322
Views
0
CrossRef citations to date
0
Altmetric
Research Articles

GABAA and serotonergic receptors participation in anxiolytic effect of chalcones in adult zebrafish

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon show all
Pages 12426-12444 | Received 14 Sep 2022, Accepted 03 Jan 2023, Published online: 16 Jan 2023

References

  • Ahmad, F., & Richardson, M. K. (2013). Exploratory behaviour in the open field test adapted for larval zebrafish: Impact of environmental complexity. Behavioural Processes, 92, 88–98. https://doi.org/10.1016/j.beproc.2012.10.014
  • Amali, M. O., Atunwa, S. A., Aiyelero, O. M., & Omotesho, Q. A. (2019). Assessment of anxiolytic potential and acute toxicity study of combretum micranthum g. don. leaves (combretaceae) in mice. IBRO Reports, 7, 42. https://doi.org/10.1016/j.ibror.2019.09.085
  • Arellano-Aguiar, O., Solis-Angeles, S., Serrano-García, L., Morales-Sierra, E., Mendez-Serrano, A., Montero-Montoya, R. (2015). Use of the Zebrafish Embryo Toxicity Test for Risk Assessment Purpose: Case Study. Fisheriessciences.com, 9(4), 052–062.
  • Arshia, A. H., Shadravan, S., Solhjoo, A., Sakhteman, A., & Sami, A. (2021). De novo design of novel protease inhibitor candidates in the treatment of SARS-CoV-2 using deep learning, docking, and molecular dynamic simulations. Computers in Biology and Medicine, 139, 104967. https://doi.org/10.1016/j.compbiomed.2021.104967
  • Bai, Q., Tan, S., Xu, T., Liu, H., Huang, J., & Yao, X. (2021). MolAICal: a soft tool for 3D drug design of protein targets by artificial intelligence and classical algorithm. Briefings in Bioinformatics, 22(3), 1–12. https://doi.org/10.1093/bib/bbaa161
  • Basak, S., Gicheru, Y., Kapoor, A., Mayer, M. L., Filizola, M., & Chakrapani, S. (2019). Molecular mechanism of setron-mediated inhibition of full-length 5-HT3A receptor. Nature Communications, 10(1), 1–11. https://doi.org/10.1038/s41467-019-11142-8
  • Batista de Andrade Neto, J., Pessoa de Farias Cabral, V., Brito Nogueira, L. F., Rocha da Silva, C., Gurgel do Amaral Valente Sá, L., Ramos da Silva, A., Barbosa da Silva, W. M., Silva, J., Marinho, E. S., Cavalcanti, B. C., Odorico de Moraes, M., & Nobre Júnior, H. V. (2021). Anti-MRSA activity of curcumin in planktonic cells and biofilms and determination of possible action mechanisms. Microbial Pathogenesis, 155, 104892. https://doi.org/10.1016/j.micpath.2021.104892
  • Benneh, C. K., Biney, R. P., Mante, P. K., Tandoh, A., Adongo, D. W., & Woode, E. (2017). Maerua angolensis stem bark extract reverses anxiety and related behaviours in zebrafish—Involvement of GABAergic and 5-HT systems. Journal of Ethnopharmacology, 207, 129–145. https://doi.org/10.1016/j.jep.2017.06.012
  • Biovia, D. S. (2016). Discovery studio modeling environment, release 2017, San Diego. Dassault Systèmes.
  • Bugel, S. M., & Tanguay, R. L. (2018). Multidimensional chemobehavior analysis of flavonoids and neuroactive compounds in zebrafish. Toxicology and Applied Pharmacology, 344(February), 23–34. https://doi.org/10.1016/j.taap.2018.02.019
  • Buxeraud, J., & Faure, S. (2019). Benzodiazepines. Actualites Pharmaceutiques, 58(591), 24-26. https://doi.org/10.1016/j.actpha.2019.09.027
  • Cachat, J., Stewart, A., Utterback, E., Hart, P., Gaikwad, S., Wong, K., Kyzar, E., Wu, N., & Kalueff, A. V. (2011). Three-dimensional neurophenotyping of adult zebrafish behavior. PLoS One, 6(3), e17597. https://doi.org/10.1371/journal.pone.0017597
  • Celada, P., Bortolozzi, A., & Artigas, F. (2013). Serotonin 5-HT1A receptors as targets for agents to treat psychiatric disorders: Rationale and current status of research. CNS Drugs, 27(9), 703–716. https://doi.org/10.1007/s40263-013-0071-0
  • Chegini, H. R., Nasehi, M., & Zarrindast, M. R. (2014). Differential role of the basolateral amygdala 5-HT3 and 5-HT4 serotonin receptors upon ACPA-induced anxiolytic-like behaviors and emotional memory deficit in mice. Behavioural Brain Research, 261, 114–126. https://doi.org/10.1016/j.bbr.2013.12.007
  • Chisholm, D., Sweeny, K., Sheehan, P., Rasmussen, B., Smit, F., Cuijpers, P., & Saxena, S. (2016). Scaling-up treatment of depression and anxiety: A global return on investment analysis. The Lancet. Psychiatry, 3(5), 415–424. https://doi.org/10.1016/S2215-0366(16)30024-4
  • Cho, S., Kim, S., Jin, Z., Yang, H., Han, D., Baek, N. I., Jo, J., Cho, C. W., Park, J. H., Shimizu, M., & Jin, Y. H. (2011). Isoliquiritigenin, a chalcone compound, is a positive allosteric modulator of GABA A receptors and shows hypnotic effects. Biochemical and Biophysical Research Communications, 413(4), 637–642. https://doi.org/10.1016/j.bbrc.2011.09.026
  • Cosacak, M. I., Bhattarai, P., Bocova, L., Dzewas, T., Mashkaryan, V., Papadimitriou, C., Brandt, K., Hollak, H., Antos, C. L., & Kizil, C. (2017). Human TAUP301L overexpression results in TAU hyperphosphorylation without neurofibrillary tangles in adult zebrafish brain. Scientific Reports, 7(1), 1–14. https://doi.org/10.1038/s41598-017-13311-5
  • Costall, B., & Naylor, R. J. (2004). 5‐ HT 3 Receptors. Current Drug Targets. CNS and Neurological Disorders, 3(1), 27–37. https://doi.org/10.1002/9780470015902.a0027709
  • Csizmadia, P. (2019). MarvinSketch and MarvinView: Molecule applets for the world wide web.The 3rd International Electronic Conference on Synthetic Organic Chemistry. https://doi.org/10.3390/ecsoc-3-01775
  • da Silva, A. W., Ferreira, M. K. A., Pereira, L. R., Rebouças, E. L., Coutinho, M. R., Dos, J., Lima, R., Guedes, M. I. F., Bandeira, P. N., Magalhães, F. E. A., Menezes, J. E. S. A. d., Marinho, M. M., Teixeira, A. M. R., Salles Trevisan, M. T., dos Santos, H. S., & Marinho, E. S. (2021). Combretum lanceolatum extract reverses anxiety and seizure behavior in adult zebrafish through GABAergic neurotransmission: an in vivo and in silico study. Journal of Biomolecular Structure and Dynamics, 40(20), 9801-9814.https://doi.org/10.1080/07391102.2021.1935322
  • da Silva, A. W., Ferreira, M. K. A., Rebouças, E. L., Mendes, F. R. S., Atilano, A. L., de Menezes, J. E. S. A., Marinho, M. M., Marinho, E. S., Santos, H. S., & Teixeira, A. M. R. (2021). Anxiolytic-like effect of natural product 2-hydroxy-3,4,6-trimethoxyacetophenone isolated from Croton anisodontus in adult zebrafish via serotonergic neuromodulation involvement of the 5-HT system. Naunyn-Schmiedeberg’s Archives of Pharmacology, 394, 2023–2032. https://doi.org/10.1007/s00210-021-02116-z
  • da Silva, P. T., Lopes, L. M. A., Xavier, J. D. C., De Carvalho, M. C. S., De Moraes, M. O., Pessoa, C., Barros-Nepomuceno, F. W. A., Bandeira, P. N., Targino, C. S. D. P. C., Teixeira, A. M. R., Fontenelle, R. O. D. S., & Santos, H. S. (2020). Cytotoxic and antifungal activity of chalcones synthesized from natural acetophenone isolated from Croton anisodontus. Revista Virtual De Química, 12(3), 712–723. https://doi.org/10.21577/1984-6835.20200057
  • DasGupta, D., Mandalaparthy, V., & Jayaram, B. (2017). A component analysis of the free energies of folding of 35 proteins: A consensus view on the thermodynamics of folding at the molecular level. Journal of Computational Chemistry, 38(32), 2791–2801. https://doi.org/10.1002/jcc.25072
  • De Campos-Buzzi, F., Padaratz, P., Meira, A. V., Corrêa, R., Nunes, R. J., & Cechinel-Filho, V. (2007). 4′-Acetamidochalcone derivatives as potential antinociceptive agents. Molecules, 12(4), 896–906. https://doi.org/10.3390/12040896
  • DeLano, W. L. (2020). The PyMOL molecular graphics system, Version 2.3. Schrödinger LLC. https://doi.org/10.1038/hr.2014.17
  • 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 and QSAR in Environmental Research, 26(1), 1–27. https://doi.org/10.1080/1062936X.2014.977819
  • Diez, M., Petuya, V., Martínez-Cruz, L. A., & Hernández, A. (2014). Insights into mechanism kinematics for protein motion simulation. BMC Bioinformatics, 15(1), 184. https://doi.org/10.1186/1471-2105-15-184
  • Farago, O. (2019). Langevin thermostat for robust configurational and kinetic sampling. Physica A: Statistical Mechanics and Its Applications, 534(122210), 122210. https://doi.org/10.1016/j.physa.2019.122210
  • Ferreira, M. K. A., da Silva, A. W., dos Santos Moura, A. L., Sales, K. V. B., Marinho, E. M., do Nascimento Martins Cardoso, J., Marinho, M. M., Bandeira, P. N., Magalhães, F. E. A., Marinho, E. S., de Menezes, J. E. S. A., & dos Santos, H. S. (2021). Chalcones reverse the anxiety and convulsive behavior of adult zebrafish. Epilepsy & Behavior: E&B, 117, 107881. https://doi.org/10.1016/j.yebeh.2021.107881
  • 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. 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 and Biophysical Research Communications, 526(2), 505–511. https://doi.org/10.1016/j.bbrc.2020.03.129
  • 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, and Behavior, 99(3), 480–486. https://doi.org/10.1016/j.pbb.2011.04.021
  • Gleeson, M. P. (2008). Generation of a set of simple, interpretable ADMET rules of thumb. Journal of Medicinal Chemistry, 51(4), 817–834. https://doi.org/10.1021/jm701122q
  • Gohlke, H., & Case, D. A. (2004). Converging free energy estimates: MM-PB(GB)SA studies on the protein-protein complex Ras-Raf. Journal of Computational Chemistry, 25(2), 238–250. https://doi.org/10.1002/jcc.10379
  • Gohlke, H., Kiel, C., & Case, D. A. (2003). Insights into protein-protein binding by binding free energy calculation and free energy decomposition for the Ras-Raf and Ras-RalGDS complexes. Journal of Molecular Biology, 330(4), 891–913. https://doi.org/10.1016/S0022-2836(03)00610-7
  • Golani, L. K., Yeunus Mian, M., Ahmed, T., Pandey, K. P., Mondal, P., Sharmin, D., Rezvanian, S., Witkin, J. M., & Cook, J. M. (2022). Rationalizing the binding and α subtype selectivity of synthesized imidazodiazepines and benzodiazepines at GABAA receptors by using molecular docking studies. Bioorganic & Medicinal Chemistry Letters, 62(July 2021), 128637. https://doi.org/10.1016/j.bmcl.2022.128637
  • Gonçalves, N. G. G., de Araújo, J. I. F., Magalhães, F. E. A., Mendes, F. R. S., Lobo, M. D. P., Moreira, A. C. d. O. M., Moreira, R., & de, A. (2020). Protein fraction from Artocarpus altilis pulp exhibits antioxidant properties and reverses anxiety behavior in adult zebrafish via the serotoninergic system. Journal of Functional Foods, 66(April 2019), 103772. https://doi.org/10.1016/j.jff.2019.103772
  • Gupta, P., Khobragade, S., Rajaram, S., & Shingatgeri, V. (2014). Assessment of locomotion behavior in adult Zebrafish after acute exposure to different pharmacological reference compounds. Drug Development and Therapeutics, 5(2), 127. https://doi.org/10.4103/2394-2002.139626
  • 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
  • Hanson, D. R., & Gottesman, I. I. (2012). Biologically flavored perspectives on Garmezian resilience. Development and Psychopathology, 24(2), 363–369. https://doi.org/10.1017/S0954579412000041
  • 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. https://doi.org/10.1186/1758-2946-4-17
  • Higgs, J., Wasowski, C., Marcos, A., Jukič, M., Paván, C. H., Gobec, S., de Tezanos Pinto, F., Colettis, N., & Marder, M. (2019). Chalcone derivatives: synthesis, in vitro and in vivo evaluation of their anti-anxiety, anti-depression and analgesic effects. Heliyon, 5(3), e01376. https://doi.org/10.1016/j.heliyon.2019.e01376
  • Hood, S., O'Neil, G., & Hulse, G. (2009). The role of flumazenil in the treatment of benzodiazepine dependence: Physiological and psychological profiles. Journal of Psychopharmacology (Oxford, England), 23(4), 401–409. https://doi.org/10.1177/0269881108100322
  • Huey, R., Morris, G. M., & Forli, S. (2012). Using AutoDock 4 and AutoDock Vina with AutoDockTools: A tutorial. The Scripps Research Institute Molecular, 2-28.
  • 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
  • Humphrey, W., Dalke, A., & Schulten, K. (1996). VMD: Visual molecular dynamics. Journal of Molecular Graphics, 14(1), 33–38. https://doi.org/10.1016/0263-7855(96)00018-5
  • 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
  • Kato, K., Nakayoshi, T., Kurimoto, E., & Oda, A. (2021). Molecular dynamics simulations for the protein–ligand complex structures obtained by computational docking studies using implicit or explicit solvents. Chemical Physics Letters, 781(139022), 139022. https://doi.org/10.1016/j.cplett.2021.139022
  • Lima, A. H., Souza, P. R. M., Alencar, N., Lameira, J., Govender, T., Kruger, H. G., Maguire, G. E. M., & Alves, C. N. (2012). Molecular Modeling of T. rangeli, T. brucei gambiense, and T. evansi sialidases in complex with the DANA Inhibitor. Chemical Biology & Drug Design, 80(1), 114–120. https://doi.org/10.1111/j.1747-0285.2012.01380.x
  • Lipinski, C. A. (2004). Lead- and drug-like compounds: The rule-of-five revolution. Drug Discovery Today. Technologies, 1(4), 337–341. https://doi.org/10.1016/j.ddtec.2004.11.007
  • Marder, M. (2012). Flavonoids as GABAA receptor ligands: the whole story? Journal of Experimental Pharmacology, 4, 9–24. https://doi.org/10.2147/jep.s23105
  • 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
  • Masiulis, S., Desai, R., Uchański, T., Serna Martin, I., Laverty, D., Karia, D., Malinauskas, T., Zivanov, J., Pardon, E., Kotecha, A., Steyaert, J., Miller, K. W., & Aricescu, A. R. (2019). GABAA receptor signalling mechanisms revealed by structural pharmacology. Nature, 565(7740), 454–459. https://doi.org/10.1038/s41586-018-0832-5
  • Maximino, C., de Brito, T. M., Colmanetti, R., Pontes, A. A. A., de Castro, H. M., de Lacerda, R. I. T., Morato, S., & Gouveia, A. (2010). Parametric analyses of anxiety in zebrafish scototaxis. Behavioural Brain Research, 210(1), 1–7. https://doi.org/10.1016/j.bbr.2010.01.031
  • Oliveira, D. R. d., Todo, A. H., Rêgo, G. M., Cerutti, J. M., Cavalheiro, A. J., Rando, D. G. G., & Cerutti, S. M. (2018). Flavones-bound in benzodiazepine site on GABAA receptor: Concomitant anxiolytic-like and cognitive-enhancing effects produced by Isovitexin and 6-C-glycoside-Diosmetin. European Journal of Pharmacology, 831(April), 77–86. https://doi.org/10.1016/j.ejphar.2018.05.004
  • Morris, G. M., Huey R Fau- Lindstrom, W., Lindstrom W Fau- Sanner, M. F., Sanner Mf Fau- Belew, R. K., Belew Rk Fau- Goodsell, D. S., Goodsell Ds Fau- Olson, A. J., Olson, A. J., & Chem, J. C. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30(16), 2785–2791.
  • Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. Journal of Immunological Methods, 65(1-2), 55–63. https://doi.org/10.1016/0022-1759(83)90303-4
  • Olivier, B., Wijngaarden, I. V., & Soudijn, W. (2000). 5-HT3 receptor antagonists and anxiety; A preclinical and clinical review. European Neuropsychopharmacology: The Journal of the European College of Neuropsychopharmacology, 10(2), 77–95. https://doi.org/10.1016/S0924-977X(99)00065-6
  • 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
  • Phillips, J. C., Braun, R., Wang, W., Gumbart, J., Tajkhorshid, E., Villa, E., Chipot, C., Skeel, R. D., Kalé, L., & Schulten, K. (2005). Scalable molecular dynamics with NAMD. Journal of Computational Chemistry, 26(16), 1781–1802. https://doi.org/10.1002/jcc.20289
  • Rajkumar, R., & Mahesh, R. (2010). The auspicious role of the 5-HT3 receptor in depression: A probable neuronal target? Journal of Psychopharmacology (Oxford, England), 24(4), 455–469. https://doi.org/10.1177/0269881109348161
  • Rani, A., Singh, A., Kaur, J., Singh, G., Bhatti, R., Gumede, N., Kisten, P., Singh, P., Kumar., & V., Sumanjit. (2021). 1H-1,2,3-triazole grafted tacrine-chalcone conjugates as potential cholinesterase inhibitors with the evaluation of their behavioral tests and oxidative stress in mice brain cells. Bioorganic Chemistry, 114(September 2020), 105053. https://doi.org/10.1016/j.bioorg.2021.105053
  • Rojas, J., Domínguez, J. N., Charris, J. E., Lobo, G., Payá, M., & Ferrándiz, M. L. (2002). Synthesis and inhibitory activity of dimethylamino-chalcone derivatives on the induction of nitric oxide synthase. European Journal of Medicinal Chemistry, 37(8), 699–705. https://doi.org/10.1016/s0223-5234(02)01387-9
  • Rose, A. S., Bradley, A. R., Valasatava, Y., Duarte, J. M., Prlic, A., & Rose, P. W. (2018). NGL viewer: Web-based molecular graphics for large complexes. Bioinformatics (Oxford, England), 34(21), 3755–3758. https://doi.org/10.1093/bioinformatics/bty419
  • Salehi, B., Quispe, C., Chamkhi, I., El Omari, N., Balahbib, A., Sharifi-Rad, J., Bouyahya, A., Akram, M., Iqbal, M., Docea, A. O., Caruntu, C., Leyva-Gómez, G., Dey, A., Martorell, M., Calina, D., López, V., & Les, F. (2021). Pharmacological properties of chalcones: A review of preclinical including molecular mechanisms and clinical evidence. Frontiers in Pharmacology, 11, 1-21. https://doi.org/10.3389/fphar.2020.592654
  • Shityakov, S., & Foerster, C. (2014). In silico predictive model to determine vector-mediated transport properties for the blood-brain barrier choline transporter. Advances and Applications in Bioinformatics and Chemistry, 2014(7), 23-36. https://doi.org/10.2147/AABC.S63749
  • Silva Mendes, F. R., Wlisses da Silva, A., Amâncio Ferreira, M. K., de Lima Rebouças, E., Marinho, E. M., Marinho, M. M., Bandeira, P. N., Rodrigues Teixeira, A. M., Silva Alencar de Menezes, J. E., Alves de Siqueira, E., Róseo Paula Pessoa Bezerra de Menezes, R., Marinho, E. S., & Silva dos Santos, H. (2022). GABAA receptor participation in anxiolytic and anticonvulsant effects of (E)-3-(furan-2-yl)-1-(2hydroxy-3,4,6-trimethoxyphenyl)prop-2-en-1-one in adult zebrafish. Neurochemistry International, 155(October 2021), 105303. https://doi.org/10.1016/j.neuint.2022.105303
  • Silva, J., Rocha, M. N. d., Marinho, E. M., Marinho, M. M., Marinho, E. S., & Santos, H. S. d (2021). Evaluation of the ADME, toxicological analysis and molecular docking studies of the anacardic acid derivatives with potential antibacterial effects against Staphylococcus aureus. Journal of Analytical & Pharmaceutical Research, 10(5), 177–194. https://doi.org/10.15406/japlr.2021.10.00384
  • Stewart, A. M., Braubach, O., Spitsbergen, J., Gerlai, R., & Kalueff, A. V. (2014). Zebrafish models for translational neuroscience research: From tank to bedside. Trends in Neurosciences, 37(5), 264–278. https://doi.org/10.1016/j.tins.2014.02.011
  • Stewart, A., Gaikwad, S., Kyzar, E., Green, J., Roth, A., & Kalueff, A. V. (2012). Modeling anxiety using adult zebrafish: A conceptual review. Neuropharmacology, 62(1), 135–143. https://doi.org/10.1016/j.neuropharm.2011.07.037
  • Stewart, A., Wu, N., Cachat, J., Hart, P., Gaikwad, S., Wong, K., Utterback, E., Gilder, T., Kyzar, E., Newman, A., Carlos, D., Chang, K., Hook, M., Rhymes, C., Caffery, M., Greenberg, M., Zadina, J., & Kalueff, A. V. (2011). Pharmacological modulation of anxiety-like phenotypes in adult zebrafish behavioral models. Progress in Neuro-Psychopharmacology & Biological Psychiatry, 35(6), 1421–1431. https://doi.org/10.1016/j.pnpbp.2010.11.035
  • 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
  • Vos, T., Lim, S. S., Abbafati, C., Abbas, K. M., Abbasi, M., Abbasifard, M., Abbasi-Kangevari, M., Abbastabar, H., Abd-Allah, F., Abdelalim, A., Abdollahi, M., Abdollahpour, I., Abolhassani, H., Aboyans, V., Abrams, E. M., Abreu, L. G., Abrigo, M. R. M., Abu-Raddad, L. J., Abushouk, A. I., … Murray, C. J. L. (2020). Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. The Lancet, 396(10258), 1204–1222. https://doi.org/10.1016/S0140-6736(20)30925-9
  • Wiatrak, B., Kubis-Kubiak, A., Piwowar, A., & Barg, E. (2020). PC12 Cell Line: Cell types, coating of culture vessels, differentiation and other culture conditions. Cells, 9(4), 958. https://doi.org/10.3390/cells9040958
  • World Health Organization. (2017). Depression and other common mental disorders. Global Health Estimates.
  • Yan, J., Zhang, G., Pan, J., & Wang, Y. (2014). α-Glucosidase inhibition by luteolin: Kinetics, interaction and molecular docking. International Journal of Biological Macromolecules, 64, 213–223. https://doi.org/10.1016/j.ijbiomac.2013.12.007
  • 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 and Modeling, 48(7), 1411–1422. https://doi.org/10.1021/ci800084x

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.