Interactions of endocannabinoid virodhamine and related analogs with human monoamine oxidase-A and -B

Biochem Pharmacol. 2018 Sep:155:82-91. doi: 10.1016/j.bcp.2018.06.024. Epub 2018 Jun 26.

Abstract

The endocannabinoid system plays an important role in the pathophysiology of various neurological disorders, such as anxiety, depression, neurodegenerative diseases, and schizophrenia; however, little information is available on the coupling of the endocannabinoid system with the monoaminergic systems in the brain. In the present study, we tested four endocannabinoids and two anandamide analogs for inhibition of recombinant human MAO-A and -B (monoamine oxidase). Virodhamine inhibited both MAO-A and -B (IC50 values of 38.70 and 0.71 μM, respectively) with ∼55-fold greater inhibition of MAO-B. Two other endocannabinoids (noladin ether and anandamide) also showed good inhibition of MAO-B with IC50 values of 18.18 and 39.98 μM, respectively. Virodhamine was further evaluated for kinetic characteristics and mechanism of inhibition of human MAO-B. Virodhamine inhibited MAO-B (Ki value of 0.258 ± 0.037 μM) through a mixed mechanism/irreversible binding and showed a time-dependent irreversible mechanism. Treatment of Neuroscreen-1 (NS-1) cells with virodhamine produced significant inhibition of MAO activity. This observation confirms potential uptake of virodhamine by neuronal cells. A molecular modeling study of virodhamine with MAO-B and its cofactor flavin adenine dinucleotide (FAD) predicted virodhamine's terminal -NH2 group to be positioned near the N5 position of FAD, but for docking to MAO-A, virodhamine's terminal -NH2 group was far away (∼6.52 Å) from the N5 position of FAD, and encountered bad contacts with nearby water molecules. This difference could explain virodhamine's higher potency and preference for MAO-B. The binding free energies for the computationally-predicted poses also showed that virodhamine was selective for MAO-B. These findings suggest potential therapeutic applications of virodhamine for the treatment of neurological disorders.

Keywords: (R)-(+)-Methanandamide (PubChem CID: 6321351); 2-Arachidonoylglycerol (PubChem CID: 5282280); 4-Hydroxyquinoline (PubChem CID: 69141); Anandamide (PubChem CID: 5281969); Clorgyline (PubChem CID: 28767); Dimethyl sulfoxide (PubChem CID: 679); Docking; Endocannabinoids; Enzyme kinetics; Kynuramine dihydrobromide (PubChem CID: 16219543); MAO-B; Noladin ether (PubChem CID: 6483057); Oleylethanolamide (PubChem CID: 5283454); Phenelzine sulfate (PubChem CID: 61100); Potassium phosphate (PubChem CID: 516951); R-(−)-Deprenyl hydrochloride (PubChem CID: 5195); Safinamide (PubChem CID: 131682); Virodhamine; Virodhamine trifluoroacetate (PubChem CID: 91691131).

Publication types

  • Research Support, N.I.H., Extramural
  • Research Support, U.S. Gov't, Non-P.H.S.

MeSH terms

  • Animals
  • Cannabinoid Receptor Modulators / chemistry
  • Cannabinoid Receptor Modulators / metabolism*
  • Cannabinoid Receptor Modulators / pharmacology
  • Cannabinoids / chemistry
  • Cannabinoids / metabolism*
  • Cannabinoids / pharmacology
  • Endocannabinoids / chemistry
  • Endocannabinoids / metabolism*
  • Endocannabinoids / pharmacology
  • Humans
  • Molecular Docking Simulation / methods
  • Monoamine Oxidase / metabolism*
  • Monoamine Oxidase Inhibitors / chemistry
  • Monoamine Oxidase Inhibitors / metabolism*
  • Monoamine Oxidase Inhibitors / pharmacology
  • PC12 Cells
  • Rats

Substances

  • Cannabinoid Receptor Modulators
  • Cannabinoids
  • Endocannabinoids
  • Monoamine Oxidase Inhibitors
  • virodhamine
  • Monoamine Oxidase
  • monoamine oxidase A, human