468
Views
0
CrossRef citations to date
0
Altmetric
Research Articles

Diterpene Sonderianin isolated from Croton blanchetianus exhibits acetylcholinesterase inhibitory action and anxiolytic effect in adult zebrafish (Danio rerio) by 5-HT system

, , , , ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , & ORCID Icon show all
Pages 13625-13640 | Received 30 May 2021, Accepted 05 Oct 2021, Published online: 25 Oct 2021

References

  • Afrikanova, T., Serruys, A. S. K., Buenafe, O. E. M., Clinckers, R., Smolders, I., de Witte, P. A. M., Crawford, A. D., & Esguerra, C. V. (2013). Validation of the zebrafish pentylenetetrazol seizure model: Locomotor versus electrographic responses to antiepileptic drugs. PLoS One, 8(1), e54166–9. https://doi.org/10.1371/journal.pone.0054166
  • Ames, B. N., Gurney, E. G., Miller, J. A., & Bartsch, H. (1972). Carcinogens as frameshift mutagens: Metabolites and derivatives of 2-acetylaminofluorene and other aromatic amine carcinogens. Proceedings of the National Academy of Sciences of the United States of America, 69(11), 3128–3132. https://doi.org/10.1073/pnas.69.11.3128
  • Baell, J. B., & Holloway, G. A. (2010). New substructure filters for removal of pan assay interference compounds (PAINS) from screening libraries and for their exclusion in bioassays. Journal of Medicinal Chemistry, 53(7), 2719–2740. https://doi.org/10.1021/jm901137j
  • Belaidi, S., Youcef, O., Salah, T., & Lanez, T. (2015). In silico approach for conformational analysis, drug-likeness properties and structure activity relationships of 12-membered macrolides. Journal of Computational & Theoretical Nanoscience, 12(11), 4855–4861. https://doi.org/10.1166/jctn.2015.4451
  • Bento, A. P., Gaulton, A., Hersey, A., Bellis, L. J., Chambers, J., Davies, M., Krüger, F. A., Light, Y., Mak, L., McGlinchey, S., Nowotka, M., Papadatos, G., Santos, R., & Overington, J. P. (2014). The ChEMBL bioactivity database: An update. Nucleic Acids Research, 42(D1), D1083–1090. https://doi.org/10.1093/nar/gkt1031
  • Bickerton, G. R., Paolini, G. V., Besnard, J., Muresan, S., & Hopkins, A. L. (2012). Quantifying the chemical beauty of drugs. Nature Chemistry, 4(2), 90–98. https://doi.org/10.1038/nchem.1243
  • Biney, R. P., Benneh, C. K., Kyekyeku, J. O., Ameyaw, E. O., Boakye-Gyasi, E., & Woode, E. (2018). Attenuation of Anxiety behaviours by xylopic acid in mice and zebrafish models of anxiety disorder. UK Journal of Pharmaceutical Biosciences, 6(3), 7–16. https://doi.org/10.20510/ukjpb/6/i3/173546
  • Biovia, D. S. (2016). Discovery studio modeling environment, Release 2017, San Diego. In Dassault systèmes.
  • Biovia, S. D. D. S. (2020). Discovery studio visualizer (20.1.0).
  • Blazina, A. R., Vianna, M. R., & Lara, D. R. (2013). The spinning task: A new protocol to easily assess motor coordination and resistance in zebrafish. Zebrafish, 10(4), 480–485. https://doi.org/10.1089/zeb.2012.0860
  • Bueno, J. (2017). Bioprospecting and drug development, parameters for a rational search and validation of biodiversity. Journal of Microbial & Biochemical Technology, 9, e128. https://doi.org/10.4172/1948-5948.1000e128
  • Claeysen, S., Bockaert, J., & Giannoni, P. (2015). Serotonin: A new hope in Alzheimer's disease? ACS Chemical Neuroscience, 6(7), 940–943. https://doi.org/10.1021/acschemneuro.5b00135
  • Csizmadia, P. (2019). MarvinSketch and MarvinView: Molecule applets for the world wide web. https://doi.org/10.3390/ecsoc-3-01775
  • Da Rosa, J. G. S., Barcellos, H. H. D. A., Idalencio, R., Marqueze, A., Fagundes, M., Rossini, M., Variani, C., Balbinoti, F., Tietböhl, T. M. H., Rosemberg, D. B., & Barcellos, L. J. G. (2017). Just keep swimming: Neuroendocrine, metabolic, and behavioral changes after a forced swimming test in zebrafish. Zebrafish, 14(1), 51–59. https://doi.org/10.1089/zeb.2016.1340
  • da Silva, W. M. B., de Oliveira Pinheiro, S., Alves, D. R., de Menezes, J. E. S. A., Magalhães, F. E. A., Silva, F. C. O., Silva, J., Marinho, E. S., & de Morais, S. M. (2020). Synthesis of quercetin–metal complexes: In vitro and In silico anticholinesterase and antioxidant evaluation, and in vivo toxicological and anxiolitic activities. Neurotoxicity Research, 37(4), 893–903. https://doi.org/10.1007/s12640-019-00142-7
  • Daina, A., Michielin, O., & Zoete, V. (2017). SwissADME: A free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small molecules. Scientific Reports, 7, 42717–42713. https://doi.org/10.1038/srep42717
  • Daina, A., & Zoete, V. (2016). A BOILED-Egg to predict gastrointestinal absorption and brain penetration of small molecules. ChemMedChem, 11(11), 1117–1121. https://doi.org/10.1002/cmdc.201600182
  • De Sousa, I. P., Sousa Teixeira, M. V., & Jacometti Cardoso Furtado, N. A. (2018). An overview of biotransformation and toxicity of diterpenes. Molecules, 23(6), 1387. https://doi.org/10.3390/molecules23061387
  • Deye, N., Vincent, F., Michel, P., Ehrmann, S., da Silva, D., Piagnerelli, M., Kimmoun, A., Hamzaoui, O., Lacherade, J.-C., de Jonghe, B., Brouard, F., Audoin, C., Monnet, X., & Laterre, P.-F, For the SRLF Trial Group (2016). Changes in cardiac arrest patients' temperature management after the 2013 "TTM" trial: Results from an international survey. Annals of Intensive Care, 6(1), 1–9. https://doi.org/10.1186/s13613-015-0104-6
  • Di, L., & Kerns, E. H. (2016). Drug-like properties: Concepts, structure design and methods from ADME to toxicity optimization. In Drug-like properties: Concepts, structure design and methods from ADME to toxicity optimization. https://doi.org/10.1016/C2013-0-18378-X
  • Diaza, R. G., Manganelli, S., Esposito, A., Roncaglioni, A., Manganaro, A., & Benfenati, E. (2015). Comparison of in silico tools for evaluating rat oral acute toxicity. SAR & QSAR in Environmental Research, 26(1), 1–27. https://doi.org/10.1080/1062936X.2014.977819
  • Dorszewska, J., Florczak-Wyspianska, J., Kowalska, M., Stanski, M., Kowalewska, A., & Kozubski, W. (2017). Serotonin in Neurological Diseases. In Serotonin – A chemical messenger between all types of living cells. https://doi.org/10.5772/intechopen.69035
  • Ekambaram, S. P., Perumal, S. S., & Pavadai, S. (2017). Anti-inflammatory effect of Naravelia zeylanica DC via suppression of inflammatory mediators in carrageenan-induced abdominal oedema in zebrafish model. Inflammopharmacology, 25(1), 147–158. https://doi.org/10.1007/s10787-016-0303-2
  • Ellman, G. L., Courtney, K. D., Andres, V., & Feather-Stone, R. M. (1961). A new and rapid colorimetric determination of acetylcholinesterase activity. Biochemical Pharmacology, 7(2), 88–95. https://doi.org/10.1016/0006-2952(61)90145-9
  • EMBL-EBI (2011). ChEMBL. In ChEMBL.
  • Ertl, P., Rohde, B., & Selzer, P. (2000). Fast calculation of molecular polar surface area as a sum of fragment-based contributions and its application to the prediction of drug transport properties. Journal of Medicinal Chemistry, 43(20), 3714–3717. https://doi.org/10.1021/jm000942e
  • Ferreira, M. K. A., da Silva, A. W., Silva, F. C. O., Holanda, C. L. A., Barroso, S. M., Lima, J., dos, R., Vieira Neto, A. E., Campos, A. R., Bandeira, P. N., dos Santos, H. S., de Lemos, T. L. G., Siqueira, S. M. C., Magalhães, F. E. A., & de Menezes, J. E. S. A. (2019). Anxiolytic-like effect of chalcone N-{(4′-[(E)-3-(4-fluorophenyl)-1-(phenyl) prop-2-en-1-one]} acetamide on adult zebrafish (Danio rerio): Involvement of the GABAergic system. Behavioural Brain Research, 374, 111871–111878. https://doi.org/10.1016/j.bbr.2019.03.040
  • Ferreira, M. K. A., da Silva, A. W., Silva, F. C. O., Vieira Neto, A. E., Campos, A. R., Alves Rodrigues Santos, S. A., Rodrigues Teixeira, A. M., da Cunha Xavier, J., Bandeira, P. N., Sampaio Nogueira, C. E., de Brito, D. H. A., Rebouças, E. L., Magalhães, F. E. A., de Menezes, J. E. S. A., & dos Santos, H. S. (2020). Anxiolytic-like effect of chalcone N-{4'[(2E)-3-(3-nitrophenyl)-1-(phenyl)prop-2-en-1-one]} acetamide on adult zebrafish (Danio rerio): Involvement of the 5-HT system. Biochemical & Biophysical Research Communications, 526(2), 505–511. https://doi.org/10.1016/j.bbrc.2020.03.129
  • Fichert, T., Yazdanian, M., & Proudfoot, J. R. (2003). A structure–permeability study of small drug-like molecules. Bioorganic & Medicinal Chemistry Letters, 13(4), 719–722. https://doi.org/10.1016/S0960-894X(02)01035-1
  • Filimonov, D. A., Lagunin, A. A., Gloriozova, T. A., Rudik, A. V., Druzhilovskii, D. S., Pogodin, P. V., & Poroikov, V. V. (2014). Prediction of the biological activity spectra of organic compounds using the pass online web resource. Chemistry of Heterocyclic Compounds, 50(3), 444–457. https://doi.org/10.1007/s10593-014-1496-1
  • García-Ayllón, M. S., Small, D. H., Avila, J., & Sáez-Valero, J. (2011). Revisiting the role of acetylcholinesterase in Alzheimer's disease: Cross-talk with P-tau and β-amyloid. Frontiers in Molecular Neuroscience, 4, 22–229. https://doi.org/10.3389/fnmol.2011.00022
  • Gebauer, D. L., Pagnussat, N., Piato, Â. L., Schaefer, I. C., Bonan, C. D., & Lara, D. R. (2011). Effects of anxiolytics in zebrafish: Similarities and differences between benzodiazepines, buspirone and ethanol. Pharmacology, Biochemistry & Behaviour, 99(3), 480–486. https://doi.org/10.1016/j.pbb.2011.04.021
  • Geldenhuys, W. J., & Van der Schyf, C. J. (2011). Role of Serotonin in Alzheimer's disease: A new therapeutic target? CNS Drugs, 25(9), 765–781. https://doi.org/10.2165/11590190-000000000-00000
  • Ghose, A. K., & Viswanadhan, V. N., & Wendoloski, J. J. (1999). A knowledge-based approach in designing combinatorial or medicinal chemistry libraries for drug discovery. 1. A qualitative and quantitative characterization of known drug databases. Journal of Combinatorial Chemistry, 1(1), 55–68. https://doi.org/10.1021/cc9800071
  • Gold, L. S., Slone, T. H., Manley, N. B., Garfinkel, G. B., Hudes, E. S., Rohrbach, L., & Ames, B. N. (1991). The Carcinogenic Potency Database: Analyses of 4000 chronic animal cancer experiments published in the general literature and by the U.S. National Cancer Institute/National Toxicology Program. Environmental Health Perspectives, 96, 11–15. https://doi.org/10.1289/ehp.919611
  • Griebel, G., & Holmes, A. (2013). 50 years of hurdles and hope in anxiolytic drug discovery. Nature Reviews: Drug Discovery, 12(9), 667–687. https://doi.org/10.1038/nrd4075
  • Gupta, P., Khobragade, S. B., & Shingatgeri, V. M. (2014). Effect of various antiepileptic drugs in zebrafish PTZ-seizure model. Indian Journal of Pharmaceutical Sciences, 76(2), 157–163.
  • Gupta, G. L., & Sharma, L. (2019). Bacopa monnieri abrogates alcohol abstinence-induced anxiety-like behavior by regulating biochemical and Gabra1, Gabra4, Gabra5 gene expression of GABAA receptor signaling pathway in rats. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie, 111, 1417–1428. https://doi.org/10.1016/j.biopha.2019.01.048
  • Hagiwara, H. (2019). Total syntheses of clerodane diterpenoids: A review. Natural Product Communications, 14(6), 1934578X1984361–1934578X1984317. https://doi.org/10.1177/1934578X19843613
  • Hakimelahi, G. H., & Khodarahmi, G. A. (2005). The identification of toxicophores for the prediction of mutagenicity, hepatotoxicity and cardiotoxicity. Journal of the Iranian Chemical Society, 2(4), 244–267. https://doi.org/10.1007/BF03245929
  • Halgren, T. A. (1996). Merck molecular force field. I. Basis, form, scope, parameterization, and performance of MMFF94. Journal of Computational Chemistry, 17(5–6), 490–519. https://doi.org/10.1002/(SICI)1096-987X(199604)17:5/6<490::AID-JCC1>3.0.CO;2-P
  • Hanwell, M. D., Curtis, D. E., Lonie, D. C., Vandermeersch, T., Zurek, E., & Hutchison, G. R. (2012). Avogadro: An advanced semantic chemical editor, visualization, and analysis platform. Journal of Cheminformatics, 4(1), 17–17. https://doi.org/10.1186/1758-2946-4-17
  • Herculano, A. M., & Maximino, C. (2014). Serotonergic modulation of zebrafish behavior: Towards a paradox. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 55, 50–66. https://doi.org/10.1016/j.pnpbp.2014.03.008
  • Howe, D. G., Bradford, Y. M., Conlin, T., Eagle, A. E., Fashena, D., Frazer, K., Knight, J., Mani, P., Martin, R., Moxon, S. A. T., Paddock, H., Pich, C., Ramachandran, S., Ruef, B. J., Ruzicka, L., Schaper, K., Shao, X., Singer, A., Sprunger, B., Van Slyke, C. E., & Westerfield, M. (2012). ZFIN, the zebrafish model organism database: Increased support for mutants and transgenics. Nucleic Acids Research, 41(D1), D854–860. https://doi.org/10.1093/nar/gks938
  • Huey, R., Morris, G. M., & Forli, S. (2012). Using autodock 4 and autodock vina with autodocktools: The Scripps Research Institute Molecular Graphics Laboratory, 105, 50, 92037. .
  • Hughes, J. D., Blagg, J., Price, D. A., Bailey, S., DeCrescenzo, G. A., Devraj, R. V., Ellsworth, E., Fobian, Y. M., Gibbs, M. E., Gilles, R. W., Greene, N., Huang, E., Krieger-Burke, T., Loesel, J., Wager, T., Whiteley, L., & Zhang, Y. (2008). Physiochemical drug properties associated with in vivo toxicological outcomes. Bioorganic & Medicinal Chemistry Letters, 18(17), 4872–4875. https://doi.org/10.1016/j.bmcl.2008.07.071
  • Ivanenkov, Y. A., Zagribelnyy, B. A., & Aladinskiy, V. A. (2019). Are we opening the door to a new era of medicinal chemistry or being collapsed to a chemical singularity? Journal of Medicinal Chemistry, 62(22), 10026–10043. https://doi.org/10.1021/acs.jmedchem.9b00004
  • Johnson, T. W., Dress, K. R., & Edwards, M. (2009). Using the Golden Triangle to optimize clearance and oral absorption. Bioorganic & Medicinal Chemistry Letters, 19(19), 5560–5564. https://doi.org/10.1016/j.bmcl.2009.08.045
  • Kalueff, A. V., Gebhardt, M., Stewart, A. M., Cachat, J. M., Brimmer, M., Chawla, J. S., Craddock, C., Kyzar, E. J., Roth, A., Landsman, S., Gaikwad, S., Robinson, K., Baatrup, E., Tierney, K., Shamchuk, A., Norton, W., Miller, N., Nicolson, T., Braubach, O., Schneider, H., Zebrafish Neuroscience Research Consortium (2013). Towards a comprehensive catalog of zebrafish behavior 1.0 and beyond. Zebrafish, 10(1), 70–86. https://doi.org/10.1089/zeb.2012.0861
  • Kalueff, A. V., Stewart, A. M., & Gerlai, R. (2014). Zebrafish as an emerging model for studying complex brain disorders. Trends in Pharmacological Sciences, 35(2), 63–75. https://doi.org/10.1016/j.tips.2013.12.002
  • Karakaya, M., Scaramuzzi, A., Macrì, S., & Porfiri, M. (2021). Acute citalopram administration modulates anxiety in response to the context associated with a robotic stimulus in zebrafish. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 108, 110172. https://doi.org/10.1016/j.pnpbp.2020.110172
  • Khan, K. M., Collier, A. D., Meshalkina, D. A., Kysil, E. V., Khatsko, S. L., Kolesnikova, T., Morzherin, Y. Y., Warnick, J. E., Kalueff, A. V., & Echevarria, D. J. (2017). Zebrafish models in neuropsychopharmacology and CNS drug discovery. British Journal of Pharmacology, 174(13), 1925–1944. https://doi.org/10.1111/bph.13754
  • Kyriakatos, A., Mahmood, R., Ausborn, J., Porres, C. P., Büschges, A., & Manira, A. E. (2011). Initiation of locomotion in adult Zebrafish. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience, 31(23), 8422–8431. https://doi.org/10.1523/JNEUROSCI.1012-11.2011
  • Kysil, E. V., Meshalkina, D. A., Frick, E. E., Echevarria, D. J., Rosemberg, D. B., Maximino, C., Lima, M. G., Abreu, M. S., Giacomini, A. C., Barcellos, L. J. G., Song, C., & Kalueff, A. V. (2017). Comparative analyses of zebrafish anxiety-like behavior using conflict-based novelty tests. Zebrafish, 14(3), 197–208. https://doi.org/10.1089/zeb.2016.1415
  • Lagunin, A. A., Zakharov, A. V., Filimonov, D. A., & Poroikov, V. V. (2007). A new approach to QSAR modelling of acute toxicity. SAR & QSAR in Environmental Research, 18(3–4), 285–298. https://doi.org/10.1080/10629360701304253
  • Li, R., Morris-Natschke, S. L., & Lee, K. H. (2016). Clerodane diterpenes: Sources, structures, and biological activities. Natural Product Reports, 33(10), 1166–1226. https://doi.org/10.1039/c5np00137d
  • Lipinski, C. A., Lombardo, F., Dominy, B. W., & Feeney, P. J. (1997). Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Advanced Drug Delivery Reviews, 23(1–3), 3–25. https://doi.org/10.1016/S0169-409X(96)00423-1
  • Madan, R., Kumar, S., Bansal, G., & Sharma, A. (2011). Plant drugs used to combat menace of anxiety disorders. Pharmacognosy Communications, 1(1), 04–51. https://doi.org/10.5530/pc.2011.1.2
  • Magalhães, F. E. A., De Sousa, C. Á. P. B., Santos, S. A. A. R., Menezes, R. B., Batista, F. L. A., Abreu, Â. O., De Oliveira, M. V., Moura, L. F. W. G., Raposo, R. D. S., & Campos, A. R. (2017). Adult zebrafish (Danio rerio): An alternative behavioral model of formalin-induced nociception. Zebrafish, 14(5), 422–429. https://doi.org/10.1089/zeb.2017.1436
  • Mansur, B. D. M., Dos Santos, B. R., Dias, C. A. G. D. M., Pinheiro, M. D. S., & Gouveia, A. (2014). Effects of the number of subjects on the dark/light preference of zebrafish (Danio rerio). Zebrafish, 11(6), 560–566. https://doi.org/10.1089/zeb.2014.0977
  • Marinho, E. M., Batista de Andrade Neto, J., Silva, J., Rocha da Silva, C., Cavalcanti, B. C., Marinho, E. S., & Nobre Júnior, H. V. (2020). Virtual screening based on molecular docking of possible inhibitors of Covid-19 main protease. Microbial Pathogenesis, 148, 104365. https://doi.org/10.1016/j.micpath.2020.104365
  • Martin, Y. C. (2005). A bioavailability score. Journal of Medicinal Chemistry, 48(9), 3164–3170. https://doi.org/10.1021/jm0492002
  • Marucci, G., Buccioni, M., Ben, D. D., Lambertucci, C., Volpini, R., & Amenta, F. (2021). Efficacy of acetylcholinesterase inhibitors in Alzheimer's disease. Neuropharmacology, 190, 108352. https://doi.org/10.1016/j.neuropharm.2020.108352
  • Melkemi, N., & Belaidi, S. (2014). Structure–property relationships and quantitative structure–activity relationship modeling of detoxication properties of some 1,2-dithiole-3-thione derivatives. Journal of Computational & Theoretical Nanoscience, 11(3), 801–806. https://doi.org/10.1166/jctn.2014.3431
  • Meza, J. C. (2010). Steepest descent. WIREs Computational Statistics, 2(6), 719–722. https://doi.org/10.1002/wics.117
  • Oliveira, I. M. M., Santos, H. S., Sena, D. M., Cruz, B. G., Teixeira, A. M. R., Freire, P. T. C., Braz-Filho, R., Sousa, J. W., Albuquerque, M. R. J. R., Bandeira, P. N., Bernardino, A. C. S. S., Gusmão, G. O. M., & Bento, R. R. F. (2015). Vibrational spectra and DFT calculations of sonderianin diterpene. Journal of Molecular Structure, 1099, 226–231. https://doi.org/10.1016/j.molstruc.2015.06.049
  • Omar Arellano-Aguilar, S. (2015). Use of the zebrafish embryo toxicity test for risk assessment purpose: Case study. Fisheriessciences.Com, 9(4), 52–62.
  • Ortiz-Mendoza, N., Zavala-Ocampo, L. M., Martínez-Gordillo, M. J., González-Trujano, M. E., Peña, F. A. B., Bazany-Rodríguez, I. J., Chávez, J. A. R., Dorazco-González, A., & Aguirre-Hernández, E. (2020). Antinociceptive and anxiolytic-like effects of a neo-clerodane diterpene from Salvia semiatrata aerial parts. Pharmaceutical Biology, 58(1), 620–629. https://doi.org/10.1080/13880209.2020.1784235
  • Pettersen, E. F., Goddard, T. D., Huang, C. C., Couch, G. S., Greenblatt, D. M., Meng, E. C., & Ferrin, T. E. (2004). UCSF Chimera: A visualization system for exploratory research and analysis. Journal of Computational Chemistry, 25(13), 1605–1612. https://doi.org/10.1002/jcc.20084
  • Picciotto, M. R., Higley, M. J., & Mineur, Y. S. (2012). Acetylcholine as a neuromodulator: Cholinergic signaling shapes nervous system function and behavior. Neuron, 76(1), 116–129. https://doi.org/10.1016/j.neuron.2012.08.036
  • Quadros, L. d C. M. d., Quevedo, L. d A., Gonçalves, H. D., Horta, B. L., Motta, J. V. D. S., & Gigante, D. P. (2020). Common mental disorders and contemporary factors: 1982 birth cohort. Revista Brasileira de Enfermagem, 73(1), 1–7. https://doi.org/10.1590/0034-7167-2018-0162
  • Sá Firmino, N. C., Alexandre, F. S. O., de Vasconcelos, M. A., Pinheiro, A. A., Arruda, F. V. S., Guedes, M. L. S., Silveira, E. R., & Teixeira, E. H. (2019). Diterpenes isolated from Croton blanchetianus Baill: Potential compounds in prevention and control of the oral Streptococci biofilms. Industrial Crops & Products, 131, 371–377. https://doi.org/10.1016/j.indcrop.2019.01.062
  • Secco, R. D. S., Cordeiro, I., De Senna-Vale, L., De Sales, M. F., De Lima, L. R., Medeiros, D., Haiad, B. D. S., De Oliveira, A. S., Caruzo, M. B. R., Carneiro-Torres, D., & Bigio, N. C. (2012). An overview of recent taxonomic studies on Euphorbiaceae s.l. in Brazil. Rodriguésia, 63(1), 227–242. https://doi.org/10.1590/S2175-78602012000100014
  • Shaikh, M. F., Sancheti, J., & Sathaye, S. (2013). Effect of Eclipta alba on acute seizure models: A GABAA-mediated effect. Indian Journal of Pharmaceutical Sciences, 75(3), 380–384. https://doi.org/10.4103/0250-474X.117432
  • Shanmugasundaram, J., Subramanian, V., Nadipelly, J., Kathirvelu, P., Sayeli, V., & Cheriyan, B. V. (2020). Anxiolytic-like activity of 5-methoxyflavone in mice with involvement of GABAergic and serotonergic systems – In vivo and in silico evidences. European Neuropsychopharmacology, 36, 100–110. https://doi.org/10.1016/j.euroneuro.2020.05.009
  • Shityakov, S., & Foerster, C. (2014). In silico predictive model to determine vector-mediated transport properties for the blood–brain barrier choline transporter. Advances & Applications in Bioinformatics & Chemistry, 7, 23–36. https://doi.org/10.2147/AABC.S63749
  • Siebel, A. M., Menezes, F. P., Capiotti, K. M., Kist, L. W., Schaefer, I. D. C., Frantz, J. Z., Bogo, M. R., Da Silva, R. S., & Bonan, C. D. (2015). Role of adenosine signaling on pentylenetetrazole-induced seizures in zebrafish. Zebrafish, 12(2), 127–136. https://doi.org/10.1089/zeb.2014.1004
  • Tabet, N. (2006). Acetylcholinesterase inhibitors for Alzheimer's disease: Anti-inflammatories in acetylcholine clothing!. Age & Ageing, 35(4), 336–338. https://doi.org/10.1093/ageing/afl027
  • Trott, O., & Olson, A. J. (2010). AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. Journal of Computational Chemistry, 31(2), 455–461. https://doi.org/10.1002/jcc.21334
  • Ulhaq, Z. S., & Kishida, M. (2018). Brain aromatase modulates serotonergic neuron by regulating serotonin levels in zebrafish embryos and larvae. Frontiers in Endocrinology, 9, 230. https://doi.org/10.3389/fendo.2018.00230
  • Veber, D. F., Johnson, S. R., Cheng, H. Y., Smith, B. R., Ward, K. W., & Kopple, K. D. (2002). Molecular properties that influence the oral bioavailability of drug candidates. Journal of Medicinal Chemistry, 45(12), 2615–2623. https://doi.org/10.1021/jm020017n
  • Veith, H., Southall, N., Huang, R., James, T., Fayne, D., Artemenko, N., Shen, M., Inglese, J., Austin, C. P., Lloyd, D. G., & Auld, D. S. (2009). Comprehensive characterization of cytochrome P450 isozyme selectivity across chemical libraries. Nature Biotechnology, 27(11), 1050–1055. https://doi.org/10.1038/nbt.1581
  • Wager, T. T., Chandrasekaran, R. Y., Hou, X., Troutman, M. D., Verhoest, P. R., Villalobos, A., & Will, Y. (2010a). Defining desirable central nervous system drug space through the alignment of molecular properties, in vitro ADME, and safety attributes. ACS Chemical Neuroscience, 1(6), 420–434. https://doi.org/10.1021/cn100007x
  • Wager, T. T., Hou, X., Verhoest, P. R., & Villalobos, A. (2010b). Moving beyond rules: The development of a central nervous system multiparameter optimization (CNS MPO) approach to enable alignment of druglike properties. ACS Chemical Neuroscience, 1(6), 435–449. https://doi.org/10.1021/cn100008c
  • Xavier, J., da, C., Ferreira, M. K. A., da Silva, A. W., de Menezes, J. E. S. A., Teixeira, A. M. R., Bandeira, P. N., Marinho, E. M., Marinho, E. S., Marinho, M. M., & Dos Santos, H. S. (2021). Anxiolytic-like and anticonvulsant effect in adult zebrafish (Danio rerio) through gabaergic system and molecular docking study of chalcone derived from natural products. Biointerface Research in Applied Chemistry, 11(6), 14021–14031. https://doi.org/10.33263/BRIAC116.1402114031
  • Xu, P., Huang, S., Zhang, H., Mao, C., Zhou, X. E., Cheng, X., Simon, I. A., Shen, D. D., Yen, H. Y., Robinson, C. V., Harpsøe, K., Svensson, B., Guo, J., Jiang, H., Gloriam, D. E., Melcher, K., Jiang, Y., Zhang, Y., & Xu, H. E. (2021). Structural insights into the lipid and ligand regulation of serotonin receptors. Nature, 592(7854), 469–473. https://doi.org/10.1038/s41586-021-03376-8
  • Yang, H., Lou, C., Sun, L., Li, J., Cai, Y., Wang, Z., Li, W., Liu, G., & Tang, Y. (2019). AdmetSAR 2.0: Web-service for prediction and optimization of chemical ADMET properties. Bioinformatics (Oxford, England), 35(6), 1067–1069. https://doi.org/10.1093/bioinformatics/bty707
  • Yusuf, D., Davis, A. M., Kleywegt, G. J., & Schmitt, S. (2008). An alternative method for the evaluation of docking performance: RSR vs RMSD. Journal of Chemical Information & Modeling, 48(7), 1411–1422. https://doi.org/10.1021/ci800084x
  • Zahno, A., Brecht, K., Morand, R., Maseneni, S., Török, M., Lindinger, P. W., & Krähenbühl, S. (2011). The role of CYP3A4 in amiodarone-associated toxicity on HepG2 cells. Biochemical Pharmacology, 81(3), 432–441. https://doi.org/10.1016/j.bcp.2010.11.002
  • Zhao, Y. H., Abraham, M. H., Le, J., Hersey, A., Luscombe, C. N., Beck, G., Sherborne, B., & Cooper, I. (2002). Rate-limited steps of human oral absorption and QSAR studies. Pharmaceutical Research, 19(10), 1446–1457. https://doi.org/10.1023/A:1020444330011

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.