Document Type : Original Paper

Authors

1 1 Department of Applied Chemistry, Faculty of Science, South Tehran Branch, Islamic Azad University, Tehran, Islamic Republic of Iran

2 2 Pharmacology Department, Medical School & Razi drug research center, Iran University of Medical Sciences, Tehran, Islamic Republic of Iran

Abstract

Based on the important interactions of donepezil with cholinesterase receptor, a series of coumarin-based N-benzyl pyridinium derivatives (5a-l) were synthesized and had in-vitro evaluation for their acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory activities. It was revealed that compound 5l with plausible IC50 values of 0.247 µM and 1.68 µM on AChE and BuChE, respectively was the most potent anticholinesterase inhibitor compared to other synthesized compounds. The enzyme kinetic assay of compound 5l was conducted on the AChE enzyme and the compound 5l was found to be a non-competitive inhibitor of the AChE (Ki= 0.356). In addition, the compound 5l remarkably protected PC12 neurons against H2O2-induced cell death. The docking study of compound 5l revealed that the inhibitor occupied both CAS and PAS binding sites of the AChE enzyme. we have synthesized 12 products in two steps reactions and high to moderate yields. The first step involves the nucleophilic substitution reaction between 4-hydroxycoumarin and pyridyl chloride (3-pyridinium and 4-pyridinium) derivatives, which produces an intermediate of 3. Following the reaction of this intermediate with benzyl chloride derivatives, led to the synthesis of final products 5. The results were compared with donepezil and tacrine as standard drugs for AChE and BuChE inhibitory assays. Based on the IC50 values, the tendency to inhibit synthetic compounds in final products for AChE is better than BuChE. Among the products in AChE inhibitory assay, the 3-pyridinium series showed more effectiveness than the 4-pyridinium series. Docking studies and product interactions with cholinesterase receptor active sites clearly show the role of 3-pyridinium derivatives in receptor binding.

Keywords

Main Subjects

  1. Eikelenboom P, Zhan SS, van Gool WA, Allsop D. Inflammatory mechanisms in Alzheimer's disease. TIPS. 1994; 15(12): 447-450.
  2. Terry AV, Buccafusco JJ. The cholinergic hypothesis of age and Alzheimer's disease-related cognitive deficits: recent challenges and their implications for novel drug development, J. Pharmacol. Exp. Ther. 2003; 306(3): 821-827.
  3. Schelterns P, Feldman H. Treatment of Alzheimer's disease; current status and new perspectives. Lancet Neurol. 2003; 2(9): 539-547.
  4. Akram M, Nawaz A. Effects of medicinal plants on Alzheimer's disease and memory deficits. Neural Regen. Res. 2017; 12(4): 660.
  5. Giacobini E. Cholinesterase inhibitors: new roles and therapeutic alternatives. Pharmacol. Res. 2004; 50(4): 433-440.
  6. Decker M, Krauth F, Lehmann J. Novel tricyclic quinazolinimines and related tetracyclic nitrogen bridgehead compounds as cholinesterase inhibitors with selectivity towards butyrylcholinesterase. Bioorg. Med. Chem. 2006; 14(6): 1966-1977.
  7. Shan WJ, Huang L, Zhou Q, Meng FC, Li XS. Synthesis, biological evaluation of 9-N-substituted berberine derivatives as multi-functional agents of antioxidant, inhibitors of acetylcholinesterase, butyrylcholinesterase and amyloid-β aggregation, ur. J. Med. Chem. 2011; 46(12): 5885-93.
  8. Lane RM, Steven GP, Enz A. Targeting acetylcholinesterase and butyrylcholinesterase in dementia, Int. J. Neuropsychopharmacol. 2006; 9 (1): 101-124.
  9. Osborn GG, Saunders AV. Current treatments for patients with Alzheimer disease, Int J Osteopath Med . 2010; 110(98): 16-26.
  10. Dinamarca MC, Weinstein D, Monasterio O, Inestrosa NC. The synaptic protein neuroligin-1 interacts with the amyloid β-peptide. Is there a role in Alzheimer’s disease?. Biochemistry. 2011; 50(38): 8127-8137.
  11. Pera M, Martinez-Otero A, Colombo L, Salmona M, Ruiz-Molina D, Badia A, Clos MV. Acetylcholinesterase as an amyloid enhancing factor in PrP82-146 aggregation process, Mol. Cell. Neurosci. 2009; 40(2): 217-224.
  12. Castro A, Martinez A. Peripheral and dual binding site acetylcholinesterase inhibitors: implications in treatment of Alzheimer's disease. Mini Rev Med Chem. 2001; 1(3): 267-272.
  13. Silman I, Sussman JL. Acetylcholinesterase: how is structure related to function? Chem. Biol. Interact. 2008; 175(1-3): 3-10.
  14. Zhou X, Wang XB, Wang T, Kong LY. Design, synthesis, and acetylcholinesterase inhibitory activity of novel coumarin analogues. Bioorg. Med. Chem. 2008; 16(17): 8011-8021.
  15. Inestrosa NC, Alvarez A, Perez CA, Moreno RD, Vicente M, Linker C, Casanueva OI, Soto C, Garrido J. Acetylcholinesterase accelerates assembly of amyloid-β-peptides into Alzheimer's fibrils: possible role of the peripheral site of the enzyme. Neuron. 1996; 16(4): 881-891.
  16. Anand P, Singh B, Singh N. A review on coumarins as acetylcholinesterase inhibitors for Alzheimer’s disease, Bioorg. Med. Chem. 2012; 20(3): 1175-1180.
  17. Sugimoto H, Ogura H, Arai Y, Iimura Y, Yamanishi Y. Research and development of donepezil hydrochloride, a new type of acetylcholinesterase inhibitor. Jpn. J. Pharmacol. 2002; 89(1): `7-20.
  18. Qizilbash N, Birks J, Lopez AJ, Lewington S, Szeto S. Tacrine for Alzheimer's disease (Withdrawn paper. 1999, art. no. CD000202). Cochrane Database Syst Rev. 2005; (1).
  19. Agid, Y, Dubois B, Anand R, Gharabawi G. International Rivastigmine Investigators.: Efficacy and tolerability of rivastigmine in patients with dementia of the Alzheimer type, Curr Ther Res Clin Exp. 1998; 59(12): 837-845.
  20. Kavanagh S, Gaudig M, Van Baelen B, Adami M, Delgado A, Guzman C, Jedenius E, Schäuble B. Galantamine and behavior in Alzheimer disease: analysis of four trials, Acta Neurol. Scand. 2011; 124(5): 302-308.
  21. Alipour M, Khoobi M, Foroumadi A, Nadri H, Moradi A, Sakhteman A, Ghandi M, Shafiee A. Novel coumarin derivatives bearing N-benzyl pyridinium moiety: potent and dual binding site acetylcholinesterase inhibitors, Bioorg. Med. Chem. 2012; 20(24): 7214-7222.
  22. Rizzo S, Bartolini M, Ceccarini L, Piazzi, L.; Gobbi, S.; Cavalli, A.; Recanatini, M.; Andrisano, V.; Rampa, A.: Targeting Alzheimer’s disease: Novel indanone hybrids bearing a pharmacophoric fragment of AP2238, Bioorg. Med. Chem. 2010; 18(5): 1749-1760.
  23. Asadi M, Ebrahimi M, Mohammadi‐Khanaposhtani M, Azizian H, Sepehri S, Nadri H, Biglar M, Amanlou M, Larijani B, Mirzazadeh R, Edraki N. Design, Synthesis, Molecular Docking, and Cholinesterase Inhibitory Potential of Phthalimide‐Dithiocarbamate Hybrids as New Agents for Treatment of Alzheimer's Disease. Chem. Biodivers. 2019; Nov;16(11):e1900370.
  24. Arab S, Sadat‐Ebrahimi SE, Mohammadi‐Khanaposhtani M, Moradi A, Nadri H, Mahdavi M, Moghimi S, Asadi M, Firoozpour L, Pirali‐Hamedani M, Shafiee A. Synthesis and Evaluation of Chroman‐4‐One Linked to N‐Benzyl Pyridinium Derivatives as New Acetylcholinesterase Inhibitors. Pharm. 2015 Sep;348(9):643-9.
  25. Huang XY, Shan ZJ, Zhai HL, Su L, Zhang XY. Study on the anticancer activity of coumarin derivatives by molecular modeling. Chem Biol Drug Des. 2011; 78(4): 651-658.
  26. Bahadır Ö, Çitoğlu GS, Özbek H, Dall'Acqua S, Hošek J, Šmejkal K. Hepatoprotective and TNF-α inhibitory activity of Zosima absinthifolia extracts and coumarin. Fitoterapia. 2011; 82(3): 454-459.
  27. Sardari S, Nishibe S, Daneshtalab M. Coumarins, the bioactive structures with antifungal property. Stud. Nat. Prod. Chem. 2000; 23: 335-393.
  28. Kawase M, Varu B, Shah A, Motohashi N, Tani S, Saito S, Debnath S, Mahapatra S, Dastidar SG, Chakrabarty AN. Antimicrobial activity of new coumarin derivatives. Arzneimittelforschung. 2001; 51(01): 67-71.
  29. Kontogiorgis C, Hadjipavlou-Litina D. Biological evaluation of several coumarin derivatives designed as possible anti-inflammatory/antioxidant agents, J Enzyme Inhib Med Chem. 2003; 18(1): 63-69.
  30. Lee SO, Choi SZ, Lee JH, Chung SH, Park SH, Kang HC, Yang EY, Cho HJ, Lee KR. Antidiabetic coumarin and cyclitol compounds from Peucedanum japonicum. Arch. Pharm. Res. 2004; 27(12): 1207-1210.
  31. Arora RB, Mathur CN. Relationship between structure and anti‐coagulant activity of coumarin derivatives, Br. j. pharmacol. chemother. 1963; 20(1): 29-35.
  32. Goplen BP, Linton JH, Bell JM. Dicoumarol studies: iii. determining tolerance limits of contamination in low coumarin sweetclover varieties using a cattle bio-assay. Can. J. Anim. Sci. 1964; 44(1): 76-86.
  33. Hoerr R, Noeldner M. Ensaculin (KA‐672. HCl): A Multitransmitter Approach to Dementia Treatment. CNS drug reviews. 2002; 8(2): 143-158.
  34. Piazzi L, Rampa A, Bisi A, Gobbi S, Belluti F, Cavalli A, Bartolini M, Andrisano V, Valenti P, Recanatini M. 3-(4-{[Benzyl (methyl) amino] methyl} phenyl)-6, 7-dimethoxy-2 H-2-chromeneone (AP2238) Inhibits Both Acetylcholinesterase and Acetylcholinesterase-Induced β-Amyloid Aggregation: A Dual Function Lead for Alzheimer's Disease Therapy, J. Med. Chem. 2003; 46(12): 2279-2282.
  35. Mogana R, Teng-Jin K, Wiart C. Anti-inflammatory, anticholinesterase, and antioxidant potential of scopoletin isolated from Canarium patentinervium Miq.(Burseraceae Kunth). eCAM. 2013.
  36. Razavi SF, Khoobi M, Nadri H, Sakhteman A, Moradi A, Emami S, Foroumadi A, Shafiee A. Synthesis and evaluation of 4-substituted coumarins as novel acetylcholinesterase inhibitors. Eur. J. Med. Chem. 2013; 64: 252-259.
  37. Yusufzai SK, Khan MS, Sulaiman O, Osman H, Lamjin DN. Molecular docking studies of coumarin hybrids as potential acetylcholinesterase, butyrylcholinesterase, monoamine oxidase A/B and β-amyloid inhibitors for Alzheimer’s disease. Chem. Cent. J. 2018; 12(1):1-57.
  38. Moya-Alvarado G, Yañez O, Morales N, González-González A, Areche C, Núñez MT, Fierro A, García-Beltrán O. Coumarin-chalcone hybrids as inhibitors of MAO-B: Biological activity and in silico studies. Molecules. 2021; 26(9): 2430.
  39. de Aquino RAN, Modolo LV, Alves RB, de Fátima Â. Synthesis, kinetic studies and molecular modeling of novel tacrine dimers as cholinesterase inhibitors. Org. Biomol. Chem. 2013; 11(48): 8395-8409.
  40. Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading, J. Comput. Chem. 2010; 31(2): 455-461.
  41. Sanner MF. Python: a programming language for software integration and development. J Mol Graph Model. 1999; 17(1): 57-61.
  42. O'Boyle NM, Banck M, James CA, Morley C, Vandermeersch T, Hutchison GR. Open Babel: An open chemical toolbox. J. Cheminformatics. 2011; 3(1): 1-14.
  43. Pettersen EF, Goddard TD, Huang CC, Couch GS, Greenblatt DM, Meng EC, Ferrin TE. UCSF Chimera—a visualization system for exploratory research and analysis, J. Comput. Chem. 2004, 25(13), 1605-1612.
  44. Stierand K, Rarey M. From Modeling to Medicinal Chemistry: Automatic Generation of Two‐Dimensional Complex Diagrams. ChemMedChem: Chemistry Enabling Drug Discovery. 2007; 2(6): 853-860.
  45. Koh SH, Kim SH, Kwon H, Park Y, Kim KS, Song CW, et al. Epigallocatechin gallate protects nerve growth factor differentiated PC12 cells from oxidative-radical-stress-induced apoptosis through its effect on phosphoinositide 3-kinase/Akt and glycogen synthase kinase-3. Mol. Brain Res. 2003; 118(1-2): 2-81.
  46. Datki Z, Juhász A, Gálfi M, Soós K, Papp R, Zádori D, Penke B. Method for measuring neurotoxicity of aggregating polypeptides with the MTT assay on differentiated neuroblastoma cells, Brain Res. Bull. 2003; 62(3): 223-229.
  47. Asadipour A, Alipour M, Jafari M, Khoobi M, Emami S, Nadri H, et al. Novel coumarin-3-carboxamides bearing N-benzylpiperidine moiety as potent acetylcholinesterase inhibitors, Eur. J. Med. Chem. 2013; 70: 623-630.
  48. Ellman GL, Courtney KD, Andres Jr V, Featherstone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem. Pharmacol. 1961; 7(2): 88-95.
  49. Branduardi D, Gervasio FL, Cavalli A, Recanatini M, Parrinello M. The role of the peripheral anionic site and cation− π interactions in the ligand penetration of the human AChE gorge. J. Am. Chem. Soc. 2005; 127(25): 9147-9155.