Next Article in Journal
Modularized Genes in an Adrenal Pathway Reveal a Novel Mechanism in Hypertension Pathogenesis
Previous Article in Journal
Dual Mutations in MSMEG_0965 and MSMEG_1380 Confer High-Level Resistance to Bortezomib and Linezolid by Both Reducing Drug Intake and Increasing Efflux in Mycobacterium smegmatis
Previous Article in Special Issue
The Sequence [RRKLPVGRS] Is a Nuclear Localization Signal for Importin 8 Binding (NLS8): A Chemical Biology and Bioinformatics Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Structural Bioinformatics Applied to Acetylcholinesterase Enzyme Inhibition

by
María Fernanda Reynoso-García
1,
Dulce E. Nicolás-Álvarez
2,*,
A. Yair Tenorio-Barajas
3 and
Andrés Reyes-Chaparro
1,*
1
Departamento de Morfología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Lázaro Cárdenas, Prolongación de Carpio y Plan de Ayala s/n, Col. Santo Tomás, Alcaldía Miguel Hidalgo, Mexico City 11340, Mexico
2
Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu S/N, Unidad Profesional Adolfo López Mateos, Mexico City 07738, Mexico
3
Laboratorio de Nanobiotecnologia, Facultad de Ciencias Físico Matemáticas, Benemerita Universidad de Puebla, Av. San Cladio y 18 Sur, Col. San Manuel, Edif. FM6-108, Ciudad Universitaria, Puebla 72570, Mexico
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(8), 3781; https://doi.org/10.3390/ijms26083781
Submission received: 21 February 2025 / Revised: 3 April 2025 / Accepted: 5 April 2025 / Published: 17 April 2025
(This article belongs to the Special Issue Molecular Advances in Bioinformatics Analysis of Protein Properties)

Abstract

:
Acetylcholinesterase (AChE) is a critical enzyme involved in neurotransmission by hydrolyzing acetylcholine at the synaptic cleft, making it a key target for drug discovery, particularly in the treatment of neurodegenerative disorders such as Alzheimer’s disease. Computational approaches, particularly molecular docking and molecular dynamics (MD) simulations, have become indispensable tools for identifying and optimizing AChE inhibitors by predicting ligand-binding affinities, interaction mechanisms, and conformational dynamics. This review serves as a comprehensive guide for future research on AChE using molecular docking and MD simulations. It compiles and analyzes studies conducted over the past five years, providing a critical evaluation of the most widely used computational tools, including AutoDock, AutoDock Vina, and GROMACS, which have significantly contributed to the advancement of AChE inhibitor screening. Furthermore, we identify PDB ID: 4EY7, the most frequently used AChE crystal structure in docking studies, and highlight Donepezil, a well-established reference molecule widely employed as a control in computational screening for novel inhibitors. By examining these key aspects, this review aims to enhance the accuracy and reliability of virtual screening approaches and guide researchers in selecting the most appropriate computational methodologies. The integration of docking and MD simulations not only improves hit identification and lead optimization but also provides deeper mechanistic insights into AChE–ligand interactions, contributing to the rational design of more effective AChE inhibitors.

1. Introduction

Acetylcholinesterase (AChE) is a key enzyme in the regulation of cholinergic neurotransmission, catalyzing the hydrolysis of acetylcholine at the synapse. Its biomedical significance lies in its role in various neurological diseases, particularly Alzheimer’s disease, where its inhibition has been the main therapeutic strategy to improve cognitive function in patients with neurodegenerative decline [1]. It is known that AChE activity can accelerate the aggregation of the β-amyloid (Aβ) peptide, increasing its neurotoxicity [2]. AChE inhibitors such as Donepezil, rivastigmine, and galantamine have shown efficacy in symptom management, although they present limitations such as side effects and a progressive loss of efficacy over time [3].
In pharmacology, AChE is also a therapeutic target for other neurological and muscular disorders, including myasthenia gravis, organophosphate poisoning, and Parkinson’s disease Recent studies have identified antiviral drugs, such as tilorone, that exhibit inhibitory activity against AChE, opening new possibilities for drug repurposing [4].
Research on AChE modulation has also explored multitarget drug design. These compounds not only inhibit AChE but also interact with other molecular targets involved in neurodegeneration, such as β-amyloid aggregation and oxidative stress [5,6,7]. This strategy aims to simultaneously address multiple pathological mechanisms, offering a potentially more effective therapeutic approach for neurodegenerative diseases. Additionally, advances in computational chemistry and molecular screening have led to the identification of new inhibitor classes with greater selectivity and lower toxicity [8,9,10]. These findings establish AChE as a target of great interest in pharmacology and biomedicine, with applications extending beyond the neurological field to new therapeutic opportunities in various diseases.
In recent years, structural bioinformatics has revolutionized the study of acetylcholinesterase (AChE) as a therapeutic target, enabling the design and optimization of novel inhibitors with greater efficacy and specificity. The availability of three-dimensional models of AChE and its interactions with different ligands has provided crucial insights into the dynamics of its active site and molecular inhibition mechanisms [11]. Using techniques such as molecular dynamics and docking studies, researchers have identified new classes of compounds capable of selectively binding to the enzyme, minimizing adverse effects, and optimizing bioavailability [12,13]. These advances have facilitated the exploration of multitarget hybrid structures, which not only inhibit AChE but also possess antioxidant or anti-inflammatory properties, opening new therapeutic avenues for neurodegenerative diseases.
One of the most significant achievements in this field has been the application of artificial intelligence and machine learning to the virtual screening of potential AChE inhibitors. Recent studies have shown that by training deep learning models with large databases of bioactive compounds, it is possible to predict with high accuracy which molecules exhibit strong affinity for the enzyme [14,15]. These techniques have significantly reduced the time and cost required for the identification of new drugs, allowing for the exploration of thousands of compounds within hours instead of years of laboratory experimentation [16,17]. Additionally, the use of computational pharmacophores and quantum simulations has enabled a more detailed characterization of key interactions between AChE and its inhibitors, enhancing the rational design of new therapies [18].
The impact of structural bioinformatics on AChE pharmacology is not limited to Alzheimer’s disease but has also extended to other biomedical applications, such as the development of antidotes against neurotoxins and organophosphate pesticides. Computational simulations have enabled the design of reversible inhibitors that can be used as rescue agents in cases of acute poisoning [19,20]. Similarly, strategies based on the chemical modification of classical inhibitors have been developed to improve their selectivity and reduce their side effects on other enzymatic systems [21,22]. Thanks to these advances, the combination of bioinformatics, computational chemistry, and structural pharmacology continues to open new possibilities for the development of safer and more effective drugs targeting AChE, reinforcing its status as a key target in precision medicine.
This article presents an analysis of recent advancements in the use of structural bioinformatics tools and computational chemistry for the design and optimization of acetylcholinesterase (AChE) inhibitors. Different approaches were reviewed, starting with the most used tools, such as molecular docking and molecular dynamics. Additionally, new approaches utilizing machine learning were considered, highlighting their impact on the identification of new compounds with greater specificity and lower toxicity. Furthermore, the role of these techniques in the development of multitarget drugs and the search for antidotes against neurotoxins is explored, consolidating the relevance of AChE as a therapeutic target in various neurodegenerative and toxicological diseases.

2. Acetylcholinesterase: Structure and Function

Acetylcholinesterase (AChE) is a hydrolase belonging to the cholinesterase family, whose primary function is the hydrolysis of the neurotransmitter acetylcholine (ACh) into choline and acetic acid, for allowing the termination of cholinergic signaling at the synapse [23]. It is classified within the serine esterase family due to the presence of a catalytic site characterized by the Ser–His–Glu catalytic triad, which is essential for its mechanism of action [24]. There are two main cholinesterases in higher organisms: acetylcholinesterase (AChE), which is highly specific for acetylcholine, and butyrylcholinesterase (BChE), which exhibits a more nonspecific activity and can hydrolyze other choline esters [25]. AChE plays a crucial role in both the central and peripheral nervous systems, where it regulates cholinergic transmission at neuromuscular junctions and in various brain regions, being essential for cognitive functions such as learning and memory [23].
From a structural perspective, AChE has been extensively characterized using X-ray crystallography, allowing for a high-resolution description of its three-dimensional structure [24,25]. The enzyme features a deep and narrow active site, known as the catalytic gorge, which consists of a catalytic site where substrate hydrolysis occurs, as well as a peripheral site that regulates acetylcholine entry (Figure 1) [26]. The active site comprises the Ser203–His447–Glu334 catalytic triad, where serine acts as a nucleophile in the hydrolysis reaction [27]. Additionally, the peripheral site contains aromatic residues that interact with the substrate and allosteric modulators, playing a key role in enzyme inhibition [27]. These structural features make AChE an attractive target for pharmacological drug design, particularly in the development of inhibitors for the treatment of neurodegenerative diseases [28].
The tissue and intracellular localization of AChE varies depending on its function and the cell type in which it is expressed. It is widely distributed in the central nervous system (CNS), specifically in the cerebral cortex, hippocampus, and brainstem, where it plays a role in regulating cholinergic neurotransmission [29]. In the peripheral nervous system, it is present at neuromuscular junctions, facilitating skeletal muscle contraction and relaxation. It is also expressed in erythrocytes, where it contributes to the regulation of responses to neurotoxic agents.
At the cellular level, AChE can be anchored to the plasma membrane, associated with the outer synaptic surface, or exist in soluble forms within intracellular compartments [30]. Its regulation occurs through gene expression mechanisms, post-translational modifications, and allosteric modulation, as well as interactions with proteins such as cholinergic components, neuropilins, and extracellular matrix proteins, which influence its activity and stability [31].

3. Structural Bioinformatics Methods in the Study of AChE

Molecular docking is a computational technique used in drug design to predict the interaction between a small molecule (ligand) and a biological macromolecule (protein or DNA) [32]. This approach is based on exploring the conformational space of the ligand and its accommodation within the active site of the target protein, evaluating binding affinity through optimization algorithms and energy functions [33]. This method allows for the identification of compounds with a high probability of specific binding, facilitating the prioritization of candidates for experimental assays. There are two main strategies in molecular docking: rigid docking, in which both the ligand and the protein maintain fixed structures, and flexible docking, which allows for the exploration of conformational changes in the ligand or protein [34]. This technique is widely used in the identification of enzyme inhibitors, the development of multitarget drugs, and the optimization of compounds in computational pharmacology.
Among the most commonly used molecular docking software, AutoDock and AutoDock Vina stand out as open-source tools widely applied in ligand–protein interaction predictions using genetic algorithms and stochastic searches [32]. Other popular programs include Molecular Operating Environment (MOE), which employs empirical energy functions to evaluate complex stability, and SwissDock, which is based on the EADock DSS engine and optimized for high-efficiency conformational exploration [35]. Additionally, Glide (Schrödinger Inc., New York, NY, USA), which incorporates quantum force models, and Gold, recognized for its accuracy in binding mode prediction, are also widely used [34]. Thanks to these tools, molecular docking has revolutionized structural pharmacology by enabling the virtual screening of thousands of compounds in significantly less time than traditional experimental methods, facilitating drug discovery and optimization.
Acetylcholinesterase (AChE) has been extensively studied using bioinformatics approaches to understand its structure, function, and interactions with potential inhibitors. These computational studies have provided valuable insights into the enzyme’s active site dynamics, facilitating the design of more effective and selective AChE inhibitors. For instance, molecular docking and molecular dynamics simulations have been employed to predict how various compounds interact with AChE, aiding in the identification of promising therapeutic candidates for neurodegenerative diseases such as Alzheimer’s disease. The continuous advancements in bioinformatics tools and techniques have significantly enhanced our ability to model AChE’s behavior and its interactions at a molecular level [36].
The importance of AChE as a pharmacological target ensures that bioinformatics studies will continue to progress in the future. As new computational methods emerge, they provide more precise modeling of AChE’s structure and its binding affinities with various ligands. These advancements not only expedite the drug discovery process but also reduce reliance on extensive laboratory experiments in the early stages of research [37]. Moreover, bioinformatics approaches enable the screening of vast chemical libraries to identify potential AChE inhibitors, thereby broadening the scope of therapeutic exploration. The integration of bioinformatics with experimental studies remains a powerful strategy for developing effective treatments targeting AChE [38].

4. Applications of Molecular Docking in AChE

Molecular docking has been extensively used to identify acetylcholinesterase (AChE) inhibitors and analyze the interaction of previously known compounds, aiming to develop more effective drugs for treating Alzheimer’s disease. Additionally, the detailed study of these interactions provides valuable insights into the activation and inhibition mechanisms of AChE. Understanding these processes at the molecular level not only allows for the optimization of existing inhibitors but also facilitates the design of new molecules with greater selectivity and efficacy in modulating enzymatic activity [39]. A quantitative analysis of research output reveals an increase in molecular docking studies focused on acetylcholinesterase over the past five years (Figure 2). This trend correlates with the mounting interest in multi-target ligands for neurodegenerative diseases.
A compilation of molecular docking studies was conducted in which acetylcholinesterase (AChE) was used as a pharmacological target. These studies aim to design and identify the reversible and partial-action inhibitors of AChE, which is crucial for developing new treatments, especially for neurodegenerative diseases such as Alzheimer’s. To achieve this, various strategies have been explored, including the design of new molecules through computational approaches and the evaluation of existing compounds with potential inhibitory activity. This comprehensive analysis evaluates 121 published studies investigating AChE inhibitors through molecular docking approaches, with a particular focus on three critical pharmacological parameters: selectivity, affinity, and reversibility (Supplementary Materials Table S1).
Among the most frequently used resources in these studies, the crystal structure 4EY7 stands out, appearing in a total of 31 records. It is followed by 4M0E with 14 records and 4EY6 with 13. The prominence of 4EY7 is notable, as this structure is co-crystallized with Donepezil, the most commonly used positive control in the assays. In contrast, 4EY6 is co-crystallized with galantamine, another widely used reference inhibitor. Most studies employ one of these inhibitors as a positive control, and in some cases, both are used. Regarding computational tools, AutoDock Vina is the most widely used software for molecular docking due to its efficiency in calculating binding energies and its ability to explore different conformations of the evaluated compounds. Moreover, it is the most widely cited open-source molecular docking software in the literature. These approaches have led to significant advancements in identifying new AChE inhibitors and optimizing compounds with therapeutic potential.
The prevalence of these studies is enabled by well-established AChE structural data, with the most frequently employed crystal structures being PDB 4EY7, 4M0E, 4EY6, 4EY5, and 1EVE. AutoDock Vina emerged as the most used tool. Docking studies incorporated subsequent molecular dynamics simulations, with simulation timescales typically ranging from 50 to 200 ns for system equilibration (Figure 3).

5. Molecular Dynamics Simulations in AChE

Molecular dynamics is a computational tool that allows for the study of the flexibility and temporal behavior of acetylcholinesterase (AChE) and its interactions with various inhibitors. The essence of this approach is to verify the stability of the predicted interactions obtained from molecular docking assays. However, it is a computationally demanding process that requires significant resources and simulation time. For this reason, molecular docking is often used to screen large databases and select the best candidates for subsequent testing with molecular dynamics. For example, Ref. [40] evaluated a library of 2270 phytochemicals, identifying three promising compounds that demonstrated stability in protein–ligand complexes during 100-nanosecond simulations, suggesting their potential as AChE inhibitors. Similarly, Ref. [41] designed and synthesized 18 new pyrrolidin-2-one derivatives; however, they only demonstrated the efficacy of two compounds through molecular dynamics simulations that formed stable complexes with AChE, indicating their potential effectiveness as inhibitors. A compilation of studies utilizing molecular dynamics with acetylcholinesterase and a test molecule is presented in Supplementary Materials Table S2.

6. Discussion

Molecular docking assays have emerged as a fundamental tool in drug development, particularly in the identification of acetylcholinesterase (AChE) inhibitors. Their application has optimized the search for compounds with high specificity and affinity towards the enzyme, which is crucial in designing therapies for neurodegenerative diseases such as Alzheimer’s. AChE inhibition is a validated therapeutic strategy, as reducing the degradation of acetylcholine enhances cholinergic neurotransmission, a key aspect in mitigating the symptoms of Alzheimer’s disease [42]. However, developing effective inhibitors faces multiple challenges, including selectivity, toxicity, and the ability to cross the blood–brain barrier. In this context, molecular docking has not only facilitated the identification of new compounds with therapeutic potential but also allowed for the structural optimization of existing drugs, improving their pharmacokinetic profile and reducing adverse effects [43]. Moreover, combining docking with advanced techniques such as molecular dynamics and machine learning has enhanced its accuracy and efficiency, enabling better predictions of drug–receptor interactions [16,44]. Given the impact of these methodologies on streamlining drug discovery, it is evident that their use will continue to evolve and solidify as key tools.
Acetylcholinesterase (AChE) is characterized by an active site located at the bottom of a deep and narrow cleft approximately 20 Å deep and 5 Å wide. This cleft is lined with 14 highly conserved aromatic residues, among which tryptophan 84 (Trp84) plays a crucial role in the binding of the acetylthiocholine [45]. Inside its active site, it presents a catalytic triad formed by serine, histidine, and glutamate, located at the bottom of the cleft. This structural arrangement poses significant challenges for molecular docking assays, where the goal is to obtain ligands that interact with the catalytic triad. Nonetheless, the primary aim is to achieve reversible inhibition, so interaction with the enzyme’s peripheral site is sufficient to cause inhibition and will favor the reversibility of the inhibition [19,23]. Additionally, AChE inhibitors that act at both the active site and the peripheral anionic site (PAS) have been developed to prevent Aβ fibril aggregation [2]. Advances are aimed at finding new, highly selective, reversible, and effective inhibitors of AChE to improve treatments for neurodegenerative diseases.
AutoDock Vina is the most widely used software for molecular docking studies with the acetylcholinesterase (AChE) enzyme. Its open-source nature, efficiency, and ease of use make it ideal for screening large databases of potential compounds. Furthermore, AutoDock Vina requires minimal computational resources and offers fast processing times, facilitating its integration into drug discovery workflows. For example, screening work can be conducted using techniques that require few computational resources, and subsequently, more expensive techniques such as molecular dynamics can be employed [46].
A direct comparison of the results obtained from different docking programs is not recommended due to variations in their algorithms and scoring functions [47]. Each software may generate different binding affinity values even when using the same PDB files and ligands [48]. For instance, it has been observed that programs like CDOCKER and ClusPro yielded higher binding affinity values compared to others (Table 1). Additionally, tools like Schrödinger have shown a greater range of variability in the results, with affinity energy results ranging from −76.3 to 0.8 Kcal/mol observed in this work. To make comparisons and ana lyze our results, it is advisable to use known molecules for which experimental data or proven tests are available.
Molecular dynamics studies are a way to verify that the complexes obtained as a result of molecular docking represent stable interactions. The most widely used software for performing molecular dynamics simulations of ligand–receptor complexes, where the receptor is typically a protein, is GROMACS. GROMACS is open-source software that enables highly efficient molecular dynamics simulations, maximizing the use of available computational resources and being compatible with several affordable GPU cards on the market [49,50].
The simulation time varies depending on the phenomenon being studied in the system. Generally, for evaluating the stability of a ligand–receptor complex, 100 ns is sufficient to confirm its stability. In some cases, if the ligand moves or adjusts within the active site during the simulation, the RMSD tends to increase (>2 nm). If the ligand readjusts, the RMSD usually decreases again; however, if the RMSD remains elevated, it provides information about the ligand’s instability in the binding site where it was initially docked through molecular docking studies.
Molecular dynamics is a method to validate docking results; however, verification can also be performed through experimental studies or complemented with other computational analyses.
The integration of experimental data with molecular docking studies is essential to validate and improve the accuracy of computational predictions in identifying acetylcholinesterase (AChE) inhibitors. For example, in a recent study, benzofuran derivatives were designed and synthesized as potential AChE inhibitors. In vitro biological assays revealed that compound 7c exhibited promising inhibitory activity, with an IC50 of 0.058 μM. These experimental findings were consistent with molecular docking results, which showed a favorable interaction of compound 7c in the active site of AChE [51]. For acetylcholinesterase, the most used molecule for comparison is Donepezil, and there is variation in the resulting binding energy (Table 2); therefore, it should be considered for inclusion as a control in each study so that docking assays are conducted under the same conditions as the molecules of interest.
The appropriate selection of crystal structures from the Protein Data Bank (PDB) is essential for obtaining accurate results in molecular docking assays. For acetylcholinesterase, it was observed that the most used crystal is 4EY7, with many more citations than the other crystals (Table 3). A key consideration for selecting a PDB crystal is the resolution of the structure; higher resolutions provide more precise details about the conformation of the active site, which improves the accuracy of docking. Additionally, it is crucial to assess the presence of co-crystallized ligands or inhibitors, as these can induce conformational changes in the protein that affect its interaction with new compounds. Crystal-structure 4EY7 stems from human acetylcholinesterase, crystallized with Donepezil, and has a resolution of 2.35 Å, an acceptable value. Being crystallized with Donepezil allows for comparing inhibition by this drug and searching for similar interactions for the new molecules to be tested [95].
Across the reviewed studies, molecular docking analyses revealed consistent and critical interactions between the tested ligands and key amino acid residues within the active or peripheral binding sites of acetylcholinesterase (AChE). Notably, π–π stacking and cation–π interactions with Trp86, Trp286, and Tyr337 emerged as highly recurrent and relevant across multiple compounds, supporting their crucial role in ligand anchoring and enzymatic inhibition (e.g., Refs. [68,96,97]). Hydrogen bonding with residues such as Ser203, Glu202, Tyr124, and His447 was also frequently observed, enhancing ligand stability within the catalytic site, as reported in Refs. [77,98,99]. Compounds targeting the peripheral anionic site (PAS), like those in Refs. [93,100], often interacted with Asp74 and Tyr341, which are known to influence β-amyloid aggregation. These overlapping interactions suggest that ligands forming multiple contacts with both CAS and PAS residues—especially those involving Trp86, Tyr337, and Phe295—may exhibit superior dual-site binding and therapeutic potential for Alzheimer’s disease treatment.
Some additional considerations to keep in mind include the presence of water molecules in the active site, which are necessary if the goal is to emulate substrates, as a water molecule is required for the chemical reaction [101]. However, most studies aim to develop a molecule that inhibits reversibly, meaning that interaction with the peripheral site is sufficient to generate enzyme inhibition. Additionally, some inhibitors bind to sites other than the active site, and understanding these mechanisms will allow for the development of molecules with greater versatility to inhibit AChE to different extents and at varying concentrations [102,103].
Molecular docking will always produce a result, as the software is designed to provide the best possible ligand conformation within the protein. However, interpretation and comparison depend on a well-designed experiment, having a reference point such as Donepezil, and understanding how the protein functions.

7. Limitations of Computational Tools

Computational tools play a crucial role in drug design and molecular docking, offering rapid analysis and reducing the costs associated with experimental methods. However, their usage comes with notable limitations.
Energy affinity depends on the algorithms employed to calculate, and this is specified according to the software used. Another limitation is the availability of protein data; in some cases, the protein of interest is partially described, and the crystal structure is incomplete. Additionally, the metabolism does not concern, so docking only considers the energy affinity at a specific point in time [104], this suggest that we need the molecular dynamic to examine the interaction in a fine way On the other hand, molecular dynamics involves ligand–protein interaction over long periods of time, which requires deep learning and machine learning methods but also high computational power due to using quantum mechanical methods to calculate the stability and fluctuation of ligand–protein binding [105,106,107].
Computational studies or in silico studies are faster than in vivo or in vitro experiments; however, they are not substitutes. In silico studies are used to predict and estimate the interaction sites during protein–ligand binding.

8. Conclusions

Molecular docking has established itself as a fundamental tool in the rational design of acetylcholinesterase (AChE) inhibitors, providing valuable insights into ligand binding interactions and selectivity. However, the complexity of AChE’s active site, its deep and narrow gorge, and the involvement of key residues in ligand recognition present challenges that require a careful and systematic approach. The selection of appropriate crystal structures, such as PDB ID: 4EY7, and the inclusion of well-characterized control molecules like Donepezil, are crucial for ensuring reliable docking studies. Furthermore, docking alone may not fully capture the dynamic nature of ligand binding; thus, molecular dynamics (MD) simulations, particularly with GROMACS, have become indispensable for validating the docking results by assessing the stability and conformational flexibility of ligand–protein complexes over time. Studies indicate that a 100 ns MD simulation is often sufficient to confirm the stability of docked ligands but longer simulations may be necessary for more complex interactions. Additionally, integrating experimental validation with computational predictions enhances the accuracy and applicability of in silico methods in drug discovery. As molecular docking and MD simulations continue to evolve, their combined application will remain a cornerstone in the development of highly selective, reversible, and effective AChE inhibitors, paving the way for improved therapeutic strategies against neurodegenerative diseases.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms26083781/s1. Table S1: Acetylcholinesterase (AChE) as target in docking studies [19,43,47,48,49,50,51,52,53,54,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164]. Table S2: Molecular dynamic studies about AChE [21,40,50,66,71,73,74,79,84,86,89,156,157,158,159,160,161,162,163,164].

Author Contributions

Conceptualization, A.R.-C. and M.F.R.-G.; validation, D.E.N.-Á., A.R.-C. and A.Y.T.-B.; formal analysis, M.F.R.-G.; investigation, A.R.-C.; writing—original draft preparation, D.E.N.-Á.; writing—review and editing, A.Y.T.-B.; visualization, A.R.-C., D.E.N.-Á., M.F.R.-G. and A.Y.T.-B.; supervision, A.R.-C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grant (20241240) Secretaria de Investigación y Posgrado (SIP)-Instituto Politécnico Nacional (IPN) and grant (20241771) SIP-IPN.

Acknowledgments

The authors thank Escuela Nacional de Ciencias Biológicas. Instituto Politécnico Nacional. Sistema Nacional de Investigadores. Secretaria de Ciencia, Humanidades, Tecnología e Innovación for membership, as well as Benemérita Universidad de Puebla.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

The following abbreviations are used in this manuscript:
AChEAcetylcholinesterase
MOEMolecular Operating Environment
LDLinear dichroism
PDBProtein Data Bank
NAData not available

References

  1. Basnet, R.; Khadka, S.; Basnet, B.B.; Gupta, R. Perspective on Acetylcholinesterase: A Potential Target for Alzheimer’s Disease Intervention. Curr. Enzym. Inhib. 2020, 16, 181–188. [Google Scholar] [CrossRef]
  2. Carvajal, F.J.; Inestrosa, N.C. Interactions of AChE with Aβ Aggregates in Alzheimer’s Brain: Therapeutic Relevance of IDN 5706. Front. Mol. Neurosci. 2011, 4, 19. [Google Scholar] [CrossRef] [PubMed]
  3. Saxena, M.; Dubey, R. Target Enzyme in Alzheimer’s Disease: Acetylcholinesterase Inhibitors. Curr. Top. Med. Chem. 2019, 19, 264–275. [Google Scholar] [CrossRef] [PubMed]
  4. Vignaux, P.A.; Minerali, E.; Lane, T.R.; Foil, D.H.; Madrid, P.B.; Puhl, A.C.; Ekins, S. The Antiviral Drug Tilorone Is a Potent and Selective Inhibitor of Acetylcholinesterase. Chem. Res. Toxicol. 2021, 34, 1296–1307. [Google Scholar] [CrossRef]
  5. García Marín, I.D.; Camarillo López, R.H.; Martínez, O.A.; Padilla-Martínez, I.I.; Correa-Basurto, J.; Rosales-Hernández, M.C. New Compounds from Heterocyclic Amines Scaffold with Multitarget Inhibitory Activity on Aβ Aggregation, AChE, and BACE1 in the Alzheimer Disease. PLoS ONE 2022, 17, e0269129. [Google Scholar] [CrossRef]
  6. Hernández-Rodríguez, M.; Correa-Basurto, J.; Martínez-Ramos, F.; Padilla-Martínez, I.I.; Benítez-Cardoza, C.G.; Mera-Jiménez, E.; Rosales-Hernández, M.C. Design of Multi-Target Compounds as AChE, BACE1, and Amyloid-Β1-42 Oligomerization Inhibitors: In Silico and In Vitro Studies. J. Alzheimer’s Dis. 2014, 41, 1073–1085. [Google Scholar] [CrossRef]
  7. Manzoor, S.; Gabr, M.T.; Rasool, B.; Pal, K.; Hoda, N. Dual Targeting of Acetylcholinesterase and Tau Aggregation: Design, Synthesis and Evaluation of Multifunctional Deoxyvasicinone Analogues for Alzheimer’s Disease. Bioorg. Chem. 2021, 116, 105354. [Google Scholar] [CrossRef]
  8. Baruah, P.; Basumatary, G.; Yesylevskyy, S.O.; Aguan, K.; Bez, G.; Mitra, S. Novel Coumarin Derivatives as Potent Acetylcholinesterase Inhibitors: Insight into Efficacy, Mode and Site of Inhibition. J. Biomol. Struct. Dyn. 2019, 37, 1750–1765. [Google Scholar] [CrossRef]
  9. Camps, P.; El Achab, R.; Morral, J.; Muñoz-Torrero, D.; Badia, A.; Baños, J.E.; Vivas, N.M.; Barril, X.; Orozco, M.; Luque, F.J. New Tacrine−Huperzine A Hybrids (Huprines): Highly Potent Tight-Binding Acetylcholinesterase Inhibitors of Interest for the Treatment of Alzheimer’s Disease. J. Med. Chem. 2000, 43, 4657–4666. [Google Scholar] [CrossRef]
  10. Lotfi, S.; Rahmani, T.; Hatami, M.; Pouramiri, B.; Kermani, E.T.; Rezvannejad, E.; Mortazavi, M.; Fathi Hafshejani, S.; Askari, N.; Pourjamali, N.; et al. Design, Synthesis and Biological Assessment of Acridine Derivatives Containing 1,3,4-Thiadiazole Moiety as Novel Selective Acetylcholinesterase Inhibitors. Bioorg. Chem. 2020, 105, 104457. [Google Scholar] [CrossRef]
  11. Berman, H.M. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235–242. [Google Scholar] [CrossRef] [PubMed]
  12. Kandiah, N.; Pai, M.-C.; Senanarong, V.; Looi, I.; Ampil, E.; Park, K.W.; Karanam, A.K.; Christopher, S. Rivastigmine: The Advantages of Dual Inhibition of Acetylcholinesterase and Butyrylcholinesterase and Its Role in Subcortical Vascular Dementia and Parkinson’s Disease Dementia. Clin. Interv. Aging 2017, 12, 697–707. [Google Scholar] [CrossRef]
  13. Sharon, N.; Ugale, V.G.; Padmaja, P.; Lokwani, D.; Salunkhe, C.; Shete, P.; Reddy, P.N.; Kulkarni, P.P. Development of Novel 9H-Carbazole-4H-Chromene Hybrids as Dual Cholinesterase Inhibitors for the Treatment of Alzheimer’s Disease. Mol. Divers. 2025, 29, 379–396. [Google Scholar] [CrossRef] [PubMed]
  14. Ojo, O.A.; Ojo, A.B.; Okolie, C.; Nwakama, M.-A.C.; Iyobhebhe, M.; Evbuomwan, I.O.; Nwonuma, C.O.; Maimako, R.F.; Adegboyega, A.E.; Taiwo, O.A.; et al. Deciphering the Interactions of Bioactive Compounds in Selected Traditional Medicinal Plants against Alzheimer’s Diseases via Pharmacophore Modeling, Auto-QSAR, and Molecular Docking Approaches. Molecules 2021, 26, 1996. [Google Scholar] [CrossRef] [PubMed]
  15. Thai, Q.M.; Pham, M.Q.; Tran, P.-T.; Nguyen, T.H.; Ngo, S.T. Searching for Potential Acetylcholinesterase Inhibitors: A Combined Approach of Multi-Step Similarity Search, Machine Learning and Molecular Dynamics Simulations. R. Soc. Open Sci. 2024, 11, 240546. [Google Scholar] [CrossRef]
  16. Son, M.; Park, C.; Rampogu, S.; Zeb, A.; Lee, K.W. Discovery of Novel Acetylcholinesterase Inhibitors as Potential Candidates for the Treatment of Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 1000. [Google Scholar] [CrossRef]
  17. Onorini, D.; Schoborg, R.; Borel, N.; Leonard, C. Beta Lactamase-Producing Neisseria Gonorrhoeae Alleviates Amoxicillin-Induced Chlamydial Persistence in a Novel in Vitro Co-Infection Model. Curr. Res. Microb. Sci. 2023, 4, 100188. [Google Scholar] [CrossRef]
  18. Bag, S.; Tulsan, R.; Sood, A.; Datta, S.; Török, M. Pharmacophore Modeling, Virtual and in Vitro Screening for Acetylcholinesterase Inhibitors and Their Effects on Amyloid-β Self- Assembly. Curr. Comput. Aided Drug Des. 2013, 9, 2–14. [Google Scholar]
  19. Eckert, S.; Eyer, P.; Worek, F. Reversible Inhibition of Acetylcholinesterase by Carbamates or Huperzine a Increases Residual Activity of the Enzyme upon Soman Challenge. Toxicology 2007, 233, 180–186. [Google Scholar] [CrossRef]
  20. Gebre, T.; Ayele, B.; Zerihun, M.; Genet, A.; Stoller, N.E.; Zhou, Z.; House, J.I.; Yu, S.N.; Ray, K.J.; Emerson, P.M.; et al. Comparison of Annual versus Twice-Yearly Mass Azithromycin Treatment for Hyperendemic Trachoma in Ethiopia: A Cluster-Randomised Trial. Lancet 2012, 379, 143–151. [Google Scholar] [CrossRef]
  21. Mishra, C.B.; Kumari, S.; Manral, A.; Prakash, A.; Saini, V.; Lynn, A.M.; Tiwari, M. Design, Synthesis, in-Silico and Biological Evaluation of Novel Donepezil Derivatives as Multi-Target-Directed Ligands for the Treatment of Alzheimer’s Disease. Eur. J. Med. Chem. 2017, 125, 736–750. [Google Scholar] [CrossRef] [PubMed]
  22. Li, Y.; Zhang, Q.; Wang, X.; Liu, Z.; Chen, H.; Su, Z.; Xu, Y.; Zhang, W.; Du, Y.; Tan, Z.; et al. Development of Novel Rivastigmine Derivatives as Selective BuChE Inhibitors for the Treatment of AD. Bioorg. Chem. 2025, 157, 108245. [Google Scholar] [CrossRef]
  23. Colovic, M.B.; Krstic, D.Z.; Lazarevic-Pasti, T.D.; Bondzic, A.M.; Vasic, V.M. Acetylcholinesterase Inhibitors: Pharmacology and Toxicology. Curr. Neuropharmacol. 2013, 11, 315–335. [Google Scholar] [CrossRef]
  24. Dvir, H.; Silman, I.; Harel, M.; Rosenberry, T.L.; Sussman, J.L. Acetylcholinesterase: From 3D Structure to Function. Chem. Biol. Interact. 2010, 187, 10–22. [Google Scholar] [CrossRef]
  25. Bourne, Y.; Taylor, P.; Bougis, P.E.; Marchot, P. Crystal Structure of Mouse Acetylcholinesterase. J. Biol. Chem. 1999, 274, 2963–2970. [Google Scholar] [CrossRef]
  26. Johnson, G.; Moore, S. The Peripheral Anionic Site of Acetylcholinesterase: Structure, Functions and Potential Role in Rational Drug Design. Curr. Pharm. Des. 2006, 12, 217–225. [Google Scholar] [CrossRef] [PubMed]
  27. Shafferman, A.; Kronman, C.; Flashner, Y.; Leitner, M.; Grosfeld, H.; Ordentlich, A.; Gozes, Y.; Cohen, S.; Ariel, N.; Barak, D. Mutagenesis of Human Acetylcholinesterase. Identification of Residues Involved in Catalytic Activity and in Polypeptide Folding. J. Biol. Chem. 1992, 267, 17640–17648. [Google Scholar] [CrossRef] [PubMed]
  28. Walczak-Nowicka, Ł.J.; Herbet, M. Acetylcholinesterase Inhibitors in the Treatment of Neurodegenerative Diseases and the Role of Acetylcholinesterase in Their Pathogenesis. Int. J. Mol. Sci. 2021, 22, 9290. [Google Scholar] [CrossRef]
  29. Fishman, E.B.; Siek, G.C.; MacCallum, R.D.; Bird, E.D.; Volicer, L.; Marquis, J.K. Distribution of the Molecular Forms of Acetylcholinesterase in Human Brain: Alterations in Dementia of the Alzheimer Type. Ann. Neurol. 1986, 19, 246–252. [Google Scholar] [CrossRef]
  30. Fernandez, H.L.; Moreno, R.D.; Inestrosa, N.C. Tetrameric (G4) Acetylcholinesterase: Structure, Localization, and Physiological Regulation. J. Neurochem. 1996, 66, 1335–1346. [Google Scholar] [CrossRef]
  31. Coleman, B.A.; Taylor, P. Regulation of Acetylcholinesterase Expression during Neuronal Differentiation. J. Biol. Chem. 1996, 271, 4410–4416. [Google Scholar] [CrossRef] [PubMed]
  32. Trott, O.; Olson, A.J. AutoDock Vina: Improving the Speed and Accuracy of Docking with a New Scoring Function, Efficient Optimization, and Multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef]
  33. Morris, G.M.; Huey, R.; Lindstrom, W.; Sanner, M.F.; Belew, R.K.; Goodsell, D.S.; Olson, A.J. AutoDock4 and AutoDockTools4: Automated Docking with Selective Receptor Flexibility. J. Comput. Chem. 2009, 30, 2785–2791. [Google Scholar] [CrossRef]
  34. Pagadala, N.S.; Syed, K.; Tuszynski, J. Software for Molecular Docking: A Review. Biophys. Rev. 2017, 9, 91–102. [Google Scholar] [CrossRef] [PubMed]
  35. Grosdidier, A.; Zoete, V.; Michielin, O. SwissDock, a Protein-Small Molecule Docking Web Service Based on EADock DSS. Nucleic Acids Res. 2011, 39, W270–W277. [Google Scholar] [CrossRef]
  36. Bermudez-Lugo, J.A.; Rosales-Hernandez, M.C.; Deeb, O.; Trujillo-Ferrara, J.; Correa-Basurto, J. In Silico Methods to Assist Drug Developers in Acetylcholinesterase Inhibitor Design. Curr. Med. Chem. 2011, 18, 1122–1136. [Google Scholar] [CrossRef] [PubMed]
  37. Pérez-Sánchez, H.; den Haan, H.; Pérez-Garrido, A.; Peña-García, J.; Chakraborty, S.; Erdogan Orhan, I.; Senol Deniz, F.S.; Villalgordo, J.M. Combined Structure and Ligand-Based Design of Selective Acetylcholinesterase Inhibitors. J. Chem. Inf. Model. 2021, 61, 467–480. [Google Scholar] [CrossRef]
  38. Pereira, G.R.C.; Gonçalves, L.M.; de Azevedo Abrahim-Vieira, B.; De Mesquita, J.F. In Silico Analyses of Acetylcholinesterase (AChE) and Its Genetic Variants in Interaction with the Anti-Alzheimer Drug Rivastigmine. J. Cell Biochem. 2022, 123, 1259–1277. [Google Scholar] [CrossRef]
  39. Fang, J.; Wu, P.; Yang, R.; Gao, L.; Li, C.; Wang, D.; Wu, S.; Liu, A.-L.; Du, G.-H. Inhibition of Acetylcholinesterase by Two Genistein Derivatives: Kinetic Analysis, Molecular Docking and Molecular Dynamics Simulation. Acta Pharm. Sin. B 2014, 4, 430–437. [Google Scholar] [CrossRef]
  40. Azmal, M.; Hossen, M.S.; Shohan, M.N.H.; Taqui, R.; Malik, A.; Ghosh, A. A Computational Approach to Identify Phytochemicals as Potential Inhibitor of Acetylcholinesterase: Molecular Docking, ADME Profiling and Molecular Dynamics Simulations. PLoS ONE 2024, 19, e0304490. [Google Scholar] [CrossRef]
  41. Gupta, M.; Kumar, A.; Prasun, C.; Nair, M.S.; Kini, S.G.; Yadav, D.; Nain, S. Design, Synthesis, Extra-Precision Docking, and Molecular Dynamics Simulation Studies of Pyrrolidin-2-One Derivatives as Potential Acetylcholinesterase Inhibitors. J. Biomol. Struct. Dyn. 2023, 41, 6282–6294. [Google Scholar] [CrossRef] [PubMed]
  42. Rees, T.M.; Brimijoin, S. The Role of Acetylcholinesterase in the Pathogenesis of Alzheimer’s Disease. Drugs Today 2003, 39, 75–83. [Google Scholar] [CrossRef] [PubMed]
  43. Galimberti, D.; Scarpini, E. Old and New Acetylcholinesterase Inhibitors for Alzheimer’s Disease. Expert. Opin. Investig. Drugs 2016, 25, 1181–1187. [Google Scholar] [CrossRef]
  44. El Khatabi, K.; El-Mernissi, R.; Aanouz, I.; Ajana, M.A.; Lakhlifi, T.; Khan, A.; Wei, D.-Q.; Bouachrine, M. Identification of Novel Acetylcholinesterase Inhibitors through 3D-QSAR, Molecular Docking, and Molecular Dynamics Simulation Targeting Alzheimer’s Disease. J. Mol. Model. 2021, 27, 302. [Google Scholar] [CrossRef] [PubMed]
  45. Sussman, J.L.; Harel, M.; Silman, I. Three-Dimensional Structure of Acetylcholinesterase and of Its Complexes with Anticholinesterase Drugs. Chem. Biol. Interact. 1993, 87, 187–197. [Google Scholar] [CrossRef]
  46. Nogara, P.A.; de Aquino Saraiva, R.; Caeran Bueno, D.; Lissner, L.J.; Lenz Dalla Corte, C.; Braga, M.M.; Rosemberg, D.B.; Rocha, J.B.T. Virtual Screening of Acetylcholinesterase Inhibitors Using the Lipinski’s Rule of Five and ZINC Databank. Biomed. Res. Int. 2015, 2015, 870389. [Google Scholar] [CrossRef]
  47. Chen, Z.; Li, H.; Zhang, Q.; Bao, X.; Yu, K.; Luo, X.; Zhu, W.; Jiang, H. Pharmacophore-Based Virtual Screening versus Docking-Based Virtual Screening: A Benchmark Comparison against Eight Targets. Acta Pharmacol. Sin. 2009, 30, 1694–1708. [Google Scholar] [CrossRef]
  48. Cross, J.B.; Thompson, D.C.; Rai, B.K.; Baber, J.C.; Fan, K.Y.; Hu, Y.; Humblet, C. Comparison of Several Molecular Docking Programs: Pose Prediction and Virtual Screening Accuracy. J. Chem. Inf. Model. 2009, 49, 1455–1474. [Google Scholar] [CrossRef]
  49. Khan, M.I.; Pathania, S.; Al-Rabia, M.W.; Ethayathulla, A.S.; Khan, M.I.; Allemailem, K.S.; Azam, M.; Hariprasad, G.; Imran, M.A. MolDy: Molecular Dynamics Simulation Made Easy. Bioinformatics 2024, 40, btae313. [Google Scholar] [CrossRef]
  50. Van Der Spoel, D.; Lindahl, E.; Hess, B.; Groenhof, G.; Mark, A.E.; Berendsen, H.J.C. GROMACS: Fast, Flexible, and Free. J. Comput. Chem. 2005, 26, 1701–1718. [Google Scholar] [CrossRef]
  51. Abd El-Karim, S.S.; Anwar, M.M.; Ahmed, N.S.; Syam, Y.M.; Elseginy, S.A.; Aly, H.F.; Younis, E.A.; Khalil, W.K.B.; Ahmed, K.A.; Mohammed, F.F.; et al. Discovery of Novel Benzofuran-Based Derivatives as Acetylcholinesterase Inhibitors for the Treatment of Alzheimer’s Disease: Design, Synthesis, Biological Evaluation, Molecular Docking and 3D-QSAR Investigation. Eur. J. Med. Chem. 2023, 260, 115766. [Google Scholar] [CrossRef] [PubMed]
  52. Homoud, Z.A.; Taha, M.; Rahim, F.; Iqbal, N.; Nawaz, M.; Farooq, R.K.; Wadood, A.; Alomari, M.; Islam, I.; Algheribe, S.; et al. Synthesis of Indole Derivatives as Alzheimer Inhibitors and Their Molecular Docking Study. J. Biomol. Struct. Dyn. 2023, 41, 9865–9878. [Google Scholar] [CrossRef] [PubMed]
  53. Azman, N.A.N.; Alhawarri, M.B.; Rawa, M.S.A.; Dianita, R.; Gazzali, A.M.; Nogawa, T.; Wahab, H.A. Potential Anti-Acetylcholinesterase Activity of Cassia Timorensis DC. Molecules 2020, 25, 4545. [Google Scholar] [CrossRef]
  54. Tsai, C.-H.; Liou, Y.-L.; Li, S.-M.; Liao, H.-R.; Chen, J.-J. Antioxidant, Anti-α-Glucosidase, Anti-Tyrosinase, and Anti-Acetylcholinesterase Components from Stem of Rhamnus Formosana with Molecular Docking Study. Antioxidants 2024, 14, 8. [Google Scholar] [CrossRef]
  55. Amir Rawa, M.S.; Nurul Azman, N.A.; Mohamad, S.; Nogawa, T.; Wahab, H.A. In Vitro and In Silico Anti-Acetylcholinesterase Activity from Macaranga Tanarius and Syzygium Jambos. Molecules 2022, 27, 2648. [Google Scholar] [CrossRef] [PubMed]
  56. Mohammadi-Farani, A.; Moradi, F.; Hosseini, A.; Aliabadi, A. Synthesis, Docking, Pharmacokinetic Prediction, and Acetylcholinesterase Inhibitory Evaluation of N-(2-(Piperidine-1-Yl)Ethyl)Benzamide Derivatives as Potential Anti-Alzheimer Agents. Res. Pharm. Sci. 2024, 19, 698–711. [Google Scholar] [CrossRef]
  57. Tran, T.S.; Le, M.T.; Nguyen, T.C.V.; Tran, T.H.; Tran, T.D.; Thai, K.M. Synthesis, in Silico and in Vitro Evaluation for Acetylcholinesterase and BACE-1 Inhibitory Activity of Some N-Substituted-4-Phenothiazine-Chalcones. Molecules 2020, 25, 3916. [Google Scholar] [CrossRef]
  58. Abbas-Mohammadi, M.; Moridi Farimani, M.; Salehi, P.; Ebrahimi, S.N.; Sonboli, A.; Kelso, C.; Skropeta, D. Molecular Networking Based Dereplication of AChE Inhibitory Compounds from the Medicinal Plant Vincetoxicum Funebre (Boiss. & Kotschy). J. Biomol. Struct. Dyn. 2022, 40, 1942–1951. [Google Scholar] [CrossRef]
  59. Amat-Ur-rasool, H.; Ahmed, M.; Hasnain, S.; Ahmed, A.; Carter, W.G. In Silico Design of Dual-Binding Site Anti-Cholinesterase Phytochemical Heterodimers as Treatment Options for Alzheimer’s Disease. Curr. Issues Mol. Biol. 2022, 44, 152–175. [Google Scholar] [CrossRef]
  60. Mella, M.; Moraga-Nicolás, F.; Machuca, J.; Quiroz, A.; Mutis, A.; Becerra, J.; Astudillo, Á.; Hormazábal, E. Acetylcholinesterase Inhibitory Activity from Amaryllis belladonna Growing in Chile: Enzymatic and Molecular Docking Studies. Nat. Prod. Res. 2022, 36, 1370–1374. [Google Scholar] [CrossRef]
  61. Kuzu, B.; Alagoz, M.A.; Demir, Y.; Gulcin, I.; Burmaoglu, S.; Algul, O. Structure-Based Inhibition of Acetylcholinesterase and Butyrylcholinesterase with 2-Aryl-6-Carboxamide Benzoxazole Derivatives: Synthesis, Enzymatic Assay, and in Silico Studies. Mol. Divers. 2025, 29, 671–693. [Google Scholar] [CrossRef]
  62. Chaichompoo, W.; Rojsitthisak, P.; Pabuprapap, W.; Siriwattanasathien, Y.; Yotmanee, P.; Haritakun, W.; Suksamrarn, A. Stephapierrines A-H, New Tetrahydroprotoberberine and Aporphine Alkaloids from the Tubers of: Stephania Pierrei Diels and Their Anti-Cholinesterase Activities. RSC Adv. 2021, 11, 21153–21169. [Google Scholar] [CrossRef] [PubMed]
  63. Hamed, A.A.; El-Shiekh, R.A.; Mohamed, O.G.; Aboutabl, E.A.; Fathy, F.I.; Fawzy, G.A.; Al-Taweel, A.M.; Elsayed, T.R.; Tripathi, A.; Al-Karmalawy, A.A. Cholinesterase Inhibitors from an Endophytic Fungus Aspergillus Niveus Fv-Er401: Metabolomics, Isolation and Molecular Docking. Molecules 2023, 28, 2559. [Google Scholar] [CrossRef]
  64. Silva, L.; Ferreira, E.; Maryam; Espejo-Román, J.; Costa, G.; Cruz, J.; Kimani, N.; Costa, J.; Bittencourt, J.; Cruz, J.; et al. Galantamine Based Novel Acetylcholinesterase Enzyme Inhibitors: A Molecular Modeling Design Approach. Molecules 2023, 28, 1035. [Google Scholar] [CrossRef] [PubMed]
  65. Haider, R.; Agnello, L.; Shah, S.M.; Sufyan, M.; Khan, N.; Nazir, A.; Ciaccio, M.; Rehman, S. Evaluating the Antioxidant, Anti-inflammatory, and Neuroprotective Potential of Fruiting Body and Mycelium Extracts from Edible Yellow Morel (Morchella esculenta L. Pers.). J. Food Sci. 2025, 90, e17619. [Google Scholar] [CrossRef]
  66. Mateev, E.; Kondeva-Burdina, M.; Georgieva, M.; Zlatkov, A. Repurposing of FDA-Approved Drugs as Dual-Acting MAO-B and AChE Inhibitors against Alzheimer’s Disease: An in Silico and in Vitro Study. J. Mol. Graph. Model. 2023, 122, 108471. [Google Scholar] [CrossRef] [PubMed]
  67. Ordoñez, W.O.C.; Palomino, N.V.; Varela, P.E.V.; Martínez, I.B.; Alves, L.B.; Giuliatti, S. Alkaloids from Caliphruria Subedentata (Amaryllidaceae) as Regulators of AChE, BuChE, NMDA and GSK3 Activity: An In Vitro and In Silico Approach for Mimicking Alzheimer’s Disease. Neurochem. Res. 2025, 50, 116. [Google Scholar] [CrossRef]
  68. Anukanon, S.; Pongpamorn, P.; Tiyabhorn, W.; Chatwichien, J.; Niwetmarin, W.; Sessions, R.B.; Ruchirawat, S.; Thasana, N. In Silico-Guided Rational Drug Design and Semi-Synthesis of C(2)-Functionalized Huperzine A Derivatives as Acetylcholinesterase Inhibitors. ACS Omega 2021, 6, 19924–19939. [Google Scholar] [CrossRef]
  69. Barakat, A.; Alshahrani, S.; Al-Majid, A.M.; Ali, M.; Altowyan, M.S.; Islam, M.S.; Alamary, A.S.; Ashraf, S.; Ul-Haq, Z. Synthesis of a New Class of Spirooxindole-Benzo[b]Thiophene-Based Molecules as Acetylcholinesterase Inhibitors. Molecules 2020, 25, 4671. [Google Scholar] [CrossRef]
  70. Chennai, H.Y.; Belaidi, S.; Bourougaa, L.; Ouassaf, M.; Sinha, L.; Samadi, A.; Chtita, S. Identification of Potent Acetylcholinesterase Inhibitors as New Candidates for Alzheimer Disease via Virtual Screening, Molecular Docking, Dynamic Simulation, and Molecular Mechanics–Poisson–Boltzmann Surface Area Calculations. Molecules 2024, 29, 1232. [Google Scholar] [CrossRef]
  71. Saeed, S.; Zahoor, A.F.; Kamal, S.; Raza, Z.; Bhat, M.A. Unfolding the Antibacterial Activity and Acetylcholinesterase Inhibition Potential of Benzofuran-Triazole Hybrids: Synthesis, Antibacterial, Acetylcholinesterase Inhibition, and Molecular Docking Studies. Molecules 2023, 28, 6007. [Google Scholar] [CrossRef] [PubMed]
  72. Li, Z.; Shi, H. Study on the Active Ingredients of Shenghui Decoction Inhibiting Acetylcholinesterase Based on Molecular Docking and Molecular Dynamics Simulation. Medicine 2023, 102, e34909. [Google Scholar] [CrossRef]
  73. Thai, Q.M.; Nguyen, T.H.; Lenon, G.B.; Thu Phung, H.T.; Horng, J.-T.; Tran, P.-T.; Ngo, S.T. Estimating AChE Inhibitors from MCE Database by Machine Learning and Atomistic Calculations. J. Mol. Graph. Model. 2025, 134, 108906. [Google Scholar] [CrossRef] [PubMed]
  74. SahIn, K.; Durdagi, S. Combined Ligand and Structure-Based Virtual Screening Approaches for Identification of Novel Ache Inhibitors. Turk. J. Chem. 2020, 44, 574–588. [Google Scholar] [CrossRef] [PubMed]
  75. Makarian, M.; Gonzalez, M.; Salvador, S.M.; Lorzadeh, S.; Hudson, P.K.; Pecic, S. Synthesis, Kinetic Evaluation and Molecular Docking Studies of Donepezil-Based Acetylcholinesterase Inhibitors. J. Mol. Struct. 2022, 1247, 131425. [Google Scholar] [CrossRef]
  76. Zhang, Y.; Zhang, Y.; Li, S.; Liu, C.; Liang, J.; Nong, Y.; Chen, M.; Sun, R. Quaternity Method for Integrated Screening, Separation, Extraction Optimization, and Bioactivity Evaluation of Acetylcholinesterase Inhibitors from Sophora flavescens Aiton. Phytochem. Anal. 2025, 36, 52–67. [Google Scholar] [CrossRef] [PubMed]
  77. Hemaida, A.Y.; Hassan, G.S.; Maarouf, A.R.; Joubert, J.; El-Emam, A.A. Synthesis and Biological Evaluation of Thiazole-Based Derivatives as Potential Acetylcholinesterase Inhibitors. ACS Omega 2021, 6, 19202–19211. [Google Scholar] [CrossRef]
  78. Talukder, M.E.K.; Akhter, S.; Ahammad, F.; Aktar, A.; Islam, M.S.; Laboni, A.A.; Afroze, M.; Khan, M.; Uddin, M.J.; Rahman, M.M. Multi-Modal Neuroprotection of Argemone mexicana L. against Alzheimer’s Disease: In Vitro and in Silico Study. Heliyon 2024, 10, e37178. [Google Scholar] [CrossRef]
  79. El-Hawwary, S.S.; Abd Almaksoud, H.M.; Saber, F.R.; Elimam, H.; Sayed, A.M.; El Raey, M.A.; Abdelmohsen, U.R. Green-Synthesized Zinc Oxide Nanoparticles, Anti-Alzheimer Potential and the Metabolic Profiling of: Sabal Blackburniana Grown in Egypt Supported by Molecular Modelling. RSC Adv. 2021, 11, 18009–18025. [Google Scholar] [CrossRef]
  80. Banu, Z.; Poduri, R.R.; Bhattamisra, S.K. Phytochemical Profiling, in Silico Molecular Docking and ADMET Prediction of Alkaloid Rich Fraction of Elaeocarpus angustifolius Blume Seeds against Alzheimer’s Disease. Nat. Prod. Res. 2025, 39, 1–9. [Google Scholar] [CrossRef]
  81. Musa, M.S.; Islam, M.T.; Billah, W.; Hossain, M.S.; Rahat, M.S.S.; Bayil, I.; Munni, Y.A.; Ganguli, S. Structure-Based Virtual Screening of Trachyspermum Ammi Metabolites Targeting Acetylcholinesterase for Alzheimer’s Disease Treatment. PLoS ONE 2024, 19, e0311401. [Google Scholar] [CrossRef]
  82. de Andrade Medeiros, S.R.; Bezerra, I.C.; Pedroza, L.A.; da Silva, A.J.; Martins, R.M.; Menezes, T.M.; de Melo, A.C.; Neves, J.L.; Gubert, P.; de Melo Filho, A.A. Evaluation of Bauhinia ungulata Essential Oil as a New Acetylcholinesterase Inhibitor from an in silico and in vitro Perspective in the Northern Amazon of Brazil. J. Oleo Sci. 2024, 73, ess23148. [Google Scholar] [CrossRef]
  83. Moussaoui, S.; Mokrani, E.H.; Kabouche, Z.; Guendouze, A.; Laribi, A.; Bradai, N.; Bensouici, C.; Yilmaz, M.A.; Cakir, O.; Tarhan, A. Evaluation of Polyphenolic Profile, Antioxidant, Anti-Cholinesterase, and Anti-Alpha-Amylase Activities of Pistacia lentiscus L. Leaves. Nat. Prod. Res. 2025, 1–14. [Google Scholar] [CrossRef] [PubMed]
  84. Kadi, I.; Seyhan, G.; Zebbiche, Z.; Sari, S.; Barut, B.; Boumoud, T.; Mermer, A.; Boulebd, H. Novel 2-Alkoxy-3-Cyanopyridine Derivatives as Cholinesterase Inhibitors: Synthesis, Biological Evaluation, and In Silico Investigations. Chem. Biodivers. 2025, 39, 1–15. [Google Scholar] [CrossRef] [PubMed]
  85. Asgarshamsi, M.H.; Fassihi, A.; Dehkordi, M.M. Design, Synthesis, Molecular Docking, and Molecular Dynamics Simulation Studies of Novel 3-Hydroxypyridine-4-one Derivatives as Potential Acetylcholinesterase Inhibitors. Chem. Biodivers. 2023, 20, e202300325. [Google Scholar] [CrossRef]
  86. Puopolo, T.; Liu, C.; Ma, H.; Seeram, N.P. Inhibitory Effects of Cannabinoids on Acetylcholinesterase and Butyrylcholinesterase Enzyme Activities. Med. Cannabis Cannabinoids 2022, 5, 85–94. [Google Scholar] [CrossRef]
  87. Negru, D.C.; Bungau, S.G.; Radu, A.; Tit, D.M.; Radu, A.-F.; Nistor-Cseppento, D.C.; Negru, P.A. Evaluation of the Alkaloids as Inhibitors of Human Acetylcholinesterase by Molecular Docking and ADME Prediction. In Vivo 2025, 39, 236–250. [Google Scholar] [CrossRef]
  88. Nair, A.C.; Benny, S.; Aneesh, T.P.; Sudheesh, M.S.; Lakshmi, P.K. Comprehensive Profiling of Traditional Herbomineral Formulation Manasamitra Vatakam in Rat Brain Following Oral Administration and In-Silico Screening of the Identified Compound for Anti-Alzheimer’s Activity. J. Ethnopharmacol. 2025, 338, 119024. [Google Scholar] [CrossRef]
  89. Singh, M.; Jindal, D.; Kumar, R.; Pancham, P.; Haider, S.; Gupta, V.; Mani, S.; R, R.; Tiwari, R.K.; Chanda, S. Molecular Docking and Network Pharmacology Interaction Analysis of Gingko Biloba (EGB761) Extract with Dual Target Inhibitory Mechanism in Alzheimer’s Disease. J. Alzheimer’s Dis. 2023, 93, 705–726. [Google Scholar] [CrossRef]
  90. Refaay, D.A.; Abdel-Hamid, M.I.; Alyamani, A.A.; Abdel Mougib, M.; Ahmed, D.M.; Negm, A.; Mowafy, A.M.; Ibrahim, A.A.; Mahmoud, R.M. Growth Optimization and Secondary Metabolites Evaluation of Anabaena Variabilis for Acetylcholinesterase Inhibition Activity. Plants 2022, 11, 735. [Google Scholar] [CrossRef]
  91. Farihi, A.; Bouhrim, M.; Chigr, F.; Elbouzidi, A.; Bencheikh, N.; Zrouri, H.; Nasr, F.A.; Parvez, M.K.; Alahdab, A.; Ahami, A.O.T. Exploring Medicinal Herbs’ Therapeutic Potential and Molecular Docking Analysis for Compounds as Potential Inhibitors of Human Acetylcholinesterase in Alzheimer’s Disease Treatment. Medicina 2023, 59, 1812. [Google Scholar] [CrossRef]
  92. Reshetnikov, D.V.; Ivanov, I.D.; Baev, D.S.; Rybalova, T.V.; Mozhaitsev, E.S.; Patrushev, S.S.; Vavilin, V.A.; Tolstikova, T.G.; Shults, E.E. Design, Synthesis and Assay of Novel Methylxanthine–Alkynylmethylamine Derivatives as Acetylcholinesterase Inhibitors. Molecules 2022, 27, 8787. [Google Scholar] [CrossRef] [PubMed]
  93. Grodner, B.; Napiórkowska, M.; Pisklak, D.M. In Vitro and in Silico Kinetic Studies of Patented 1,7-diethyl and 1,7-dimethyl Aminoalkanol Derivatives as New Inhibitors of Acetylcholinesterase. Int. J. Mol. Sci. 2022, 23, 270. [Google Scholar] [CrossRef] [PubMed]
  94. Drozdowska, D.; Maliszewski, D.; Wróbel, A.; Ratkiewicz, A.; Sienkiewicz, M. New Benzamides as Multi-Targeted Compounds: A Study on Synthesis, AChE and BACE1 Inhibitory Activity and Molecular Docking. Int. J. Mol. Sci. 2023, 24, 14901. [Google Scholar] [CrossRef] [PubMed]
  95. Cheung, J.; Rudolph, M.J.; Burshteyn, F.; Cassidy, M.S.; Gary, E.N.; Love, J.; Franklin, M.C.; Height, J.J. Structures of Human Acetylcholinesterase in Complex with Pharmacologically Important Ligands. J. Med. Chem. 2012, 55, 10282–10286. [Google Scholar] [CrossRef]
  96. El Khatabi, K.; Aanouz, I.; El-Mernissi, R.; Singh, A.K.; Ajana, M.A.; Lakhlifi, T.; Kumar, S.; Bouachrine, M. Integrated 3D-QSAR, Molecular Docking, and Molecular Dynamics Simulation Studies on 1,2,3-Triazole Based Derivatives for Designing New Acetylcholinesterase Inhibitors. Turk. J. Chem. 2021, 45, 647–660. [Google Scholar] [CrossRef]
  97. Bilginer, S.; Anil, B.; Koca, M.; Demir, Y.; Gülçin, I. Novel Mannich Bases with Strong Carbonic Anhydrases and Acetylcholinesterase Inhibition Effects: 3-(Aminomethyl)-6-{3-[4-(Trifluoromethyl)Phenyl]Acryloyl}-2(3H)-Benzoxazolones. Turk. J. Chem. 2021, 45, 805–818. [Google Scholar] [CrossRef]
  98. Suwanhom, P.; Saetang, J.; Khongkow, P.; Nualnoi, T.; Tipmanee, V.; Lomlim, L. Synthesis, Biological Evaluation, and in Silico Studies of New Acetylcholinesterase Inhibitors Based on Quinoxaline Scaffold. Molecules 2021, 26, 4895. [Google Scholar] [CrossRef]
  99. Zarei, S.; Shafiei, M.; Firouzi, M.; Firoozpour, L.; Divsalar, K.; Asadipour, A.; Akbarzadeh, T.; Foroumadi, A. Design, Synthesis and Biological Assessment of New 1-Benzyl-4-((4-Oxoquinazolin-3(4H)-Yl)Methyl) Pyridin-1-Ium Derivatives (BOPs) as Potential Dual Inhibitors of Acetylcholinesterase and Butyrylcholinesterase. Heliyon 2021, 7, e06683. [Google Scholar] [CrossRef]
  100. Chowdhury, S.; Rahman, A.; Hussain, F.; Rahman, S.M.A. Synthesis, characterization and in vitro, in vivo, in silico biological evaluations of substituted benzimidazole derivatives. Saudi J. Biol. Sci. 2022, 29, 239–250. [Google Scholar] [CrossRef]
  101. Ramos, A.S.F.; Techert, S. Influence of the Water Structure on the Acetylcholinesterase Efficiency. Biophys. J. 2005, 89, 1990–2003. [Google Scholar] [CrossRef]
  102. Bondžić, A.M.; Lazarević-Pašti, T.D.; Leskovac, A.R.; Petrović, S.Ž.; Čolović, M.B.; Parac-Vogt, T.N.; Janjić, G.V. A New Acetylcholinesterase Allosteric Site Responsible for Binding Voluminous Negatively Charged Molecules—The Role in the Mechanism of AChE Inhibition. Eur. J. Pharm. Sci. 2020, 151, 105376. [Google Scholar] [CrossRef] [PubMed]
  103. Hopkins, T.J.; Rupprecht, L.E.; Hayes, M.R.; Blendy, J.A.; Schmidt, H.D. Galantamine, an Acetylcholinesterase Inhibitor and Positive Allosteric Modulator of Nicotinic Acetylcholine Receptors, Attenuates Nicotine Taking and Seeking in Rats. Neuropsychopharmacology 2012, 37, 2310–2321. [Google Scholar] [CrossRef]
  104. Ekins, S.; Mestres, J.; Testa, B. In Silico Pharmacology for Drug Discovery: Methods for Virtual Ligand Screening and Profiling. Br. J. Pharmacol. 2007, 152, 9–20. [Google Scholar] [CrossRef] [PubMed]
  105. Zhao, J.; Cao, Y.; Zhang, L. Exploring the Computational Methods for Protein-Ligand Binding Site Prediction. Comput. Struct. Biotechnol. J. 2020, 18, 417–426. [Google Scholar] [CrossRef] [PubMed]
  106. Dutkiewicz, Z. Computational Methods for Calculation of Protein-Ligand Binding Affinities in Structure-Based Drug Design. Phys. Sci. Rev. 2022, 7, 933–968. [Google Scholar] [CrossRef]
  107. Agu, P.C.; Afiukwa, C.A.; Orji, O.U.; Ezeh, E.M.; Ofoke, I.H.; Ogbu, C.O.; Ugwuja, E.I.; Aja, P.M. Molecular Docking as a Tool for the Discovery of Molecular Targets of Nutraceuticals in Diseases Management. Sci. Rep. 2023, 13, 13398. [Google Scholar] [CrossRef]
  108. Khare, N.; Maheshwari, S.K.; Jha, A.K. Screening and Identification of Secondary Metabolites in the Bark of Bauhinia variegata to Treat Alzheimer’s Disease by Using Molecular Docking and Molecular Dynamics Simulations. J. Biomol. Struct. Dyn. 2021, 39, 5988–5998. [Google Scholar] [CrossRef]
  109. Yi, P.; Zhang, Z.; Huang, S.; Huang, J.; Peng, W.; Yang, J. Integrated Meta-Analysis, Network Pharmacology, and Molecular Docking to Investigate the Efficacy and Potential Pharmacological Mechanism of Kai-Xin-San on Alzheimer’s Disease. Pharm. Biol. 2020, 58, 932–943. [Google Scholar] [CrossRef]
  110. Shen, Y.; Zhang, B.; Pang, X.; Yang, R.; Chen, M.; Zhao, J.; Wang, J.; Wang, Z.; Yu, Z.; Wang, Y.; et al. Network Pharmacology-Based Analysis of Xiao-Xu-Ming Decoction on the Treatment of Alzheimer’s Disease. Front. Pharmacol. 2020, 11, 595254. [Google Scholar] [CrossRef]
  111. Hussein, R.A.; Afifi, A.H.; Soliman, A.A.F.; El Shahid, Z.A.; Zoheir, K.M.A.; Mahmoud, K.M. Neuroprotective Activity of Ulmus pumila L. in Alzheimer’s Disease in Rats; Role of Neurotrophic Factors. Heliyon 2020, 6, e05678. [Google Scholar] [CrossRef]
  112. Kareti, S.R.; Pharm, S.M. In Silico Molecular Docking Analysis of Potential Anti-Alzheimer’s Compounds Present in Chloroform Extract of Carissa Carandas Leaf Using Gas Chromatography MS/MS. Curr. Ther. Res. Clin. Exp. 2020, 93, 100615. [Google Scholar] [CrossRef] [PubMed]
  113. Kong, X.P.; Ren, H.Q.; Liu, E.Y.L.; Leung, K.W.; Guo, S.C.; Duan, R.; Dong, T.T.X.; Tsim, K.W.K. The Cholinesterase Inhibitory Properties of Stephaniae Tetrandrae Radix. Molecules 2020, 25, 5914. [Google Scholar] [CrossRef]
  114. Heo, J.H.; Eom, B.H.; Ryu, H.W.; Kang, M.G.; Park, J.E.; Kim, D.Y.; Kim, J.H.; Park, D.; Oh, S.R.; Kim, H. Acetylcholinesterase and Butyrylcholinesterase Inhibitory Activities of Khellactone Coumarin Derivatives Isolated from Peucedanum Japonicum Thurnberg. Sci. Rep. 2020, 10, 21695. [Google Scholar] [CrossRef]
  115. Azevedo, R.D.S.; Falcão, K.V.G.; Assis, C.R.D.; Martins, R.M.G.; Araújo, M.C.; Yogui, G.T.; Neves, J.L.; Seabra, G.M.; Maia, M.B.S.; Amaral, I.P.G.; et al. Effects of Pyriproxyfen on Zebrafish Brain Mitochondria and Acetylcholinesterase. Chemosphere 2021, 263, 128029. [Google Scholar] [CrossRef] [PubMed]
  116. Ogata, N.; Tagishi, H.; Tsuji, M. Inhibition of Acetylcholinesterase by Wood Creosote and Simple Phenolic Compounds. Chem. Pharm. Bull. 2020, 68, 1193–1200. [Google Scholar] [CrossRef]
  117. Kocakaya, S.O.; Ertas, A.; Yener, I.; Ercan, B.; Oral, E.V.; Akdeniz, M.; Kaplaner, E.; Topcu, G.; Kolak, U. Selective In-Vitro Enzymes’ Inhibitory Activities of Fingerprints Compounds of Salvia Species and Molecular Docking Simulations. Iran. J. Pharm. Res. 2020, 19, 187–198. [Google Scholar] [CrossRef]
  118. Karasova, J.Z.; Mzik, M.; Kucera, T.; Vecera, Z.; Kassa, J.; Sestak, V. Interaction of Cucurbit [7]Uril with Oxime K027, Atropine, and Paraoxon: Risky or Advantageous Delivery System? Int. J. Mol. Sci. 2020, 21, 7883. [Google Scholar] [CrossRef] [PubMed]
  119. Adalat, B.; Rahim, F.; Taha, M.; Alshamrani, F.J.; Anouar, E.H.; Uddin, N.; Shah, S.A.A.; Ali, Z.; Zakaria, Z.A. Synthesis of Benzimidazole–Based Analogs as Anti Alzheimer’s Disease Compounds and Their Molecular Docking Studies. Molecules 2020, 25, 4828. [Google Scholar] [CrossRef]
  120. Pitchai, A.; Rajaretinam, R.K.; Mani, R.; Nagarajan, N. Molecular Interaction of Human Acetylcholinesterase with Trans-Tephrostachin and Derivatives for Alzheimer’s Disease. Heliyon 2020, 6, e04930. [Google Scholar] [CrossRef]
  121. Tran, T.S.; Tran, T.D.; Tran, T.H.; Mai, T.T.; Nguyen, N.L.; Thai, K.M.; Le, M.T. Synthesis, in Silico and in Vitro Evaluation of Some Flavone Derivatives for Acetylcholinesterase and BACE-1 Inhibitory Activity. Molecules 2020, 25, 4064. [Google Scholar] [CrossRef]
  122. Jusril, N.A.; Juhari, A.N.N.M.; Bakar, S.I.A.; Saad, W.M.M.; Adenan, M.I. Combining in Silico and in Vitro Studies to Evaluate the Acetylcholinesterase Inhibitory Profile of Different Accessions and the Biomarker Triterpenes of Centella Asiatica. Molecules 2020, 25, 3353. [Google Scholar] [CrossRef] [PubMed]
  123. Kausar, N.; Murtaza, S.; Arshad, M.N.; Zaib Saleem, R.S.; Asiri, A.M.; Kausar, S.; Altaf, A.A.; Tatheer, A.; Elnaggar, A.Y.; El-Bahy, S.M. Design, Synthesis, Crystal Structure, in Vitro Cytotoxicity Evaluation, Density Functional Theory Calculations and Docking Studies of 2-(Benzamido) Benzohydrazide Derivatives as Potent AChE and BChE Inhibitors. RSC Adv. 2021, 12, 154–167. [Google Scholar] [CrossRef]
  124. Krátký, M.; Štěpánková, Š.; Konečná, K.; Svrčková, K.; Maixnerová, J.; Švarcová, M.; Jand’ourek, O.; Trejtnar, F.; Vinšová, J. Novel Aminoguanidine Hydrazone Analogues: From Potential Antimicrobial Agents to Potent Cholinesterase Inhibitors. Pharmaceuticals 2021, 14, 1229. [Google Scholar] [CrossRef]
  125. Iqbal, D.; Khan, M.S.; Waiz, M.; Rehman, M.T.; Alaidarous, M.; Jamal, A.; Alothaim, A.S.; AlAjmi, M.F.; Alshehri, B.M.; Banawas, S.; et al. Exploring the Binding Pattern of Geraniol with Acetylcholinesterase through In Silico Docking, Molecular Dynamics Simulation, and In Vitro Enzyme Inhibition Kinetics Studies. Cells 2021, 10, 3533. [Google Scholar] [CrossRef] [PubMed]
  126. Mostafa, N.M.; Mostafa, A.M.; Ashour, M.L.; Elhady, S.S. Neuroprotective Effects of Black Pepper Cold-Pressed Oil on Scopolamine-Induced Oxidative Stress and Memory Impairment in Rats. Antioxidants 2021, 10, 1993. [Google Scholar] [CrossRef] [PubMed]
  127. Onikanni, A.S.; Lawal, B.; Olusola, A.O.; Olugbodi, J.O.; Sani, S.; Ajiboye, B.O.; Ilesanmi, O.B.; Alqarni, M.; Mostafahedeab, G.; Obaidullah, A.J.; et al. Sterculia Tragacantha Lindl Leaf Extract Ameliorates STZ-Induced Diabetes, Oxidative Stress, Inflammation and Neuronal Impairment. J. Inflamm. Res. 2021, 14, 6749–6764. [Google Scholar] [CrossRef] [PubMed]
  128. Zhu, Q.; Lin, M.; Zhuo, W.; Li, Y. Chemical Constituents from the Wild Atractylodes Macrocephala Koidz and Acetylcholinesterase Inhibitory Activity Evaluation as Well as Molecular Docking Study. Molecules 2021, 26, 7299. [Google Scholar] [CrossRef]
  129. Johnson, T.O.; Ojo, O.A.; Ikiriko, S.; Ogunkua, J.; Akinyemi, G.O.; Rotimi, D.E.; Oche, J.R.; Adegboyega, A.E. Biochemical Evaluation and Molecular Docking Assessment of Cymbopogon Citratus as a Natural Source of Acetylcholine Esterase (AChE)- Targeting Insecticides. Biochem. Biophys. Rep. 2021, 28, 101175. [Google Scholar] [CrossRef]
  130. Upadhyay, S.P.; Singh, V.; Sharma, R.; Zhou, J.; Thapa, P.; Johnson, D.K.; Keightley, A.; Chen, M.; Suo, W.; Sharma, M. Influence of Ligand Geometry on Cholinesterase Enzyme—A Comparison of 1-Isoindolinone Based Structural Analog with Donepezil. J. Mol. Struct. 2022, 1247, 131385. [Google Scholar] [CrossRef]
  131. Kiziltas, H.; Goren, A.C.; Alwasel, S.H.; Gulcin, İ. Sahlep (Dactylorhiza Osmanica): Phytochemical Analyses by LC-HRMS, Molecular Docking, Antioxidant Activity, and Enzyme Inhibition Profiles. Molecules 2022, 27, 6907. [Google Scholar] [CrossRef]
  132. Lolak, N.; Akocak, S.; Durgun, M.; Duran, H.E.; Necip, A.; Türkeş, C.; Işık, M.; Beydemir, Ş. Novel Bis-Ureido-Substituted Sulfaguanidines and Sulfisoxazoles as Carbonic Anhydrase and Acetylcholinesterase Inhibitors. Mol. Divers. 2023, 27, 1735–1749. [Google Scholar] [CrossRef] [PubMed]
  133. Hussain, R.; Rahim, F.; Ullah, H.; Khan, S.; Sarfraz, M.; Iqbal, R.; Suleman, F.; Al-Sadoon, M.K. Design, Synthesis, In Vitro Biological Evaluation and In Silico Molecular Docking Study of Benzimidazole-Based Oxazole Analogues: A Promising Acetylcholinesterase and Butyrylcholinesterase Inhibitors. Molecules 2023, 28, 7015. [Google Scholar] [CrossRef]
  134. Jaśkiewicz, A.; Budryn, G.; Carmena-Bargueño, M.; Pérez-Sánchez, H. Evaluation of Activity of Sesquiterpene Lactones and Chicory Extracts as Acetylcholinesterase Inhibitors Assayed in Calorimetric and Docking Simulation Studies. Nutrients 2022, 14, 3633. [Google Scholar] [CrossRef] [PubMed]
  135. Atanasova, M.; Dimitrov, I.; Ivanov, S.; Georgiev, B.; Berkov, S.; Zheleva-Dimitrova, D.; Doytchinova, I. Virtual Screening and Hit Selection of Natural Compounds as Acetylcholinesterase Inhibitors. Molecules 2022, 27, 3139. [Google Scholar] [CrossRef] [PubMed]
  136. Zeng, F.; Lu, T.; Wang, J.; Nie, X.; Xiong, W.; Yin, Z.; Peng, D. Design, Synthesis and Bioactivity Evaluation of Coumarin–BMT Hybrids as New Acetylcholinesterase Inhibitors. Molecules 2022, 27, 2142. [Google Scholar] [CrossRef]
  137. Xiao, Y.; Liang, W.; Liu, D.; Zhang, Z.; Chang, J.; Zhu, D. Isolation and Acetylcholinesterase Inhibitory Activity of Asterric Acid Derivatives Produced by Talaromyces Aurantiacus FL15, an Endophytic Fungus from Huperzia Serrata. 3 Biotech 2022, 12, 60. [Google Scholar] [CrossRef]
  138. Rahim, F.; Ullah, H.; Taha, M.; Hussain, R.; Sarfraz, M.; Iqbal, R.; Iqbal, N.; Khan, S.; Ali Shah, S.A.; Albalawi, M.A.; et al. Synthesis of New Triazole-Based Thiosemicarbazone Derivatives as Anti-Alzheimer’s Disease Candidates: Evidence-Based In Vitro Study. Molecules 2022, 28, 21. [Google Scholar] [CrossRef]
  139. Liao, Y.; Hu, X.; Pan, J.; Zhang, G. Inhibitory Mechanism of Baicalein on Acetylcholinesterase: Inhibitory Interaction, Conformational Change, and Computational Simulation. Foods 2022, 11, 168. [Google Scholar] [CrossRef]
  140. Tarabasz, D.; Szczeblewski, P.; Laskowski, T.; Płaziński, W.; Baranowska-Wójcik, E.; Szwajgier, D.; Kukula-Koch, W.; Meissner, H.O. The Distribution of Glucosinolates in Different Phenotypes of Lepidium Peruvianum and Their Role as Acetyl- and Butyrylcholinesterase Inhibitors—In Silico and In Vitro Studies. Int. J. Mol. Sci. 2022, 23, 4858. [Google Scholar] [CrossRef]
  141. da Silva Mesquita, R.; Kyrylchuk, A.; Cherednichenko, A.; Costa Sá, I.S.; Macedo Bastos, L.; Moura Araújo da Silva, F.; Saraiva Nunomura, R.d.C.; Grafov, A. In Vitro and In Silico Evaluation of Cholinesterase Inhibition by Alkaloids Obtained from Branches of Abuta Panurensis Eichler. Molecules 2022, 27, 3138. [Google Scholar] [CrossRef]
  142. Wu, T.; Hou, W.; Liu, C.; Li, S.; Zhang, Y. Efficient Combination of Complex Chromatography, Molecular Docking and Enzyme Kinetics for Exploration of Acetylcholinesterase Inhibitors from Poria Cocos. Molecules 2023, 28, 1228. [Google Scholar] [CrossRef] [PubMed]
  143. Eltahawy, N.A.; Ali, A.I.; Ibrahim, S.A.; Nafie, M.S.; Sindi, A.M.; Alkharobi, H.; Almalki, A.J.; Badr, J.M.; Elhady, S.S.; Abdelhameed, R.F.A. Analysis of Marrubiin in Marrubium alysson L. Extract Using Advanced HPTLC: Chemical Profiling, Acetylcholinesterase Inhibitory Activity, and Molecular Docking. Metabolites 2023, 14, 27. [Google Scholar] [CrossRef] [PubMed]
  144. Jamal, Q.M.S.; Khan, M.I.; Alharbi, A.H.; Ahmad, V.; Yadav, B.S. Identification of Natural Compounds of the Apple as Inhibitors against Cholinesterase for the Treatment of Alzheimer’s Disease: An In Silico Molecular Docking Simulation and ADMET Study. Nutrients 2023, 15, 1579. [Google Scholar] [CrossRef]
  145. Pishgouii, F.; Lotfi, S.; Sedaghati, E. Anti-AChE and Anti-BuChE Screening of the Fermentation Broth Extracts from Twelve Aspergillus Isolates and GC-MS and Molecular Docking Studies of the Most Active Extracts. Appl. Biochem. Biotechnol. 2023, 195, 5199–5216. [Google Scholar] [CrossRef]
  146. Belaiba, M.; Aldulaijan, S.; Messaoudi, S.; Abedrabba, M.; Dhouib, A.; Bouajila, J. Evaluation of Biological Activities of Twenty Flavones and In Silico Docking Study. Molecules 2023, 28, 2419. [Google Scholar] [CrossRef]
  147. Laghchioua, F.E.; da Silva, C.F.M.; Pinto, D.C.G.A.; Cavaleiro, J.A.S.; Mendes, R.F.; Paz, F.A.A.; Faustino, M.A.F.; Rakib, E.M.; Neves, M.G.P.M.S.; Pereira, F.; et al. Design of Promising Thiazoloindazole-Based Acetylcholinesterase Inhibitors Guided by Molecular Docking and Experimental Insights. ACS Chem. Neurosci. 2024, 15, 2853–2869. [Google Scholar] [CrossRef]
  148. Raturi, A.; Yadav, V.; Hoda, N.; Subbarao, N.; Chaudhry, S.A. In Silico Identification of Colchicine Derivatives as Novel and Potential Inhibitors Based on Molecular Docking and Dynamic Simulations Targeting Multifactorial Drug Targets Involved in Alzheimer’s Disease. J. Biomol. Struct. Dyn. 2024, 42, 11555–11573. [Google Scholar] [CrossRef] [PubMed]
  149. Faloye, K.O.; Mahmud, S.; Fakola, E.G.; Oyetunde, Y.M.; Fajobi, S.J.; Ugwo, J.P.; Olusola, A.J.; Famuyiwa, S.O.; Olajubutu, O.G.; Oguntade, T.I.; et al. Revealing the Acetylcholinesterase Inhibitory Potential of Phyllanthus amarus and Its Phytoconstituents: In Vitro and in Silico Approach. Bioinform. Biol. Insights 2022, 16, 1–11. [Google Scholar] [CrossRef]
  150. Kurbanova, M.; Maharramov, A.; Safarova, A.; Ahmad, S.; El Bakri, Y. Molecular Docking Study and Molecular Dynamics Simulation of Ethyl 3,5-diphenyl-1 H. -pyrrole-2-carboxylate and (Z)-ethyl-2-(3-oxo-1,3-diphenylprop-1-enylamino)Acetate. J. Biochem. Mol. Toxicol. 2022, 36, e23013. [Google Scholar] [CrossRef]
  151. Gholami, A.; Minai-Tehrani, D.; Eriksson, L.A. In Silico and in Vitro Studies Confirm Ondansetron as a Novel Acetylcholinesterase and Butyrylcholinesterase Inhibitor. Sci. Rep. 2023, 13, 643. [Google Scholar] [CrossRef]
  152. de Almeida, R.B.M.; Barbosa, D.B.; do Bomfim, M.R.; Amparo, J.A.O.; Andrade, B.S.; Costa, S.L.; Campos, J.M.; Cruz, J.N.; Santos, C.B.R.; Leite, F.H.A.; et al. Identification of a Novel Dual Inhibitor of Acetylcholinesterase and Butyrylcholinesterase: In Vitro and In Silico Studies. Pharmaceuticals 2023, 16, 95. [Google Scholar] [CrossRef] [PubMed]
  153. Mendes, G.O.; Pita, S.S.d.R.; Carvalho, P.B.d.; Silva, M.P.d.; Taranto, A.G.; Leite, F.H.A. Molecular Multi-Target Approach for Human Acetylcholinesterase, Butyrylcholinesterase and β-Secretase 1: Next Generation for Alzheimer’s Disease Treatment. Pharmaceuticals 2023, 16, 880. [Google Scholar] [CrossRef] [PubMed]
  154. Arumugam, N.; Darshan, V.M.D.; Venketesh, V.; Pradhan, S.S.; Garg, A.; Sivaramakrishnan, V.; Kanchi, S.; Mahalingam, S.M. Synthesis, Computational Docking and Molecular Dynamics Studies of a New Class of Spiroquinoxalinopyrrolidine Embedded Chromanone Hybrids as Potent Anti-Cholinesterase Agents. RSC Adv. 2024, 14, 18815–18831. [Google Scholar] [CrossRef]
  155. Saha, B.; Das, A.; Jangid, K.; Kumar, A.; Kumar, V.; Jaitak, V. Identification of Coumarin Derivatives Targeting Acetylcholinesterase for Alzheimer’s Disease by Field-Based 3D-QSAR, Pharmacophore Model-Based Virtual Screening, Molecular Docking, MM/GBSA, ADME and MD Simulation Study. Curr. Res. Struct. Biol. 2024, 7, 100124. [Google Scholar] [CrossRef]
  156. Keçeci Sarıkaya, M.; Yıldırım, Ş.; Kocyigit, U.M.; Ceylan, M.; Yırtıcı, Ü.; Eyüpoğlu, V. Novel Aminothiazole–Chalcone Analogs: Synthesis, Evaluation Acetylcholinesterase Activity, In Silico Analysis. Chem. Biodivers. 2025, e202402777. [Google Scholar] [CrossRef]
  157. Rawat, K.; Tewari, D.; Bisht, A.; Chandra, S.; Tiruneh, Y.K.; Hassan, H.M.; Al-Emam, A.; Sindi, E.R.; Al-Dies, A.-A.M. Identification of AChE Targeted Therapeutic Compounds for Alzheimer’s Disease: An in-Silico Study with DFT Integration. Sci. Rep. 2024, 14, 30356. [Google Scholar] [CrossRef]
  158. Emam, M.; El-Newary, S.A.; Aati, H.Y.; Wei, B.; Seif, M.; Ibrahim, A.Y. Anti-Alzheimer’s Potency of Rich Phenylethanoid Glycosides Extract from Marrubium vulgare L.: In Vitro and In Silico Studies. Pharmaceuticals 2024, 17, 1282. [Google Scholar] [CrossRef]
  159. Al-Maqtari, H.M.; Hasan, A.H.; Suleiman, M.; Ahmad Zahidi, M.A.; Noamaan, M.A.; Alexyuk, P.; Alexyuk, M.; Bogoyavlenskiy, A.; Jamalis, J. Benzyloxychalcone Hybrids as Prospective Acetylcholinesterase Inhibitors against Alzheimer’s Disease: Rational Design, Synthesis, In Silico ADMET Prediction, QSAR, Molecular Docking, DFT, and Molecular Dynamic Simulation Studies. ACS Omega 2024, 9, 32901–32919. [Google Scholar] [CrossRef]
  160. Szeleszczuk, Ł.; Pisklak, D.M.; Grodner, B. Thiamine and Thiamine Pyrophosphate as Non-Competitive Inhibitors of Acetylcholinesterase—Experimental and Theoretical Investigations. Molecules 2025, 30, 412. [Google Scholar] [CrossRef]
  161. Zhu, J.; Xu, Z.; Liu, X. Chemical Composition, Antioxidant Activities, and Enzyme Inhibitory Effects of Lespedeza bicolour Turcz. Essential Oil. J. Enzym. Inhib. Med. Chem. 2025, 40, 2460053. [Google Scholar] [CrossRef]
  162. Khedraoui, M.; Abchir, O.; Nour, H.; Yamari, I.; Errougui, A.; Samadi, A.; Chtita, S. An In Silico Study Based on QSAR and Molecular Docking and Molecular Dynamics Simulation for the Discovery of Novel Potent Inhibitor against AChE. Pharmaceuticals 2024, 17, 830. [Google Scholar] [CrossRef] [PubMed]
  163. Tabbiche, A.; Bouchama, A.; Fadli, K.; Ahmad, B.; Kumar, N.; Chiter, C.; Yahiaoui, M.; Zaidi, F.; Boudjemaa, K.; Dege, N.; et al. Development of New Benzil-Hydrazone Derivatives as Anticholinesterase Inhibitors: Synthesis, X-Ray Analysis, DFT Study and in Vitro/in Silico Evaluation. J. Biomol. Struct. Dyn. 2025, 43, 2518–2533. [Google Scholar] [CrossRef] [PubMed]
  164. Żołek, T.; Purgatorio, R.; Kłopotowski, Ł.; Catto, M.; Ostrowska, K. Coumarin Derivative Hybrids: Novel Dual Inhibitors Targeting Acetylcholinesterase and Monoamine Oxidases for Alzheimer’s Therapy. Int. J. Mol. Sci. 2024, 25, 12803. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Representation of acetylcholinesterase and its active site in different visualizations. (A) Upper view of the pocket in AChE PDB ID: 4EY7 monomer A (in green surface) and Donepezil (in blue sticks). (B) Side view slice of the pocket 15 Å deep in AChE PDB ID: 4EY7 monomer A (in green surface) and Donepezil (in blue sticks). (C) Side view rotated 90 degrees. Three main amino acids in contact with Donepezil are shown: D72, Y121, and W286; Donepezil is represented by blue sticks and the AChE protein by green transparent surface to better observe the pocket. (D) Catalytic triad at the bottom of the gorge composed of H447, E202, and S203; Y337 and F338 are observed in the middle of the gorge.
Figure 1. Representation of acetylcholinesterase and its active site in different visualizations. (A) Upper view of the pocket in AChE PDB ID: 4EY7 monomer A (in green surface) and Donepezil (in blue sticks). (B) Side view slice of the pocket 15 Å deep in AChE PDB ID: 4EY7 monomer A (in green surface) and Donepezil (in blue sticks). (C) Side view rotated 90 degrees. Three main amino acids in contact with Donepezil are shown: D72, Y121, and W286; Donepezil is represented by blue sticks and the AChE protein by green transparent surface to better observe the pocket. (D) Catalytic triad at the bottom of the gorge composed of H447, E202, and S203; Y337 and F338 are observed in the middle of the gorge.
Ijms 26 03781 g001
Figure 2. Annual publication trends in molecular docking studies related to AChE (2020–2025). The bar graph illustrates a steady increase in research output, reflecting growing interest in computational approaches for AChE inhibition and drug discovery. Data source from PubMed consulted up to March 2025.
Figure 2. Annual publication trends in molecular docking studies related to AChE (2020–2025). The bar graph illustrates a steady increase in research output, reflecting growing interest in computational approaches for AChE inhibition and drug discovery. Data source from PubMed consulted up to March 2025.
Ijms 26 03781 g002
Figure 3. Acetylcholinesterase (AchE) as a target in docking studies. Systematic analysis of 121 Molecular Docking Studies Targeting Acetylcholinesterase (AChE) in Alzheimer’s disease therapeutics. The color-coded bar chart (green to red gradient) illustrates software usage frequency, revealing that AutoDock Vina and GROMACS emerged as the most used tools.
Figure 3. Acetylcholinesterase (AchE) as a target in docking studies. Systematic analysis of 121 Molecular Docking Studies Targeting Acetylcholinesterase (AChE) in Alzheimer’s disease therapeutics. The color-coded bar chart (green to red gradient) illustrates software usage frequency, revealing that AutoDock Vina and GROMACS emerged as the most used tools.
Ijms 26 03781 g003
Table 1. Compilation of results from molecular docking assays, grouped by software used.
Table 1. Compilation of results from molecular docking assays, grouped by software used.
SoftwareNo. Refs.Binding Affinity [Kcal/mol]
MeanMinMaxSTDV
Vina33−9.76−19.3−3.7−2.45
AutoDock420−7.68−12.7581.6111.47
Schrödinger Suite17−13.40−114.60.818.10
MOE14−8.25−14.7−4.42.36
Glide8−9.92−15.2−4.72.44
Biovia Discovery Studio6−13.04−32.2−4.68.40
CDOCKER4−3.51−46.384.545.53
GOLD314.27−65.860.154.95
DSHC2−28.08−54.5−16.610.90
FlexX3−24.63−36.5−11.84.52
Smina2−10.92−15.0−8.61.70
Achilles Docking Server1−7.79−8.5−6.90.68
ArgusLab 4.01−9.72−12.7−8.90.98
ClusPro y Pymol1−883.10−974.0−792.2128.55
DOCK1−30.71−52.4−14.012.42
ICM Pro Molsoft1−15.79−18.4−12.71.68
iGEMDOCK1−87.49−92.4−81.13.73
PyVSvina1−10.60−10.60−10.600.0
Surflex-Dock1−8.96−114.618.112.847
Vina + Umbrella Sampling simulation1−37.14−53.3−18.514.53
Table 2. Docking studies conducted with the ligand Donepezil in different research works.
Table 2. Docking studies conducted with the ligand Donepezil in different research works.
DonepezilGalantamine
PDBAffinity Energy
(kcal/mol)
SoftwareCitePDBAffinity Energy
(kcal/mol)
SoftwareCite
1ACL−6.3226MOE[52]1W6R−9.63AutoDock4[53]
1C2B−11.7Vina[54]−8.68[55]
1EVE−12.74ArgusLab 4.0[56]1DX6−28.53Vina[57]
−9.81Glide[58]4EY5−7.7Vina[59]
−9.811C2B−7.9Blind docking[60]
−6.49Schrödinger suite[61]4EY6−7.91AutoDock4[62]
1OCE−8.04MOE[63]−9.9[64]
4BDT−8.5[65]−11.54Glide[66]
4EY5−10.5Vina[59]59.74GOLD[67]
−12.42AutoDock4[68]−9.28MOE[69]
4EY7−10.8Biovia Discovery studio[70]−7.07[71]
4EY7−31.26CDOCKER[72]−14.2Smina[73]
−5.552Glide[74]−9.61Vina[15]
−17.7ICM Pro Molsoft[75]−9.1[76]
−15.5MOE[77]4EY7−9.268Glide[74]
−10.171Schrodinger suite[78]−9Vina[79]
−18.909[80]−8.9[81]
−8.7Vina[79]−10.5[82]
−11.94[15]4M0E−21.2FlexX[83]
−11.8[84]−7Glide[84]
−10.5[85]5HFA−8.2Biovia Discovery Studio[86]
−11.7[87]6O4W−10.4Vina[88]
−18.1[73]6O4X−8.02Glide[89]
4M0E−45.18CDOCKER[90]NA
−8.271Schrödinger suite[81]
4PQE−8.6Biovia Discovery Studio[91]
6O4W−14.817Glide[92]
6O4X−11.1Vina[93]
73EH−11.6[94]
Table 3. Compilation of acetylcholinesterase crystals used in molecular docking studies.
Table 3. Compilation of acetylcholinesterase crystals used in molecular docking studies.
No. ReferencesPDB ID
314EY7
144M0E
134EY6
81EVE
64EY5
44PQE, 6O4W
31C2B, 4BDT, 6O4X, 7D9P
21ACJ, 1DX6, 1OCE, 1W6R, 3LII, 4EY4, 6H12, 7D90, 7D9Q, 7XN1
11C2O, 1EA5, 1EEA, 1F8U, 1GQS, 1H23, 1O86, 1QON, 2ACK, 3I6M, 3I6Z, 5FPQ, 5FUM, 5HF5, 5HFA, 6CQV, 6CQZ, 6EUC, 6EYF, 6NTL, 6NTO, 6O50, 6O69, 6U37, 6WO4, 6WUZ, 6WVO, 6WVQ, 6XYU, 73EH, 7E3H
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Reynoso-García, M.F.; Nicolás-Álvarez, D.E.; Tenorio-Barajas, A.Y.; Reyes-Chaparro, A. Structural Bioinformatics Applied to Acetylcholinesterase Enzyme Inhibition. Int. J. Mol. Sci. 2025, 26, 3781. https://doi.org/10.3390/ijms26083781

AMA Style

Reynoso-García MF, Nicolás-Álvarez DE, Tenorio-Barajas AY, Reyes-Chaparro A. Structural Bioinformatics Applied to Acetylcholinesterase Enzyme Inhibition. International Journal of Molecular Sciences. 2025; 26(8):3781. https://doi.org/10.3390/ijms26083781

Chicago/Turabian Style

Reynoso-García, María Fernanda, Dulce E. Nicolás-Álvarez, A. Yair Tenorio-Barajas, and Andrés Reyes-Chaparro. 2025. "Structural Bioinformatics Applied to Acetylcholinesterase Enzyme Inhibition" International Journal of Molecular Sciences 26, no. 8: 3781. https://doi.org/10.3390/ijms26083781

APA Style

Reynoso-García, M. F., Nicolás-Álvarez, D. E., Tenorio-Barajas, A. Y., & Reyes-Chaparro, A. (2025). Structural Bioinformatics Applied to Acetylcholinesterase Enzyme Inhibition. International Journal of Molecular Sciences, 26(8), 3781. https://doi.org/10.3390/ijms26083781

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop