In-silico screening and ADMET evaluation of therapeutic MAO-B inhibitors against Parkinson disease

Abduljelil Ajala, Wafa Ali Eltayb, Terungwa Michael Abatyough, Stephen Ejeh, Mohamed El fadili, Habiba Asipita Otaru, Emmanuel Israel Edache, A. Ibrahim Abdulganiyyu, Omole Isaac Areguamen, Shashank M. Patil, Ramith Ramu

PDF(3614 KB)
PDF(3614 KB)
Intelligent Pharmacy ›› 2024, Vol. 2 ›› Issue (4) : 554-564. DOI: 10.1016/j.ipha.2023.12.008

In-silico screening and ADMET evaluation of therapeutic MAO-B inhibitors against Parkinson disease

Author information +
History +

Abstract

MAOs are flavoenzymes that aid in the oxidative deamination of neurotransmitters such as dopamine, serotonin, and epinephrine. MAO inhibitors are antidepressants that act by inhibiting neurotransmitter breakdown in the brain and controlling mood. MAO inhibitors with the chlorophenyl-chromone-carboxamide structure have been shown in investigations to be extremely effective. The current study employs in-silico screening, MD simulation, and drug kinetics evaluation, all of which are evaluated using different criteria. The study comprised 37 ligands, and three stood out as the best, with greater binding scores above the threshold value. Docking analysis found that compound 34 had the highest docking score in the series (-13.60 kcal/mol) and interacted with the important amino acids TYR 435, CYS 397, CYS 172, PHE 343, TYR 398, and LYS 296 required for MAO inhibitory activity. The ADMET study revealed that the compounds had drug-like properties. The results of this study could be used to develop chromone drugs that target the MAO inhibitor. The top three ligands with the highest force and work were then simulated using molecular dynamics. The protein-ligand complexes had steady trajectories throughout the 100 ns simulation, according to the data. Furthermore, the drug likeliness predicted by ADMET analysis findings indicated that the top three lead compounds had strong inhibitory efficiency, superior pharmacokinetics, and were non-toxic under physiological settings. As a result, these compounds have the potential to be exploited as possible treatment medications for PD.

Keywords

MAO-B inhibitors / Parkinsonism disorder / Molecular docking / Molecular dynamics simulation / Pharmacokinetic

Cite this article

Download citation ▾
Abduljelil Ajala, Wafa Ali Eltayb, Terungwa Michael Abatyough, Stephen Ejeh, Mohamed El fadili, Habiba Asipita Otaru, Emmanuel Israel Edache, A. Ibrahim Abdulganiyyu, Omole Isaac Areguamen, Shashank M. Patil, Ramith Ramu. In-silico screening and ADMET evaluation of therapeutic MAO-B inhibitors against Parkinson disease. Intelligent Pharmacy, 2024, 2(4): 554‒564 https://doi.org/10.1016/j.ipha.2023.12.008

References

[1]
Alexander GE. Biology of Parkinson’s disease: pathogenesis and pathophysiology of a multisystem neurodegenerative disorder. Dialogues Clin Neurosci. 2022.
[2]
Espay AJ, Vizcarra JA, Marsili L, et al. Revisiting protein aggregation as pathogenic in sporadic Parkinson’s and Alzheimer’s diseases. Neurology. 2019;92(7):329–337.
[3]
Ragab OA, Elheneedy YA, Bahnasy WS. Non-motor symptoms in newly diagnosed Parkinson’s disease patients. Egyptian J Neurol, Psychiatr Neurosurg. 2019;55(1):1–7.
CrossRef Google scholar
[4]
Schaeffer E, Berg D. Dopaminergic therapies for non-motor symptoms in Parkinson’s disease. CNS Drugs. 2017;31(7):551–570.
CrossRef Google scholar
[5]
Radhakrishnan DM, Goyal V. Parkinson’s disease: a review. Neurol India. 2018;66(7): 26.
CrossRef Google scholar
[6]
Khanfar MA, Al-Qtaishat S, Habash M, Taha MO. Discovery of potent adenosine A2a antagonists as potential anti-Parkinson disease agents. Non-linear QSAR analyses integrated with pharmacophore modeling. Chem Biol Interact. 2016;254:93–101.
CrossRef Google scholar
[7]
Sebastián-Pérez V, Martínez MJ, Gil C, Campillo NE, Martínez A, Ponzoni I. QSAR Modelling to identify LRRK2 inhibitors for Parkinson’s disease. J Integrat Bioinformatic. 2019;16(1).
CrossRef Google scholar
[8]
R Munteanu C, Fernández-Blanco E, A Seoane J, et al. Drug discovery and design for complex diseases through QSAR computational methods. Curr Pharmaceut Des. 2010; 16(24):2640–2655.
CrossRef Google scholar
[9]
Nikolic K, Mavridis L, Djikic T, et al. Drug design for CNS diseases: polypharmacological profiling of compounds using cheminformatic, 3D-QSAR and virtual screening methodologies. Front Neurosci. 2016;10:265.
CrossRef Google scholar
[10]
De P, Roy K. QSAR modeling of PET imaging agents for the diagnosis of Parkinson’s disease targeting dopamine receptor. Theor Chem Acc. 2020;139(12):1–12.
CrossRef Google scholar
[11]
Helguera AM, Pérez-Garrido A, Gaspar A, et al. Combining QSAR classification models for predictive modeling of human monoamine oxidase inhibitors. Eur J Med Chem. 2013;59:75–90.
CrossRef Google scholar
[12]
Tong JB, Wang J, Luo D, et al. QSAR study, molecular docking, and ADMET prediction of vinyl sulfone-containing Nrf2 activator derivatives for treating Parkinson disease. Struct Chem. 2022;33(4):1109–1131.
CrossRef Google scholar
[13]
Pandey S, Srivanitchapoom P. Levodopa-induced dyskinesia: clinical features, pathophysiology, and medical management. Ann Indian Acad Neurol. 2017;20(3): 190.
CrossRef Google scholar
[14]
Seppi K, Ray Chaudhuri K, Coelho M, et al. Update on treatments for nonmotor symptoms of Parkinson’s disease—an evidence-based medicine review. Mov Disord. 2019;34(2):180–198.
CrossRef Google scholar
[15]
Armstrong MJ, Okun MS. Diagnosis and treatment of Parkinson disease: a review. JAMA. 2020;323(6):548–560.
CrossRef Google scholar
[16]
Tran TN, Vo TN, Frei K, Truong DD. Levodopa-induced dyskinesia: clinical features, incidence, and risk factors. J Neural Transm. 2018;125(8):1109–1117.
CrossRef Google scholar
[17]
Tripathi AC, Upadhyay S, Paliwal S, Saraf SK. Privileged scaffolds as MAO inhibitors: retrospect and prospects. Eur J Med Chem. 2018;145:445–497.
CrossRef Google scholar
[18]
Behl T, Kaur I, Sehgal A, et al. The footprint of kynurenine pathway in neurodegeneration: Janus-faced role in Parkinson’s disorder and therapeutic implications. Int J Mol Sci. 2021;22(13):6737.
CrossRef Google scholar
[19]
Hussain S, Ullah F, Ayaz M, et al. In silico, cytotoxic and antioxidant potential of novel ester, 3-hydroxyoctyl-5-trans-docosenoate isolated from anchusa arvensis (L.) m. bieb. against hepg-2 cancer cells. Drug Des Dev Ther. 2019;13:4195.
CrossRef Google scholar
[20]
Er-Rajy M, Kara M, Assouguem A, et al. QSAR, ADMET in silico pharmacokinetics, molecular docking and molecular dynamics studies of novel Bicyclo (Aryl methyl) benzamides as potent GlyT1 inhibitors for the treatment of Schizophrenia. Pharmaceuticals. 2022;15(6):670.
CrossRef Google scholar
[21]
Khan A, Pervaiz A, Ansari B, et al. Phytochemical profiling, anti-inflammatory, anti-oxidant and in-silico approach of cornus macrophylla bioss (Bark). Molecules. 2022; 27(13):4081.
CrossRef Google scholar
[22]
Ullah N, Zeb S, Rukh S, et al. Bioassay’s directed isolation-structure elucidation and molecular docking of triterpenes from persea duthiei against biologically important microbial proteins. Evid base Compl Alternative Med. 2022;2022.
CrossRef Google scholar
[23]
Bari WU, Zahoor M, Zeb A, et al. Anticholinesterase, antioxidant potentials, and molecular docking studies of isolated bioactive compounds from Grewia optiva. Int J Food Prop. 2019;22(1):1386–1396.
CrossRef Google scholar
[24]
Shahid F, Ali R, Badshah SL, et al. Identification of Potential HCV Inhibitors Based on the interaction of epigallocatechin-3-gallate with viral envelope proteins. Molecules. 2021;26(5):1257.
CrossRef Google scholar
[25]
Ali R, Badshah SL, Faheem M, et al. Identification of potential inhibitors of Zika virus NS5 RNA-dependent RNA polymerase through virtual screening and molecular dynamic simulations. Saudi Pharmaceut J. 2020;28(12):1580–1591.
CrossRef Google scholar
[26]
dan M, Djikic T, Obradovic D, Nikolic K. Application of in vitro PAMPA technique and in silico computational methods for blood-brain barrier permeability prediction of novel CNS drug candidates. Eur J Pharmaceut Sci. 2022;168:106056.
CrossRef Google scholar
[27]
RaSaini N, Akhtar A, Chauhan M, Dhingra N, Sah SP. Protective effect of Indole-3-carbinol, an NF-κB inhibitor in experimental paradigm of Parkinson’s disease: in silico and in vivo studies. Brain Behav Immun. 2020;90:108–137.
CrossRef Google scholar
[28]
Cruz-Vicente P, Passarinha LA, Silvestre S, Gallardo E. Recent developments in new therapeutic agents against Alzheimer and Parkinson diseases: in-silico approaches. Molecules. 2021;26(8):2193.
CrossRef Google scholar
[29]
Patil SM, Martiz RM, Ramu R, et al. Evaluation of flavonoids from banana pseudostem and flower (quercetin and catechin) as potent inhibitors of α-glucosidase: an in silico perspective. J Biomol Struct Dyn. 2021;7:1–5.
[30]
Reis J, Manzella N, Cagide F, et al. Tight-binding inhibition of human monoamine oxidase B by chromone analogs: a kinetic, crystallographic, and biological analysis. J Med Chem. 2018 May 10;61(9):4203–4212.
CrossRef Google scholar
[31]
Ganavi D, Ramu R, Kumar V, et al. In vitro and in silico studies of fluorinated 2,3-disubstituted thiazolidinone-pyrazoles as potential α-amylase inhibitors and antioxidant agents. Arch Pharm. 2021;12:e2100342.
[32]
Secci D, Carradori S, Petzer A, et al. 4-(3-Nitrophenyl) thiazol-2-ylhydrazone derivatives as antioxidants and selective hMAO-B inhibitors: synthesis, biological activity and computational analysis. J Enzym Inhib Med Chem. 2019;34(1):597–612.
CrossRef Google scholar
[33]
Patil SM, Martiz RM, Ramu R, et al. In silico identification of novel benzophenonecoumarin derivatives as SARS-CoV-2 RNAdependent RNA polymerase (RdRp) inhibitors. J Biomol Struct Dyn. 2021;11:1–17.
[34]
Poojary B, Kumar V, Arunodaya HS, et al. Potential fluorinated anti-MRSA thiazolidinone derivatives with antibacterial, antitubercular activity and molecular docking studies. Chem Biodivers. 2021;19:e202100532.
CrossRef Google scholar
[35]
Maradesha T, Patil SM, Al-Mutairi KA, Ramu R, Madhunapantula SV, Alqadi T. Inhibitory effect of polyphenols from the whole green jackfruit flour against α-glucosidase, α-amylase, aldose reductase and glycation at multiple stages and their interaction: inhibition kinetics and molecular simulations. Molecules. 2022;27:1888.
CrossRef Google scholar
[36]
Gurupadaswamy HD, Ranganatha VL, Ramu R, Patil SM, Khanum SA. Competent synthesis of biaryl analogs via asymmetric Suzuki–Miyaura cross-coupling for the development of anti-inflammatory and analgesic agents. J Iran Chem Soc. 2022;1: 1–16.
[37]
Martiz RM, Patil SM, Abdulaziz M, et al. Defining the role of isoeugenol from ocimum tenuiflorum against diabetes mellitus-linked alzheimer’s disease through network pharmacology and computational methods. Molecules. 2022;27:2398.
CrossRef Google scholar
[38]
Martiz RM, Patil SM, Ramu R, et al. Discovery of novel benzophenone integrated derivatives as anti-Alzheimer’s agents targeting presenilin-1 and presenilin-2 inhibition: a computational approach. PLoS One. 2022;17:e0265022.
CrossRef Google scholar
[39]
Kumar V, Ramu R, Shirahatti PS, et al. α-Glucosidase; α-amylase inhibition; kinetics and docking studies of novel (2-Chloro-6-(trifluoromethyl) benzyloxy) arylidene) based rhodanine and rhodanine acetic acid derivatives. Chem Select. 2021;6: 9637–9644.
CrossRef Google scholar
[40]
Kumar V, Shetty P, Arunodaya HS, et al. Potential fluorinated anti-MRSA thiazolidinone derivatives with antibacterial, antitubercular activity and molecular docking studies. Chem Biodivers. 2022;19:e202100532.
CrossRef Google scholar
[41]
Patil SM, Maruthi KR, Bajpe NS, et al. Comparative molecular docking and simulation analysis of molnupiravir and remdesivir with SARS-CoV-2 RNA dependent RNA polymerase (RdRp). Bioinformation. 2021;7:932–939.
CrossRef Google scholar
[42]
Zhao Z, Ukidve A, Kim J, Mitragotri S. Targeting strategies for tissue-specific drug delivery. Cell. 2020;181(1):151–167.
CrossRef Google scholar
[43]
Tiwary P, Mondal J, Berne BJ. How and when does an anticancer drug leave its binding site? Sci Adv. 2017;3(5):e1700014.
CrossRef Google scholar
[44]
Nagar PR, Gajjar ND, Dhameliya TM. In search of SARS CoV-2 replication inhibitors: virtual screening, molecular dynamics simulations and ADMET analysis. J Mol Struct. 2021;1246:131190.
CrossRef Google scholar
[45]
Mishra S, Dahima R. In vitro ADME studies of TUG-891, a GPR-120 inhibitor using SWISS ADME predictor. J Drug Deliv Therapeut. 2019;9(2-s):366–369.
[46]
Lucas AJ, Sproston JL, Barton P, Riley RJ. Estimating human ADME properties, pharmacokinetic parameters and likely clinical dose in drug discovery. Expet Opin Drug Discov. 2019;14(12):1313–1327.
CrossRef Google scholar
[47]
Ban F, Dalal K, Li H, LeBlanc E, Rennie PS, Cherkasov A. Best practices of computer-aided drug discovery: lessons learned from the development of a preclinical candidate for prostate cancer with a new mechanism of action. J Chem Inf Model. 2017;57(5):1018–1028.
CrossRef Google scholar
[48]
Basu A, Sarkar A, Maulik U. Molecular docking study of potential phytochemicals and their effects on the complex of SARS-CoV2 spike protein and human ACE2. Sci Rep. 2020;10(1):1–15.
CrossRef Google scholar
[49]
Dhiman P, Malik N, Khatkar A. Lead optimization for promising monoamine oxidase inhibitor from eugenol for the treatment of neurological disorder: synthesis and in silico based study. BMC Chem. 2019 Dec;13(1):1–20.
CrossRef Google scholar
[50]
Grychowska K, Olejarz-Maciej A, Blicharz K, et al. Overcoming undesirable hERG affinity by incorporating fluorine atoms: a case of MAO-B inhibitors derived from 1 H-pyrrolo-[3, 2-c] quinolines. Eur J Med Chem. 2022 Jun 5;236:114329.
CrossRef Google scholar
[51]
Mateev E, Georgieva M, Zlatkov A. Improved molecular docking of MAO-B inhibitors with glide. 2022;13(2):159.
CrossRef Google scholar
[52]
Catalano R, Procopio F, Chavarria D, et al. Molecular modeling and experimental evaluation of non-chiral components of bergamot essential oil with inhibitory activity against human monoamine oxidases. Molecules. 2022 Apr 11;27(8):2467.
CrossRef Google scholar
[53]
Santos LHS, Ferreira RS, Caffarena ER. Integrating molecular docking and molecular dynamics simulations. Methods Mol Biol. 2019;2053:13–34.
CrossRef Google scholar
[54]
Chaurasiya ND, Zhao J, Pandey P, Doerksen RJ, Muhammad I, Tekwani BL. Selective inhibition of human monoamine oxidase B by acacetin 7-methyl ether isolated from turnera diffusa (Damiana). Molecules. 2019 Jan;24(4):810.
CrossRef Google scholar
[55]
Abdalla MA, Li F, Wenzel-Storjohann A, Sulieman S, Tasdemir D, Mühling KH. Comparative metabolite profile, biological activity and overall quality of three lettuce (Lactuca sativa L., Asteraceae) cultivars in response to sulfur nutrition. Pharmaceutics. 2021;13(5):713.
CrossRef Google scholar
[56]
Olasupo SB, Uzairu A, Shallangwa GA, Uba S. Computer-aided drug design and in silico pharmacokinetics predictions of some potential antipsychotic agents. Scientific African. 2021;12:e00734.
CrossRef Google scholar
[57]
Olasupo SB, Uzairu A, Shallangwa GA, Uba S. Unveiling novel inhibitors of dopamine transporter via in silico drug design, molecular docking, and bioavailability predictions as potential antischizophrenic agents. Fut J Pharmaceut Sci. 2021;7:1–10.
CrossRef Google scholar

RIGHTS & PERMISSIONS

2024 2024 The Authors. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co. Ltd.
AI Summary AI Mindmap
PDF(3614 KB)

Accesses

Citations

Detail

Sections
Recommended

/