259
Views
6
CrossRef citations to date
0
Altmetric
Review

Cheminformatics and virtual screening studies of COMT inhibitors as potential Parkinson’s disease therapeutics

, , ORCID Icon, &
Pages 53-62 | Received 12 Mar 2019, Accepted 06 Nov 2019, Published online: 20 Nov 2019

References

  • Axelrod J, Tomchick R. Enzymatic O-methylation of epinephrine and other catechols. J Biol Chem. 1958;233:702–705.
  • Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev. 1975;27:135–206.
  • Mannisto PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective comt inhibitors. Pharmacol Rev. 1999;51:593–628.
  • Lotta T, Vidgren J, Tilgmann C, et al. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme. Biochemistry. 1995;34:4202–4210.
  • Tenhunen J, Salminen M, Lundström K, et al. Genomic organization of the human catechol O-methyltransferase gene and its expression from two distinct promoters. Eur J Biochem. 1994;223:1049–1059.
  • Karhunen T, Tilgmann C, Ulmanen I, et al. Distribution of catechol-O-methyltransferase enzyme in rat tissues. J Histochem Cytochem. 1994;42:1079–1090.
  • Orłowski A, St-Pierre JF, Magarkar A, et al. Properties of the membrane binding component of catechol-o-methyltransferase revealed by atomistic molecular dynamics simulations. J Phys Chem B. 2011;115:13541–13550.
  • Kostic VS. COMT inhibition in the treatment of Parkinson’s disease: neuroprotection and future perspectives. Adv Exp Med Biol. 2004;541:75–90.
  • Bonifacio JM, Palma N, Almeida L, et al. Catechol-O-methyltransferase and its inhibitors in Parkinson’s disease. CNS Drug Rev. 2007;13:352–379.
  • Bakke OM. O-methylation of simple phenols in the rat. Acta Pharmacol Toxicol. 1970;28(1):28–38.
  • Bonifacio MJ, Torrao L, Wright L, et al. Membrane-bound COMT inhibition by nebicapone. Faseb J. 2007;21(804):3.
  • Dingemanse J, Jorga K, Zurcher G, et al. Pharmacokinetic-pharmacodynamic interaction between the COMT inhibitor tolcapone and single-dose levodopa. Br J Pharmacol. 1995;40:253–262.
  • Haasio C. Toxicology and safety of COMT inhibitors. Int Rev Neurobiol. 2010;95:163–189.
  • Lerner C, Ruf A, Gramlich V, et al. X-ray crystal structure of a bisubstrate inhibitor bound to the enzyme catechol-O-methyltransferase: A dramatic effect of inhibitor preorganization on binding affinity. Angew Chem Int Ed Engl. 2001;40:4040–4042.
  • Masjost B, Ballmer P, Borroni E, et al. Structure-based design, synthesis, and in vitro evaluation of bisubstrate inhibitors for catechol O-methyltransferase (COMT). Chemistry. 2000;6:971–982.
  • Vidgren J, Svensson LA, Liljas A. Crystal structure of catechol Omethyltransferase. Nature. 1994;368:354–358.
  • Rutherford K, Le Trong I, Stenkamp RE, et al. Crystal structures of human 108 V and 108 M catechol O-methyltransferase. J Mol Biol. 2008;380:120–130.
  • Bonifacio MJ, Archer M, Rodrigues ML, et al. Kinetics and crystal structure of catechol-O-methyltransferase complex with co-substrate and a novel inhibitor with potential therapeutic application. Mol Pharmacol. 2002;62:795–805.
  • Zheng YJ, Bruice TC. A theoretical examination of the factors controlling the catalytic efficiency of a transmethylation enzyme: catechol O-methyltransferase. J Am Chem Soc. 1997;119:8137–8145.
  • Woodard RW, Tsai MD, Floss HG, et al. Stereochemical course of the transmethylation catalyzed by catechol-O-methyltransferase. J Biol Chem. 1980;255:9124–9127.
  • Vidgren J. X-ray crystallography of catechol O-methyltransferase: perspectives for target-based drug development. Adv Pharmacol. 1997;42:238–331.
  • Jeffery DR, Roth JA. Kinetic reaction mechanism for magnesium binding to membrane-bound and soluble catechol-O-methyltransferase. Biochemistry. 1987;26:2955–2958.
  • Lau EY, Bruice TC. Importance of correlated motions in forming highly reactive near attack conformations in catechol-O-methyltransferase. J Am Chem Soc. 1998;120:12387–12394.
  • Kuhn B, Kollman PA. QM-FE and molecular dynamics calculations on catechol-O-methyltransferase: free energy of activation in the enzyme and in aqueous solution and regioselectivity of the enzyme-catalyzed reaction. J Am Chem Soc. 2000;122:2586–2596.
  • Lautala P, Ulmanen I, Taskinen J. Molecular mechanisms controlling the rate and specificity of catechol-O-methylation by human soluble catechol-O-methyltransferase. Mol Pharmacol. 2001;59:393–402.
  • Chen D, Wang CY, Lambert DJ, et al. Inhibition of human liver catechol-O-methyltransferase by tea catechins and their metabolites: structure–activity relationship and molecular-modeling studies. Biochem Pharmacol. 2005;69:1523–1531.
  • Azam F, Mohamed N, Alhussen F. Molecular interaction studies of green tea catechins as multitarget drug candidates for the treatment of Parkinson’s disease: computational and structural insights. Network Comput Neural Syst. 2015;26:97–115.
  • Cao Y, Chen ZJ, Jiang HD, et al. computational studies of the regioselectivities of comt-catalyzed meta-/para-O methylations of luteolin and quercetin. J Phys Chem B. 2014;118:470−481.
  • Zhu BT, Ezell EL, Liehr JG. Catechol-O- methyltransferase-catalyzed rapid O-methylation of mutagenic flavonoids. Metabolic inactivation as a possible reason for their lack of carcinogenicity in vivo. J. Biol. Chem. 1994;269:292−299.
  • Nagappan P, Krishnamurthy V. structural prediction and comparative molecular docking studies of hesperidin and l-dopa on α-synuclein, MAO-B, COMT and UCHL-1 inhibitors. Int J Pharm Clin Res. 2015;7:221–225.
  • Monteiro A, Viana J, Nayarisseri A, et al. computational studies applied to flavonoids against Alzheimer’s and Parkinson’s diseases. Hindawi Oxid Med Cell Longevity. 2018;2:1–21.
  • Palma PN, Bonifacio MJ, Loureiro AI, et al. Molecular modeling and metabolic studies of the interaction of catechol-O-methyltransferase and a new nitrocatechol inhibitor. Drug Metab Dispos. 2003;31:250–258.
  • Soares-da-Silva P, Vieira-Coelho MA, Parada A. Catechol-O-methyltransferase inhibition in erythrocytes and liver by BIA 3–202 (1-[3,4-dibydroxy-5-nitrophenyl]-2-phenyl-ethanone). Pharmacol Toxicol. 2003;92:272–278.
  • Palma PN, Rodrigues ML, Archer M, et al. Comparative study of ortho- and meta-nitrated inhibitors of catechol-O-methyltransferase: interactions with the active site and regioselectivity of O-methylation. Mol Pharmacol. 2006;70:143–153.
  • Learmonth DA, Palma PN, Vieira-Coelho MA, et al. Synthesis, biological evaluation, and molecular modeling studies of a novel, peripherally selective inhibitor of catechol-O-methyltransferase. J Med Chem. 2004;47:6207–6217.
  • Lee JY, Baek S, Kim Y. Receptor-oriented pharmacophore-based in silico screening of human catechol O-methyltransferase for the design of antiparkinsonian drug. B Korean Chem Soc. 2007;28:379–385.
  • Paulini R, Lerner C, Jakob-Roetne R, et al. Bisubstrate inhibitors of the enzyme catechol O-methyltransferase (COMT): efficient inhibition despite the lack of a nitro group. Chembiochem. 2004;5:1270–1274.
  • Smith KS, Smith PL, Heady TN, et al. In vitro metabolism of tolcapone to reactive intermediates: relevance to tolcapone liver toxicity. Chem Res Toxicol. 2003;16:123–128.
  • Ellermann M, Jakob-Roetne R, Lerner C, et al. Molecular recognition at the active site of catechol-O-methyltransferase: energetically favorable replacement of a water molecule imported by a bisubstrate inhibitor. Angew Chem Int Ed Engl. 2009;48:9092–9096.
  • Ellermann M, Paulini R, Jakob-Roetne R, et al. Molecular recognition at the active site of catechol-O-methyltransferase (COMT): adenine replacements in bisubstrate inhibitors. Chemistry. 2011;17:6369–6381.
  • Mazumber KM, Bhaltacharjee N, Borah A. Garcinol prevents hyperhomocysteinemia and enhances bioavailability of L-DOPA by inhibiting catechol-O-methyltransferase: an in silico approach. Med Chem Res. 2016;25:116–122.
  • Masullo M, Bassarello C, Suzuki H, et al. Polyisoprenylated benzophenones and an unusual polyisopreny- lated tetracyclic xanthone from the fruits of Garcinia cambogia. J Agric Food Chem. 2008;56:5205–5210.
  • Hong J, Sang S, Park HJ, et al. Modulation of arachidonic acid metabolism and nitric oxide synthesis by garcinol and its derivatives. Carcinogenesis. 2006;27:278–286.
  • Liao CH, Ho CT. Lin JK effects of garcinol on free radical generation and NO production in embryonic rat cortical neurons and astrocytes. Biochem Biophys Res Commun. 2005;329:1306–1314.
  • Deba S, Phukana BC, Mazumder KM, et al. Garcinol, a multifaceted sword for the treatment of Parkinson’s disease. Neurochem Int. 2019;128:50–57.
  • Troen AM. The central nervous system in animal models of hyperhomocysteinemia. Prog Neuropsychopharmacol Biol Psychiatry. 2005;29:1140–1151.
  • Tsuji E, Okazaki K, Takeda K. Crystal structures of rat catechol-O-methyltransferase complexed with coumarine-based inhibitor. Biochem Biophys Res Commun. 2009;378:494–497.
  • Kiss LE, Ferreira HS, Torrao L, et al. Discovery of a long-acting, peripherally selective inhibitor of catechol-O-methyltransferase. J Med Chem. 2010;53:3396–3411.
  • Jatana N, Sharma A, Latha N. Pharmacophore modeling and virtual screening studies to design potential COMT inhibitors as new leads. J Mol Graph Model. 2013;39:145–164.
  • Palma PN, Bonifacio MJ, Loureiro AI, et al. Computation of the binding affinities of catechol-O-methyltransferase inhibitors: multisubstate relative free energy calculations. J Comput Chem. 2012;33:970–986.
  • Magarkar A, Parkkila P, Viitala T, et al. Membrane bound COMT isoform is an interfacial enzyme: general mechanism, new drug design paradigm. Chem Comm. 2018;54:3440–3443.
  • Savelieff MG, Nam G, Kang J, et al. Development of multifunctional molecules as potential therapeutic candidates for Alzheimer’s disease, Parkinson’s disease, and amyotrophic lateral sclerosis in the last decade. Chem Rev. 2019;119:1221–1332.
  • Aarthy M, Panwar U, Selvaraj C, et al. Advantages of structure-based drug design approaches in neurological disorders. Curr Neuropharmacol. 2017;15:1136–1155.
  • Patel N, Georrge J, Modi K, et al. Pharmacophore-based virtual screening of catechol-o-methyltransferase (COMT) inhibitors to combat Alzheimer’s disease. J Biomol Struct Dyn. 2017;36:3938–3957.
  • Fredriksson A, Archer T. Synergetic interactions between COMT-/MAO- inhibitors and L-Dopa in MPTP-treated mice. J Neural Transm Gen. 1995;102:19–34.
  • Lyytinen J, Kaakkola S, Ahtila S, et al. Simultaneous MAO-B and COMT inhibition in L-DOPA- treated patients with Parkinson’s disease. Mov Disord. 1997;12:497–505.
  • Talati R, Reinhart K, Baker W, et al. Pharmacological treatment of advanced Parkinson’s desease: A meta-analysis of COMT inhibitors and MAO-B inhibitors. Parkinsonism Related Dicord. 2009;15:500–505.
  • Khromova I, Voronina T, Kraineva VA, et al. Effects of selective catechol-O-methyltransferase inhibitors on single-trial passive avoidance retention in male rats. Behav Brain Res. 1997;86:49–57.
  • Moreau JL, Borgulya J, Jenck F, et al. Tolcapone: a potential new antidepressant detected in a novel animal model of depression. Behav Pharmacol. 1994;5:344–350.
  • Gasparini M, Fabrizio E, Bonifati V, et al. Cognitive improvement during tolcapone treatment in parkinson’s disease. J Neural Transm. 1997;104:887–894.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.