442
Views
0
CrossRef citations to date
0
Altmetric
Research articles

A new bio-active asymmetric-Schiff base: synthesis and evaluation of calf thymus DNA interaction, topoisomerase IIα inhibition, in vitro antiproliferative activity, SEM analysis and molecular docking studies

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 2804-2822 | Received 31 Mar 2021, Accepted 02 Feb 2022, Published online: 18 Feb 2022

References

  • Aboafia, S. A., Elsayed, S. A., El-Sayed, A. K. A., & El-Hendawy, A. M. (2018). New transition metal complexes of 2,4-dihydroxybenzaldehyde benzoylhydrazone Schiff base (H2dhbh): Synthesis, spectroscopic characterization, DNA binding/cleavage and antioxidant activity. Journal of Molecular Structure, 1158, 39–50. https://doi.org/10.1016/j.molstruc.2018.01.008
  • Afsan, Z., Roisnel, T., Tabassum, S., & Arjmand, F. (2020). Structure elucidation {spectroscopic, single crystal X-ray diffraction and computational DFT studies} of new tailored benzenesulfonamide derived Schiff base copper(II) intercalating complexes: Comprehensive biological profile {DNA binding, pBR322 DNA cleavage, Topo I inhibition and cytotoxic activity}. Bioorganic Chemistry, 94, 103427. https://doi.org/10.1016/j.bioorg.2019.103427
  • Ali, I., Mahmood, L. M. A., Mehdar, Y. T. H., Aboul-Enein, H. Y., & Said, M. A. (2020). Synthesis, characterization, simulation, DNA binding and anticancer activities of Co(II), Cu(II), Ni(II) and Zn(II) complexes of a Schiff base containing o-hydroxyl group nitrogen ligand. Inorganic Chemistry Communications, 118, 108004.) https://doi.org/10.1016/j.inoche.2020.108004
  • Al-Khathami, N. D., Al-Rashdi, K. S., Babgi, B. A., Hussien, M. A., Nadeem Arshad, M., Eltayeb, N. E., Elsilk, S. E., Lasri, J., Basaleh, A. S., & Al-Jahdali, M. (2019). Spectroscopic and biological properties of platinum complexes derived from 2-pyridyl Schiff bases. Journal of Saudi Chemical Society, 23(7), 903–915. https://doi.org/10.1016/j.jscs.2019.03.004
  • Arjmand, F., Parveen, S., Afzal, M., Toupet, L., & Hadda, T. B. (2012). Molecular drug design, synthesis and crystal structure determination of CuII-SnIV heterobimetallic core: DNA binding and cleavage studies. European Journal of Medicinal Chemistry, 49, 141–150. https://doi.org/10.1016/j.ejmech.2012.01.005
  • Arjmand, F., Sharma, G. C., Muddassir, M., & Tabassum, S. (2011). Synthesis and enantiopreferential DNA-binding profile of late 3d transition metal R- and S-enantiomeric complexes derived from N,N-bis-(1-benzyl-2-ethoxyethane): Validation of R-enantiomer of copper(II) complex as a human topoisomerase II inhibitor. Chirality, 23(7), 557–567. https://doi.org/10.1002/chir.20970
  • Arslantas, A., & Agirtas, M. S. (2017). Investigation of DNA-Binding Activities of Zinc(II) and Cobalt(II) Phthalocyanine Compounds with 3,4,5-Trimethoxybenzyloxy Substituents. ChemistrySelect, 2(35), 11659–11665. https://doi.org/10.1002/slct.201702362
  • Baginski, M., Fogolari, F., & Briggs, J. M. (1997). Electrostatic and non-electrostatic contributions to the binding free energies of anthracycline antibiotics to DNA. Journal of Molecular Biology, 274(2), 253–267.
  • Bailon-Moscoso, N., Romero-Benavides, J. C., & Ostrosky-Wegman, P. (2014). Development of anticancer drugs based on the hallmarks of tumor cells. Tumour Biology : The Journal of the International Society for Oncodevelopmental Biology and Medicine, 35(5), 3981–3995. https://doi.org/10.1007/s13277-014-1649-y
  • Barros, F. W., Bandeira, P. N., Lima, D. J., Meira, A. S., de Farias, S. S., Albuquerque, M. R. J., Santos, J. R. M., Lemos, S. H., Morais, G. L. T., Costa-Lotufo, O. M., & Pessoa, C. d O. (2011). Amyrin esters induce cell death by apoptosis in HL-60 leukemia cells. Bioorganic & Medicinal Chemistry, 19(3), 1268–1276. https://doi.org/10.1016/j.bmc.2010.12.016
  • BIOVIA. (2021). Dassault Systèmes, discovery studio, release. Dassault Systèmes.
  • Borowska, J., Sierant, M., Sochacka, E., Sanna, D., & Lodyga-Chruscinska, E. (2015). DNA binding and cleavage studies of copper(II) complexes with 2'-deoxyadenosine modified histidine moiety. Journal of Biological Inorganic Chemistry : JBIC : a Publication of the Society of Biological Inorganic Chemistry, 20(6), 989–1004. https://doi.org/10.1007/s00775-015-1282-2
  • Drew, H. R., Wing, R. M., Takano, T., Broka, C., Tanaka, S., Itakura, K., & Dickerson, R. E. (1981). Structure of a B-DNA dodecamer: conformation and dynamics. Proceedings of the National Academy of Sciences of the United States of America, 78(4), 2179–2183. https://doi.org/10.1073/pnas.78.4.2179
  • Fenech, M. (2000). The in vitro micronucleus technique. Mutation Research, 455(1-2), 81–95. https://doi.org/10.1016/S0027-5107(00)00065-8
  • Fenech, M. (2007). Cytokinesis-block micronucleus cytome assay. Nature Protocols, 2(5), 1084–1104. https://doi.org/10.1038/nprot.2007.77
  • Fenech, M., Kirsch-Volders, M., Natarajan, A. T., Surralles, J., Crott, J. W., Parry, J., Norppa, H., Eastmond, D. A., Tucker, J. D., & Thomas, P. (2011). Molecular mechanisms of micronucleus, nucleoplasmic bridge and nuclear bud formation in mammalian and human cells. Mutagenesis, 26(1), 125–132. https://doi.org/10.1093/mutage/geq052
  • Forli, S., Huey, R., Pique, M. E., Sanner, M. F., Goodsell, D. S., & Olson, A. J. (2016). Computational protein-ligand docking and virtual drug screening with the AutoDock suite. Nature Protocols, 11(5), 905–919. https://doi.org/10.1038/nprot.2016.051
  • Guan, F., Ding, Y., Zhang, Y., Zhou, Y., Li, M., & Wang, C. (2016). Curcumin suppresses proliferation and migration of MDA-MB-231 breast cancer cells through autophagy-dependent akt degradation. PloS One, 11(1), e0146553. https://doi.org/10.1371/journal.pone.0146553
  • Guerrant, W., Patil, V., Canzoneri, J. C., & Oyelere, A. K. (2012). Dual targeting of histone deacetylase and topoisomerase II with novel bifunctional inhibitors. Journal of Medicinal Chemistry, 55(4), 1465–1477. https://doi.org/10.1021/jm200799p
  • Gultneh, Y., Khan, A. R., Blaise, D., Chaudhry, S., Ahvazi, B., Marvey, B. B., & Butcher, R. J. (1999). Syntheses and structures of and catalysis of hydrolysis by Zn(II) complexes of chelating pyridyl donor ligands. Journal of Inorganic Biochemistry, 75(1), 7–18. https://doi.org/10.1016/S0162-0134(99)00025-2
  • Güngör, Ö. Ö. (2017). Intramolecular proton transfer equilibrium in salicylidene- and naphthalene-based tetraimine Schiff bases. Gazi University Journal of Science, 30, 191–214.
  • Güngör, O., & Gürkan, P. (2010). Synthesis and spectroscopic properties of novel asymmetric Schiff bases. Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 77(1), 304–311. https://doi.org/10.1016/j.saa.2010.05.027
  • Güngör, Ö., & Gürkan, P. (2019). Potentiometric and antimicrobial studies on the asymmetric Schiff bases and their binuclear Ni(II) and Fe(III) complexes; synthesis and characterization of the complexes. Arabian Journal of Chemistry, 12(8), 2244–2256. https://doi.org/10.1016/j.arabjc.2015.02.009
  • Hernandez, A. M., Rodriguez, N., Gonzalez, J. E., Reyes, E., Rondon, T., Grinan, T., Macias, A., Alfonso, S., Vazquez, M. A., & Perez, R. (2011). Anti-NeuGcGM3 antibodies, actively elicited by idiotypic vaccination in nonsmall cell lung cancer patients, induce tumor cell death by an oncosis-like mechanism. The Journal of Immunology, 186(6), 3735–3744. https://doi.org/10.4049/jimmunol.1000609
  • Ibrahim, M. N., Sharif, S. A. I., & Chem, E.-J. (2011). E-Journal of Chemistry, 8(1), 180–184. https://doi.org/10.1155/2011/821616
  • Jarvinen, T. A., & Liu, E. T. (2006). Simultaneous amplification of HER-2 (ERBB2) and topoisomerase IIalpha (TOP2A) genes-molecular basis for combination chemotherapy in cancer. Current Cancer Drug Targets, 6(7), 579–602. https://doi.org/10.2174/156800906778742497
  • Jaswal, S., Nehra, B., Kumar, S., & Monga, V. (2020). Recent advancements in the medicinal chemistry of bacterial type II topoisomerase inhibitors. Bioorganic Chemistry, 104, 104266. https://doi.org/10.1016/j.bioorg.2020.104266
  • Jeyalakshmi, K., Selvakumaran, N., Bhuvanesh, N. S. P., Sreekanth, A., & Karvembu, R. (2014). DNA/protein binding and cytotoxicity studies of copper(ii) complexes containing N,N′,N′′-trisubstituted guanidine ligands. RSC Advances, 4(33), 17179–17195. https://doi.org/10.1039/c4ra01459f
  • Jun, K. Y., Lee, E. Y., Jung, M. J., Lee, O. H., Lee, E. S., Choo, H. Y. P., Na, Y., & Kwon, Y. (2011). Synthesis, biological evaluation, and molecular docking study of 3-(3'-heteroatom substituted-2'-hydroxy-1'-propyloxy) xanthone analogues as novel topoisomerase IIα catalytic inhibitor. European Journal of Medicinal Chemistry, 46(6), 1964–1971. https://doi.org/10.1016/j.ejmech.2011.01.011
  • Karatas, E., Foto, E., Ertan-Bolelli, T., Yalcin-Ozkat, G., Yilmaz, S., Ataei, S., Zilifdar, F., & Yildiz, I. (2021). Discovery of 5-(or 6)-benzoxazoles and oxazolo[4,5-b]pyridines as novel candidate antitumor agents targeting hTopo IIα. Bioorganic Chemistry, 112, 104913. https://doi.org/10.1016/j.bioorg.2021.104913
  • Keypour, H., Forouzandeh, F., Salehzadeh, S., Hajibabaei, F., Feizi, S., Karamian, R., Ghiasi, N., & Gable, R. W. (2019). DNA binding studies and antibacterial properties of a new Schiff base ligand containing homopiperazine and products of its reaction with Zn(II), Cu(II) and Co(II) metal ions: X-ray crystal structure of Cu(II) and Zn(II) complexes. Polyhedron, 170, 584–592. https://doi.org/10.1016/j.poly.2019.06.023
  • Konkolova, E., Janockova, J., Perjesi, P., Vaskova, J., & Kozurkova, M. (2018). Selected ferrocenyl chalcones as DNA/BSA-interacting agents and inhibitors of DNA topoisomerase I and II activity. Journal of Organometallic Chemistry, 861, 1–9.
  • Kraicheva, I., Tsacheva, I., Vodenicharova, E., Tashev, E., Tosheva, T., Kril, A., Topashka-Ancheva, M., Iliev, I., Gerasimova, T., & Troev, K. (2012). Synthesis, antiproliferative activity and genotoxicity of novel anthracene-containing aminophosphonates and a new anthracene-derived Schiff base. Bioorganic & Medicinal Chemistry, 20(1), 117–124. https://doi.org/10.1016/j.bmc.2011.11.024
  • Kumar, A., Bansal, D., Bajaj, K., Sharma, S., & Srivastava, V. K. (2003). Synthesis of some newer derivatives of 2-amino benzoic acid as potent anti-inflammatory and analgesic agents. Bioorganic & Medicinal Chemistry, 11, 5281–5291. https://doi.org/10.1016/S0968-0896(03)00529-7
  • Larsson, D. E., Wickström, M., Hassan, S., Öberg, K., & Granberg, D. A. N. (2010). The cytotoxic agents NSC-95397, brefeldin A, bortezomib and sanguinarine induce apoptosis in neuroendocrine tumors in vitro. Anticancer Research, 30(1), 149–156.
  • Lee, M. P., Sander, M., & Hsieh, T. S. (1989). Single strand DNA cleavage reaction of duplex DNA by Drosophila topoisomerase II. The Journal of Biological Chemistry, 264(23), 13510–13518. https://doi.org/10.1016/S0021-9258(18)80026-6
  • Liang, Y. S., Qi, W. T., Guo, W., Wang, C. L., Hu, Z. B., & Li, A. K. (2018). Genistein and daidzein induce apoptosis of colon cancer cells by inhibiting the accumulation of lipid droplets. Food & Nutrition Research, 62, 1384.
  • Li, D. D., Tian, J. L., Gu, W., Liu, X., Zeng, H. H., & Yan, S. P. (2011). DNA binding, oxidative DNA cleavage, cytotoxicity, and apoptosis-inducing activity of copper(II) complexes with 1,4-tpbd (N,N,N',N'-tetrakis(2-yridylmethyl)benzene-1,4-diamine) ligand. Journal of Inorganic Biochemistry, 105(6), 894–901. https://doi.org/10.1016/j.jinorgbio.2011.03.012
  • Liu, F., Gao, S., Yang, Y., Zhao, X., Fan, Y., Ma, W., Yang, D., Yang, A., & Yu, Y. (2017). Curcumin induced autophagy anticancer effects on human lung adenocarcinoma cell line A549. Oncology Letters, 14(3), 2775–2782. https://doi.org/10.3892/ol.2017.6565
  • Liu, Y., Ma, X. Q., Jin, P., Li, H. T., Zhang, R. R., Ren, X. L., Wang, H. B., Tang, D., & Tian, W. R. (2011). Apoptosis induced by hematoporphyrin monomethyl ether combined with He-Ne laser irradiation in vitro on canine breast cancer cells. Veterinary Journal (London, England : 1997), 188(3), 325–330. https://doi.org/10.1016/j.tvjl.2010.05.013
  • Li, Q., Yang, P., Wang, H., & Guo, M. (1996). Diorganotin(IV) antitumor agent. (C2H5)2SnCl2 (phen)/nucleotides aqueous and solid-state coordination chemistry and its DNA binding studies. Journal of Inorganic Biochemistry, 64(3), 181–195. https://doi.org/10.1016/0162-0134(96)00039-6
  • Mariappan, M., Suenaga, M., Mukhopadhyay, A., & Maiya, B. G. (2012). Synthesis, structure, DNA binding and photonuclease activity of a nickel(II) complex with a N,N′-Bis(salicylidene)-9-(3,4-diaminophenyl)acridine ligand. Inorganica Chimica Acta, 390, 95–104. https://doi.org/10.1016/j.ica.2012.04.016
  • Marmur, J. (1961). A procedure for the isolation of deoxyribonucleic acid from micro-organisms. Journal of Molecular Biology, 3(2), 208–218. https://doi.org/10.1016/S0022-2836(61)80047-8
  • Marvin. (20.2). 1.0, 2020, ChemAxon (http://www.chemaxon.com)
  • Morita, H., He, F., Fuse, T., Ouwehand, A. C., Hashimoto, H., Hosoda, M., Mizumachi, K., & Kurisaki, J. I. (2002). Adhesion of lactic acid bacteria to caco-2 cells and their effect on cytokine secretion. Microbiology and Immunology, 46(4), 293–297. https://doi.org/10.1111/j.1348-0421.2002.tb02698.x
  • Morris, G. M., Huey, R., Lindstrom, W., Sanner, M. F., Belew, R. K., Goodsell, D. S., & Olson, A. J. (2009). AutoDock4 and AutoDockTools4: Automated docking with selective receptor flexibility. Journal of Computational Chemistry, 30(16), 2785–2791. https://doi.org/10.1002/jcc.21256
  • 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
  • Nartop, D., Gürkan, P., Sarı, N., & Çete, S. (2008). Tetradentate asymmetric Schiff bases and their Ni(II) and Fe(III) complexes. Journal of Coordination Chemistry, 61(21), 3516–3524. https://doi.org/10.1080/00958970802090130
  • Nartop, D., & Öğütcü, H. (2020). Novel unsymmetric diimines: Synthesis and biological evaluation against pathogenic microorganisms. Journal of Medicinal and Chemical Sciences, 3, 219–227.
  • Nartop, D., & Öğütçü, H. (2020). Synthesis of new unsymmetrical Schiff bases as potential antimicrobial agents. Sinop University Journal of Natural Sciences, 5, 13–25.
  • Nartop, D., Özdemir, Ö., & Gürkan, P. (2017). Synthesis, characterization and investigation of tautomeric, potentiometric and antimicrobial properties of a novel unsymmetric Schiff base and its Fe(III) and Ni(II) complexes. Moroccan Journal of Chemistry, 5, 560–572.
  • Nartop, D., Özkan, E. H., Kızıl, H. E., Ağar, G., Sarı, N., Düzce, U. J., & Sci Technol, 8. (2020). Evulation of antimutagenic activity of Ni(II) complexes with unsymmetric Schiff bases. Düzce University Journal of Science Technology, 8, 608–616. https://doi.org/10.29130/dubited.575747
  • O’Boyle, N. M., Banck, M., James, C. A., Morley, C., Vandermeersch, T., & Hutchison, G. R. (2011). Open Babel: An open chemical toolbox. Journal of Cheminformatics, 3, 33.
  • OECD. (2010). OECD guideline for testing of chemicals. No. 487: In vitro mammalian cell micronucleus test. Organization for Economic Cooperation and Development. (French).
  • Özdemir, Ö., Gürkan, P., Şimay Demir, Y. D., & Ark, M. (2020). Novel palladium(II) complexes of N-(5-nitro-salicylidene)-Schiff bases: Synthesis, spectroscopic characterization and cytotoxicity investigation. Journal of Molecular Structure, 1207, 127852. https://doi.org/10.1016/j.molstruc.2020.127852
  • Özdemir, Ö., Gürkan, P., Şimay Demir, Y. D., & Ark, M., (2020). Antioxidant and cytotoxic activity studies in series of higher amino acid Schiff bases. Gazi University Journal of Science, 33, 646–660.
  • Özyurt, N., Candan, S., & Suludere, Z. (2013). The morphology and histology of the male reproductive system in Dolycoris baccarum Linnaeus 1758 (Heteroptera: Pentatomidae)-light and scanning electron micoscope studies. Micron (Oxford, England: 1993), 44, 101–106. https://doi.org/10.1016/j.micron.2012.04.017
  • Peng, C., Muthusamy, S., Xia, Q., Lal, V., Denison, M. S., & Ng, J. C. (2015). Micronucleus formation by single and mixed heavy metals/loids and PAH compounds in HepG2 cells. Mutagenesis, 30(5), 593–602. https://doi.org/10.1093/mutage/gev021
  • Pepe, G., Grossi, M. R., Berni, A., Filippi, S., Shanmugakani, R. K., Papeschi, C., Mosesso, P., Natarajan, A. T., & Palitti, F. (2013). Effect of blueberries (BB) on micronuclei induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) and 7,12-dimethylbenz(a)anthracene (DMBA) in mammalian cells, assessed in in vitro and in vivo assays. Mutation Research. Genetic Toxicology and Environmental Mutagenesis, 758(1-2), 6–11. https://doi.org/10.1016/j.mrgentox.2013.07.012
  • Pravin, N., Utthra, P. P., Kumaravel, G., & Raman, N. (2016). Effective DNA binding and cleaving tendencies of malonic acid coupled transition metal complexes. Journal of Molecular Structure, 1123, 162–170. https://doi.org/10.1016/j.molstruc.2016.06.034
  • Proudfoot, E. M., Mackay, J. P., & Karuso, P. (2001). Probing Site Specificity of DNA Binding Metallointercalators by NMR Spectroscopy and Molecular Modeling. Biochemistry, 40(15), 4867–4878. ehttps://doi.org/10.1021/bi001655f
  • Pyle, A. M., Rehmann, J. P., Meshoyrer, R., Kumar, C. V., Turro, N. J., & Barton, J. K. (1989). Mixed-ligand complexes of ruthenium(II): factors governing binding to DNA. Journal of the American Chemical Society, 111(8), 3051–3058. https://doi.org/10.1021/ja00190a046
  • Ramesh, G., Daravath, S., Ganji, N., Rambabu, A., & Venkateswarlu, K. (2020). Facile synthesis, structural characterization, DNA binding, incision evaluation, antioxidant and antimicrobial activity studies of Cobalt(II), Nickle(II) and Copper(II) complexes of 3-amino-5-(4-fluorophenyl)isoxazole derivatives. Journal of Molecular Structure, 1202, 127338. https://doi.org/10.1016/j.molstruc.2019.127338
  • Rao, J., Xu, D. R., Zheng, F. M., Long, Z. J., Huang, S. S., Wu, X., Zhou, W. H., Huang, R. W., & Liu, Q. (2011). Curcumin reduces expression of Bcl-2, leading to apoptosis in daunorubicin-insensitive CD34+ acute myeloid leukemia cell lines and primary sorted CD34+ acute myeloid leukemia cells. Journal of Translational Medicine, 9, 71. https://doi.org/10.1186/1479-5876-9-71
  • Reytman, L., Braitbard, O., & Tshuva, E. Y. (2012). Highly cytotoxic vanadium(V) complexes of salan ligands; insights on the role of hydrolysis. Dalton Transactions (Cambridge, England: 2003), 41(17), 5241–5247. https://doi.org/10.1039/c2dt11514j
  • Rubner, G., Bensdorf, K., Wellner, A., Bergemann, S., Ott, I., & Gust, R. (2010). [Cyclopentadienyl]metalcarbonyl complexes of acetylsalicylic acid as neo-anticancer agents. European Journal of Medicinal Chemistry, 45(11), 5157–5163. https://doi.org/10.1016/j.ejmech.2010.08.028
  • Sandhaus, S., Taylor, R., Edwards, T., Huddleston, A., Wooten, Y., Venkatraman, R., Weber, R. T., Gonzalez-Sarrias, A., Martin, P. M., Cagle, P., Tse-Dinh, Y. C., Beebe, S. J., Seeram, N., & Holder, A. A. (2016). A novel copper(II) complex identified as a potent drug against colorectal and breast cancer cells and as a poison inhibitor for human topoisomerase IIα. Inorganic Chemistry Communications, 64, 45–49. https://doi.org/10.1016/j.inoche.2015.12.013
  • Scheffler, H., You, Y., & Ott, I. (2010). Comparative studies on the cytotoxicity, cellular and nuclear uptake of a series of chloro gold(I) phosphine complexes. Polyhedron, 29(1), 66–69. https://doi.org/10.1016/j.poly.2009.06.007
  • Sedighipoor, M., Kianfar, A. H., Sabzalian, M. R., & Abyar, F. (2018). Synthesis and characterization of new unsymmetrical Schiff base Zn (II) and Co (II) complexes and study of their interactions with bovin serum albumin and DNA by spectroscopic techniques. Spectrochimica Acta. Part A, Molecular and Biomolecular Spectroscopy, 198, 38–50. https://doi.org/10.1016/j.saa.2018.02.050
  • Shahabadi, N., Kashanian, S., & Darabi, F. (2009). In vitro study of DNA interaction with a water-soluble dinitrogen schiff base. DNA and Cell Biology, 28(11), 589–596. https://doi.org/10.1089/dna.2009.0881
  • Shahabadi, N., Kashanian, S., & Fatahi, A. (2011). Identification of Binding Mode of a Platinum (II) Complex, PtCl(2)(DIP), and Calf Thymus DNA. Bioinorganic Chemistry and Applications, 2011, 687571. https://doi.org/10.1155/2011/687571
  • Shi, S., Liu, J., Li, J., Zheng, K. C., Huang, X. M., Tan, C. P., Chen, L. M., & Ji, L. N. (2006). Synthesis, characterization and DNA-binding of novel chiral complexes delta- and lambda-[Ru(bpy)2L]2+ (L = o-mopip and p-mopip). Journal of Inorganic Biochemistry, 100(3), 385–395. https://doi.org/10.1016/j.jinorgbio.2005.12.005
  • Shinojima, N., Yokoyama, T., Kondo, Y., & Kondo, S. (2007). Roles of the Akt/mTOR/p70S6K and ERK1/2 signaling pathways in curcumin-induced autophagy. Autophagy, 3(6), 635–637. https://doi.org/10.4161/auto.4916
  • Shivananjappa, M. M., & Joshi, M. K. (2011). Influence of aqueous extract of Arjuna (Terminalia arjuna) on growth and antioxidant defense system of human hepatoma cell line (HepG2). Journal of Medicinal Plants Research, 5, 1711–1721.
  • Stockert, A., Kinder, D., Christ, M., Amend, K., & Aulthouse, A. (2014). Improving the efficacy of cisplatin in colon cancer HT-29 cells via combination therapy with selenium. Austin Journal of Pharmacology and Therapeutics, 2, id1013.
  • Suludere, Z., Canbulat, S., & Candan, S. (2009). External morphology of eggs of Macronemurus bilineatus and Megistopus flavicornis (Neuroptera, Myrmeleontidae): a scanning electron microscopy study. Turkish Journal of Zoology, 33, 387–392.
  • Suzuki, K., & Uyeda, M. (2002). Inhibitory properties of antitumor prostaglandins against topoisomerases. Bioscience, Biotechnology, and Biochemistry, 66(8), 1706–1712. https://doi.org/10.1271/bbb.66.1706
  • Symonds, E. L., Konczak, I., & Fenech, M. (2013). The Australian fruit Illawarra plum (Podocarpus elatus Endl., Podocarpaceae) inhibits telomerase, increases histone deacetylase activity and decreases proliferation of colon cancer cells. British Journal of Nutrition, 109(12), 2117–2125. https://doi.org/10.1017/S0007114512004333
  • Tarantini, A., Lanceleur, R., Mourot, A., Lavault, M. T., Casterou, G., Jarry, G., Hogeveen, K., & Fessard, V. (2015). Toxicity, genotoxicity and proinflammatory effects of amorphous nanosilica in the human intestinal Caco-2 cell line. Toxicology in Vitro: An International Journal Published in Association with BIBRA, 29(2), 398–407. https://doi.org/10.1016/j.tiv.2014.10.023
  • Thompson, K. H., & Orvig, C. (2006). Metal complexes in medicinal chemistry: new vistas and challenges in drug design. Dalton Transactions, (6), 761–764. https://doi.org/10.1039/B513476E
  • Thongrakard, V., Titone, R., Follo, C., Morani, F., Suksamrarn, A., Tencomnao, T., & Isidoro, C. (2014). Turmeric toxicity in A431 epidermoid cancer cells associates with autophagy degradation of anti-apoptotic and anti-autophagic p53 mutant. Phytotherapy Research : PTR, 28(12), 1761–1769. https://doi.org/10.1002/ptr.5196
  • Titenko-Holland, N., Windham, G., Kolachana, P., Reinisch, F., Parvatham, S., Osorio, A. M., & Smith, M. T. (1997). Genotoxicity of malathion in human lymphocytes assessed using the micronucleus assay in vitro and in vivo: A study of malathion-exposed workers. Mutation Research/Genetic Toxicology and Environmental Mutagenesis, 388(1), 85–95. https://doi.org/10.1016/S1383-5718(96)00140-4
  • Tserkezidou, C., Hatzidimitriou, A. G., & Psomas, G. (2016). Nickel(II) complexes of flufenamic acid: Characterization, structure and interaction with DNA and albumins. Polyhedron, 117, 184–192. https://doi.org/10.1016/j.poly.2016.05.044
  • Vamsikrishna, N., Daravath, S., Ganji, N., & Pasha, N. (2020). Synthesis, structural characterization, DNA interaction, antibacterial and cytotoxicity studies of bivalent transition metal complexes of 6-aminobenzothiazole Schiff base. Inorganic Chemistry Communications, 113, 107767. https://doi.org/10.1016/j.inoche.2020.107767
  • Varna, D., Kapetanaki, E., Koutsari, A., Hatzidimitriou, A. G., Psomas, G., Angaridis, P., Papi, R., Pantazaki, A. A., & Aslanidis, P. (2018). Heterocyclic thioamide/phosphine mixed-ligand silver(I) complexes: Synthesis, molecular structures, DNA-binding properties and antibacterial activity. Polyhedron, 151, 131–140. https://doi.org/10.1016/j.poly.2018.05.020
  • Venkatesan, P., Bhutia, S. K., Singh, A. K., Das, S. K., Dash, R., Chaudhury, K., Sarkar, D., Fisher, B. P., & Mandal, M. (2012). AEE788 potentiates celecoxib-induced growth inhibition and apoptosis in human colon cancer cells. Life Sciences, 91(15-16), 789–799. https://doi.org/10.1016/j.lfs.2012.08.024
  • Venugopal, N., Krishnamurthy, G., Bhojyanaik, H. S., & Giridhar, M. (2019). Novel bioactive azo-azomethine based Cu (II), Co (II) and Ni(II) complexes, structural determination and biological activity. Journal of Molecular Structure, 1191, 85–94. https://doi.org/10.1016/j.molstruc.2019.04.022
  • Vidhya, N., & Devaraj, S. N. (2011). Induction of apoptosis by eugenol in human breast cancer cells. Indian Journal of Experimental Biology, 49(11), 871–878.
  • Wahab, S. I. A., Abdul, A. B., Alzubairi, A. S., Elhassan, M. M., & Mohan, S. (2009). In vitro ultramorphological assessment of apoptosis induced by zerumbone on (HeLa). Journal of Biomedicine and Biotechnology, 2009.
  • Wang, Y.-R., Chen, S.-F., Wu, C.-C., Liao, Y.-W., Lin, T.-S., Liu, K.-T., Chen, Y.-S., Li, T.-K., Chien, T.-C., & Chan, N.-L. (2017). Producing irreversible topoisomerase II-mediated DNA breaks by site-specific Pt(II)-methionine coordination chemistry. Nucleic Acids Research, 45(18), 10861–10871. https://doi.org/10.1093/nar/gkx742
  • White, E. (2012). Deconvoluting the context-dependent role for autophagy in cancer. Nature Reviews. Cancer, 12(6), 401–410. https://doi.org/10.1038/nrc3262
  • Wolfe, A., Shimer, G. H., Jr., & Meehan, T. (1987). Polycyclic aromatic hydrocarbons physically intercalate into duplex regions of denatured DNA. Biochemistry, 26(20), 6392–6396. https://doi.org/10.1021/bi00394a013
  • Xia, Y., Liu, X., Zhang, L., Zhang, J., Li, C., Zhang, N., Xu, H., & Li, Y. (2019). A new Schiff base coordinated copper(II) compound induces apoptosis and inhibits tumor growth in gastric cancer. Cancer Cell International, 19, 81. https://doi.org/10.1186/s12935-019-0801-6
  • Xiao, K., Jiang, J., Guan, C., Dong, C., Wang, G., Bai, L., Sun, J., Hu, C., & Bai, C. (2013). Curcumin induces autophagy via activating the AMPK signaling pathway in lung adenocarcinoma cells. Journal of Pharmacological Sciences, 123(2), 102–109. https://doi.org/10.1254/jphs.13085fp
  • Xue, X., Qu, X. J., Gao, Z. H., Sun, C. C., Liu, H. P., Zhao, C. R., Cheng, Y. N., & Lou, H. X. (2012). Riccardin D, a novel macrocyclic bisbibenzyl, induces apoptosis of human leukemia cells by targeting DNA topoisomerase II. Investigational New Drugs, 30(1), 212–222. https://doi.org/10.1007/s10637-010-9554-8
  • Zeglis, B. M., Divilov, V., & Lewis, J. S. (2011). Role of metalation in the topoisomerase IIα inhibition and antiproliferation activity of a series of α-heterocyclic-N4-substituted thiosemicarbazones and their Cu(II) complexes. Journal of Medicinal Chemistry, 54(7), 2391–2398. https://doi.org/10.1021/jm101532u
  • Zhao, G., Lin, H., Zhu, S., Sun, H., & Chen, Y. (1998). Dinuclear palladium(II) complexes containing two monofunctional [Pd(en)(pyridine)Cl]+ units bridged by Se or S. Synthesis, characterization, cytotoxicity and kinetic studies of DNA-binding. Journal of Inorganic Biochemistry, 70(3-4), 219–226. https://doi.org/10.1016/S0162-0134(98)10019-3
  • Zhivotovsky, B., & Kroemer, G. (2004). Apoptosis and genomic instability. Nature Reviews. Molecular Cell Biology, 5(9), 752–762. https://doi.org/10.1038/nrm1443

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.