202
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Molecular docking, molecular dynamics, MM/PBSA approaches and bioactivity studies of nepetanudoside B isolated from endemic Nepeta aristata

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Received 14 Aug 2023, Accepted 18 Jan 2024, Published online: 30 Jan 2024

References

  • AbdElmoniem, N., H Abdallah, M., M Mukhtar, R., Moutasim, F., Rafie Ahmed, A., Edris, A., Ibraheem, W., Makki, A. A., M Elshamly, E., Elhag, R., Osman, W., A Mothana, R., & Alzain, A. A. (2023). Identification of novel natural dual HDAC and Hsp90 inhibitors for metastatic TNBC using e-pharmacophore modeling, molecular docking, and molecular dynamics studies. Molecules (Basel, Switzerland), 28(4), 1771. https://doi.org/10.3390/molecules28041771
  • Addar, L., Bensouici, C., Zennia, S. S. A., Haroun, S. B., & Mati, A. (2019). Antioxidant, tyrosinase and urease inhibitory activities of camel αS-casein and its hydrolysate fractions. Small Ruminant Research, 173, 30–35. https://doi.org/10.1016/j.smallrumres.2019.01.015
  • Aksu, F. (2018). Ordu Ilinden Toplanan Yabani Ve Yenilebilir Mantar Türlerinin Antioksidan Aktivitelerinin Ve Kolinesteraz. Tirosinaz Ve Üreaz Inhibisyon Potansiyellerinin Belirlenmesi. Yüksek Lisans Tezi, Ordu Üniversitesi.
  • Ali, H., Houghton, P., & Soumyanath, A. (2006). α-Amylase inhibitory activity of some Malaysian plants used to treat diabetes; with particular reference to Phyllanthus amarus. Journal of Ethnopharmacology, 107(3), 449–455. https://doi.org/10.1016/j.jep.2006.04.004
  • Alyar, S., Özmen, Ü. Ö., Adem, Ş., Alyar, H., Bilen, E., & Kaya, K. (2021). Synthesis, spectroscopic characterizations, carbonic anhydrase II inhibitory activity, anticancer activity and docking studies of new Schiff bases of sulfa drugs. Journal of Molecular Structure, 1223, 128911. https://doi.org/10.1016/j.molstruc.2020.128911
  • Amtul, Z., Rahman, A. U., Siddiqui, R. A., & Choudhary, M. I. (2002). Chemistry and mechanism of urease inhibition. Current Medicinal Chemistry, 9(14), 1323–1348. https://doi.org/10.2174/0929867023369853
  • Aničić, N., Gašić, U., Lu, F., Ćirić, A., Ivanov, M., Jevtić, B., Dimitrijević, M., Anđelković, B., Skorić, M., Nestorović Živković, J., Mao, Y., Liu, J., Tang, C., Soković, M., Ye, Y., & Mišić, D. (2021). Antimicrobial and ımmunomodulating activities of two endemic nepeta species and their major ıridoids ısolated from natural sources. Pharmaceuticals (Basel, Switzerland), 14(5), 414. https://www.mdpi.com/1424-8247/14/5/414 https://doi.org/10.3390/ph14050414
  • Baiseitova, A., Jenis, J., Kim, J. Y., Li, Z. P., & Park, K. H. (2019). Phytochemical analysis of aerial part of Ikonnikovia kaufmanniana and their protection of DNA damage. Natural Product Research, 35(5), 880–883. https://doi.org/10.1080/14786419.2019.1607858
  • Başar, Y., Yenigün, S., İpek, Y., Behçet, L., Gül, F., Özen, T., & Demirtaş, İ. (2023). DNA protection, molecular docking, enzyme inhibition and enzyme kinetic studies of 1,5,9-epideoxyloganic acid isolated from Nepeta aristata with bio-guided fractionation. Journal of Biomolecular Structure & Dynamics, 42, 1–14. https://doi.org/10.1080/07391102.2023.2250461
  • Berendsen, H.,Van Der Spoel, D., & Drunen, R. (1995). GROMACS: A message-passing parallel molecular dynamics implementation. Computer Physics Communications, 91(1-3), 43–56. https://doi.org/10.1016/0010-4655(95)00042-E
  • Berman, H. M., Westbrook, J., Feng, Z., Gilliland, G., Bhat, T. N., Weissig, H., Shindyalov, I. N., & Bourne, P. E. (2000). The protein data bank. Nucleic Acids Research, 28(1), 235–242. https://doi.org/10.1093/nar/28.1.235
  • Birari, R. B., & Bhutani, K. K. (2007). Pancreatic lipase inhibitors from natural sources: Unexplored potential. Drug Discovery Today, 12(19-20), 879–889. https://doi.org/10.1016/j.drudis.2007.07.024
  • Bussi, G.,Donadio, D., &Parrinello, M. (2007). Canonical sampling through velocity rescaling. The Journal of Chemical Physics, 126(1), 014101. https://doi.org/10.1063/1.2408420. PMC: 17212484
  • Chanda, J., Mukherjee, P. K., Biswas, R., Biswas, S., Tiwari, A. K., & Pargaonkar, A. (2019). UPLC‐QTOF‐MS analysis of a carbonic anhydrase‐inhibiting extract and fractions of Luffa acutangula (L.) Roxb (ridge gourd). Phytochemical Analysis: PCA, 30(2), 148–155. https://doi.org/10.1002/pca.2800
  • Chang, T.-S. (2009). An updated review of tyrosinase inhibitors. International Journal of Molecular Sciences, 10(6), 2440–2475. https://doi.org/10.3390/ijms10062440
  • Conforti, F., Statti, G., Loizzo, M. R., Sacchetti, G., Poli, F., & Menichini, F. (2005). In vitro antioxidant effect and inhibition of α-amylase of two varieties of Amaranthus caudatus seeds. Biological & Pharmaceutical Bulletin, 28(6), 1098–1102. https://doi.org/10.1248/bpb.28.1098
  • Copeland, R. A., Harpel, M. R., & Tummino, P. J. (2007). Targeting enzyme inhibitors in drug discovery. Expert Opinion on Therapeutic Targets, 11(7), 967–978. https://doi.org/10.1517/14728222.11.7.967
  • 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
  • Ercan, P., & El, S. N. (2016). Inhibitory effects of chickpea and Tribulus terrestris on lipase, α-amylase and α-glucosidase. Food Chemistry, 205, 163–169. https://doi.org/10.1016/j.foodchem.2016.03.012
  • Feldhammer, M., Uetani, N., Miranda-Saavedra, D., & Tremblay, M. L. (2013). PTP1B: A simple enzyme for a complex world. Critical Reviews in Biochemistry and Molecular Biology, 48(5), 430–445. https://doi.org/10.3109/10409238.2013.819830
  • Formisano, C., Rigano, D., & Senatore, F. (2011). Chemical constituents and biological activities of Nepeta species. Chemistry & Biodiversity, 8(10), 1783–1818. https://doi.org/10.1002/cbdv.201000191
  • Galano, A., Tan, D.-X., & Reiter, R. J. (2018). Melatonin: A versatile protector against oxidative DNA damage. Molecules (Basel, Switzerland), 23(3), 530. https://doi.org/10.3390/molecules23030530
  • Gerrard, J. A., Prince, M. J., & Abell, A. D. (2000). Kinetic characterisation of ene-diol-based inhibitors of α-amylase. Bioorganic & Medicinal Chemistry Letters, 10(14), 1575–1576. https://doi.org/10.1016/s0960-894x(00)00288-2
  • Gonzales, A. L., Huang, S. K.-H., Sevilla, U. T. A., Hsieh, C.-Y., & Tsai, P.-W. (2023). In silico analysis of anti-ınflammatory and antioxidant properties of bioactive compounds from Crescentia cujete L. Molecules (Basel, Switzerland), 28(8), 3547. https://www.mdpi.com/1420-3049/28/8/3547 https://doi.org/10.3390/molecules28083547
  • Güvenalp, Z., Özbek, H., Kuruüzüm-Uz, A. Y. ŞE., Kazaz, C., & Demirezer, L. Ö. (2009). Secondary metabolites from Nepeta heliotropifolia. Turkish Journal of Chemistry, 33, 667–675. https://doi.org/10.3906/kim-0812-60
  • Hamdan, I., & Afifi, F. (2004). Studies on the in vitro and in vivo hypoglycemic activities of some medicinal plants used in treatment of diabetes in Jordanian traditional medicine. Journal of Ethnopharmacology, 93(1), 117–121. https://doi.org/10.1016/s0378-8741(04)00157-6
  • Hansen, N., & Van Gunsteren, W. F. (2014). Practical aspects of free-energy calculations: A review. Journal of Chemical Theory and Computation, 10(7), 2632–2647. https://doi.org/10.1021/ct500161f
  • Hopkins, A. L., & Groom, C. R. (2002). The druggable genome. Nature Reviews. Drug Discovery, 1(9), 727–730. https://doi.org/10.1038/nrd892
  • Irfandi, R., Raya, I., Ahmad, A., Fudholi, A., Santi, S., Puspa Azalea, W., Ratih Tirto Sari, D., Jarre, S., Eka Putri, S., & Kartina, D. (2023). Anticancer potential of Cu (II) prolinedithiocarbamate complex: Design, synthesis, spectroscopy, molecular docking, molecular dynamic, ADMET, and in-vitro studies. Journal of Biomolecular Structure & Dynamics, 41(22), 12938–12950. https://doi.org/10.1080/07391102.2023.2169764
  • Karas, V. O., Westerlaken, I., & Meyer, A. S. (2013). Application of an in vitro DNA protection assay to visualize stress mediation properties of the Dps protein. Journal of Visualized Experiments, 75, e50390. https://doi.org/10.3791/50390
  • Kassamali, R., & Sica, D. A. (2011). Acetazolamide: A forgotten diuretic agent. Cardiology in Review, 19(6), 276–278. https://doi.org/10.1097/CRD.0b013e31822b4939
  • Kollman, P. A.,Massova, I.,Reyes, C.,Kuhn, B.,Huo, S.,Chong, L.,Lee, M.,Lee, T.,Duan, Y.,Wang, W.,Donini, O.,Cieplak, P.,Srinivasan, J.,Case, D. A., &Cheatham, T. E. (2000). Calculating structures and free energies of complex molecules: combining molecular mechanics and continuum models. Accounts of Chemical Research, 33(12), 889–897. https://doi.org/10.1021/ar000033j 11123888
  • Kumari, R.,Kumar, R., &Lynn, A. (2014). g_mmpbsa-a GROMACS tool for high-throughput MM-PBSA calculations. Journal of Chemical Information and Modeling, 54(7), 1951–1962. https://doi.org/10.1021/ci500020m 24850022
  • Liu, X. G., Lv, M. C., Huang, M. Y., Sun, Y. Q., Gao, P. Y., & Li, D. Q. (2019). A network pharmacology study on the triterpene saponins from Medicago sativa L. for the treatment of Neurodegenerative diseases. Journal of Food Biochemistry, 43(8), e12955. https://doi.org/10.1111/jfbc.12955
  • Mayur, B., Sandesh, S., Shruti, S., & Sung-Yum, S. (2010). Antioxidant and α-glucosidase inhibitory properties of Carpesium abrotanoides L. Journal of Medicinal Plants Research, 4(15), 1547–1553. https://doi.org/10.5897/JMPR.9000218
  • Miceli, N., Taviano, M., Giuffrida, D., Trovato, A., Tzakou, O., & Galati, E. (2005). Anti-inflammatory activity of extract and fractions from Nepeta sibthorpii Bentham. Journal of Ethnopharmacology, 97(2), 261–266. https://doi.org/10.1016/j.jep.2004.11.024
  • Moreno, M., Lombardi, A., De Lange, P., Silvestri, E., Ragni, M., Lanni, A., & Goglia, F. (2003). Fasting, lipid metabolism, and triiodothyronine in rat gastrocnemius muscle: İnterrelated roles of uncoupling protein 3, mitochondrial thioesterase, and coenzyme Q. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 17(9), 1112–1114. https://doi.org/10.1096/fj.02-0839fje
  • Motamed, S. M., & Naghibi, F. (2010). Antioxidant activity of some edible plants of the Turkmen Sahra region in northern Iran. Food Chemistry, 119(4), 1637–1642. https://doi.org/10.1016/j.foodchem.2009.09.057
  • Mukherjee, P. K., Kumar, V., Mal, M., & Houghton, P. J. (2007). Acetylcholinesterase inhibitors from plants. Phytomedicine: İnternational Journal of Phytotherapy and Phytopharmacology, 14(4), 289–300. https://doi.org/10.1016/j.phymed.2007.02.002
  • Muttaqin, F. Z. (2019). Molecular docking and molecular dynamic studies of stilbene derivative compounds as sirtuin-3 (Sirt3) histone deacetylase inhibitor on melanoma skin cancer and their toxicities prediction. Journal of Pharmacopolium, 2(2), 112-121. https://doi.org/10.36465/jop.v2i2.489
  • Nasiri, R., Moghimi, A., Alijanianzadeh, M., & Rabiee, S. (2013). Inhibition of mushroom tyrosinase by phenol derivative (HDNOS) ligand. International Journal of Chemistry and Technology, 4, 57–65.
  • Özyürek, M., Bektaşoğlu, B., Güçlü, K., & Apak, R. (2008). Hydroxyl radical scavenging assay of phenolics and flavonoids with a modified cupric reducing antioxidant capacity (CUPRAC) method using catalase for hydrogen peroxide degradation. Analytica Chimica Acta, 616(2), 196–206. https://doi.org/10.1016/j.aca.2008.04.033
  • Parrinello, M., &Rahman, A. (1981). Polymorphic transitions in single crystals: A new molecular dynamics method. Journal of Applied Physics, 52(12), 7182–7190. https://doi.org/10.1063/1.328693
  • Robinson, P. K. (2015). Enzymes: Principles and biotechnological applications. Essays in Biochemistry, 59, 1–41. https://doi.org/10.1042/bse0590001
  • Russo, A., Izzo, A. A., Borrelli, F., Renis, M., & Vanella, A. (2003). Free radical scavenging capacity and protective effect of Bacopa monniera L. on DNA damage. Phytotherapy Research: PTR, 17(8), 870–875. https://doi.org/10.1002/ptr.1061
  • Sciú, M. L., Sebastián-Pérez, V., Martinez-Gonzalez, L., Benitez, R., Perez, D. I., Pérez, C., Campillo, N. E., Martinez, A., & Moyano, E. L. (2019). Computer-aided molecular design of pyrazolotriazines targeting glycogen synthase kinase 3. Journal of Enzyme İnhibition and Medicinal Chemistry, 34(1), 87–96. https://doi.org/10.1080/14756366.2018.1530223
  • Sevgi, K., Tepe, B., & Sarikurkcu, C. (2015). Antioxidant and DNA damage protection potentials of selected phenolic acids. Food and Chemical Toxicology: An İnternational Journal Published for the British Industrial Biological Research Association, 77, 12–21. https://doi.org/10.1016/j.fct.2014.12.006
  • Sinha, P., & Yadav, A. K. (2023). Identification of 3, 4-dihydroxy complexes as potential antiviral via DFT, molecular docking, molecular dynamics and MM/PBSA against rabies and dengue receptors. Journal of Biomolecular Structure & Dynamics, 42, 1–17. https://doi.org/10.1080/07391102.2023.2246572
  • Smith, H. J., & Simons, C. (2004). Enzymes and their inhibitors: Drug development (Vol. 2). CRC Press.
  • Takeda, Y., Yagi, T., Matsumoto, T., Honda, G., Tabata, M., Fujita, T., Shingu, T., Otsuka, H., Sezik, E., & Yesilada, E. (1996). Nepetanudosides and iridoid glucosides having novel stereochemistry from Nepeta nuda ssp. albiflora. Phytochemistry, 42(4), 1085–1088. https://doi.org/10.1016/0031-9422(96)00074-x
  • Tepe, B., Degerli, S., Arslan, S., Malatyali, E., & Sarikurkcu, C. (2011). Determination of chemical profile, antioxidant, DNA damage protection and antiamoebic activities of Teucrium polium and Stachys iberica. Fitoterapia, 82(2), 237–246. https://doi.org/10.1016/j.fitote.2010.10.006
  • Thomsen, R., & Christensen, M. H. (2006). MolDock: A new technique for high-accuracy molecular docking. Journal of Medicinal Chemistry, 49(11), 3315–3321. https://doi.org/10.1021/jm051197e
  • Torella, R., Moroni, E., Caselle, M., Morra, G., & Colombo, G. (2010). Investigating dynamic and energetic determinants of protein nucleic acid recognition: Analysis of the zinc finger zif268-DNA complexes. BMC Structural Biology, 10(1), 42. https://doi.org/10.1186/1472-6807-10-42
  • Torres, P. H. M., Sodero, A. C. R., Jofily, P., & Silva, F. P. Jr, (2019). Key topics in molecular docking for drug design. International Journal of Molecular Sciences, 20(18), 4574. https://www.mdpi.com/1422-0067/20/18/4574 https://doi.org/10.3390/ijms20184574
  • Trentin, R., Custódio, L., Rodrigues, M. J., Moschin, E., Sciuto, K., da Silva, J. P., & Moro, I. (2020). Exploring Ulva australis Areschoug for possible biotechnological applications: In vitro antioxidant and enzymatic inhibitory properties, and fatty acids contents. Algal Research, 50, 101980. https://doi.org/10.1016/j.algal.2020.101980
  • Trott, O., & Olson, A. J. (2010). AutoDock Vina: İmproving 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
  • Tundis, R., Nadjafi, F., & Menichini, F. (2013). Angiotensin-converting enzyme inhibitory activity and antioxidant properties of Nepeta crassifolia Boiss & Buhse and Nepeta binaludensis Jamzad. Phytotherapy Research: PTR, 27(4), 572–580. https://doi.org/10.1002/ptr.4757
  • Wang, C., Gong, X., Bo, A., Zhang, L., Zhang, M., Zang, E., Zhang, C., & Li, M. (2020). Iridoids: Research advances in their phytochemistry, biological activities, and pharmacokinetics. Molecules (Basel, Switzerland), 25(2), 287(1-24). https://doi.org/10.3390/molecules25020287
  • Wu, Z., Shen, L., Han, Q., Lu, J., Tang, H., Xu, X., Xu, H., Huang, F., Xie, J., He, Z., Zeng, Z., & Hu, Z. (2017). Mechanism and nature of inhibition of trypsin by ligupurpuroside A, a Ku-Ding tea extract, studied by spectroscopic and docking methods. Food Biophysics, 12(1), 78–87. https://doi.org/10.1007/s11483-016-9465-0
  • Yenigün, S., Başar, Y., İpek, Y., Behçet, L., Özen, T., & Demirtaş, İ. (2023). Determination of antioxidant, DNA protection, enzyme inhibition potential and molecular docking studies of a biomarker ursolic acid in Nepeta species. Journal of Biomolecular Structure & Dynamics, 42, 1–18. https://doi.org/10.1080/07391102.2023.2229440
  • Zare, F., Solhjoo, A., Sadeghpour, H., Sakhteman, A., & Dehshahri, A. (2023). Structure-based virtual screening, molecular docking, molecular dynamics simulation and MM/PBSA calculations towards identification of steroidal and non-steroidal selective glucocorticoid receptor modulators. Journal of Biomolecular Structure & Dynamics, 41(16), 7640–7650. https://doi.org/10.1080/07391102.2022.2123392
  • Zhang, L., Mulrooney, S. B., Leung, A. F., Zeng, Y., Ko, B. B., Hausinger, R. P., & Sun, H. (2006). Inhibition of urease by bismuth (III): İmplications for the mechanism of action of bismuth drugs. Biometals: An İnternational Journal on the Role of Metal İons in Biology, Biochemistry, and Medicine, 19(5), 503–511. https://doi.org/10.1007/s10534-005-5449-0

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.