Volume 11, Issue 1 (7-2023)                   Jorjani Biomed J 2023, 11(1): 3-8 | Back to browse issues page

XML Print


Download citation:
BibTeX | RIS | EndNote | Medlars | ProCite | Reference Manager | RefWorks
Send citation to:

pirmoradi S. Ligand-based pharmacophore modeling to identify plant-derived acetylcholinesterase inhibitor natural compounds in Alzheimer’s disease. Jorjani Biomed J 2023; 11 (1) :3-8
URL: http://goums.ac.ir/jorjanijournal/article-1-952-en.html
Department of Biochemistry, Shahid Chamran University of Ahvaz, Ahvaz, Iran. , pirmoradi150@gmail.com
Abstract:   (839 Views)
Background: Alzheimer’s disease (AD) is a neurodegenerative disease characterized by decreased cognitive function in patients due to forming Aβ peptides and neurofibrillary tangles (NFT) in the brain. Therefore, the need to develop new treatments can reduce this risk. Acetylcholinesterase is one of the targets used in the design of new drugs for the treatment of AD. The researchers obtain new inhibitory ligands based on natural compounds from various medicinal plants, such as the family of Asteroideace, Malvacea, Zingbracea, Hypericacea, and Ebenacea, for treating Alzheimer’s disease. 
Methods: After selecting the reference compound of the enzyme acetylcholinesterase inhibitor with the help of bioinformatics tools such as pharmit and ZINCPHARMER for virtual search through the structural and pharmacologic properties of the reference inhibitor compound, several thousand natural structures of several serious ligands were obtained. Then, the ligands were compared by examining the docking process with the acetylcholinesterase enzyme, and their interactions were visualized with the help of Discovery Studio. Then, the top selected ligands in terms of toxicity, allergy, toxicity, and ADME prediction were evaluated with tools such as molsoft, PKCSM, ADMEtlab2.0, Swiss ADME.
Results: The results revealed that these obtained ligands, like donepezil, have the ability of favorable interactions with different amino acids, the crucial of which are HIS381, TRP385, and GLN527 of AChE, and they all fall in the active site or binding pocket of the active site. The present docking supports this hypothesis that these compounds are possible and valuable small molecule ligands for targeting/inhibiting acetylcholinesterases. Indicatively, according to the binding free energy calculation results, it can be concluded that these ligands can compete with donepezil and affect the formation of acetylcholinesterase complexes. Cholinesterase/donepezil can have an excellent competitive inhibitory effect on it.
Conclusion: On the other hand, the study on the designed ligands showed that with favorable interactions and lower binding energy, they form more stable complexes with acetylcholinesterase and can be proposed as inhibitors competing with donepezil in a bind to this enzyme.
Full-Text [PDF 979 kb]   (477 Downloads) |   |   Full-Text (HTML)  (110 Views)  
Type of Article: Original article | Subject: Basic Medical Sciences
Received: 2023/01/25 | Accepted: 2023/06/11 | Published: 2023/07/1

References
1. Mohammadzadeh Honarvar N, Saedisomeolia A, Abdolahi M, Shayeganrad A, Taheri Sangsari G, Hassanzadeh Rad B, et al. Molecular anti-inflammatory mechanisms of retinoids and carotenoids in Alzheimer's disease: A review of current evidence. J Mol Neurosci. 2017;61(3):289-304. [view at publisher] [DOI] [PMID] [Google Scholar]
2. Waldemar G, Dubois B, Emre M, Georges J, McKeith IG, Rossor M, Scheltens P, Tariska P, Winblad B. Recommendations for the diagnosis and management of Alzheimer's disease and other disorders associated with dementia: EFNS guideline. Eur J Neurol. 2007;14(1):e1-26. [view at publisher] [DOI] [PMID] [Google Scholar]
3. Sugimoto H. Development of anti-Alzheimer's disease drug based on beta-amyloid hypothesis. Yakugaku zasshi: Journal of the Pharmaceutical Society of Japan. 2010;130(4):521-6. [view at publisher] [DOI] [PMID] [Google Scholar]
4. Kennedy DO, Dodd FL, Robertson BC, Okello EJ, Reay JL, Scholey AB, Haskell CF. Monoterpenoid extract of sage (Salvia lavandulaefolia) with cholinesterase inhibiting properties improves cognitive performance and mood in healthy adults. J Psychopharmacol. 2011;25(8):1088-100. [view at publisher] [DOI] [PMID] [Google Scholar]
5. Akhtar MN, Lam KW, Abas F, Ahmad S, Shah SA, Choudhary MI, Lajis NH. New class of acetylcholinesterase inhibitors from the stem bark of Knema laurina and their structural insights. Bioorg Med Chem Lett. 2011;21(13):4097-103. [view at publisher] [DOI] [PMID] [Google Scholar]
6. Xu Y, Colletier JP, Jiang H, Silman I, Sussman JL, Weik M. Induced‐fit or preexisting equilibrium dynamics? Lessons from protein crystallography and MD simulations on acetylcholinesterase and implications for structure‐based drug design. Protein Sci. 2008;17(4):601-5. [view at publisher] [DOI] [PMID] [Google Scholar]
7. Ballard CG, Greig NH, Guillozet-Bongaarts AL, Enz A, Darvesh S. Cholinesterases: roles in the brain during health and disease. Curr Alzheimer Res. 2005;2(3):307-18. [view at publisher] [DOI] [PMID] [Google Scholar]
8. Guevara-Salazar JA, Espinoza-Fonseca M, Beltrán HI, Correa-Basurto J, Quintana Zavala D, Trujillo-Ferrara JG. The electronic influence on the active site-directed inhibition of acetylcholinesterase by N-aryl-substituted succinimides. Journal of the Mexican Chemical Society. 2007;51(4):222-7. [view at publisher] [Google Scholar]
9. Grover A, Shandilya A, Agrawal V, Bisaria VS, Sundar D. Computational evidence to inhibition of human acetyl cholinesterase by withanolide a for Alzheimer treatment. J Biomol Struct Dyn. 2012;29(4):651-62. [view at publisher] [DOI] [PMID] [Google Scholar]
10. Camps P, Formosa X, Galdeano C, Gómez T, Muñoz-Torrero D, Ramírez L, et al. Tacrine-based dual binding site acetylcholinesterase inhibitors as potential disease-modifying anti-Alzheimer drug candidates. Chem Biol Interact. 2010;187(1-3):411-5. [view at publisher] [DOI] [PMID] [Google Scholar]
11. Stefanou V, Matiadis D, Melagraki G, Afantitis A, Athanasellis G, Igglessi-Markopoulou O, et al. Functionalized 4-hydroxy coumarins: novel synthesis, crystal structure and DFT calculations. Molecules. 2011;16(1):384-402. [view at publisher] [DOI] [PMID] [Google Scholar]
12. dos Santos Pisoni D, da Costa JS, Gamba D, Petzhold CL, de Amorim Borges AC, Ceschi MA, et al. Synthesis and AChE inhibitory activity of new chiral tetrahydroacridine analogues from terpenic cyclanones. Eur J Med Chem. 2010;45(2):526-35. [view at publisher] [DOI] [PMID] [Google Scholar]
13. Gurovic MS, Castro MJ, Richmond V, Faraoni MB, Maier MS, Murray AP. Triterpenoids with acetylcholinesterase inhibition from Chuquiraga erinacea D. Don. subsp. erinacea (Asteraceae). Planta Med. 2010;76(6):607-10. [view at publisher] [DOI] [PMID] [Google Scholar]
14. Carpinella MC, Andrione DG, Ruiz G, Palacios SM. Screening for acetylcholinesterase inhibitory activity in plant extracts from Argentina. Phytother Res: An International Journal Devoted to Pharmacological and Toxicological Evaluation of Natural Product Derivatives. 2010;24(2):259-63. [view at publisher] [DOI] [PMID] [Google Scholar]
15. Peng XW, Dong KL. Clinical observation on acupuncture combined with Yizhi Jiannao granules for treatment of Alzheimer's disease. Zhongguo Zhen jiu: Chinese Acupuncture & Moxibustion. 2009;29(4):269-71. [view at publisher] [PMID] [Google Scholar]
16. Mah SH, Teh SS, Ee GC. Anti-inflammatory, anti-cholinergic and cytotoxic effects of Sida rhombifolia. Pharm Biol. 2017;55(1):920-8. [view at publisher] [DOI] [PMID] [Google Scholar]
17. Tappayuthpijarn P, Itharat A, Makchuchit S. Acetylcholinesterase inhibitory activity of Thai traditional nootropic remedy and its herbal ingredients. J Med Assoc Thai: Chotmaihet thangphaet. 2011;94(Suppl7):S183-9. [view at publisher] [PMID] [Google Scholar]
18. de Castro AA, da Cunha EF, Pereira AF, Soares FV, Leal DH, Kuca K, et al. Insights into the drug repositioning applied to the Alzheimer's disease treatment and future perspectives. Curr Alzheimer Res. 2018;15(12):1161-78. [view at publisher] [DOI] [PMID] [Google Scholar]
19. Huang SW, Wang W, Zhang MY, Liu QB, Luo SY, Peng Y, et al. The effect of ethyl acetate extract from persimmon leaves on Alzheimer's disease and its underlying mechanism. Phytomedicine. 2016;23(7):694-704. [view at publisher] [DOI] [PMID] [Google Scholar]
20. Pirmoradi S, Darvishkhadem M, Esmaeillashgarian H. Design of an inhibitory ligand based on interleukin 6 receptor for disruption and inhibition of interleukin 6-dependent inflammatory signaling pathway in Covid-19 patients. Yafte. 2021;23:344-55. [view at publisher] [Google Scholar]
21. Erdős G, Dosztányi Z. Analyzing protein disorder with IUPred2A. Curr Protoc Bioinformatics. Current Protocols in Bioinformatics. 2020;70(1):e99. [view at publisher] [DOI] [PMID] [Google Scholar]
22. Refsgaard HH, Jensen BF, Brockhoff PB, Padkjær SB, Guldbrandt M, Christensen MS. In silico prediction of membrane permeability from calculated molecular parameters. J Med Chem. 2005;48(3):805-11. [view at publisher] [DOI] [PMID] [Google Scholar]
23. Debnath AK. Pharmacophore mapping of a series of 2, 4-diamino-5-deazapteridine inhibitors of Mycobacterium avium complex dihydrofolate reductase. J Med Chem. 2002;45(1):41-53. [view at publisher] [DOI] [PMID] [Google Scholar]
24. Veber DF, Johnson SR, Cheng HY, Smith BR, Ward KW, Kopple KD. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002;45(12):2615-23. [view at publisher] [DOI] [PMID] [Google Scholar]
25. Boonyaketgoson S, Rukachaisirikul V, Phongpaichit S, Trisuwan K. Naphthoquinones from the leaves of Rhinacanthus nasutus having acetylcholinesterase inhibitory and cytotoxic activities. Fitoterapia. 2018;124:206-10. [view at publisher] [DOI] [PMID] [Google Scholar]
26. Ritchie TJ, Ertl P, Lewis R. The graphical representation of ADME-related molecule properties for medicinal chemists. Drug Discov Today. 2011;16(1-2):65-72. [view at publisher] [DOI] [PMID] [Google Scholar]
27. Pirmoradi S, Jafari H. Design of Immunosuppressive Structure Based on Spike Protein (s) virus Corona. J Arak Uni Med Sci. 2021;24(4):596-615. [view at publisher] [DOI] [Google Scholar]
28. Seniya C, Khan GJ, Misra R, Vyas V, Kaushik S. In-silico modelling and identification of a possible inhibitor of H1N1 virus. Asian Pacific Journal of Tropical Disease. 2014;4(suppl1):S467-76. [view at publisher] [DOI] [Google Scholar]
29. de Castro AA, da Cunha EF, Pereira AF, Soares FV, Leal DH, Kuca K, et al. Insights into the drug repositioning applied to the Alzheimer's disease treatment and future perspectives. Curr Alzheimer Res. 2018;15(12):1161-78. [view at publisher] [DOI] [PMID] [Google Scholar]
30. Wilkinson D, Doody R, Helme R, Taubman K, Mintzer J, Kertesz A, et al. Donepezil in vascular dementia: a randomized, placebo-controlled study. Neurology. 2003;61(4):479-86. [view at publisher] [DOI] [PMID] [Google Scholar]
31. Conforti F, Rigano D, Formisano C, Bruno M, Loizzo MR, Menichini F, et al. Metabolite profile and in vitro activities of Phagnalon saxatile (L.) Cass. relevant to treatment of Alzheimer's disease. J Enzyme Inhib Med Chem. 2010;25(1):97-104. [view at publisher] [DOI] [PMID] [Google Scholar]
32. Duan S, Guan X, Lin R, Liu X, Yan Y, Lin R, Zhang T, Chen X, Huang J, Sun X, Li Q. Silibinin inhibits acetylcholinesterase activity and amyloid β peptide aggregation: a dual-target drug for the treatment of Alzheimer's disease. Neurobiol Aging. 2015;36(5):1792-807. [view at publisher] [DOI] [PMID] [Google Scholar]
33. Bakthir H, Ali NA, Arnold N, Teichert A, Wessjohann L. Anticholinesterase activity of endemic plant extracts from Soqotra. Afr J Tradit Complement Altern Med. 2011;8(3):296-9. [view at publisher] [DOI] [PMID] [Google Scholar]
34. Chougouo RD, Nguekeu YM, Dzoyem JP, Awouafack MD, Kouamouo J, Tane P, et al. Anti-inflammatory and acetylcholinesterase activity of extract, fractions and five compounds isolated from the leaves and twigs of Artemisia annua growing in Cameroon. Springerplus. 2016;5(1):1-7. [view at publisher] [DOI] [PMID] [Google Scholar]
35. Zhao Y, Dou J, Wu T, Aisa HA. Investigating the antioxidant and acetylcholinesterase inhibition activities of Gossypium herbaceam. Molecules. 2013;18(1):951-62. [view at publisher] [DOI] [PMID] [Google Scholar]
36. Mah SH, Teh SS, Ee GC. Anti-inflammatory, anti-cholinergic and cytotoxic effects of Sida rhombifolia. Pharm Biol. 2017;55(1):920-8. [view at publisher] [DOI] [PMID] [Google Scholar]
37. Hendrayani SF, Al-Harbi B, Al-Ansari MM, Silva G, Aboussekhra A. The inflammatory/cancer-related IL-6/STAT3/NF-κB positive feedback loop includes AUF1 and maintains the active state of breast myofibroblasts. Oncotarget. 2016;7(27):41974. [view at publisher] [DOI] [PMID] [Google Scholar]
38. Zhang W, Zhao Y, Zhang F, Wang Q, Li T, Liu Z, et al. The use of anti-inflammatory drugs in the treatment of people with severe coronavirus disease 2019 (COVID-19): The Perspectives of clinical immunologists from China. Clinical immunology. 2020;214:108393. [view at publisher] [DOI] [PMID] [Google Scholar]
39. Mohanty SK, Satapathy A, Naidu MM, Mukhopadhyay S, Sharma S, Barton LM, et al. Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) and coronavirus disease 19 (COVID-19)-anatomic pathology perspective on current knowledge. Diagnostic pathology. 2020;15(1):1-7. [view at publisher] [DOI] [PMID] [Google Scholar]
40. Pirmoradi S. In-silico Designing of Immunogenic Construct Based on Peptide Epitopes Using Immuno-informatics Tools Against Tuberculosis. Iran J Med Microbiol. 2022;16(6):506-19. [view at publisher] [DOI] [PMID] [Google Scholar]
41. Iranshahi M, Javadi B. Neurological and Neuroprotective effects of Melissa officinalis L. Navid No. 2019;22(69):60-71. [view at publisher] [DOI] [PMID] [Google Scholar]

Add your comments about this article : Your username or Email:
CAPTCHA

Send email to the article author


Rights and permissions
Creative Commons License This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.

© 2024 CC BY-NC 4.0 | Jorjani Biomedicine Journal

Designed & Developed by : Yektaweb