263
Views
0
CrossRef citations to date
0
Altmetric
Research Article

Isolation, characterization and evaluation of oxypeucedanin and osthol from local endemic Prangos aricakensis Behçet and Yapar root as antioxidant, enzyme inhibitory, antibacterial and DNA protection: molecular docking and DFT approaches

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Received 13 Sep 2023, Accepted 03 Jan 2024, Published online: 12 Jan 2024

References

  • Abbas-Mohammadi, M., Farimani, M. M., Salehi, P., Ebrahimi, S. N., Sonboli, A., Kelso, C., & Skropeta, D. (2018). Acetylcholinesterase-inhibitory activity of Iranian plants: Combined HPLC/bioassay-guided fractionation, molecular networking and docking strategies for the dereplication of active compounds. Journal of Pharmaceutical and Biomedical Analysis, 158, 471–479. https://doi.org/10.1016/j.jpba.2018.06.026
  • Aćimović, M. G., Kostadinović, L. M., Popović, S. J., & Dojčinović, N. S. (2015). Apiaceae seeds as functional food. Journal of Agricultural Sciences (Belgrade), 60(3), 237–246. https://doi.org/10.2298/JAS1503237A
  • 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
  • Albayrak, G., Demir, S., Koyu, H., & Baykan, S. (2022). Anticholinesterase compounds from endemic Prangos uechtritzii. Chemistry & Biodiversity, 19(11), e202200557. https://doi.org/10.1002/cbdv.202200557
  • Aloui, L., Kossentini, M., Geffroy-Rodier, C., Guillard, J., & Zouari, S. (2015). Phytochemical investigation, isolation and characterization of coumarins from aerial parts and roots of Tunisian Pituranthos chloranthus (Apiaceae). Pharmacognosy Communications, 5(4), 237–243. https://doi.org/10.5530/pc.2015.4.4
  • 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
  • Andrews, J. M. (2001). Determination of minimum inhibitory concentrations. Journal of Antimicrobial Chemotherapy, 48(suppl_1), 5–16. https://doi.org/10.1093/jac/dkf083
  • Bağlan, M., Gören, K., & Yildiko, Ü. (2023). HOMO–LUMO, NBO, NLO, MEP analysis and molecular docking using DFT calculations in DFPA molecule. International Journal of Chemistry and Technology, 7(1), 38–47. https://doi.org/10.32571/ijct.1135173
  • 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
  • Becke, A. D. (1988). Density-functional exchange-energy approximation with correct asymptotic behavior. Physical Review. A, General Physics, 38(6), 3098–3100. https://doi.org/10.1103/PhysRevA.38.3098
  • Behçet, L., Yapar, Y., & Olgun, Ş. (2019). Prangos aricakensis (Apiaceae), a new species from eastern Turkey. Phytotaxa, 401(1), 55–63. https://doi.org/10.11646/phytotaxa.401.1.5
  • BİOVİA, D. S. (2019). BIOVIA Discovery Studio Visualize 2019, version 2019. Dassault Systèmes BIOVIA.
  • Blois, M. S. (1958). Antioxidant determinations by the use of a stable free radical. Nature, 181(4617), 1199–1200. https://doi.org/10.1038/1811199a0
  • Bruno, M., Ilardi, V., Lupidi, G., Quassinti, L., Bramucci, M., Fiorini, D., Venditti, A., & Maggi, F. (2019). The nonvolatile and volatile metabolites of Prangos ferulacea and their biological properties. Planta Medica, 85(11–12), 815–824. https://doi.org/10.1055/a-0873-8622
  • Christensen, L. P., & Brandt, K. (2006). Bioactive polyacetylenes in food plants of the Apiaceae family: Occurrence, bioactivity and analysis. Journal of Pharmaceutical and Biomedical Analysis, 41(3), 683–693. https://doi.org/10.1016/j.jpba.2006.01.057
  • Costin, G.-E., & Hearing, V. J. (2007). Human skin pigmentation: Melanocytes modulate skin color in response to stress. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 21(4), 976–994. https://doi.org/10.1096/fj.06-6649rev
  • Cutler, H. G., & Cutler, S. J. (1999). Biologically active natural products: Agrochemicals. CRC Press.
  • de Almeida, S. M. V., Lafayette, E. A., da Silva, L. P. B. G., Amorim, C. A. D C., de Oliveira, T. B., Ruiz, A. L. T. G., de Carvalho, J. E., de Moura, R. O., Beltrão, E. I. C., de Lima, M. D C. A., & de Carvalho Júnior, L. B. (2015). Synthesis, DNA binding, and antiproliferative activity of novel acridine-thiosemicarbazone derivatives. International Journal of Molecular Sciences, 16(6), 13023–13042. https://doi.org/10.3390/ijms160613023
  • Dechayont, B., Ruamdee, P., Poonnaimuang, S., Mokmued, K., & Chunthorng-Orn, J. (2017). Antioxidant and antimicrobial activities of Pogostemon cablin (Blanco) Benth. Journal of Botany, 2017, 1–6. https://doi.org/10.1155/2017/8310275
  • Desjardins, A. E., Spencer, G. F., & Plattner, R. D. (1989). Tolerance and metabolism of furanocoumarins by the phytopathogenic fungus Gibberella pulicaris (Fusarium sambucinum). Phytochemistry, 28(11), 2963–2969. https://doi.org/10.1016/0031-9422(89)80262-6
  • 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
  • El‐Rayyes, A. A., & Umar, Y. (2021). Theoretical geometry and ıntramolecular hydrogen bonding in 8‐hydroxy‐1‐naphthaldehyde. Bulletin of the Korean Chemical Society, 42(10), 1310–1318. https://doi.org/10.1002/bkcs.12367
  • Fang, R., Jiang, C. H., Wang, X. Y., Zhang, H. M., Liu, Z. L., Zhou, L., Du, S. S., & Deng, Z. W. (2010). Insecticidal activity of essential oil of Carum carvi fruits from China and its main components against two grain storage insects. Molecules (Basel, Switzerland), 15(12), 9391–9402. https://doi.org/10.3390/molecules15129391
  • Farooq, Saleem, Dangroo, Nisar Ahmad, Priya, Dev, Banday, Javid Ahmad, Sangwan, Pyare Lal, Qurishi, Mushtaq Ahmad, Koul, Surrinder, Saxena, Ajit Kumar, Shakeel-u-Rehman, (2014). Isolation, cytotoxicity evaluation and HPLC-quantification of the chemical constituents from Prangos pabularia. Plos One, 9(10), e108713. https://doi.org/10.1371/journal.pone.0108713
  • Farozi, A., Shah, S. A., & Banday, J. A. (2016). Structural and optical studies of 7-methoxy-8-(3-metrhylbut-2-enyl)-2-chromenone (osthol), a plant based coumarin. Optik, 127(5), 2802–2805. https://doi.org/10.1016/j.ijleo.2015.11.224
  • Fernández, J., Bert, F., & Nicolas-Chanoine, M.-H. (2016). The challenges of multi-drug-resistance in hepatology. Journal of Hepatology, 65(5), 1043–1054. https://doi.org/10.1016/j.jhep.2016.08.006
  • 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
  • Ge, R.-G., Wang, D.-X., Hao, M.-C., & Sun, X.-S. (2013). Nickel trafficking system responsible for urease maturation in Helicobacter pylori. World Journal of Gastroenterology, 19(45), 8211–8218. https://doi.org/10.3748/wjg.v19.i45.8211
  • Gholivand, M. B., Yamini, Y., Dayeni, M., & Shokoohinia, Y. (2015). The influence of the extraction mode on three coumarin compounds yield from Prangos ferulacea (L.) Lindl roots. Journal of the Iranian Chemical Society, 12(4), 707–714. https://doi.org/10.1007/s13738-014-0529-0
  • Hajlaoui, H., Mighri, H., Noumi, E., Snoussi, M., Trabelsi, N., Ksouri, R., & Bakhrouf, A. (2010). Chemical composition and biological activities of Tunisian Cuminum cyminum L. essential oil: A high effectiveness against Vibrio spp. strains. Food and Chemical Toxicology: An İnternational Journal Published for the British Industrial Biological Research Association, 48(8–9), 2186–2192. https://doi.org/10.1016/j.fct.2010.05.044
  • Hearing, V. J. (1999). Biochemical control of melanogenesis and melanosomal organization. Journal of Investigative Dermatology Symposium Proceedings: Elsevier, 4(1), 24–28. https://doi.org/10.1038/sj.jidsp.5640176
  • Hopkins, A. L., & Groom, C. R. (2002). The druggable genome. Nature Reviews. Drug Discovery, 1(9), 727–730. https://doi.org/10.1038/nrd892
  • Jalilian, F., Moieni-Arya, M., Hosseinzadeh, L., & Shokoohinia, Y. (2022). Oxypeucedanin and isoimperatorin extracted from Prangos ferulacea (L.) Lindl protect PC12 pheochromocytoma cells from oxidative stress and apoptosis induced by doxorubicin. Research in Pharmaceutical Sciences, 17(1), 12–21. https://doi.org/10.4103/1735-5362.329922
  • Jelodarian, Z., Shokoohinia, Y., Rashidi, M., Ghiasvand, N., Hosseinzadeh, L., & Iranshahi, M. (2017). New polyacetylenes from Echinophora cinerea (Boiss.) Hedge et Lamond. Natural Product Research, 31(19), 2256–2263. https://doi.org/10.1080/14786419.2017.1300797
  • Kansız, S., Azam, M., Basılı, T., Meral, S., Aktaş, F. A., Yeşilbağ, S., Min, K., Ağar, A. A., & Dege, N. (2022). Synthesis, structural studies, Hirshfeld surface analysis, and molecular docking studies of a thiophene-based Schiff base compound. Journal of Molecular Structure, 1265(, 133477. https://doi.org/10.1016/j.molstruc.2022.133477
  • Karakaya, S., Bingol, Z., Koca, M., Dagoglu, S., Pınar, N. M., Demirci, B., Gulcin, İ., Brestic, M., & Sytar, O. (2020). Identification of non-alkaloid natural compounds of Angelica purpurascens (Avé-Lall.) Gilli.(Apiaceae) with cholinesterase and carbonic anhydrase inhibition potential. Saudi Pharmaceutical Journal: SPJ: The Official Publication of the Saudi Pharmaceutical Society, 28(1), 1–14. https://doi.org/10.1016/j.jsps.2019.11.001
  • Khalid, M., Hussain, R., Hussain, A., Ali, B., Jaleel, F., Imran, M., Assiri, M. A., Khan, M. U., Ahmed, S., Abid, S., Haq, S., Saleem, K., Majeed, S., & Tariq, C. J. (2019). Electron donor and acceptor ınfluence on the nonlinear optical response of diacetylene-functionalized organic materials (DFOMs): Density functional theory calculations. Molecules (Basel, Switzerland), 24(11), 2096. https://doi.org/10.3390/molecules24112096
  • Kinghorn, A. D., Pan, L., Fletcher, J. N., & Chai, H. (2011). The relevance of higher plants in lead compound discovery programs. Journal of Natural Products, 74(6), 1539–1555. https://doi.org/10.1021/np200391c
  • Kitchen, D. B., Decornez, H., Furr, J. R., & Bajorath, J. (2004). Docking and scoring in virtual screening for drug discovery: Methods and applications. Nature Reviews. Drug Discovery, 3(11), 935–949. https://doi.org/10.1038/nrd1549
  • Koppula, S., & Choi, D. K. (2011). Cuminum cyminum extract attenuates scopolamine-induced memory loss and stress-induced urinary biochemical changes in rats: A noninvasive biochemical approach. Pharmaceutical Biology, 49(7), 702–708. https://doi.org/10.3109/13880209.2010.541923
  • Kumar, S., Saini, V., Maurya, I. K., Sindhu, J., Kumari, M., Kataria, R., & Kumar, V. (2018). Design, synthesis, DFT, docking studies and ADME prediction of some new coumarinyl linked pyrazolylthiazoles: Potential standalone or adjuvant antimicrobial agents. PloS One, 13(4), e0196016. https://doi.org/10.1371/journal.pone.0196016
  • Lee, C., Yang, W., & Parr, R. G. (1988). Development of the Colle-Salvetti correlation-energy formula into a functional of the electron density. Physical Review. B, Condensed Matter, 37(2), 785–789. 101103/physrevb.37.785. https://doi.org/10.1103/physrevb.37.785
  • Frisch, M. J., Schlegel, H. B., Scuseria, G. E., Robb, M. A., Cheeseman, J. R., Montgomery, J. A., Jr., Vreven, T., Kudin, K. N., Burant, J. C., Millam, J. M., Iyengar, S. S., Tomasi, J., Barone, V., Mennucci, B., Cossi, M., Scalmani, G., Rega, N., Petersson, G. A., Nakatsuji, H., Hada, M., Ehara, M., Toyota, K., Fukuda, R., Hasegawa, J., Ishida, M., Nakajima, T., Honda, Y., Kitao, O., Nakai, H., Klene, M., Li, X., Knox, J. E., Hratchian, H. P., Cross, J. B., Bakken, V., Adamo, C., Jaramillo, J., Gomperts, R., Stratmann, R. E., Yazyev, O., Austin, A. J., Cammi, R., Pomelli, C., Ochterski, J. W., Ayala, P. Y., Morokuma, K., Voth, G. A., Salvador, P., Dannenberg, J. J., Zakrzewski, V. G., Dapprich, S., Daniels, A. D., Strain, M. C., Farkas, O., Malick, D. K., Rabuck, A. D., Raghavachari, K., Foresman, J. B., Ortiz, J. V., Cui, Q., Baboul, A. G., Clifford, S., Cioslowski, J., Stefanov, B. B., Liu, G., Liashenko, A., Piskorz, P., Komaromi, I., Martin, R. L., Fox, D. J., Keith, T., Al-Laham, M. A., Peng, C. Y., Nanayakkara, A., Challacombe, M., Gill, P. M. W., Johnson, B., Chen, W., Wong, M. W., … Gonzalez, C. (2004). and J. A. Pople. Gaussian 03, Revision C.02
  • Maeda, K., & Fukuda, M. (1991). In vitro effectiveness of several whitening cosmetic components in human melanocytes. Journal of the Society of Cosmetic Chemists, 42(6), 361–368.
  • Massoulié, J., Pezzementi, L., Bon, S., Krejci, E., & Vallette, F.-M. (1993). Molecular and cellular biology of cholinesterases. Progress in Neurobiology, 41(1), 31–91. https://doi.org/10.1016/0301-0082(93)90040-y
  • Mettai, M., Daoud, I., Mesli, F., Kenouche, S., Melkemi, N., Kherachi, R., & Belkadi, A. (2023). Molecular docking/dynamics simulations, MEP analysis, bioisosteric replacement and ADME/T prediction for identification of dual targets inhibitors of Parkinson’s disease with novel scaffold. In Silico Pharmacology, 11(1), 3. https://doi.org/10.1007/s40203-023-00139-3
  • Mir, F. A., Bhat, G., Asokan, K., Batoo, K., & Banday, J. A. (2014). Crystal structure, morphological, optical and electrical investigations of Oxypeucedanin microcrystals: An isolated compound from a plant. Journal of Materials Science: Materials in Electronics, 25(1), 431–437. https://doi.org/10.1007/s10854-013-1606-3
  • Mir, F. A., & Khan, S. H. (2016). UV photoprotection of osthol. Radiation Effects and Defects in Solids, 171(11–12), 943–950. https://doi.org/10.1080/10420150.2016.1259240
  • Mottaghipisheh, J. (2021). Oxypeucedanin: Chemotaxonomy, isolation, and bioactivities. Plants, 10(8), 1577. https://doi.org/10.3390/plants10081577
  • Mrdaković, M., Ilijin, L., Vlahović, M., Matić, D., Gavrilović, A., Mrkonja, A., & Perić-Mataruga, V. (2016). Acetylcholinesterase (AChE) and heat shock proteins (Hsp70) of gypsy moth (Lymantria dispar L.) larvae in response to long-term fluoranthene exposure. Chemosphere, 159, 565–569. https://doi.org/10.1016/j.chemosphere.2016.06.059
  • 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
  • Murray, J. S., & Politzer, P. (2011). The electrostatic potential: An overview. WIREs Computational Molecular Science, 1(2), 153–163. https://doi.org/10.1002/wcms.19
  • Newman, D. J., & Cragg, G. M. (2012). Natural products as sources of new drugs over the 30 years from 1981 to 2010. Journal of Natural Products, 75(3), 311–335. https://doi.org/10.1021/np200906s
  • Numonov, S., Bobakulov, K., Numonova, M., Sharopov, F., Setzer, W. N., Khalilov, Q., Begmatov, N., Habasi, M., & Aisa, H. A. (2018). New coumarin from the roots of Prangos pabularia. Natural Product Research, 32(19), 2325–2332. https://doi.org/10.1080/14786419.2017.1413558
  • Oke Altuntas, F., Aslim, B., & Duman, H. (2016). The anti-lipid peroxidative, metal chelating, and radical scavenging properties of the fruit extracts from endemic prangos meliocarpoides Boiss var. meliocarpoides. Gazi University Journal of Science, 29(3), 537–542.
  • Orhan, I. E., Tosun, F., Deniz, F. S. S., Eren, G., Mıhoğlugil, F., Akalgan, D., & Miski, M. (2021). Butyrylcholinesterase-inhibiting natural coumarin molecules as potential leads. Phytochemistry Letters, 44, 48–54. https://doi.org/10.1016/j.phytol.2021.05.001
  • Özek, G., Bedir, E., Tabanca, N., Ali, A., Khan, I. A., Duran, A., Başer, K. H., & Özek, T. (2018). Isolation of eudesmane type sesquiterpene ketone from Prangos heyniae H. Duman & MF Watson essential oil and mosquitocidal activity of the essential oils. Open Chemistry, 16(1), 453–467. https://doi.org/10.1515/chem-2018-0051
  • Pinzi, L., & Rastelli, G. (2019). Molecular docking: Shifting paradigms in drug discovery. International Journal of Molecular Sciences, 20(18), 4331. https://doi.org/10.3390/ijms20184331
  • Politzer, P., Murray, J. S., & Peralta, I. Z. (2001). Molecular surface electrostatic potentials in relation to noncovalent interactions in biological systems. International Journal of Quantum Chemistry, 85(6), 676–684. https://doi.org/10.1002/qua.1706
  • Raftani, M., Abram, T., Azaid, A., Kacimi, R., Bennani, M. N., & Bouachrine, M. (2021). Theoretical design of new organic compounds based on diketopyrrolopyrrole and phenyl for organic bulk heterojunction solar cell applications: DFT and TD-DFT study. Materials Today: Proceedings, 45, 7334–7343. https://doi.org/10.1016/j.matpr.2020.12.1228
  • Rather, Z., Banday, J., & Chisti, H. (2021). Synthesis, characterization, and cytotoxic evaluation of new triazole derivatives of osthol. Russian Journal of Organic Chemistry, 57(6), 986–993. https://doi.org/10.1134/S1070428021060154
  • Rather, Z., Banday, J. A., & Chisti, H. T. N. (2020). Spectroscopic, X‐ray crystal, DFT and ın vitro analysis of 3‐(2, 4‐dimethoxy‐3‐(3‐methylbut‐2‐en‐1‐yl) phenyl) acrylic acid. ChemistrySelect, 5(26), 7849–7854.
  • Razavi, S. M., Nazemiyeh, H., Delazar, A., Hajiboland, R., Rahman, M. M., Gibbons, S., Nahar, L., & Sarker, S. D. (2008). Coumarins from the roots of Prangos uloptera. Phytochemistry Letters, 1(3), 159–162. https://doi.org/10.1016/j.phytol.2008.07.009
  • Razavi, S. M., Zahri, S., Motamed, Z., & Ghasemi, G. (2010). Bioscreening of oxypeucedanin, a known furanocoumarin.
  • Re, R., Pellegrini, N., Proteggente, A., Pannala, A., Yang, M., & Rice-Evans, C. (1999). Antioxidant activity applying an improved ABTS radical cation decolorization assay. Free Radical Biology & Medicine, 26(9–10), 1231–1237. https://doi.org/10.1016/s0891-5849(98)00315-3
  • Rossolini, G. M., Arena, F., Pecile, P., & Pollini, S. (2014). Update on the antibiotic resistance crisis. Current Opinion in Pharmacology, 18, 56–60. https://doi.org/10.1016/j.coph.2014.09.006
  • Saadon, K. E., Taha, N. M. H., Mahmoud, N. A., Elhagali, G. A. M., & Ragab, A. (2022). Synthesis, characterization, and in vitro antibacterial activity of some new pyridinone and pyrazole derivatives with some in silico ADME and molecular modeling study. Journal of the Iranian Chemical Society, 19(9), 3899–3917. https://doi.org/10.1007/s13738-022-02575-y
  • Sajadi, S., Zeynivand, H., & Shokouhinia, Y. (2009). Isolation and identification of osthol from the fruits and essential oil composition of the leaves of Prangos asperula Boiss.
  • Salih, K. S. (2021). Synthesis, characterization, surface analysis, optical activity and solvent effects on the electronic absorptions of Schiff base-functionalized amino thiophene derivatives: Experimental and TD-DFT investigations. Journal of Molecular Structure, 1244, 131267. https://doi.org/10.1016/j.molstruc.2021.131267
  • Saravanan, R. R., Seshadri, S., Gunasekaran, S., Mendoza-Meroño, R., & Garcia-Granda, S. (2015). Conformational analysis, X-ray crystallographic, FT-IR, FT-Raman, DFT, MEP and molecular docking studies on 1-(1-(3-methoxyphenyl) ethylidene) thiosemicarbazide. Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 139, 321–328. https://doi.org/10.1016/j.saa.2014.12.026
  • Schwöbel, J., Ebert, R.-U., Kühne, R., & Schüürmann, G. (2009). Prediction of the intrinsic hydrogen bond acceptor strength of chemical substances from molecular structure. The Journal of Physical Chemistry. A, 113(37), 10104–10112. https://doi.org/10.1021/jp904812b
  • Seiberg, M., Paine, C., Sharlow, E., Eisinger, M., Shapiro, S. S., Andrade-Gordon, P., & Costanzo, M. (2000). Inhibition of melanosome transfer results in skin lightening1. The Journal of İnvestigative Dermatology, 115(2), 162–167. https://doi.org/10.1046/j.1523-1747.2000.00035.x
  • Sevin, G., Alan, E., Demir, S., Albayrak, G., Demiroz, T., Yetik-Anacak, G., & Baykan, S. (2022). Comparative evaluation of relaxant effects of three Prangos species on mouse corpus cavernosum: Chemical characterization and the relaxant mechanisms of action of P. pabularia and (+)-oxypeucedanin. Journal of Ethnopharmacology, 284, 114823. https://doi.org/10.1016/j.jep.2021.114823
  • Sharma, D., & Tiwari, S. N. (2016). Comparative computational analysis of electronic structure, MEP surface and vibrational assignments of a nematic liquid crystal: 4-n-methyl-4’-cyanobiphenyl. Journal of Molecular Liquids, 214, 128–135. https://doi.org/10.1016/j.molliq.2015.11.045
  • Shokoohinia, Y., Hosseinzadeh, L., Alipour, M., Mostafaie, A., & Mohammadi-Motlagh, H.-R. (2014). Comparative evaluation of cytotoxic and apoptogenic effects of several coumarins on human cancer cell lines: Osthole induces apoptosis in p53-deficient H1299 cells. Advances in Pharmacological Sciences, 2014, 847574–847578. https://doi.org/10.1155/2014/847574
  • Shokoohinia, Y., Jafari, F., Mohammadi, Z., Bazvandi, L., Hosseinzadeh, L., Chow, N., Bhattacharyya, P., Farzaei, M. H., Farooqi, A. A., Nabavi, S. M., Yerer, M. B., & Bishayee, A. (2018). Potential anticancer properties of osthol: A comprehensive mechanistic review. Nutrients, 10(1), 36. https://doi.org/10.3390/nu10010036
  • Shokoohinia, Y., Sajjadi, S.-E., Gholamzadeh, S., Fattahi, A., & Behbahani, M. (2014). Antiviral and cytotoxic evaluation of coumarins from Prangos ferulacea. Pharmaceutical Biology, 52(12), 1543–1549. https://doi.org/10.3109/13880209.2014.907322
  • Stavri, M., & Gibbons, S. (2005). The antimycobacterial constituents of dill (Anethum graveolens). Phytotherapy Research, 19(11), 938–941. https://doi.org/10.1002/ptr.1758
  • Taslimi, P., Caglayan, C., Farzaliyev, V., Nabiyev, O., Sujayev, A., Turkan, F., Kaya, R., & Gulçin, İ. (2018). Synthesis and discovery of potent carbonic anhydrase, acetylcholinesterase, butyrylcholinesterase, and α‐glycosidase enzymes inhibitors: The novel N, N′‐bis‐cyanomethylamine and alkoxymethylamine derivatives. Journal of Biochemical and Molecular Toxicology, 32(4), e22042. https://doi.org/10.1002/jbt.22042
  • 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
  • Velavan, S. (2015). Phytochemical techniques-a review. World Journal of Science and Research, 1(2), 80–91.
  • Wei, Y., & Ito, Y. (2006). Preparative isolation of imperatorin, oxypeucedanin and isoimperatorin from traditional Chinese herb “bai zhi” Angelica dahurica (Fisch. ex Hoffm) Benth. et Hook using multidimensional high-speed counter-current chromatography. Journal of Chromatography. A, 1115(1–2), 112–117. https://doi.org/10.1016/j.chroma.2006.02.081
  • WHO. (2013). WHO traditional medicine strategy: 2014–2023. World Health Organization.
  • Xiao, J., Wang, J., Yuan, L., Hao, L., & Wang, D. (2019). Study on the mechanism and intervention strategy of sunitinib induced nephrotoxicity. European Journal of Pharmacology, 864, 172709. https://doi.org/10.1016/j.ejphar.2019.172709
  • Yakan, H., Cakmak, S., Kutuk, H., Yenigun, S., & Ozen, T. (2020). Synthesis, characterization, antioxidant, and antibacterial activities of new 2, 3-dimethoxy and 3-acetoxy-2-methyl benzamides. Research on Chemical Intermediates, 46(5), 2767–2787. https://doi.org/10.1007/s11164-020-04118-7
  • Yazici-Tütüniş, S., Eruçar, F. M., Öztaş, E., Akalin, E., Özhan, G., Miski, M., & Tan, N. (2021). Chemical composition and cytotoxic effect of Prangos turcica A. Duran, M. Sagiroglu & H. Duman. Records of Natural Products, 15(6), 503–512. https://doi.org/10.25135/rnp.235.21.02.1991
  • Yenigün, S., Başar, Y., İpek, Y., Behçet, L., Özen, T., & Demirtaş, İ. (2023a). 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, 1–18. https://doi.org/10.1080/07391102.2023.2229440 (Early Access)
  • Yenigun, S., Ipek, Y., Marah, S., Demirtas, I., & Ozen, T. (2023b). DNA protection, molecular docking, antioxidant, antibacterial, enzyme inhibition, and enzyme kinetic studies for parietin, isolated from Xanthoria parietina (L.) Th. Fr. Journal of Biomolecular Structure & Dynamics, 1–15. https://doi.org/10.1080/07391102.2023.2196693 (Early Access)
  • 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): implications 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
  • Zheng, M.-C., Tang, W.-T., Yu, L.-L., Qian, X.-J., Ren, J., Li, J.-J., Rong, W.-W., Li, J.-X., & Zhu, Q. (2022). Preclinical pharmacokinetics and bioavailability of oxypeucedanin in rats after single ıntravenous and oral administration. Molecules (Basel, Switzerland), 27(11), 3570. https://doi.org/10.3390/molecules27113570

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.