High-throughput Virtual Screening- and Molecular Docking-based Prediction for Acetylcholinesterase Inhibitors and Exploring its Mechanisms against Alzheimer’s Disease based on Network Pharmacology

Authors

Keywords:

Virtual screening, Acetylcholinesterase inhibitors, Molecular docking, Drug-likeness properties, BBB permeability, Pharmacokinetic properties, Network Pharmacology

Abstract

Objective  This study aimed to identify promising ligands for inhibiting acetylcholinesterase (AChE) activity using virtual screening (VS).

Methods  VS was used to identify potential AChE inhibitors from the PubChem database. Ligands with favorable binding pocket interactions were selected. SwissADME and pkCSM tools were used to assess drug-likeness and pharmacokinetic properties. Molecular dynamic (MD) simulations provided insights into binding interactions. Network pharmacology was used to explore interactions between the target molecule and AD-related genes to determine its mechanism of action.

Results VS identified promising AChE inhibitor candidates with acridone, carbazole, and xanthone scaffolds. Docking simulations showed strong binding with AChE. These ligands displayed favorable drug-likeness and ADMET properties, with one (M5) lacking predicted hepatotoxicity. MD simulations suggested stable binding of M5 to AChE, potentially affecting both catalytic and peripheral sites, hinting at dual inhibition. M5’s interactions, especially near His440, appeared more favorable than donepezil. Network analysis implicated M5 in targeting multiple pathways in AD, with potential focus on neuroinflammation.

Conclusions This study identified promising AChE inhibitor candidates through virtual screening. Ligand M5 emerged as particularly promising due to its favorable binding characteristics, lack of predicted hepatotoxicity, and potential for targeting multiple pathways in AD. However, further in vitro and in vivo validation is essential for clinical development. 

References

United Nations, Department of Economic and Social Affairs, Population Division. World population ageing 2019: Highlights. New York: United Nations; 2019.

Hasselmo M. The Role of acetylcholine in learning and memory. Curr Opin Neurobiol. 2006;16:710-5.

Chen Z, Huang J, Yang S, Hong F. Role of cholinergic signaling in Alzheimer’s disease. Molecules. 2022;27:1816. PubMed PMID: 35335180

Giacobini E. Is anti-cholinesterase therapy of Alzheimer’s disease delaying progression? Aging (Milano). 2001;13:247-54.

Čolović M, Krstić D, Lazarević-Pašti T, Bondžić A, Vasić V. Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol. 2013;11:315-35.

Budd Haeberlein S, Aisen P, Barkhof F, Chalkias S, Chen T, Cohen S, et al. Two randomized phase 3 studies of aducanumab in early Alzheimer’s disease. J Prev Alzheimers Dis. 2022;9:197-210.

Kapetanovic I. Computer-aided drug discovery and development (CADDD): in silico-chemico-biological approach. Chem Biol Interact. 2008;171:165-76.

Mohs R, Greig N. Drug discovery and development: role of basic biological research. Alzheimers Dement (N Y). 2017;3:651-7.

Muegge I, Oloff S. Advances in virtual screening. Drug Discovery Today: Technologies. 2006;3:405-11.

Wu F, Zhou Y, Li L, Shen X, Chen G, Wang X, Liang X, Tan M, Huang Z. Computational approaches in preclinical studies on drug discovery and development. Front Chem. 2020;8:726. PubMed PMID: 33062633.

Durrant J, McCammon J. Molecular dynamics simulations and drug discovery. BMC Biology. 2011;9:71. PubMed PMID: 22035460.

Daina A, Michielin O, Zoete V. SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Sci Rep. 2017;7:42717. PubMed PMID: 28256516.

Pires D, Blundell T, Ascher D. pkCSM: predicting small-molecule pharmacokinetic and toxicity properties using graph-based signatures. J Med Chem. 2015;58:4066-72.

Eastman P, Swails J, Chodera J, McGibbon R, Zhao Y, Beauchamp K, et al. OpenMM 7: rapid development of high performance algorithms for molecular dynamics. PLoS Comput Biol. 2017;13:e1005659. PubMed PMID: 28746339.

Tian C, Kasavajhala K, Belfon K, Raguette L, Huang H, Migues A, et al. ff19SB: amino-acid-specific protein backbone parameters trained against quantum mechanics energy surfaces in solution. J Chem Theory Comput. 2020;16:528-52.

Lipinski C, Lombardo F, Dominy B, Feeney P. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev. 2001;46:3-26.

Tiikkainen P, Franke L. Analysis of commercial and public bioactivity databases. J Chem Inf Model. 2012; 52:319-26.

Tetko I, Engkvist O, Koch U, Reymond J, Chen H. BIGCHEM: challenges and opportunities for big data analysis in chemistry. Molecular Informatics. 2016; 35:615-21.

Kim S, Thiessen PA, Bolton EE, Chen J, Fu G, Gindulyte A, et al. PubChem substance and compound databases. Nucleic Acids Res. 2016;44(Database issue): D1202-13.

Marucci G, Buccioni M, Ben D, Lambertucci C, Volpini R, Amenta F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology. 2021; 190:108352. PubMed PMID: 33035532.

Ogura H, Kosasa T, Kuriya Y, Yamanishi Y. Comparison of inhibitory activities of donepezil and other cholinesterase inhibitors on acetylcholinesterase and butyrylcholinesterase in vitro. Methods Find Exp Clin Pharmacol. 2000;22:609-13.

Khawli LA, Prabhu S. Drug delivery across the blood–brain barrier. Mol Pharmaceutics. 2013;10:1471-2.

Bajda M, Więckowska A, Hebda M, Guzior N, Sotriffer C, Malawska B. Structure-based search for new inhibitors of cholinesterases. IJMS. 2013;14:5608-32.

Kadry H, Noorani B, Cucullo L. A blood-brain barrier overview on structure, function, impairment, and biomarkers of integrity. Fluids Barriers CNS. 2020;17:69. PubMed PMID: 33208141.

Zanger U, Schwab M. Cytochrome P450 enzymes in drug metabolism: regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol Ther. 2013;138:103-41.

Bhatia S. Concise guide to drug interaction principles of medical practice: cytochrome P450s, UGTs, P-Glycoproteins, Second Edition. J Am Acad Child Adolesc Psychiatry. 2005;44:607-8.

Lynch T, Price A. The effect of cytochrome P450 metabolism on drug response, interactions, and adverse effects. Am Fam Physician. 2007;76:391-6.

Gade AC, Murahari M, Pavadai P, Kumar MS. Virtual screening of a marine natural product database for in silico identification of a potential acetylcholinesterase inhibitor. Life. 2023;13:1298. PubMed PMID: 37374081.

Thomas PD. The gene ontology and the meaning of biological function. Methods Mol Biol. 2017;1446:15-24.

Cooper GM. Structure of the plasma membrane. In: the cell: a molecular approach 2nd edition [Internet]. Sinauer Associates; 2000 [cited 2024 Jan 25]. Available from: https://www.ncbi.nlm.nih.gov/books/NBK9898/

Ray S, Kassan A, Busija A, Rangamani P, Patel H. The plasma membrane as a capacitor for energy and metabolism. Am J Physiol Cell Physiol. 2016;310:C181-92.

Cho I, Jackson M, Swift J. Roles of cross-membrane transport and signaling in the maintenance of cellular homeostasis. Cell Mol Bioeng. 2016;9:234-46.

Lauss M, Kriegner A, Vierlinger K, Noehammer C. Characterization of the drugged human genome. Pharmacogenomics. 2007;8:1063-73.

DeTure M, Dickson D. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegener. 2019;14:32. PubMed PMID: 31375134.

Kinney J, Bemiller S, Murtishaw A, Leisgang A, Salazar A, Lamb B. Inflammation as a central mechanism in Alzheimer’s disease. Alzheimers Dement (N Y). 2018;4: 575-90.

Pietronigro E, Della Bianca V, Zenaro E, Constantin G. NETosis in Alzheimer’s disease. Front Immunol. 2017;8:211. PubMed PMID: 28303140.

Turner R, Sharp F. Implications of MMP9 for blood brain barrier disruption and hemorrhagic transformation following ischemic stroke. Front Cell Neurosci. 2016;10:56. PubMed PMID: 26973468.

Chen Y, Williams V, Filippova M, Filippov V, Duerksen-Hughes P. Viral carcinogenesis: factors inducing dna damage and virus integration. Cancers (Basel). 2014;6:2155-86.

Pezone A, Olivieri F, Napoli MV, Procopio A, Avvedimento EV, Gabrielli A. Inflammation and DNA damage: cause, effect or both. Nat Rev Rheumatol. 2023;19: 200-11.

Papassotiropoulos A, de Quervain D. Failed drug discovery in psychiatry: time for human genome-guided solutions. Trends Cogn Sci. 2015;19:183-7.

Zitti B, Bryceson Y. Natural killer cells in inflammation and autoimmunity. Cytokine Growth Factor Rev. 2018;42:37-46.

Harjunpää H, Llort Asens M, Guenther C, Fagerholm S. Cell adhesion molecules and their roles and regulation in the immune and tumor microenvironment. Front Immunol. 2019;10:1078. PubMed PMID: 31231358.

Sharma P, Kumar P, Sharma R. Natural killer cells - their role in tumour immunosurveillance. J Clin Diagn Res. 2017;11:BE01-5.

Qi C, Liu Q. Natural killer cells in aging and age-related diseases. Neurobiol Dis. 2023;183:106156. PubMed PMID: 37209924

Baechle JJ, Chen N, Makhijani P, Winer S, Furman D, Winer DA. Chronic inflammation and the hallmarks of aging. Mol Metab. 2023;74:101755. PubMed PMID: 37329949

Downloads

Published

2024-06-14

How to Cite

1.
Takomthong P, Waiwut P, Ballatore C, Choowongkomon K, Arsito PN, Chulikhit Y, Boonyarat C. High-throughput Virtual Screening- and Molecular Docking-based Prediction for Acetylcholinesterase Inhibitors and Exploring its Mechanisms against Alzheimer’s Disease based on Network Pharmacology. BSCM [Internet]. 2024 Jun. 14 [cited 2024 Jun. 30];63(3). Available from: https://he01.tci-thaijo.org/index.php/CMMJ-MedCMJ/article/view/269932

Issue

Section

Original Article