Skip to main content

Advertisement

Log in

Predicting targets of compounds against neurological diseases using cheminformatic methodology

  • Published:
Journal of Computer-Aided Molecular Design Aims and scope Submit manuscript

Abstract

Recently developed multi-targeted ligands are novel drug candidates able to interact with monoamine oxidase A and B; acetylcholinesterase and butyrylcholinesterase; or with histamine N-methyltransferase and histamine H3-receptor (H3R). These proteins are drug targets in the treatment of depression, Alzheimer’s disease, obsessive disorders, and Parkinson’s disease. A probabilistic method, the Parzen–Rosenblatt window approach, was used to build a “predictor” model using data collected from the ChEMBL database. The model can be used to predict both the primary pharmaceutical target and off-targets of a compound based on its structure. Molecular structures were represented based on the circular fingerprint methodology. The same approach was used to build a “predictor” model from the DrugBank dataset to determine the main pharmacological groups of the compound. The study of off-target interactions is now recognised as crucial to the understanding of both drug action and toxicology. Primary pharmaceutical targets and off-targets for the novel multi-target ligands were examined by use of the developed cheminformatic method. Several multi-target ligands were selected for further study, as compounds with possible additional beneficial pharmacological activities. The cheminformatic targets identifications were in agreement with four 3D-QSAR (H3R/D1R/D2R/5-HT2aR) models and by in vitro assays for serotonin 5-HT1a and 5-HT2a receptor binding of the most promising ligand (71/MBA-VEG8).

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8

Similar content being viewed by others

Abbreviations

AD:

Alzheimer’s disease

AChE:

Acetylcholinesterase

BuChE:

Butyrylcholinesterase

CFP:

Circular fingerprint

3D-QSAR:

3D-Quantitive structure–activity relationship

EDTA:

Ethylenediaminetetraacetic acid

FP:

False positive

GSK-3:

Glycogen synthase kinase 3

HMT:

Histamine N-methyltransferase

H3R:

Histamine H3-receptor

5-HT1a :

5-Hydroxytryptamine-1a (serotonin)

5-HT2a :

5-Hydroxytryptamine-2a (serotonin)

5-HT2c :

5-Hydroxytryptamine-2c (serotonin)

MAO-A:

Monoamine oxidase A

MAO-B:

Monoamine oxidase B

MCC:

Matthews correlation coefficient

MTDL:

Multi-target-directed ligand

NMDA receptors:

N-Methyl-d-aspartate receptor

nAChRs:

Nicotinic acetylcholine receptors

8-OH-DPAT:

(±)-8-Hydroxy-2-dipropylaminotetralin

PDE-4:

Phosphodiesterase 4

PD:

Parkinson’s disease

RMSEE:

Root main square error of estimation

RMSEP:

Root main square error of prediction

SERT:

Serotonin transporter

TP:

True positive

Tris:

Tris(hydroxymethyl)aminomethane

WADA:

World Anti-Doping Agency

References

  1. Goedert M, Spillantini MGA (2006) A century of Alzheimer’s disease. Science 314:777–781

    Article  CAS  Google Scholar 

  2. Humbert-Claude M, Morisset S, Gbahou F, Arrang JM (2007) Histamine H3 and dopamine D2 receptor-mediated [35S]GTPγ[S] binding in rat striatum: evidence for additive effects but lack of interactions. Biochem Pharmacol 73:1172–1181

    Article  CAS  Google Scholar 

  3. Garduno-Torres B, Trevino M, Gutierrez R, Arias-Montano JA (2007) Presynaptic histamine H3 receptors regulate glutamate, but not GABA release in rat thalamus. Neuropharmacology 52:527–535

    Article  CAS  Google Scholar 

  4. Dai H, Fu Q, Shen Y, Hu W, Zhang Z, Timmerman H, Leurs R, Chen Z (2007) The histamine H3 receptor antagonist clobenpropit enhances GABA release to protect against NMDA induced excitotoxicity through the cAMP/protein kinase A pathway in cultured cortical neurons. Eur J Pharmacol 563:117–123

    Article  CAS  Google Scholar 

  5. Threlfell S, Cragg SJ, Imre K, Turi GF, Coen CW, Greenfield SA (2004) Histamine H3 receptors inhibit serotonin release in substantia nigra pars reticulata. J Neurosci 24:8704–8710

    Article  CAS  Google Scholar 

  6. Gemkow MJ, Davenport AJ, Harich S, Ellenbroek BA, Cesura A, Hallett D (2009) The histamine H3 receptor as a therapeutic drug target for CNS disorders. Drug Discov Today 14:509–515

    Article  CAS  Google Scholar 

  7. León R, Garcia AG, Marco-Contelles J (2013) Recent advances in the multitarget-directed ligands approach for the treatment of Alzheimer’s disease. Med Res Rev 33:139–189

    Article  Google Scholar 

  8. Millan MJ (2014) On ‘polypharmacy’ and multi-target agents, complementary strategies for improving the treatment of depression: a comparative appraisal. Int J Neuropsychopharmacol 17:1009–1037

    Article  CAS  Google Scholar 

  9. Anighoro A, Bajorath J, Rastelli G (2014) Polypharmacology: challenges and opportunities in drug discovery. J Med Chem 57:7874–7887

    Article  CAS  Google Scholar 

  10. Morphy R, Rankovic Z (2005) Designed multiple ligands. An emerging drug discovery paradigm. J Med Chem 48:6523–6543

    Article  CAS  Google Scholar 

  11. Roth BL, Sheffler DJ, Kroeze WK (2004) Magic shotguns versus magic bullets: selectively non-selective drugs for mood disorders and schizophrenia. Nat Rev Drug Discov 3:353–359

    Article  CAS  Google Scholar 

  12. Lipina TV, Palomo V, Gil C, Martinez A, Roder JC (2013) Dual inhibitor of PDE7 and GSK-3-VP1.15 acts as antipsychotic and cognitive enhancer in C57BL/6J mice. Neuropharmacology 64:205–214

    Article  CAS  Google Scholar 

  13. Lipina TV, Wang M, Liu F, Roder JC (2012) Synergistic interactions between PDE4B and GSK-3: DISC1 mutant mice. Neuropharmacology 62:1252–1262

    Article  CAS  Google Scholar 

  14. Millan MJ (2006) Multi-target strategies for the improved treatment of depressive states: conceptual foundation and neuronal substrates, drug discovery and therapeutic application. Pharmacol Ther 110:135–370

    Article  CAS  Google Scholar 

  15. Meltzer HY, Massey BW, Horiguchi M (2012) Serotonin receptors as targets for drugs useful to treat psychosis and cognitive impairment in schizophrenia. Curr Pharm Biotechnol 13:1572–1586

    Article  CAS  Google Scholar 

  16. Quesseveur G, Nguyen HT, Gardier AM, Guiard BP (2012) 5-HT2 ligands in the treatment of anxiety and depression. Expert Opin Investig Drugs 21:1701–1725

    Article  CAS  Google Scholar 

  17. Youdim MBH, Buccafusco JJ (2005) Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders. Trends Pharmacol Sci 26:27–35

    Article  CAS  Google Scholar 

  18. Tahtouh T, Elkins JM, Filippakopoulos P, Soundararajan M, Burgy G, Durieu E, Cochet C, Schmid RS, Lo DC, Delhommel F, Oberholzer AE, Pearl LH, Carreaux F, Bazureau JP, Knapp S, Meijer L (2012) Selectivity, cocrystal structures, and neuroprotective properties of leucettines, a family of protein kinase inhibitors derived from the marine sponge alkaloid leucettamine B. J Med Chem 55:9312–9330

    Article  CAS  Google Scholar 

  19. Rosini M, Antonello A, Cavalli A, Bolognesi ML, Minarini A, Marucci G, Poggesi E, Leonardi A, Melchiorre C (2003) Prazosin-related compounds. Effect of transforming the piperazinylquinazoline moiety into an aminomethyltetrahydroacridine system on the affinity for α1-adrenoreceptors. J Med Chem 46:4895–4903

    Article  CAS  Google Scholar 

  20. Fang L, Appenroth D, Decker M, Kiehntopf M, Roegler C, Deufel T, Fleck C, Peng S, Zhang Y, Lehmann J (2008) Synthesis and biological evaluation of NO-donor-tacrine hybrids as hepatoprotective anti-Alzheimer drug candidates. J Med Chem 51:713–716

    Article  CAS  Google Scholar 

  21. Stosel A, Schlenk M, Hinz S, Kuppers P, Heer J, Gutschow M, Muller CE (2013) Dual targeting of adenosine A2A receptors and monoamine oxidase B by 4H-3,1-benzothiazin-4-ones. J Med Chem 56:4580–4596

    Article  Google Scholar 

  22. Fang L, Kraus B, Lehmann J, Heilmann J, Zhang Y, Decker M (2008) Design and synthesis of tacrine–ferulic acid hybrids as multi-potent anti-Alzheimer drug candidates. Bioorg Med Lett 18:2905–2909

    Article  CAS  Google Scholar 

  23. Jiménez JJ, Mendes E, Galdeano C, Martins C, Silva DB, Marco-Contelles J, Carmo Carreiras M, Luque FJ, Ramsay RR (2014) Exploring the structural basis of the selective inhibition of monoamine oxidase A by dicarbonitrile aminoheterocycles: role of Asn181 and Ile335 validated by spectroscopic and computational studies. Biochim Biophys Acta 1844:389–397

    Article  Google Scholar 

  24. Bautista-Aguileraa OM, Estebanb G, Bolea I, Nikolic K, Agbaba D, Moraleda I, Iriepa I, Samadi A, Soriano E, Unzeta M, Marco-Contelles J (2014) Design, synthesis, pharmacological evaluation, QSAR analysis, molecular modeling and ADMET of novel donepezil–indolyl hybrids as multipotent cholinesterase/monoamine oxidase inhibitors for the potential treatment of Alzheimer’s disease. Eur J Med Chem 75:82–95

    Article  Google Scholar 

  25. Samadi A, Chioua M, Bolea I, de los Ríos C, Iriepa I, Moraleda I, Bastida A, Esteban G, Unzeta M, Gálvez E, Marco-Contelles J (2011) Synthesis, biological assessment and molecular modeling of new multipotent MAO and cholinesterase inhibitors as potential drugs for the treatment of Alzheimer’s disease. Eur J Med Chem 46:4665–4668

    Article  CAS  Google Scholar 

  26. Bolea I, Juárez-Jiménez J, Ríos C, Chioua M, Pouplana R, Javier Luque F, Unzeta M, Marco-Contelles J, Samadi A (2011) Synthesis, biological evaluation, and molecular modeling of donepezil and N-[(5-(benzyloxy)-1-methyl-1H-indol-2-yl)methyl]-N-methylprop-2-yn-1-amine hybrids as new multipotent cholinesterase/monoamine oxidase inhibitors for the treatment of Alzheimer’s disease. J Med Chem 54:8251–8270

    Article  CAS  Google Scholar 

  27. Pérez V, Marco-Contelles J, Fernández-Álvarez E, Unzeta M (1999) Relevance of benzyloxy group in 2-indolyl methylamines in the selective MAO-B inhibition. Br J Pharmacol 127:869–876

    Article  Google Scholar 

  28. Marco-Contelles J, Leon R, Rıos C, Guglietta A, Terencio J, Lopez MG, Garcıa AG, Villarroya M (2006) Novel multipotent tacrine–dihydropyridine hybrids with improved acetylcholinesterase inhibitory and neuroprotective activities as potential drugs for the treatment of Alzheimer’s disease. J Med Chem 49(26):7607–7610

  29. Marco-Contelles J, Leon R, Rıos C, Samadi A, Bartolini M, Andrisano V, Huertas O, Barril X, Luque FJ, Rodrıguez-Franco MI, Lopez B, Lopez MG, Garcıa AG, Carmo Carreiras M, Villarroya M (2009) Tacripyrines, the first tacrine–dihydropyridine hybrids, as multitarget-directed ligands for the treatment of Alzheimer’s disease. J Med Chem 52:2724–2732

    Article  CAS  Google Scholar 

  30. Apelt J, Ligneau X, Pertz H, Arrang JM, Ganellin CR, Schwartz JC, Schunack W, Holger S (2002) Development of a new class of nonimidazole histamine H(3) receptor ligands with combined inhibitory histamine N-methyltransferase activity. J Med Chem 45:1128–1141

    Article  CAS  Google Scholar 

  31. Petroianu G, Arafat K, Sasse BC, Stark H (2006) Multiple enzyme inhibitions by histamine H3 receptor antagonists as potential procognitive agents. Pharmazie 61:179–182

    CAS  Google Scholar 

  32. Grassmann S, Apelt J, Sippl W, Ligneau X, Pertz HH, Zhao YH, Arrang JM, Ganellin CR, Schwartz JC, Schunack W, Stark H (2003) Imidazole derivatives as a novel class of hybrid compounds with inhibitory histamine N-methyltransferase potencies and histamine H3 receptor affinities. Bioorg Med Chem 11:2163–2174

    Article  CAS  Google Scholar 

  33. Grassmann S, Apelt J, Ligneau X, Pertz HH, Arrang JM, Schwartz JC, Schunack W, Stark H (2004) Search for histamine H(3) receptor ligands with combined inhibitory potency at histamine N-methyltransferase: omega-piperidinoalkanamine derivatives. Arch Pharm Med Chem 337:533–545

    Article  CAS  Google Scholar 

  34. Apelt J, Grassmann S, Ligneau X, Pertz HH, Ganellin CR, Arrang JM, Schwartz JC, Schunack W, Stark H (2005) Search for histamine H3 receptor antagonists with combined inhibitory potency at Ntau-methyltransferase: ether derivatives. Pharmazie 60:97–106

    CAS  Google Scholar 

  35. Ligneau X, Lin JS, Vanni-Mercier G, Jouvet M, Muir JL, Ganellin CR, Stark H, Elz S, Schunack W, Schwartz JC (1998) Neurochemical and behavioural effects of ciproxifan, a potent histamine H3-receptor antagonist. J Pharmacol Exp Ther 287:658–666

    CAS  Google Scholar 

  36. Sander K, Kottke T, Stark H (2008) Histamine H3 receptor antagonists go to clinics. Biol Pharm Bull 31:2163–2181

    Article  CAS  Google Scholar 

  37. Esbenshade TA, Browman KE, Bitner RS, Strakhova M, Cowart MD, Brioni JD (2008) The histamine H3 receptor: an attractive target for the treatment of cognitive disorders. Br J Pharmacol 154:1166–1181

    Article  CAS  Google Scholar 

  38. Di Giovanni G, Di Matteo V, Pierucci M, Esposito E (2008) Serotonin–dopamine interaction: electrophysiological evidence. Prog Brain Res 172:45–71

    Article  Google Scholar 

  39. Di Matteo V, Di Giovanni G, Pierucci M, Esposito E (2008) Serotonin control of central dopaminergic function: focus on in vivo microdialysis studies. Prog Brain Res 172:7–44

    Article  Google Scholar 

  40. Carlsson M, Carlsson A (1990) Interactions between glutamatergic and monoaminergic systems within the basal ganglia—implications for schizophrenia and Parkinson ‘s disease. Trends Neurosci 13:272–276

    Article  CAS  Google Scholar 

  41. Millan MJ (2005) N-Methyl-d-aspartate receptors as a target for improved antipsychotic agents: novel insights and clinical perspectives. Psychopharmacology 179:30–53

    Article  CAS  Google Scholar 

  42. Rogawski MA, Wenk GL (2003) The neuropharmacological basis for the use of memantine in the treatment of Alzheimer’s disease. CNS Drug Rev 9:275–308

    Article  CAS  Google Scholar 

  43. Cummings JL, Morstorf T, Zhong K (2014) Alzheimer’s disease drug-development pipeline: few candidates, frequent failures. Alzheimers Res Ther 6:37

    Article  Google Scholar 

  44. Zheng H, Youdim MBH, Fridkin M (2009) Site-activated multifunctional chelator with acetylcholinesterase and neuroprotective-neurorestorative moieties for Alzheimer’s therapy. J Med Chem 52:4095–4098

  45. Cavalli A, Bolognesi ML, Minarini M, Rosini V, Tumiatti M, Recanatini C, Melchiorre C (2008) Multi-target-directed ligands to combat neurodegenerative diseases. J Med Chem 51:347–372

    Article  CAS  Google Scholar 

  46. Cannon EO, Bender A, Palmer DS, Mitchell JBO (2006) Chemoinformatics-based classification of prohibited substances employed for doping in sport. J Chem Inf Model 5:2369–2380

    Article  Google Scholar 

  47. Cannon EO, Mitchell JBO (2006) Classifying the World Anti-Doping Agency’s 2005 prohibited list using the chemistry development kit fingerprint. Lect Notes Bioinform 5:173–182

    Google Scholar 

  48. Cannon EO, Nigsch F, Mitchell JBO (2008) Novel hybrid ultrafast shape descriptor method for use in virtual screening. Chem Central J 5:3

    Article  Google Scholar 

  49. Paolini VG, Shapland RHB, Van Hoorn WP, Mason JS, Hopkins AL (2006) Global mapping of pharmacological space. Nat Biotechnol 5:805–815

    Article  Google Scholar 

  50. Bender A, Scheiber J, Glick M, Davies JW, Azzaoui K, Hamon J, Urban L, Whitebread S, Jenkins JL (2007) Analysis of pharmacology data and the prediction of adverse drug reactions and off-target effects from chemical structure. Chem Med Chem 5:861–873

    Article  Google Scholar 

  51. Nigsch F, Mitchell JBO (2008) Toxicological relationships between proteins obtained from protein target predictions of large toxicity databases. Toxicol Appl Pharmacol 5:225–234

    Article  Google Scholar 

  52. Nigsch F, Bender A, Jenkins JL, Mitchell JBO (2008) Ligand-target prediction using winnow and naive bayesian algorithms and the implications of overall performance statistics. J Chem Inf Model 5:2313–2325

    Article  Google Scholar 

  53. Keiser MJ, Setola V, Irwin JJ, Laggner C, Abbas AI, Hufeisen SJ, Jensen NH, Kuijer MB, Matos RC, Tran TB, Whaley R, Glennon RA, Hert J, Thomas KLH, Edwards DD, Shoichet BK, Roth BL (2009) Predicting new molecular targets for known drugs. Nature 5:175–181

    Article  Google Scholar 

  54. Niijima S, Yabuuchi H, Okuno Y (2011) Cross-target view to feature selection: identification of molecular interaction features in ligand-target space. J Chem Inf Model 5:15–24

    Article  Google Scholar 

  55. Lounkine E, Keiser MJ, Whitebread S, Mikhailov D, Hamon J, Jenkins JL, Lavan P, Weber E, Doak AK, Cote S, Shoichet BK, Urban L (2012) Large-scale prediction and testing of drug activity on side-effect targets. Nature 5:361–367

    Google Scholar 

  56. Perez-Nueno VI, Venkatraman V, Mavridis L, Ritchie DW (2012) Detecting drug promiscuity using gaussian ensemble screening. J Chem Inf Model 5:1948–1961

    Article  Google Scholar 

  57. Glen RC, Bender A, Arnby CH, Carlsson L, Boyer S, Smith J (2006) Circular fingerprints: flexible molecular descriptors with applications from physical chemistry to ADME. IDrugs 9:199–204

    CAS  Google Scholar 

  58. Mavridis L, Mitchell JBO (2013) Predicting the protein targets for athletic performance-enhancing substances. J Cheminform 5:31

    Article  CAS  Google Scholar 

  59. Gaulton A, Bellis LJ, Bento PA, Chambers J, Davies M, Hersey A, Light Y, McGlinchey S, Michalovich D, Al-Lazikani B, Overington JP (2012) ChEMBL: a large-scale bioactivity database for drug discovery. Nucl Acids Res 5:D1100–D1107

    Article  Google Scholar 

  60. Knox C, Law V, Jewison T, Liu P, Ly S, Frolkis A, Pon A, Banco K, Mak C, Neveu V, Djoumbou Y, Eisner R, Guo AC, Whishart DS (2011) DrugBank 3.0: a comprehensive resource for ‘omics’ research on drugs. Nucl Acids Res 5:D1035–D1041

    Article  Google Scholar 

  61. Mavridis L, Nath N, Mitchell JBO (2013) PFClust: a novel parameter free clustering algorithm. BMC Bioinform 14:213

    Article  Google Scholar 

  62. Rogers DJ, Tanimoto TT (1960) A computer program for classifying plants. Science 5:1115–1118

    Article  Google Scholar 

  63. Parzen E (1962) On estimation of a probability density function and mode. Ann Math Statist 5:1065–1076

    Article  Google Scholar 

  64. Rosenblatt M (1956) Remarks on some nonparametric estimates of a density function. Ann Math Statist 5:832–837

    Article  Google Scholar 

  65. Matthews BW (1975) Comparison of the predicted and observed secondary structure of T4 phage lysozyme. Biochim Biophys Acta 5:442–451

    Article  Google Scholar 

  66. Hamacher A, Weigt M, Wiese M, Hoefgen B, Lehmann J, Kassack MU (2006) Dibenzazecine compounds with a novel dopamine/5HT2A receptor profile and 3D-QSAR analysis. BMC Pharmacol 6:11

    Article  Google Scholar 

  67. ChemAxon MarvinSketch 5.5.1.0 program (2011) Budapest, Hungary. www.chemaxon.com/products.html

  68. Froese Fischer CF (1977) The Hartree–Fock method for atoms: a numerical approach. Wiley, New York

    Google Scholar 

  69. Frisch MJ et al (1998) Gaussian 98 (Revision A.7). Gaussian Inc., Pittsburgh

  70. Pentacle, Version 1.0.6. (2009) Molecular Discovery Ltd., Perugia, Italy. http://www.moldiscovery.com/soft_pentacle.php

  71. Eriksson L, Johansson E, Kettaneh-Wold N, Trygg J, Wikstrom C, Wold S (eds) (2001) Multi-and megavariate data analysis. Basic principles and applications I, 2nd edn. Umetrics Academy, Umeå

    Google Scholar 

  72. Tropsha A (2010) Best practices for QSAR model development, validation, and exploitation. Mol Inf 29:476–488

    Article  CAS  Google Scholar 

  73. Cheng YC, Prusoff WH (1973) Relationship between the inhibition constant (Ki) and the concentration of inhibitor which causes 50 per cent inhibition (IC50) of an enzymatic reaction. Biochem Pharmacol 22:3099–3108

    Article  CAS  Google Scholar 

  74. Scatchard G (1949) The attraction of proteins for small molecules and ions. Ann NY Acad Sci 51:660–672

    Article  CAS  Google Scholar 

  75. Schlegel JR, Peroutka SJ (1986) Nucleotide interactions with 5-HT1A binding sites directly labeled by [3H]-8-hydroxy-2-(di-n-propylamino)tetralin ([3H]-8-OH-DPAT). Biochem Pharmacol 35:1943–1949

    Article  CAS  Google Scholar 

  76. Herndon JL, Ismaiel A, Ingher SP, Teitler M, Glennon RA (1992) Ketanserin analogues: structure–affinity relationships for 5-HT2 and 5-HT1C serotonin receptor binding. J Med Chem 35:4903–4910

    Article  CAS  Google Scholar 

  77. Buhot MC, Martin S, Segu L (2000) Role of serotonin in memory impairment. Ann Med 32:210–221

    Article  CAS  Google Scholar 

  78. Roth BL, Hanizavareh SM, Blum AE (2004) Serotonin receptors represent highly favorable molecular targets for cognitive enhancement in schizophrenia and other disorders. Psychopharmacology 174:17–24

    Article  CAS  Google Scholar 

  79. Gray JA, Roth BL (2007) The pipeline and future of drug development in schizophrenia. Mol Psychiatry 12:904–922

    Article  CAS  Google Scholar 

  80. Nakamura M, Ogasa M, Guarino J, Phillips D, Severs J, Cucchiaro J, Loebel A (2009) Lurasidone in the treatment of acute schizophrenia: a double-blind, placebo-controlled trial. J Clin Psychiatry 70:829–836

    Article  CAS  Google Scholar 

  81. Patat A, Parks V, Raje S, Plotka A, Dietrich B (2005) Age–gender study of SRA-333, a novel 5-HT1A antagonist. Clin Pharmacol Ther 77:P29

    Article  Google Scholar 

  82. Pitsikas N, Tsitsirigou S, Zisopoulou S, Sakellaridis N (2005) The 5-HT1A receptor and recognition memory. Possible modulation of its behavioral effects by the nitrergic system. Behav Brain Res 159:287–293

    Article  CAS  Google Scholar 

  83. Schechter LE, Smith DL, Rosenzweig-Lipson S, Sukoff SJ, Dawson LA, Marquis K, Jones D, Piesla M, Andree T, Nawoschik S, Harder JA, Womack MD, Buccafusco J, Terry AV, Hoebel B, Rada P, Kelly M, Abou-Gharbia M, Barrett JE, Childers W (2005) Lecozotan (SRA-333): a selective serotonin 1A receptor antagonist that enhances the stimulated release of glutamate and acetylcholine in the hippocampus and possesses cognitive-enhancing properties. J Pharmacol Exp Ther 314:1274–1289

    Article  CAS  Google Scholar 

  84. Ballanger B, Klinger H, Eche J, Lerond J, Vallet AE, Le Bars D, Tremblay L, Sgambato-Faure V, Broussolle E, Thobois S (2012) Role of serotonergic 1A receptor dysfunction in depression associated with Parkinson’s disease. Mov Disord 27:84–89

    Article  CAS  Google Scholar 

  85. Wallace TL, Ballard TM, Pouzet B, Riedel WJ, Wettstein JG (2011) Drug targets for cognitive enhancement in neuropsychiatric disorders. Pharmacol Biochem Behav 99:130–145

    Article  CAS  Google Scholar 

  86. Terry AV Jr, Buccafusco JJ, Bartoszyk GD (2005) Selective serotonin 5-HT2A receptor antagonist EMD 281014 improves delayed matching performance in young and aged rhesus monkeys. Psychopharmacology 179:725–732

    Article  CAS  Google Scholar 

  87. Onogi H, Ishigaki S, Nakagawasai O, Arai-Kato Y, Arai Y, Watanabe H, Miyamoto A, Tan-No K, Tadano T (2009) Influence of memantine on brain monoaminergic neurotransmission parameters in mice: neurochemical and behavioral study. Biol Pharm Bull 32:850–855

    Article  CAS  Google Scholar 

  88. Reisberg B, Doody R, Stöffler A, Schmitt F, Ferris S, Möbius HJ (2006) A 24-week open-label extension study of memantine in moderate to severe Alzheimer disease. Arch Neurol 63:49–54

    Article  Google Scholar 

  89. Kim HS, Park IS, Park WK (1998) NMDA receptor antagonists enhance 5-HT2 receptor-mediated behavior, head-twitch response, in mice. Life Sci 63:2305–2311

    Article  CAS  Google Scholar 

  90. Nakagawasai O, Arai Y, Satoh SE, Satoh N, Neda M, Hozumi M, Oka R, Hiraga H, Tadano T (2004) Monoamine oxidase and head-twitch response in mice mechanisms of α-methylated substrate derivatives. Neurotoxicology 25:223–232

    Article  CAS  Google Scholar 

  91. Raddatz R, Tao M, Hudkins RL (2010) Histamine H3 antagonists for treatment of cognitive deficits in CNS diseases. Curr Top Med Chem 10:153–169

    Article  CAS  Google Scholar 

Download references

Acknowledgments

This project has been carried out with the support of WADA. We also acknowledge financial support from the Scottish Universities Life Sciences Alliance (SULSA). OMBA and JMC thenk MINECO (Spain) for a fellowship, and support (SAF2012-33304), respectively. KN and DA acknowledge project supported by the Ministry of Education and Science of the Republic of Serbia, Contract No. 172033. Further supports by Else Kröner-Fresenius-Stiftung, Translational Research Innovation—Pharma (TRIP), Fraunhofer-Projektgruppe für Translationale Medizin und Pharmakologie (TMP) (to HS) and the European COST Actions BM1007, CM1103 (including STSM 10295 to KN), and CM1207 are also gratefully acknowledged.

Conflict of interest

The authors (JBOM, LM) have received funding from WADA. Other then this sponsorship, the authors declare no conflict of interest.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Katarina Nikolic.

Electronic supplementary material

Below is the link to the electronic supplementary material.

10822_2014_9816_MOESM1_ESM.tif

Supplementary Figure 1: Ligand-pharmacological group associations for all examined compounds (1134), obtained by querying the 134 compounds against the refined DrugBank dataset (TIFF 14384 kb)

10822_2014_9816_MOESM2_ESM.xls

Supplementary Table 1: Ligand-target associations for all examined compounds (1134), obtained by querying the 134 compounds against the refined ChEMBL dataset (XLS 466 kb)

10822_2014_9816_MOESM3_ESM.xls

Supplementary Table 2: Ligand-pharmacological group associations for all examined compounds (1134), obtained by querying the 134 compounds against the refined DrugBank dataset (XLS 546 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nikolic, K., Mavridis, L., Bautista-Aguilera, O.M. et al. Predicting targets of compounds against neurological diseases using cheminformatic methodology. J Comput Aided Mol Des 29, 183–198 (2015). https://doi.org/10.1007/s10822-014-9816-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10822-014-9816-1

Keywords

Navigation