868
Views
26
CrossRef citations to date
0
Altmetric
Research Articles

Determination of anticancer properties and inhibitory effects of some metabolic enzymes including acetylcholinesterase, butyrylcholinesterase, alpha-glycosidase of some compounds with molecular docking study

ORCID Icon, ORCID Icon, , , , , , & ORCID Icon show all
Pages 3693-3702 | Received 20 Apr 2020, Accepted 09 May 2020, Published online: 04 Jun 2020

References

  • Abdalrazaq, S. M., Cabir, B., Gümüş, S., & Ağırtaş, M. S. (2016). Synthesis of metallophthalocyanines with four oxy-2, 2-diphenylacetic acid substituents and their structural and electronic properties. Heterocyclic Communications, 22(5), 275–280. https://doi.org/10.1515/hc-2016-0120
  • Ağırtaş, M. S. (2017). A new method for the reaction of cross-coupling: Preparation of 5, 5′-bi (1, 10-phenanthroline). Heterocyclic Communications, 23(1), 7–8. https://doi.org/10.1515/hc-2016-0225
  • Ağırtaş, M. S., Solğun, D. G., Özdemir, S., & İzgi, M. S. (2018). Synthesis of tetra 3,4‐dimethoxyphenethoxy peripheral substituted metallophthalocyanines and investigation of some properties. ChemistrySelect, 3(12), 3523–3528. https://doi.org/10.1002/slct.201703122
  • Ahmad, S. S., Waheed, T., Rozeen, S., Mahmood, S., & Kamal, M. A. (2019). Therapeutic study of phytochemicals against cancer and Alzheimer’s disease management. Current Drug Metabolism, 20(13), 1006–1013. https://doi.org/10.2174/1389200221666200103092719
  • Ali, I., Haque, A., Wani, W. A., Saleem, K., & Al Za'abi, M. (2013). Analyses of anticancer drugs by capillary electrophoresis: A review. Biomedical Chromatography: BMC, 27(10), 1296–1311. https://doi.org/10.1002/bmc.2953
  • Ali, I., Wani, W. A., Saleem, K., & Hsieh, M.-F. (2014). Anticancer metallodrugs of glutamic acid sulphonamides: In silico, DNA binding, hemolysis and anticancer studies. RSC Advances, 4(56), 29629–29641. https://doi.org/10.1039/C4RA02570A
  • Altındağ, F. D., Sağlık, B. N., Acar Çevik, U., Işıkdağ, İ., Özkay, Y., & Karaca Gençer, H. (2019). Novel imidazole derivatives as antifungal agents: Synthesis, biological evaluation, ADME prediction and molecular docking studies. Phosphorus, Sulfur, and Silicon and the Related Elements, 194(9), 887–894. https://doi.org/10.1080/10426507.2019.1565761
  • Aras, A., Bursal, E., Türkan, F., Tohma, H., Kılıç, Ö., Gülçin, İ., & Köksal, E. (2019). Phytochemical content, antidiabetic, anticholinergic, and antioxidant activities of endemic lecokia cretica extracts. Chemistry & Biodiversity, 16(10), e1900341. https://doi.org/10.1002/cbdv.201900341
  • Buldurun, K., Turan, N., Aras, A., Mantarcı, A., Türkan, F., & Bursal, E. (2019). Spectroscopic and structural characterization, enzyme inhibitions, and antioxidant effects of new Ru(II) and Ni(II) complexes of Schiff base. Chemistry & Biodiversity, 16(8), e1900243. https://doi.org/10.1002/cbdv.201900243
  • Buldurun, K., Turan, N., Bursal, E., Mantarcı, A., Türkan, F., Taslimi, P., & Gülçin, İ. (2020). Synthesis, spectroscopic properties, crystal structures, antioxidant activities and enzyme inhibition determination of Co (II) and Fe (II) complexes of Schiff base. Research on Chemical Intermediates, 46(1), 283–297. https://doi.org/10.1007/s11164-019-03949-3
  • Burmaoglu, S., Yilmaz, A. O., Polat, M. F., Kaya, R., Gulcin, İ., & Algul, O. (2019). Synthesis and biological evaluation of novel tris-chalcones as potent carbonic anhydrase, acetylcholinesterase, butyrylcholinesterase and α-glycosidase inhibitors. Bioorganic Chemistry, 85, 191–197. https://doi.org/10.1016/j.bioorg.2018.12.035
  • Bursal, E., Aras, A., Kılıç, Ö., Taslimi, P., Gören, A. C., & Gülçin, İ. (2019). Phytochemical content, antioxidant activity, and enzyme inhibition effect of Salvia eriophora Boiss. & Kotschy against acetylcholinesterase, α-amylase, butyrylcholinesterase, and α-glycosidase enzymes. Journal of Food Biochemistry, 43(3), e12776. https://doi.org/10.1111/jfbc.12776
  • Cheung, J., Rudolph, M. J., Burshteyn, F., Cassidy, M. S., Gary, E. N., Love, J., Franklin, M. C., & Height, J. J. (2012). Structures of human acetylcholinesterase in complex with pharmacologically important ligands. Journal of Medicinal Chemistry, 55(22), 10282–10286. https://doi.org/10.1021/jm300871x
  • Du, S., Zhang, K., Yao, X., & Du, J. (2019). Investigation on the fungicide resistance mechanism against Botrytis cinerea β-tubulin inhibitor zoxamide by computational study. Journal of Biomolecular Structure and Dynamics, 1–9. https://doi.org/10.1080/07391102.2019.1671230
  • El-Sayed, N. A.-E., Farag, A. E.-S., Ezzat, M. A. F., Akincioglu, H., Gülçin, İ., & Abou-Seri, S. M. (2019). Design, synthesis, in vitro and in vivo evaluation of novel pyrrolizine-based compounds with potential activity as cholinesterase inhibitors and anti-Alzheimer’s agents. Bioorganic Chemistry, 93, 103312. https://doi.org/10.1016/j.bioorg.2019.103312
  • Erdemir, F., Celepci, D. B., Aktaş, A., Gök, Y., Kaya, R., Taslimi, P., Demir, Y., & Gulçin, İ. (2019). Novel 2-aminopyridine liganded Pd(II) N-heterocyclic carbene complexes: Synthesis, characterization, crystal structure and bioactivity properties. Bioorganic Chemistry, 91, 103134. https://doi.org/10.1016/j.bioorg.2019.103134
  • Friesner, R. A., Murphy, R. B., Repasky, M. P., Frye, L. L., Greenwood, J. R., Halgren, T. A., Sanschagrin, P. C., & Mainz, D. T. (2006). Extra precision glide: Docking and scoring incorporating a model of hydrophobic enclosure for protein-ligand complexes. Journal of Medicinal Chemistry, 49(21), 6177–6196. https://doi.org/10.1021/jm051256o
  • Frisch, M., Trucks, G., Schlegel, H., Scuseria, G., Robb, E., Cheeseman, J., Scalmani, G., Barone, V., Mennucci, B., Petersson, G., & Fox, D. J. (2009). Gaussian 09. Gaussian Inc., Wallingford, CT.
  • Gondolova, G., Taslimi, P., Medjidov, A., Farzaliyev, V., Sujayev, A., Huseynova, M., Şahin, O., Yalçın, B., Türkan, F., & Gulçin, İ. (2018). Synthesis, crystal structure and biological evaluation of spectroscopic characterization of Ni(II) and Co(II) complexes with N-salicyloil-N′-maleoil-hydrazine as anticholinergic and antidiabetic agents. Journal of Biochemical and Molecular Toxicology, 32(9), e22197. https://doi.org/10.1002/jbt.22197
  • Gulçin, İ., Taslimi, P., Aygün, A., Sadeghian, N., Bastem, E., Kufrevioglu, O. I., Türkan, F., & Şen, F. (2018). Antidiabetic and antiparasitic potentials: Inhibition effects of some natural antioxidant compounds on α-glycosidase, α-amylase and human glutathione S-transferase enzymes. International Journal of Biological Macromolecules, 119, 741–746. https://doi.org/10.1016/j.ijbiomac.2018.08.001
  • Günsel, A., Kırbaç, E., Tüzün, B., Erdoğmuş, A., Bilgiçli, A. T., & Yarasir, M. N. (2019). Selective chemosensor phthalocyanines for Pd2+ ions; Synthesis, characterization, quantum chemical calculation, photochemical and photophysical properties. Journal of Molecular Structure, 1180, 127–138. https://doi.org/10.1016/j.molstruc.2018.11.094
  • Jayarajan, R., Satheeshkumar, R., Kottha, T., Subbaramanian, S., Sayin, K., & Vasuki, G. (2020). Water mediated synthesis of 6-amino-5-cyano-2-oxo-N-(pyridin-2-yl)-4-(p-tolyl)-2H-[1,2′-bipyridine]-3-carboxamide and 6-amino-5-cyano-4-(4-fluorophenyl)-2-oxo-N-(pyridin-2-yl)-2H-[1,2′-bipyridine]-3-carboxamide: An experimental and computational studies with non-linear optical (NLO) and molecular docking analyses . Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 229, 117861. https://doi.org/10.1016/j.saa.2019.117861
  • Kaya, S., Tüzün, B., Kaya, C., & Obot, I. B. (2016). Determination of corrosion inhibition effects of amino acids: Quantum chemical and molecular dynamic simulation study. Journal of the Taiwan Institute of Chemical Engineers, 58, 528–535. https://doi.org/10.1016/j.jtice.2015.06.009
  • Košak, U., Brus, B., Knez, D., Žakelj, S., Trontelj, J., Pišlar, A., Šink, R., Jukič, M., Živin, M., Podkowa, A., Nachon, F., Brazzolotto, X., Stojan, J., Kos, J., Coquelle, N., Sałat, K., Colletier, J.-P., & Gobec, S. (2018). The magic of crystal structure-based inhibitor optimization: Development of a butyrylcholinesterase inhibitor with picomolar affinity and in vivo activity. Journal of Medicinal Chemistry, 61(1), 119–139. https://doi.org/10.1021/acs.jmedchem.7b01086
  • Kosiha, A., Lo, K. M., Parthiban, C., & Elango, K. P. (2019). Studies on the interaction of mononuclear metal(II) complexes of amino-naphthoquinone with bio-macromolecules . Materials Science & Engineering. C, Materials for Biological Applications, 94, 778–787. https://doi.org/10.1016/j.msec.2018.10.021
  • Li, Q., Yang, H., Chen, Y., & Sun, H. (2017). Recent progress in the identification of selective butyrylcholinesterase inhibitors for Alzheimer’s disease. European Journal of Medicinal Chemistry, 132, 294–309. https://doi.org/10.1016/j.ejmech.2017.03.062
  • Lovering, A. L., Lee, S. S., Kim, Y.-W., Withers, S. G., & Strynadka, N. C. (2005). Mechanistic and structural analysis of a family 31 alpha-glycosidase and its glycosyl-enzyme intermediate . The Journal of Biological Chemistry, 280(3), 2105–2115. https://doi.org/10.1074/jbc.M410468200
  • Mamedova, G., Mahmudova, A., Mamedov, S., Erden, Y., Taslimi, P., Tüzün, B., Tas, R., Farzaliyev, V., Sujayev, A., Alwasel, S. H., & Gulçin, İ. (2019). Novel tribenzylaminobenzolsulphonylimine based on their pyrazine and pyridazines: Synthesis, characterization, antidiabetic, anticancer, anticholinergic, and molecular docking studies. Bioorganic Chemistry, 93, 103313. https://doi.org/10.1016/j.bioorg.2019.103313
  • Mermer, A., Demirbas, N., Uslu, H., Demirbas, A., Ceylan, S., & Sirin, Y. (2019). Synthesis of novel Schiff bases using green chemistry techniques; antimicrobial, antioxidant, antiurease activity screening and molecular docking studies. Journal of Molecular Structure, 1181, 412–422. https://doi.org/10.1016/j.molstruc.2018.12.114
  • 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
  • Ozalp, L., Erdem, S. S., Yüce-Dursun, B., Mutlu, Ö., & Özbil, M. (2018). Computational insight into the phthalocyanine-DNA binding via docking and molecular dynamics simulations. Computational Biology and Chemistry, 77, 87–96. https://doi.org/10.1016/j.compbiolchem.2018.09.009
  • Prince, M., Ali, G., Guerchet, M., Prina, A., Albanese, E., & Wu, Y. (2016). Recent global trends in the prevalence and incidence of dementia, and survival with dementia. Alzheimer’s Research & Therapy, 8(1), 23. https://doi.org/10.1186/s13195-016-0188-8
  • Sağlık, B. N., Çavuşoğlu, B. K., Osmaniye, D., Levent, S., Çevik, U. A., Ilgın, S., Özkay, Y., Kaplancıklı, Z. A., & Öztürk, Y. (2019). In vitro and in silico evaluation of new thiazole compounds as monoamine oxidase inhibitors. Bioorganic Chemistry, 85, 97–108. https://doi.org/10.1016/j.bioorg.2018.12.019
  • Sari, S., Barut, B., Özel, A., Kuruüzüm-Uz, A., & Şöhretoğlu, D. (2019). Tyrosinase and α-glucosidase inhibitory potential of compounds isolated from Quercus coccifera bark: In vitro and in silico perspectives. Bioorganic Chemistry, 86, 296–304. https://doi.org/10.1016/j.bioorg.2019.02.015
  • Sastry, G. M., Adzhigirey, M., Day, T., Annabhimoju, R., & Sherman, W. (2013). Protein and ligand preparation: Parameters, protocols, and influence on virtual screening enrichments. Journal of Computer-Aided Molecular Design, 27(3), 221–234. https://doi.org/10.1007/s10822-013-9644-8
  • Sayin, K., & Üngördü, A. (2019). Investigations of structural, spectral and electronic properties of enrofloxacin and boron complexes via quantum chemical calculation and molecular docking. Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 220, 117102https://doi.org/10.1016/j.saa.2019.05.007
  • Schrödinger. (2017). Schrödinger release 2017-2: LigPrep. Schrödinger, LLC, New York, NY.
  • Schrödinger. (2019). Small-molecule drug discovery suite 2019-3. Schrödinger, LLC, New York, NY.
  • Singh, P., & Bast, F. (2014). In silico molecular docking study of natural compounds on wild and mutated epidermal growth factor receptor. Medicinal Chemistry Research, 23(12), 5074–5085. https://doi.org/10.1007/s00044-014-1090-1
  • Taslimi, P., Turhan, K., Türkan, F., Karaman, H. S., Turgut, Z., & Gulcin, İ. (2020). Cholinesterases, α-glycosidase, and carbonic anhydrase inhibition properties of 1H-pyrazolo[1,2-b]phthalazine-5,10-dione derivatives: Synthetic analogues for the treatment of Alzheimer’s disease and diabetes mellitus . Bioorganic Chemistry, 97, 103647. https://doi.org/10.1016/j.bioorg.2020.103647
  • Tekin, S., Erden, Y., Sandal, S., & Yilmaz, B. (2015). Is irisin an anticarcinogenic peptide? Medicine Science, 4, 2172–2180. https://doi.org/10.5455/medscience.2014.03.8210
  • Türe, A., Kahraman, D. C., Cetin-Atalay, R., Helvacıoğlu, S., Charehsaz, M., & Küçükgüzel, İ. (2019). Synthesis, anticancer activity, toxicity evaluation and molecular docking studies of novel phenylaminopyrimidine-(thio)urea hybrids as potential kinase inhibitors. Computational Biology and Chemistry, 78, 227–241. https://doi.org/10.1016/j.compbiolchem.2018.12.003
  • Türkan, F. (2019a). Investigation of inhibition effects of some natural phenolic compounds on glutathione S-transferase (GST), acetylcholinesterase (AChE), butyrylcholinesterase (BChE), α-amylase, and α-glycosidase: Antidiabetic, anticholinergics, antiparasitic study. Journal - Chemical Society of Pakistan, 41(4), 714–721.
  • Türkan, F. (2019b). Investigation of the toxicological and inhibitory effects of some benzimidazole agents on acetylcholinesterase and butyrylcholinesterase enzymes. Archives of Physiology and Biochemistry, 1–5. https://doi.org/10.1080/13813455.2019.1618341
  • Türkan, F., Atalar, M. N., Aras, A., Gülçin, İ., & Bursal, E. (2020). ICP-MS and HPLC analyses, enzyme inhibition and antioxidant potential of Achillea schischkinii Sosn. Bioorganic Chemistry, 94, 103333. https://doi.org/10.1016/j.bioorg.2019.103333
  • Tüzün, B., & Kaya, C. (2018). Investigation of DNA–RNA molecules for the efficiency and activity of corrosion inhibition by DFT and molecular docking. Journal of Bio- and Tribo-Corrosion, 4, 69.
  • Üngördü, A., & Sayin, K. (2019). Quantum chemical calculations on sparfloxacin and boron complexes. Chemical Physics Letters, 733, 136677. https://doi.org/10.1016/j.cplett.2019.136677
  • Wang, J.-L., Li, L., Hu, M.-B., Wu, B., Fan, W.-X., Peng, W., Wei, D.-N., & Wu, C.-J. (2019). In silico drug design of inhibitor of nuclear factor kappa B kinase subunit beta inhibitors from 2-acylamino-3-aminothienopyridines based on quantitative structure-activity relationships and molecular docking. Computational Biology and Chemistry, 78, 297–305. https://doi.org/10.1016/j.compbiolchem.2018.12.021
  • Yates, K., Pohl, F., Busch, M., Mozer, A., Watters, L., Shiryaev, A., & Lin, P. K. T. (2019). Determination of sinapine in rapeseed pomace extract: Its antioxidant and acetylcholinesterase inhibition properties. Food Chemistry, 276, 768–775. https://doi.org/10.1016/j.foodchem.2018.10.045
  • Zengin, G., Stefanucci, A., Rodrigues, M. J., Mollica, A., Custodio, L., Aumeeruddy, M. Z., & Mahomoodally, M. F. (2019). Scrophularia lucida L. as a valuable source of bioactive compounds for pharmaceutical applications: In vitro antioxidant, anti-inflammatory, enzyme inhibitory properties, in silico studies, and HPLC profiles. Journal of Pharmaceutical and Biomedical Analysis, 162, 225–233. https://doi.org/10.1016/j.jpba.2018.09.035

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.