Next Article in Journal
Chromo-Fluorogenic Detection of Cyanide Ion with a Cyanine Probe
Previous Article in Journal
Synthesis of C60 Fullerene Hexakis Adducts Based on Strained Polycyclic Hydrocarbons and Their Application in Organic Electronics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Proceeding Paper

In Silico Approaches for the Study of New Anti-Parkinson’s Agents †

1
Group of Computational and Medicinal Chemistry, LMCE Laboratory, University of Mohamed KhiderBiskra, Biskra 07000, Algeria
2
Department of Chemistry, Faculty of Sciences, University of Mohamed Khider Biskra, Biskra 07000, Algeria
3
Department of Chemistry, Laboratory of Natural Substances and Bioactive (LASNABIO), University of Abou Bekr Belkaid, Tlemcen 13000, Algeria
*
Author to whom correspondence should be addressed.
Presented at the 27th International Electronic Conference on Synthetic Organic Chemistry (ECSOC-27), 15–30 November 2023; Available online: https://ecsoc-27.sciforum.net/.
Chem. Proc. 2023, 14(1), 55; https://doi.org/10.3390/ecsoc-27-16067
Published: 15 November 2023

Abstract

:
Parkinson’s disease (PD) is the second most common neurodegenerative chronic disorder in older people, caused by the loss of dopaminergic neurons in the brain, which leads to dopamine depletion. Among its symptoms are resting tremor, rigidity, and dementia. Monoamine oxidase B (MAO-B) is a key enzyme in PD. The inhibition of it increases the level of dopamine in the brain by preventing its degradation, especially in the early stages of the disease. In the present work, we combined molecular docking with an ADME analysis of a new class of ten derivatives of indanone in order to reduce the metabolic depletion of dopamine through inhibiting the MAO-B. A molecular docking study was performed to clarify the bonding modes and affinities between the active site residues of MAO-B with the new class derivatives of indanone, followed by the ADME prediction to check the drug-likeness properties of the best ligands obtained using the method above. Molecular docking results show that the ligands L3 and L5 presented a high negative score energy (−8.809 kcal/mol, −9.276 kcal/mol), respectively, with good RMSD values (1.419 Å, 1.560 Å), respectively. Moreover, the ADME prediction gave promising results that those ligands have an oral bioavailability and a high level of gastrointestinal absorption, which means they can be good inhibitors against Parkinson’s disease.

1. Introduction

The prevalence of neurodegenerative disorders (NDs) is increasing, making them a global health concern of significant proportions. The aging population, especially in wealthy countries, has seen an elevation in the rate at which chronic diseases are spreading in recent years [1]. Parkinson’s disease (PD) is the second most commonneurodegenerative disease [2], caused by the death of dopaminergic neurons in the substantia nigra, which depletes dopamine (Figure 1). Motor symptoms include resting tremor, and rigidity, while non-motor symptoms include disorganization and coherence distortion [3]. MAO-B (monoamine oxidase B) (Figure 2) is a major target in PD. Inhibition of this enzyme boosts dopamine levels in the brain by preventing its breakdown, particularly in the early stages of illness [2].
This research is a structure-based drug design investigation of a new family of indanone derivatives to diminish the metabolic depletion of dopamine by inhibiting the Parkinson’s disease-related enzyme MAO-B.

2. Material and Methods

In order to understand the interactions and affinities between this new class of derivatives and the active site residues of the MAO-B target, we conducted a molecular docking analysis using the following parameters: the scoring energy, the RSMD value of the generated complexes, and the interaction kinds and distances. A computation of ADME-T characteristics was performed to investigate the drug-likeness proprieties of the best compounds generated using molecular modeling simulation.
MAO-B (PDB ID: 2C65) crystal structure were retrieved from the RCSB Database (www.rcsb.org/pdb, accessed on.14 October 2023). Compound structures were designed using the ChemDraw professional 16.0 program; they were then optimized using the Molecular Operating Environment (MOE) software (Version 2014.09) and the semi-empirical energy function AM1.
To facilitate the molecular docking process, cofactors, ions, and glycols have been removed from the chosen enzyme. The compounds were designed to be flexible, whereas the target was inflexible. All complexes’ molecular docking data were acquired using the same software. The ADME calculations were performed using the SWISSADME web-based program.

3. Results and Discussion

According to the results of the molecular docking studies (Figure 3), the formation of the complexes that are the most stable occurs when molecules L3: 5-(2-(Pyrrolidin-1-yl)ethoxy)-2,3-dihydro-1H-inden-1-one and L5: 5-(3-(Piperidin-1-yl)propoxy)-2,3-dihydro-1H-inden-1-one are incorporated into MAO-B (PDB: 2C65). From Table 1, both L3 and L5 have negative energy scores of −8.809 and −9.276 (kcal/mol) and good RMSD values of 1.419 (Å), 1.560 (Å), respectively; have passed all drug-likeness rules (Table 2) without any violations; and have a bioavailability score of 0.55 (Table 3), which indicates that they have excellent pharmacokinetic characteristics and good oral and gastrointestinal absorption; this means they are also considered excellent candidates for further study. The low values of the synthetic accessibility score (SA score) for these ligands (SA2) provide additional evidence for the ease with which they can be synthesized.

4. Conclusions

In light of these findings and due to their low negative score energy, MAO-B-L3 and MAO-B-L5 complexes are the most stable.
Compounds L1: 6-(2-(Pyrrolidin-1-yl)ethoxy)-2,3-dihydro-1H-inden-1-one and L2: 6-(3-(Piperidin-1-yl)propoxy)-2,3-dihydro-1H-inden-1-one were also shown to be pharmacologically active, suggesting they could be useful as lead candidates for the cure or inhibition of Parkinson’s disease.

Author Contributions

Data collection, software, formal analysis, and first draft of the manuscript were prepared by M.M. All authors commented on previous versions of the manuscript. I.D. contributed to the conceptualization and supervision of the study. N.M. and I.D. contributed to the interpretation of docking and ADME-T studies. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare that they have no conflict of interest.

References

  1. Mettai, M.; Daoud, I.; Mesli, F.; Kenouche, S.; Melkemi, N.; Kherachi, R.; Belkadi, A. Molecular docking/dynamics simulations, MEP analysis, bioisosteric replacement and ADME/T prediction for identification of dual targets inhibitors of Parkinson’s disease with novel scaffold. In Silico Pharmacol. 2023, 11, 3. [Google Scholar] [CrossRef] [PubMed]
  2. Koszła, O.; Stępnicki, P.; Zięba, A.; Grudzińska, A.; Matosiuk, D.; Kaczor, A.A. Current approaches and tools used in drug development against parkinson’s disease. Biomolecules 2021, 11, 897. [Google Scholar] [CrossRef] [PubMed]
  3. Saleem, U.; Shabana, B.; Muhammad, A.S.; Bashir, A.; Ammara, S.; Zunera, C.; Fareeha, A.; Nimra, J.; Sundas, H.; Muhammad, F.A.; et al. Anti-Parkinson’s Evaluation of Brassica Juncea Leaf Extract and Underlying Mechanism of Its Phytochemicals. Front. Biosci.-Landmark 2021, 26, 1031–1051. [Google Scholar]
Figure 1. Healthy and depreciated substantianigra because of Parkinson’s disease.
Figure 1. Healthy and depreciated substantianigra because of Parkinson’s disease.
Chemproc 14 00055 g001
Figure 2. 3D structure of MAO-B.
Figure 2. 3D structure of MAO-B.
Chemproc 14 00055 g002
Figure 3. 2D and 3D interaction of the best compounds into MAO-B.
Figure 3. 2D and 3D interaction of the best compounds into MAO-B.
Chemproc 14 00055 g003
Table 1. S-score (energy) and interactions between compounds and the active site residues of hMAO-B target.
Table 1. S-score (energy) and interactions between compounds and the active site residues of hMAO-B target.
LigandsS-Score (kcal/mol)RMSD
(Å)
Bonds between Atoms and hMAO-B (PDB ID:2C65) Active Site Residues
Atom of LigandInvolved Receptor AtomsInvolved Receptor ResiduesType of Interaction BondDistance
(Å)
L3−8.8091.4196-ringCAILE199(A)Pi-H4.60
L5−9.2761.560/////
Table 2. ADME properties of the best ligands.
Table 2. ADME properties of the best ligands.
LigandsPhysico-Chemical PropertiesLipophilcityPharmacokenitics
TPAS (Å)MW
(g/mol)
Num Rotatable BondsNum H-Bond AcceptorsNum H-Bond DonorsMLOGPWLOGPGI AbsorptionBBB Permeant
L329.54245.324301.831.91HighYes
L529.54273.375302.332.69HighYes
Table 3. Drug-likeness, bioavailability score and synthetic accessibility of the best ligands.
Table 3. Drug-likeness, bioavailability score and synthetic accessibility of the best ligands.
LigandsDrug-likenessBiovailability ScoreSynthetic Accessibility
LipinskiGhoseVeberEgan
L3Yes
0 violation
YesYesYes0.552.09
L5Yes
0 violation
YesYesYes0.552.30
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

Mettai, M.; Daoud, I.; Melkemi, N. In Silico Approaches for the Study of New Anti-Parkinson’s Agents. Chem. Proc. 2023, 14, 55. https://doi.org/10.3390/ecsoc-27-16067

AMA Style

Mettai M, Daoud I, Melkemi N. In Silico Approaches for the Study of New Anti-Parkinson’s Agents. Chemistry Proceedings. 2023; 14(1):55. https://doi.org/10.3390/ecsoc-27-16067

Chicago/Turabian Style

Mettai, Merzaka, Ismail Daoud, and Nadjib Melkemi. 2023. "In Silico Approaches for the Study of New Anti-Parkinson’s Agents" Chemistry Proceedings 14, no. 1: 55. https://doi.org/10.3390/ecsoc-27-16067

Article Metrics

Back to TopTop