EVALUATION OF ANTAGONIST ACTIVITY OF IFENPRODIL AND THEIR ANALOGOUS AGAINST GLUN1/GLUN2B USING IN SILICO MOLECULAR DOCKING AND ABSORPTION-DISTRIBUTIONMETABOLISM- EXCRETION TOXICITY

Authors

  • MALLARI PRAVEEN Department of Zoology, Indira Gandhi National Tribal University, Amarkantak, Madhya Pradesh, India. https://orcid.org/0000-0003-0896-400X

DOI:

https://doi.org/10.22159/ajpcr.2022.v15i5.44248

Keywords:

Ifenprodil and analogous, molecular docking, Absorption-distribution-metabolism-excretion-toxicity, GluN1/GluN2B

Abstract

Objectives: High Ca2+ permeability represents a characteristic feature of N-methyl-D-aspartate (NMDA) receptors that in extreme amounts affects physiological functions such as reduced neural development, synaptic plasticity, and learning and memory. The study aims to elucidate the potent inhibitory ifenprodil and their eleven analogues, retrieved from the PubChem database, which act as ligands to the target Glun1/GluN2B subunit of the NMDA receptor.

Methods: In silico methods such as molecular docking performed using AutoDock Vina and absorption-distribution-metabolism-excretion-toxicity (ADMET) were SwissADME and OSIRIS carried out to elucidate the potent antagonist ligand against the target.

Results: Molecular docking showed that six of the compounds had significant binding affinities (7.8–9.0 kcal/mol) for the target. The ADMET study revealed that three (PubChemID: 12613159, 12613162, and 6604117) of six compounds with good binding affinity obeyed Lipinski’s rule of five.

Conclusion: This study revealed three good antagonists of GluN1/GluN2B, namely, 4- [(1R, 2R)-2-(4-benzylpiperidin-1-yl)-1- hydroxy propyl] phenol (A2), 4-[2-(4-benzylpiperidin-1-yl)-1-hydrooxypropyl] phenol hydrobromide (A4), and 4- [(1R, 2S)-2-(4-benzylpiperidin-1-yl)-1-hydroxypropyl] phenol (A7) that can be further exploited for wet lab studies.

Downloads

Download data is not yet available.

References

Lodge D. The history of the pharmacology and cloning of ionotropic glutamate receptors and development of idiosyncartic nomenclature, neuropharmacol. Neuropharmocology 2009;56:6-21.

Bräuner-Osborne H, Egebjeg J, Nielson EO, Madsen U, Krogagaard- Larsen P. Ligands for glutamate receptors: Design and therapeutic prospects. J Med Chem 2000;43:2609-45.

Traynelis SF, Wollmuth LP, McBrain CJ, Menniti FS, Vance KM, Odgen KK, et al. Glutamate receptor ion channels: Structure, regulation, and function. Phamacology 2010;62:405-96.

Kew JN, Kemp JA. Ionotropic and metabotropic glutamate receptor structure and pharmacology. Psychopharmacology 2005;179:4-29.

Meldrum BS. Glutamate as a neurotransmitter in the brain: Review of physiology and pathology. J Nutr 2000;130:1007-50.

Westbrook GL, Mayer ML. The physiology of excitatory amino acids in the vertebrate central nervous system. Prog Neurobiol 1987;28:197-276.

Lynch DR, Lawrence JJ, Lenz S, Anegawa NJ, Ditcher M, Pritchett DB. Pharmacological characterization of heterodimeric NMDA receptors composed of NR 1a and 2B subunits: Differences with receptors formed from NR 1a and 2A. J Neurochem 1995;64:1462-8.

Köhr G. NMDA receptor function: Subunit composition versus spatial distribution. Cell Tissue Res 2006;326:439-46.

Neyton J, Paoletti P. NMDA receptor subunits: Function and pharmacology. Curr Opin Pharmacol 2007;7:39-47.

Bellone C, Zhou Q, Paoletti P. NMDA receptor subunit diversity: Impact on receptor properties, synaptic plasticity and disease. Nat Rev Neurosci 2013;14:383-400.

Bear MF, Kleinschmidt A, Gu QA, Singer W. Disruption of experience-dependent synaptic modifications in striate cortex by infusion of an NMDA receptor antagonist. J Neurosci 1990;10:909-25.

Longdon RB, Sur M, Hahm JO. Disruption of retinogeniculate afferent segregation by antagonists to NMDA receptors. Nature 1991;351:508-10.

Fox K, Schlagger BL, Glazewski S, O’Leary DD. Glutamate receptor blockade at cortical synapses disrupts development of thalamocortical and columnar organization in somatosensory cortex. Proc Natl Acad Sci U S A 1996;93:5584-9.

Simorowski N, Furukawia H, Karakas E. Subunit arrangement and phenylethanolamine binding in GluN1/GluN2B NMDA receptors. Nature 2011;475:249-53.

Buhl DL, Knafels JD, Chanda PK, Green M. Sciabda S, Mong L, et al. A novel binding mode reveals two distinct classes of NMDA receptor GluN2B-selective antagonists. Mol Pharmacol 2016;89:541-51.

Furukawa H, Karakas E. Crystal structure of a heterotetrameric NMDA receptor ion channel. Science (New York NY) 2014;30:992-7.

Lu W, Michel JC, Goehring A, Du J, Song X, Gouaux E, et al. NMDA receptor structures reveal subunit arrangement and pore architecture. Nature 2014;511:191-7.

Barber PA, Buchan AM, Hill MD, Hoyte L. The rise and fall of NMDA antagonists for ischemic stroke. Curr Med Chem 2004;4:131-6.

Manabe T, Irino O, Saitoh K. The mode for the manifestation of the inhibitory effects of ifenprodil tartrate on platelet aggregation in vivo and ex vivo. Nihon Yakurigaku Zasshi 1988;91:105-9.

Ramsden DB, Davies S. Huntington’s disease. Mol Pathol 2001;54:409-13.

Caro M, Hadzimichalis N, Lakhan SE. NMDA receptor activity in neuropsychiatric disorders. Front Psychiatry 2013;10:52.

Sheng M, Zhou Q. NMDA receptors in nervous system diseases. Neuropharmacology 2013;74:69-75.

Mikhaylova M, Ronicke S, Meinhardt J, Schroder UH, Fandrich M, Reiser G, et al. Early neuronal dysfunction by amyloid β oligomers depends on activation of NR2B-containing NMDA receptors. Neurobiol Aging 2011;32:2219-28.

Dannhardt G, Kohl BK. The NMDA receptor complex: A promising target for novel antiepileptic strategies. Curr Med Chem 2001;8:1275-89.

Morris GM, Olson AJ, Goodsell DS. Automated docking of flexible ligands: Applications of AutoDock. J Mol Recogn 1996;9:1-5.

Thiessen PA, Botton EE, Chen J, Fu G, Gindullyte A, Han I, et al. PubChem substance and compound databases. Nucleic Acids Res 2016;44:1202-13.

Banck M, James CA, Morley C, Vandermursch T, Hutchison GR, O’Boyle NM. Open babel: An open chemical toolbox. J Cheminform 2011;3:33.

Huey R, Lindstrom WL, Morris GM, et al. AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. J Comput Chem 2009;3:2785-91.

Olson AJ, Trott O. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010;31:455-61.

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

Michielin O, Zoete V, Daina A. iLOGP: A simple, robust, and efficient description of n-octanol/water partition coefficient for drug design using the GB/SA approach. J Chem Inf Model 2014;54:3284-301.

Zoete V, Daina A. A BOILED-egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem 2016;11:1117-21.

Dominy BW, Feeney PJ, Lipinski CA. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 1997;23:3-25.

Lipinski CA. Lead- and drug-like compounds: The rule-of-five revolution. Drug Discov Today Technol 2004;1:337-41.

Rhode B, Ertl P. Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. J Med Chem 2000;43:3714-7.

Lipinski CA. Drug-like properties and the causes of poor solubility and poor permeability. J Pharmacol Toxicol Methods 2000;44:235-49.

Published

07-05-2022

How to Cite

PRAVEEN, M. “EVALUATION OF ANTAGONIST ACTIVITY OF IFENPRODIL AND THEIR ANALOGOUS AGAINST GLUN1/GLUN2B USING IN SILICO MOLECULAR DOCKING AND ABSORPTION-DISTRIBUTIONMETABOLISM- EXCRETION TOXICITY”. Asian Journal of Pharmaceutical and Clinical Research, vol. 15, no. 5, May 2022, pp. 34-40, doi:10.22159/ajpcr.2022.v15i5.44248.

Issue

Section

Original Article(s)