Document Type : Original Article

Authors

1 Department of Natural and Applied Sciences, School of Science and Technology, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh 247121, India

2 Department of Applied Science, Dr. K. N. Modi University, Tonk, Rajasthan 304021, India

3 School of Pharmacy, Glocal University, Mirzapur Pole, Saharanpur, Uttar Pradesh 247121 India

4 Division of Chemistry, Department of Basic Sciences, School of Basic and Applied Sciences, Galgotias University, Greater Noida, Uttar Pradesh 203201, India

5 CSIR-CBRI Roorkee, Uttarakhand 247667, India

Abstract

In the present scenario of eco-preservation and eco-safe utilization, researchers globally have been attracted to the utilization of raw and sustainable products having significant therapeutic potential that allow safety, modality, and biological activeness with environmental compatibility. Ar-turmerone has various pharmacological actions, including antidepressant, antiepileptic, anti-dermatophyte, antivenom, anticancer, antiplatelet activity, etc. In the present work, we investigated Ar-Turmerone (Ar-Tume), one of the chief phytoconstituents present in Curcuma longa for human anticholinesterase (AChE) inhibitor (4PQE) and human salivary alpha-Amylase dimer (1XV8) hydrolase inhibitor as a natural product-based emerging scaffold. Our study reveals that the selected compound Ar-Tume showed remarked biological, ADMET profiling, and superior docking scores/negative binding energies (-7.9 against 4PQE and -6.7 against 1XV8) concerning the reference drugs, which attributed to the strong hydrogen-bonding interactions both towards both anti-Alzheimers and antidiabetic capabilities.

Graphical Abstract

Docking and ADMET Study of Ar-Turmerone: Emerging Scaffold for Acetylcholine Esterase Inhibition and Antidiabetic Target

Keywords

Introduction

In the 21st century, computer-aided drug design (CADD) enables a better understanding of synthetic protocols that provide the opportunity to transform drug development towards developing novel drugs with superior clinical properties. In this regard, many repurposed drugs have been developed using computational approaches and approved by FDA at a faster quicker approach to the clinical trials to treat several diseases such as inflammation, hypertension, obesity, type-2 diabetes, and Alzheimer’s disease (Figure 1) [1, 2]. The CADD approach further helps assess whether natural products/phytoconstituents cope with biological activity [3]. Natural products contain a range of phytochemical classes such as indole, quinolizidine, isoquinoline, stilbenes, piperidine, phenolics, flavonoids, and terpenoids [4-6], which have profound effects to exhibit excellent eco-friendly therapeutic potential along with better biodegradation with high environmental compatibility possessing the excellent bioactive functionalities owing to their core moieties [7].

Figure 1. Some FDA-approved drugs to treat Alzheimer's disease and diabetes type II

Turmeric (Curcuma longa), often used as a spice and affects the nature, color, and taste of foods, under the family Zingiberaceae, is commonly called turmeric or Haldi, typically cultivated and propagated in the tropical part of Asia, including India, China, Thailand, Iran, Malaysia, etc. The chief medicinally important part is its rhizome which contains curcuminoids, phenolics, aromatic quinone, quinines, and other bioactive compounds. The extract of Haldi is traditionally known for its therapeutic and biological engagements, for example, antioxidant, cardiovascular diseases, diabetic, immune-boosting, anti-inflammatory, anticancer, etc. Turmerones, the principal sesquiterpenes occurred naturally in turmeric, are α-turmerone, aromatic-turmerone, and β-turmerone, out of which Ar-turmerone is a major bioactive phytoconstituent [8]. Ar-turmerone is one of the main compounds contained in turmeric essential oil. Ar-turmerone also has diverse biological activities, as well as curcumin. Hucklenbroich et al. reported that Ar-turmerone inhibited microglia activation indicating its effectiveness in treating neurodegenerative diseases [9]. In a previous study, we found that curcuminoids and other turmeric constituents had significant in silico ADME and COX-2 inhibitory activity [10]. Ar-turmerone has powerful antivenom action against snake bites [11]. Likewise, Ar-turmerone has anti-inflammatory, anti-aging, anti-plasmodial, and neuroprotective activities [12-14]. In addition, with a certain dose, ar-turmerone has biological activities such as antiepileptic, antidepressant, anti-dermatophyte, anticancer, and antiplatelet effects [15]. Various biological action of ar-turmerone has led to the development of research on ar-turmerone.

In continuation, we investigated Ar-Turmerone (Ar-Tume) for Acetylcholine Esterase (AChE) inhibition and antidiabetic target as a natural product-based emerging scaffold.

 Experimental

Materials and methods

Preparation of receptor human Alzheimer’s disease inhibitors and ligands

The crystal structure of human anticholinesterase (AChE) inhibitor (4PQE) under a resolution of 2.90 Å and the crystal structure of human salivary alpha-Amylase dimer (1XV8) hydrolase inhibitor under a resolution of 3.00 Å in PDB format was downloaded from Protein Data Bank (https://www.rcsb.org/). Galantamine (Gala, ChemSpider ID: 9272) and Voglibose (Vogl, ChemSpider ID: 392046) were taken from ChemSpider online platform (http://www.chemspider.com/) as a standard reference drugs. Ligands 1-3 (Table 1) were prepared using ChemDraw Ultra (Cambridge Soft Corporation USA) and obtained SMILES and .mol2 files were validated via Avogadro Software v1.2.0.

Table 1. Selected reference drugs and Ar-Tumeroneas ligand

Biological characteristics evaluation and ADMET profile assay

Molinspiration biological characteristics of the selected compounds 1-3 were evaluated using Molinspiration Cheminformatics Online Server (https://www.molinspiration.com/) [16]. To evaluate ADMET properties, Swiss ADME algorithm (http://www.swissadme.ch) and pkCSM online servers (http://biosig.unimelb.edu.au/pkcsm/) were used.

Molecular docking studies against human anticholinesterase and antidiabetic hydrolase inhibitors

The Vinadock automation-assisted prediction of binding energies and interactive 3D visualization of results towards human anticholinesterase (AChE) inhibitor (4PQE: water molecules were deleted from the uploaded protein structure before docking) was used CB-Dock Online platform; server2 (http://cadd.labshare.cn/cb-dock2/) [10,17] concerning reference drugs.

Results and Discussion

Biological characteristics evaluation and prediction of ADMET properties

The biological activities of Gala, Vogl, and Ar-Tume were evaluated and presented in Table 2. Concerning the standard drug, Ar-Tume has shown the pronounced biological action profiles. For a novel drug discovery using pharmacokinetic and pharmacodynamics profiles, the validation of the drug is essentially recommended by researchers [17]. Therefore, it fundamentally required the safety criterion with significant efficacy [10,18].

Table 2. Molinspiration-predicted biological characteristics of selected drugs

The ADMET profiles for Gala, Vogl, and Ar-Tumehave been found to have significant positive characteristics concerning physicochemical properties, lipophilicity, drug-likeness, and medicinal chemistry parameters (Table 3) for targeted drugs calculated with Swissdock ADME and pkCSM platforms capped with biological-logarithms. Ar-Tume exhibited remarked performance and characteristics to a significant extent, similar to standard drugs. Table 4 lists that Ar-Tume shows acceptable absorption parameters compared with the reference drug concerning Caco-2 permeability, Caco-2 permeability, human intestinal absorption, and skin permeability. Furthermore, Table 5 provides considerable human volume of distribution and human fraction unbound (Fu) values, BBB, and CNS permeability for Ar-Tume. Table 6 also explains that Ar-Tumeexhibitedgood effectiveness towards CYP2D6 substrate, CYP3A4 substrate, CYP1A2 inhibitor, CYP2C19 inhibitor, CYP2C9 inhibitor, CYP2D6 inhibitor, and CYP3A4 inhibitor. Table 7 demonstrates non-hepatotoxic, and no toxicity profiling was observed for Ar-Tume.

Table 3. Physicochemical properties, lipophilicity, drug-likeness, and medicinal chemistry parameters for selected drugs calculated with Swissdock ADME and pkCSM

 

 

 

 

 

Table 4. Absorption parameters for selected drugs calculated with pkCSM

Table 5. Distribution and excretion parameters for selected drugs calculated with pkCSM

Table 6. Metabolism parameters for selected drugs calculated with pkCSM

Table 7. Toxicity parameters for selected drugs calculated with pkCSM

Molecular docking studies

Molecular docking is used to predict the orientation, type of interaction, and binding energy of selected molecular ligands in the interior of the binding site. Figure 2 depicts the docking score for the active linkage of Gala, Vogl, and Ar-Tume against human anticholinesterase (AChE) inhibitor (4PQE) and human salivary alpha-Amylase dimer (1XV8) hydrolase inhibitor (Table 1) and as a result, the binding energies directs a significant ratio concerning the reference drug (Table 1, Figures 2 and 3). Plant-derived natural products are potentially important because of their inherent biological activities like antioxidant, antimicrobial, antidiabetic, etc. with environmental compatibility [15-17, 19, 20]. This study demonstrates that the selected compound Ar-Tume remarked to have mobile superior docking scores/negative binding energies concerning the reference drugs attributed to the strong hydrogen-bonding interactions in both inter and intra assemblies towards both anti-Alzheimers and antidiabetic capabilities.

Figure 2. A comparative molecular docking score of Gala, Vogl, and Ar-Tume compounds

Figure 3. Molecular docking pattern of Gala, Vogland Ar-Tume ligands

Conclusion

Turmeric contains various natural phytoconstituents and has been found effective to have biological and therapeutic potential such as antimicrobial, anticancer, antidiabetic, and other health-related ailments. In the present study, Ar-Turmerone, one of the chief phytoconstituents present in Curcuma longa for human anticholinesterase (AChE) inhibitor (4PQE) and human salivary alpha-Amylase dimer (1XV8) hydrolase inhibitor as an emerging scaffold. This study reveals that the selected compound Ar-Tume showed remarked biological, ADMET profiling, and superior docking scores/ negative binding energies (-7.9 against 4PQE and -6.7 against 1XV8) concerning the reference drugs attributed to the strong hydrogen-bonding interactions both inter and intra assemblies towards both anti-Alzheimer's and antidiabetic capabilities which might further be used as oral therapeutics after clinical trials and these derivatives would be the initial step towards the exploration for biomedical applications with promising drug candidature to support in the treatment of neurologic disorders and diabetes in future.

Acknowledgments

Prof. S. A. Ahmed, Vice-Chancellor of Glocal University, is acknowledged for his moral support and critical suggestions.

Orcids

Mohd Yusuf:

https://orcid.org/0000-0003-0927-8490

Sukhvinder Pal:

https://orcid.org/0000-0002-3277-0566

Mohammad Shahid:

https://orcid.org/0000-0001-9787-2881

Mohammad Asif:

https://orcid.org/0000-0002-9352-3462

Shafat Ahmad Khan:

https://orcid.org/0000-0003-3744-6391

Rakhi Tyagi:

https://orcid.org/0000-0001-8084-7655

Citation M. Yusuf, S. Pal, M. Shahid, M. Asif*, S. Ahmad Khan, R. Tyagi. Docking and ADMET Study of Ar-Turmerone: Emerging Scaffold for Acetylcholine Esterase Inhibition and Antidiabetic Target. J. Appl. Organomet. Chem., 2023, 3(1), 52-60.

 

[1]. O. Benek, J. Korabecny, O. Soukup, Trends Pharmacol Sci., 2020, 41, 434-445. [Crossref], [Google Scholar], [Publisher]
[2]. E. Gourgari, E.E. Wilhelm, H. Hassanzadeh, V.R. Aroda, I. Shoulson, J. Diabetes Complicat., 2017, 31, 1719-1727. [Crossref], [Google Scholar], [Publisher]
[3]. J.J. Kellogg, M.F. Paine, J.S. McCune, N.H. Oberlies, N.B. Cech, Nat. Prod. Rep., 2019, 36, 1196-1221. [Crossref], [Google Scholar], [Publisher]
[4]. A. Parihar, S.S. Ahmed, P. Sharma, N.K. Choudhary, F. Akter, M. Ali, Z.F. Sonia, R. Khan, Mol. Simul., 2022, 1-13. [Crossref], [Google Scholar], [Publisher]
[5]. M. Aijaz, N. Keserwani, M. Yusuf, N.H. Ansari, R. Ushal, P. Kalia, Biointerface Res. J. Appl. Chem., 2022, 13, 324. [Crossref], [Google Scholar]
[6]. M. Yusuf, U. Chawla, N.H. Ansari, M. Sharma, M. Asif, Adv. J. Chem. A, 2023, 6, 31-49. [Crossref], [Publisher]
[7]. V.K. Sharma, M. Yusuf, P. Kumar, M. Sharma, V.C. Waila, S.A. Khan, N.H. Ansari, S. Zaphar, J. Pharm. Res. Int., 2021, 33, 216-230 [Crossref], [Google Scholar], [Publisher]
[8]. M. Yusuf, B.A. Sadiya, M. Gulfishan. Biointerface Res Appl Chem. 2022, 12, 7177-204. [Crossref], [Google Scholar]
[9]. J. Hucklenbroich, R. Klein, B. Neumaier, R. Graf, G.R. Fink, M. Schroeter, M.A. Rueger, Stem Cell Res. Ther., 2014, 5, 100. [Crossref], [Google Scholar], [Publisher]
[10]. M. Yusuf, S.A. Khan, Biointerface Res Appl Chem., 2021, 13, 1-23. [Crossref], [Google Scholar]
[11]. G.S. Husein, S. Hamdani, K. Rinaldi. Lett Appl Nano Bio Sci. 2023, 12, 53. [Crossref], [Google Scholar], [Publisher]
[12]. S. Yang, J. Liu, J. Jiao, L. Jiao, Inflammation, 2020, 43, 478-486. [Crossref], [Google Scholar], [Publisher]
[13]. A.H. Ali, H.K. Agustar, N.I. Hassan, J. Latip, N. Embi, H.M. Sidek, Data Brief., 2020, 33, 106592. [Crossref], [Google Scholar], [Publisher]
[14]. Y. Saga, Y. Hatakenaka, M. Matsumoto, Y. Yoshioka, S. Matsumura, N. Zaima, Y. Konishi, Neuro Report., 2020, 31, 1302-1307. [Crossref], [Google Scholar], [Publisher]
[15]. Y. Zheng, C. Pan, Z. Zhang, W. Luo, X. Liang, Y. Shi, L. Liang, X. Zheng, L. Zhang, Z. Du., Microchem J., 2020, 154, 104608. [Crossref], [Google Scholar], [Publisher]
[16]. Y. Liu. M. Grimm, W.T. Dai, M.C. Hou, Z.X. Xiao, Y. Cao, Acta Pharmacol Sin., 2020, 41, 138-144. [Crossref], [Google Scholar], [Publisher]
[17]. J.M. Korth-Bradley, J Clin Pharmacol., 2022, 62, S15-S26. [Crossref], [Google Scholar], [Publisher]
[18]. M.M. Almehmadi, M. Halawi, M. Kamal, M. Yusuf, U. Chawla, M. Asif, Lat Am JPharm., 2022, 41, 1428-1432. [Crossref], [Google Scholar]
[19]. S. Chadorshabi, S. Hallaj-Nezhadi, Z. Ghasempour, Food Chem., 2022, 386, 132737. [Crossref], [Google Scholar], [Publisher]
[20]. M. Yusuf, M. Shabbir, F. Mohammad. Nat Prod Bioprospect. 2017, 7, 123-145. [Crossref], [Google Scholar], [Publisher]