmacother. 2019, 111, 765–777. [Google Scholar] [CrossRef] [PubMed]
  3. Huang, C.; Chu, H.; Ma, Y.; Zhou, Z.; Dai, C.; Huang, X.; Fang, L.; Ao, Q.; Huang, D. The neuroprotective effect of deep brain stimulation at nucleus basalis of Meynert in transgenic mice with Alzheimer’s disease. Brain Stimul. 2019, 12, 161–174. [Google Scholar] [CrossRef] [PubMed]
  4. Burmaoglu, S.; Yilmaz, A.O.; Polat, M.F.; Kaya, R.; Gulcin, İ.; Algul, O. Synthesis and Biological Evaluation of Novel Tris-Chalcones as Potent Carbonic Anhydrase, Acetylcholinesterase, Butyrylcholinesterase and α-Glycosidase Inhibitors. Bioorg. Chem. 2019, 85, 191–197. [Google Scholar] [CrossRef] [PubMed]
  5. Ibrahim, M.M.; Gabr, M.T. Multitarget therapeutic strategies for Alzheimer’s disease. Neural. Regen. Res. 2019, 14, 437. [Google Scholar] [PubMed]
  6. Pilipenko, V.; Narbute, K.; Beitnere, U.; Rumaks, J.; Pupure, J.; Jansone, B.; Klusa, V. Very low doses of muscimol and baclofen ameliorate cognitive deficits and regulate protein expression in the brain of a rat model of streptozocin-induced Alzheimer’s disease. Eur. J. Pharmacol. 2018, 818, 381–399. [Google Scholar] [CrossRef] [PubMed]
  7. Gu, J.; Anumala, U.R.; Heyny-von Haußen, R.; Hölzer, J.; Goetschy-Meyer, V.; Mall, G.; Hılger, I.; Czech, C.; Schmidt, B. Design, synthesis and biological evaluation of trimethine cyanine dyes as fluorescent probes for the detection of tau fibrils in Alzheimer’s disease brain and olfactory epithelium. Chem. Med. Chem. 2013, 8, 891–897. [Google Scholar] [CrossRef] [PubMed]
  8. Javed, H.; Khan, A.; Vaibhav, K.; Khan, M.M.; Ahmad, A.; Ahmad, M.E.; Ahmad, A.; Tabassum, R.; Islam, F.; Safhi, M.M.; et al. Taurine ameliorates neurobehavioral, neurochemical and immunohistochemical changes in sporadic dementia of Alzheimer’s type (SDAT) caused by intracerebroventricular streptozotocin in rats. Neurol. Sci. 2013, 34, 2181–2192. [Google Scholar] [CrossRef] [PubMed]
  9. Kumar, D.; Gupta, S.K.; Ganeshpurkar, A.; Gutti, G.; Krishnamurthy, S.; Modi, G.; Singh, S.K. Development of Piperazinediones as dual inhibitor for treatment of Alzheimer’s disease. Eur. J. Med. Chem. 2018, 150, 87–101. [Google Scholar] [CrossRef] [PubMed]
  10. Saeed, A.; Zaib, S.; Ashraf, S.; Iftikhar, J.; Muddassar, M.; Zhang, K.Y.; Iqbal, J. Synthesis, cholinesterase inhibition and molecular modelling studies of coumarin linked thiourea derivatives. Bioorg. Chem. 2015, 63, 58–63. [Google Scholar] [CrossRef] [PubMed]
  11. Najafi, Z.; Mahdavi, M.; Saeedi, M.; Karimpour-Razkenari, E.; Edraki, N.; Sharifzadeh, M.; Khanavi, M.; Akbarzadeh, T. Novel tacrine-coumarin hybrids linked to 1, 2, 3-triazole as anti-Alzheimer’s compounds: In vitro and in vivo biological evaluation and docking study. Bioorg. Chem. 2019, 83, 303–316. [Google Scholar] [CrossRef] [PubMed]
  12. Nicolet, Y.; Lockridge, O.; Masson, P.; Fontecilla-Camps, J.C.; Nachon, F. Crystal structure of human butyrylcholinesterase and of its complexes with substrate and products. J. Biol. Chem. 2003, 278, 41141–41147. [Google Scholar] [CrossRef] [PubMed]
  13. Mallender, W.D.; Szegletes, T.; Rosenberry, T.L. Acetylthiocholine binds to Asp74 at the peripheral site of human acetylcholinesterase as the first step in the catalytic pathway. Biochemistry 2000, 39, 7753–7763. [Google Scholar] [CrossRef] [PubMed]
  14. Mehfooz, H.; Saeed, A.; Sharma, A.; Albericio, F.; Larik, F.A.; Jabeen, F.; Channar, P.A.; Flörke, U. Dual Inhibition of AChE and BChE with the C-5 Substituted Derivative of Meldrum’s Acid: Synthesis, Structure Elucidation, and Molecular Docking Studies. Crystals 2017, 7, 211. [Google Scholar] [CrossRef]
  15. Guevara, J.A.; Trujillo, J.G.; Quintana, D.; Jiménez, H.A.; Arellano, M.G.; Bahena, J.R.; Tamay, F.; Ciprés, F.J. Acetylcholinesterase inhibition by products generated in situ from the transformation of N-arylisomaleimides. Med. Chem. Res. 2018, 27, 989–1003. [Google Scholar] [CrossRef]
  16. Kim, J.; Malpani, Y.R.; Lee, J.; Shin, J.S.; Han, S.B.; Jung, Y.S. Novel tacrine-pyridinium hybrid reactivators of organophosphorus-inhibited acetylcholinesterase: Synthesis, molecular docking, and in vitro reactivation study. Bioorg. Med. Chem. Lett. 2018, 28, 3784–3786. [Google Scholar] [CrossRef] [PubMed]
  17. Özkay, Ü.D.; Can, Ö.D.; Sağlık, B.N.; Cevik, U.A.; Levent, S.; Özkay, Y.; Ilgın, S.; Atlı, Ö. Design, synthesis, and AChE inhibitory activity of new benzothiazole–piperazines. Bioorg. Med. Chem. Lett. 2016, 26, 5387–5394. [Google Scholar] [CrossRef] [PubMed]
  18. Gul, H.; Demirtas, A.; Ucar, G.; Taslimi, P.; Gulcin, I. Synthesis of Mannich bases by two different methods and evaluation of their acetylcholine esterase and carbonic anhydrase inhibitory activities. Lett. Drug Des. Discov. 2017, 14, 573–580. [Google Scholar] [CrossRef]
  19. Lima, L.M.; Barreiro, E.J. Bioisosterism: A useful strategy for molecular modification and drug design. Curr. Med. Chem. 2005, 12, 23–49. [Google Scholar] [CrossRef] [PubMed]
  20. Patani, G.A.; LaVoie, E.J. Bioisosterism: A rational approach in drug design. Chem. Rev. 1996, 96, 3147–3176. [Google Scholar] [CrossRef] [PubMed]
  21. Havrylyuk, D.; Heidary, D.K.; Nease, L.; Parkin, S.; Glazer, E.C. Photochemical Properties and Structure-Activity Relationships of RuII Complexes with Pyridylbenzazole Ligands as Promising Anticancer Agents. Eur. J. Inorg. Chem. 2017, 12, 1687–1694. [Google Scholar] [CrossRef] [PubMed]
  22. Hiremathad, A.; Keri, R.S.; Esteves, A.R.; Cardoso, S.M.; Chaves, S.; Santos, M.A. Novel Tacrine-Hydroxyphenylbenzimidazole hybrids as potential multitarget drug candidates for Alzheimer’s disease. Eur. J. Med. Chem. 2018, 148, 255–267. [Google Scholar] [CrossRef] [PubMed]
  23. Salehi, N.; Mirjalili, B.B.F.; Nadri, H.; Abdolahi, Z.; Forootanfar, H.; Samzadeh-Kermani, A.; Küçükkılınç, T.T.; Ayazgok, B.; Emami, S.; Haririan, I.; et al. Synthesis and biological evaluation of new N-benzylpyridinium-based benzoheterocycles as potential anti-Alzheimer’s agents. Bioorg. Chem. 2019, 83, 559–568. [Google Scholar] [CrossRef] [PubMed]
  24. Santana, M.V.; Castro, H.C.; Abreu, P.A. Molecular Modeling of Benzimidazole Derivatives: A Promising Series of GluN2B Selective NMDA Receptor Antagonists. Curr. Drug Therapy 2018, 13, 152–161. [Google Scholar] [CrossRef]
  25. Murtaza, S.; Tatheer, A. 4-Acetamidobenzaldehyde derivatives as biological active candidates; synthesis, anti-oxidant, Anti-Alzheimer and DNA binding studies. Lett. Drug Des. Discov. 2018, 15, 957–968. [Google Scholar] [CrossRef]
  26. Turan-Zitouni, G.; Hussein, W.; Saglik, B.N.; Baysal, M.; Kaplancikli, Z.A. Fighting Against Alzheimer’s Disease: Synthesis of New Pyrazoline and Benzothiazole Derivatives as New Acetylcholinesterase and MAO Inhibitors. Lett. Drug Des. Discov. 2018, 15, 414–427. [Google Scholar] [CrossRef]
  27. Chaves, S.; Hiremathad, A.; Tomás, D.; Keri, R.S.; Piemontese, L.; Santos, M.A. Exploring the chelating capacity of 2-hydroxyphenyl-benzimidazole based hybrids with multi-target ability as anti-Alzheimer’s agents. New J. Chem. 2018, 42, 16503–16515. [Google Scholar] [CrossRef]
  28. Pouramiri, B.; Moghimi, S.; Mahdavi, M.; Nadri, H.; Moradi, A.; Tavakolinejad-Kermani, E.; Firoozpour, L.; Asadipour, A.; Foroumadi, A. Synthesis and anticholinesterase activity of new substituted benzo [d] oxazole-based derivatives. Chem. Biol. Drug Des. 2017, 89, 783–789. [Google Scholar] [CrossRef] [PubMed]
  29. Sarıkaya, G.; Çoban, G.; Parlar, S.; Tarikogullari, A.H.; Armagan, G.; Erdoğan, M.A.; Alptüzün, V.; Alpan, A.S. Multifunctional cholinesterase inhibitors for Alzheimer’s disease: Synthesis, biological evaluations, and docking studies of o/p-propoxyphenylsubstituted-1H-benzimidazole derivatives. Arch. Pharm. 2018, 351, 1800076. [Google Scholar]
  30. Unsal-Tan, O.; Ozadali-Sari, K.; Ayazgok, B.; Küçükkılınç, T.T.; Balkan, A. Novel 2-Arylbenzimidazole derivatives as multi-targeting agents to treat Alzheimer’s disease. Med. Chem. Res. 2017, 26, 1506–1515. [Google Scholar] [CrossRef]
  31. Ozadali-Sari, K.; Küçükkılınç, T.T.; Ayazgok, B.; Balkan, A.; Unsal-Tan, O. Novel multi-targeted agents for Alzheimer’s disease: Synthesis, biological evaluation, and molecular modeling of novel 2-[4-(4-substitutedpiperazin-1-yl) phenyl] benzimidazoles. Bioorg. Chem. 2017, 72, 208–214. [Google Scholar] [CrossRef] [PubMed]
  32. Alpan, A.S.; Parlar, S.; Carlino, L.; Tarikogullari, A.H.; Alptüzün, V.; Güneş, H.S. Synthesis, biological activity and molecular modeling studies on 1H-benzimidazole derivatives as acetylcholinesterase inhibitors. Bioorg. Med. Chem. 2013, 21, 4928–4937. [Google Scholar] [CrossRef] [PubMed]
  33. Karaburun, A.Ç.; Kaya Çavuşoğlu, B.; Acar Çevik, U.; Osmaniye, D.; Sağlık, B.N.; Levent, S.; Özkay, Y.; Atlı, Ö.; Koparal, A.S.; Kaplancıklı, Z.A. Synthesis and Antifungal Potential of Some Novel Benzimidazole-1,3,4-Oxadiazole Compounds. Molecules 2019, 24, 191. [Google Scholar] [CrossRef] [PubMed]
  34. Karaca Gençer, H.; Acar Çevik, U.; Levent, S.; Sağlık, B.N.; Korkut, B.; Özkay, Y.; Ilgın, S.; Öztürk, Y. New Benzimidazole-1,2,4-Triazole Hybrid Compounds: Synthesis, Anticandidal Activity and Cytotoxicity Evaluation. Molecules 2017, 22, 507. [Google Scholar] [CrossRef] [PubMed]
  35. Can, Ö.D.; Osmaniye, D.; Demir Özkay, Ü.; Sağlık, B.N.; Levent, S.; Ilgın, S.; Baysal, M.; Özkay, Y.; Kaplancıklı, Z.A. MAO enzymes inhibitory activity of new benzimidazole derivatives including hydrazone and propargyl side chains. Eur. J. Med. Chem. 2017, 131, 92–106. [Google Scholar] [CrossRef] [PubMed]
  36. Cheung, J.; Rudolph, M.J.; Burshteyn, F.; Cassidy, M.S.; Gary, E.N.; Love, J.; Height, J.J. Structures of human acetylcholinesterase in complex with pharmacologically important ligands. J. Med. Chem. 2012, 55, 10282–10286. [Google Scholar] [CrossRef] [PubMed]
  37. Sağlık, B.N.; Ilgın, S.; Özkay, Y. Synthesis of new donepezil analogues and investigation of their effects on cholinesterase enzymes. Eur. J. Med. Chem. 2016, 124, 1026–1040. [Google Scholar] [CrossRef] [PubMed]
  38. Hussein, W.; Sağlık, B.N.; Levent, S.; Korkut, B.; Ilgın, S.; Özkay, Y.; Kaplancıklı, Z.A. Synthesis and biological evaluation of new cholinesterase inhibitors for Alzheimer’s disease. Molecules 2018, 23, 2033. [Google Scholar] [CrossRef] [PubMed]
  39. Tok, F.; Koçyiğit Kaymakçıoğlu, B.; Sağlık, B.N.; Levent, S.; Özkay, Y.; Kaplancıklı, Z.A. Synthesis and biological evaluation of new pyrazolone Schiff bases as monoamine oxidase and cholinesterase inhibitors. Bioorg. Chem. 2019, 84, 41–50. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds 3ap are available from the authors.
Scheme 1. Synthesis way of the compounds 1ad, 3ap.
Scheme 1. Synthesis way of the compounds 1ad, 3ap.
Molecules 24 00861 sch001

CompoundsR1R2R3R4Yield (%)
1a–Cl–CH3--81
1b–Cl–C2H5--78
1d–F–CH3--77
1c–F–C2H5--75
3a–Cl–CH3–H–CN84
3b–Cl–CH3–H–Br82
3c–Cl–CH3–H–CH379
3d–Cl–CH3–OH–OH76
3e–Cl–C2H5–H–CN81
3f–Cl–C2H5–H–Br80
3g–Cl–C2H5–H–CH374
3h–Cl–C2H5–OH–OH83
3i–F–CH3–H–CN76
3j–F–CH3–H–Br73
3k–F–CH3–H–CH371
3l–F–CH3–OH–OH77
3m–F–C2H5–H–CN70
3n–F–C2H5–H–Br73
3o–F–C2H5–H–CH382
3p–F–C2H5–OH–OH81
Figure 1. (A) Lineweaver-Burk plots for the inhibition of AChE by compound 3d. [S], substrate concentration (μM); V, reaction velocity (abs/min)−1. (B) Secondary plot for calculation of steady-state inhibition constant (Ki) of compound 3d. Ki was calculated as 0.0262 μM.
Figure 1. (A) Lineweaver-Burk plots for the inhibition of AChE by compound 3d. [S], substrate concentration (μM); V, reaction velocity (abs/min)−1. (B) Secondary plot for calculation of steady-state inhibition constant (Ki) of compound 3d. Ki was calculated as 0.0262 μM.
Molecules 24 00861 g001
Figure 2. (A) Lineweaver-Burk plots for the inhibition of AChE by compound 3h. [S], substrate concentration (μM); V, reaction velocity (abs/min)−1. (B) Secondary plot for calculation of steady-state inhibition constant (Ki) of compound 3h. Ki was calculated as 0.0248 μM.
Figure 2. (A) Lineweaver-Burk plots for the inhibition of AChE by compound 3h. [S], substrate concentration (μM); V, reaction velocity (abs/min)−1. (B) Secondary plot for calculation of steady-state inhibition constant (Ki) of compound 3h. Ki was calculated as 0.0248 μM.
Molecules 24 00861 g002
Figure 3. The superimposition pose of compounds 3d and 3h (red and green colored, respectively) in the enzyme active site (AChE PDB Code: 4EY7).
Figure 3. The superimposition pose of compounds 3d and 3h (red and green colored, respectively) in the enzyme active site (AChE PDB Code: 4EY7).
Molecules 24 00861 g003
Figure 4. Two-dimensional interaction mode of compound 3d in the enzyme active site of AChE.
Figure 4. Two-dimensional interaction mode of compound 3d in the enzyme active site of AChE.
Molecules 24 00861 g004
Figure 5. The interacting mode of compound 3d in the active region of AChE. The inhibitor, colored with red, and the important residues, colored with white, in the active site of the enzyme are presented by tube model.
Figure 5. The interacting mode of compound 3d in the active region of AChE. The inhibitor, colored with red, and the important residues, colored with white, in the active site of the enzyme are presented by tube model.
Molecules 24 00861 g005
Figure 6. Two-dimensional interaction mode of compound 3h in the enzyme active site of AChE.
Figure 6. Two-dimensional interaction mode of compound 3h in the enzyme active site of AChE.
Molecules 24 00861 g006
Figure 7. The interacting mode of compound 3h in the active region of AChE. The inhibitor, colored with green, and the important residues, colored with white, in the active site of the enzyme are presented by tube model.
Figure 7. The interacting mode of compound 3h in the active region of AChE. The inhibitor, colored with green, and the important residues, colored with white, in the active site of the enzyme are presented by tube model.
Molecules 24 00861 g007
Table 1. % Inhibition of all compounds 5ap against AChE and BChE enzymes.
Table 1. % Inhibition of all compounds 5ap against AChE and BChE enzymes.
Compounds% Inhibition
AChEBChE
10−3 M10−4 M10−3 M10−4 M
3a68.18 ± 1.0547.26 ± 0.6240.28 ± 0.9825.13 ± 0.56
3b44.20 ± 0.8726.18 ± 0.6328.15 ± 0.9718.42 ± 0.88
3c39.18 ± 0.7117.89 ± 0.5426.11 ± 0.7420.21 ± 0.61
3d98.55 ± 1.1793.99 ± 1.1051.19 ± 0.9938.12 ± 0.41
3e77.29 ± 1.0641.22 ± 0.8438.22 ± 0.6322.41 ± 0.47
3f47.22 ± 0.6724.08 ± 0.4829.15 ± 0.5917.88 ± 0.40
3g37.20 ± 0.6818.33 ± 0.3831.89 ± 0.9924.75 ± 0.95
3h98.25 ± 1.1294.55 ± 1.0861.26 ± 0.9035.20 ± 0.42
3i61.22 ± 1.0238.28 ± 0.6743.51 ± 0.7818.49 ± 0.50
3j48.51 ± 0.8818.39 ± 0.6029.64 ± 0.8721.44 ± 0.70
3k42.29 ± 0.7127.88 ± 0.6736.17 ± 0.9321.16 ± 0.97
3l94.58 ± 1.7483.20 ± 1.1355.43 ± 0.4729.88 ± 0.38
3m59.03 ± 0.8839.26 ± 0.4648.11 ± 0.3820.66 ± 0.45
3n49.27 ± 0.5530.21 ± 0.4927.10 ± 0.5318.77 ± 0.42
3o28.06 ± 0.3319.22 ± 0.3025.99 ± 0.3820.08 ± 0.46
3p87.26 ± 1.1781.25 ± 1.8853.19 ± 0.4631.44 ± 0.38
Donepezil99.42 ± 1.8597.26 ± 1.24--
Tacrine--98.52 ± 1.6695.50 ± 1.74
Table 2. IC50 values of selected compounds (3d, 3h, 3l and 3p) and donepezil, as a reference drug, on AChE enzyme.
Table 2. IC50 values of selected compounds (3d, 3h, 3l and 3p) and donepezil, as a reference drug, on AChE enzyme.
Compounds% Inhibition of AChEAChE IC50 (nM)
10−5 M10−6 M10−7 M10−8 M10−9 M
3d87.26 ± 1.2878.85 ± 1.1967.18 ± 0.9245.32 ± 0.8215.75 ± 0.4831.9 ± 0.1
3h90.23 ± 1.0381.25 ± 1.0865.32 ± 0.9744.28 ± 0.7117.20 ± 0.5829.5 ± 1.2
3l73.18 ± 1.1768.20 ± 0.9748.11 ± 0.6132.29 ± 0.5818.77 ± 0.37169.4 ± 7.2
3p77.48 ± 1.9871.62 ± 1.0847.83 ± 0.9739.33 ± 0.6715.47 ± 0.64139.9 ± 5.1
Donepezil94.11 ± 1.7190.17 ± 1.0275.65 ± 1.1535.74 ± 0.5817.89 ± 0.4221.8 ± 0.9

Share and Cite

MDPI and ACS Style

Acar Cevik, U.; Saglik, B.N.; Levent, S.; Osmaniye, D.; Kaya Cavuşoglu, B.; Ozkay, Y.; Kaplancikli, Z.A. Synthesis and AChE-Inhibitory Activity of New Benzimidazole Derivatives. Molecules 2019, 24, 861. https://doi.org/10.3390/molecules24050861

AMA Style

Acar Cevik U, Saglik BN, Levent S, Osmaniye D, Kaya Cavuşoglu B, Ozkay Y, Kaplancikli ZA. Synthesis and AChE-Inhibitory Activity of New Benzimidazole Derivatives. Molecules. 2019; 24(5):861. https://doi.org/10.3390/molecules24050861

Chicago/Turabian Style

Acar Cevik, Ulviye, Begüm Nurpelin Saglik, Serkan Levent, Derya Osmaniye, Betul Kaya Cavuşoglu, Yusuf Ozkay, and Zafer Asim Kaplancikli. 2019. "Synthesis and AChE-Inhibitory Activity of New Benzimidazole Derivatives" Molecules 24, no. 5: 861. https://doi.org/10.3390/molecules24050861

Article Metrics

Back to TopTop