Novel Isatin-based activator of p53 transcriptional functions in tumor cells

Document Type : Original article

Authors

1 Kazan Federal University, Kazan, Russian Federation

2 Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russian Federation

Abstract

Bioinorganic medicinal chemistry remains a hot field for research aimed at developing novel anti-cancer treatments. Discovery of metal complexes as potent antitumor chemotherapeutics such as cisplatin led to a significant shift of focus toward organometallic/ bioinorganic compounds containing transition metals and their chelates as novel scaffolds for drug discovery. In that way, transition metal complexes coordinated to essential biological scaffolds represent a highly promising class of compounds for design of novel target-specific therapeutics. Here, we report novel data on p53 activating Isatin-based Cu(II) complex exhibiting cytotoxic properties towards HCT116 and MCF7 tumor cell lines, as confirmed by cell viability assay and flow cytometry analysis of apoptosis. Furthermore, putative p53-mediated mechanism of action of this compound is supported by quantitative analysis of TP53, MDM2 and PUMA genes expression, as well as luciferase-based p53 pathway activation assay. Multiplex immunoassay analysis of inflammatory markers revealed potential modulation of several cytokines and chemokines.

Keywords


1. Bykov VJN, Eriksson SE, Bianchi J, Wiman KG. Targeting mutant p53 for efficient cancer therapy. Nat Rev Cancer 2018;18:89-102.
2. Muller PA, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 2014;25:304-317.
3. Hainaut P, Hollstein M. p53 and human cancer: the first ten thousand mutations. Adv Cancer Res 2000;77:81-137.
4. Danovi D, Meulmeester E, Pasini D, Migliorini D, Capra M, Frenk R, de Graaf P, Francoz S, Gasparini P, Gobbi A, Helin K, Pelicci PG, Jochemsen AG, Marine JC. Amplification of Mdmx (or Mdm4) directly contributes to tumor formation by inhibiting p53 tumor suppressor activity. Mol Cell Biol 2004;24:5835-5843.
5. Duffy MJ, Synnott NC, Crown J. Mutant p53 as a target for cancer treatment. Eur J Cancer 2017;83:258-265.
6. Bykov VJN, Issaeva N, Shilov A, Hultcrantz M, Pugacheva E, Chumakov P, Bergman J, Wiman KG, Selivanova G. Restoration of the tumor suppressor function to mutant p53 by a low-molecular-weight compound. Nat Med 2002;8:282-288.
7. Joerger AC, Ang HC, Fersht AR. Structural basis for understanding oncogenic p53 mutations and designing rescue drugs. Proc Natl Acad Sci USA 2006;103:15056-15061.
8. Bauer MR, Jones RN, Baud MGJ, Wilcken R, Boeckler FM, Fersht AR, Joerger AC, Spencer J. Harnessing fluorine-sulfur contacts and multipolar interactions for the design of p53 mutant Y220C rescue drugs. ACS Chem Biol 2016;11:2265-2274.
9. Miller JJ, Orvain C, Jozi S, Clarke RM, Smith JR, Blanchet A, Gaiddon C, Warren JJ, Storr T. Multifunctional compounds for activation of the p53-Y220C mutant in cancer. Chemistry 2018;24:17734-17742.
10. Baud MGJ, Bauer MR, Verduci L, Dingler FA, Patel KJ, Horil Roy D, oerger AC, Fersht AR. Aminobenzothiazole derivatives stabilize the thermolabile p53 cancer mutant Y220C and show anticancer activity in p53-Y220C cell lines. Eur J Med Chem 2018;152:101-114.
11. Bulatov E, Zagidullin A, Valiullina A, Sayarova R, Rizvanov A. Small molecule modulators of RING-type E3 ligases: MDM and ullin families as targets. Front Pharmacol 2018;9:450.
12. Bulatov E, Valiullina A, Sayarova R, Rizvanov A. Promising new therapeutic targets for regulation of inflammation and immunity: RING-type E3 ubiquitin ligases. Immunol Lett 2018;202:44-51.
13. Gadd MS, Bulatov E, Ciulli A. Serendipitous SAD solution for DMSO-Soaked SOCS2-elonginC-elonginB crystals using covalently incorporated dimethylarsenic: Insights into substrate receptor conformational flexibility in cullin RING ligases. PLoS One 2015;10: e0131218.
14. Bulatov E, Khaiboullina S, Reis dos HJ, Palotás A, Venkataraman K, Vijayalakshmi M, Rizvanov A. Ubiquitin-proteasome system: Promising therapeutic targets in autoimmune and neurodegenerative diseases. BioNanoSci 2016;6:341-344.
15.Frezza M, Hindo S, Chen D, Davenport A, Schmitt S, Tomco D, Dou QP. Novel Metals and Metal Complexes as Platforms for Cancer Therapy. Curr Pharm Des. 2010;16: 1813-1825.
16. Ng CH, Kong SM, Tiong YL, Maah MJ, Sukram N, Ahmad M, Khoo AS. Selective anticancer copper(II)-mixed ligand complexes: targeting of ROS and proteasomes. Metallomics 2014;6: 892-906.
17. Zuo J, Bi C, Fan Y, Buac D, Nardon C, Daniel KG, Dou QP. Cellular and computational studies of proteasome inhibition and apoptosis induction in human cancer cells by amino acid Schiff base-copper complexes. J Inorg Biochem. 2013;118:83-93.
18. Milacic V, Chen D, Giovagnini L, Diez A, Fregona D, Dou QP. Pyrrolidine dithiocarbamate-zinc(II) and -copper(II) complexes induce apoptosis in tumor cells by inhibiting the proteasomal activity. Toxicol Appl Pharmacol 2008;231:24-33.
19. Zhang P, Bi C, Schmitt SM, Li X, Fan Y, Zhang N, Dou QP. Metal-based 2,3-indolinedione derivatives as proteasome inhibitors and inducers of apoptosis in human cancer cells. Int J Mol Med 2014;34:870-879.
20. Miri R, Razzaghi-asl N, Mohammadi MK. QM study and conformational analysis of an isatin Schiff base as a potential cytotoxic agent. J Mol Model 2013;19:727-735.
21. Al-Resayes SI, Shakir M, Abbasi A, Amin KMY, Lateef A. Synthesis, spectroscopic characterization and biological activities of N4O2 schiff base ligand and its metal complexes of Co(II), Ni(II), Cu(II) and Zn(II). Spectrochim Acta A Mol Biomol Spectrosc 2012;93:86-94.
22. Shakir M, Hanif S, Sherwani MA, Mohammad O, Azam M, Al-Resayes SI. Pharmacophore hybrid approach of new modulated bis-diimine Cu(II)/Zn(II) complexes based on 5-chloro Isatin Schiff base derivatives: Synthesis, spectral studies and comparative biological assessment. J Photochem Photobiol B: Biology 2016;157:39-56.
23. Liang C, Xia J, Lei D, Li X, Yao Q, Gao J. Synthesis, in vitro and in vivo antitumor activity of symmetrical bis-Schiff base derivatives of isatin. Eur J Med Chem 2014;74:742-750.
24. da Silveira VC, Luz JS, Oliveira CC, Graziani I, Ciriolo MR, Da Costa Ferreira AM. Double-strand DNA cleavage induced by oxindole-Schiff base copper(II) complexes with potential antitumor activity. J Inorg Biochem. 2008;102:1090-1103.
25. Cerchiaro G, Aquilano K, Filomeni G, Rotilio G, Ciriolo MR, Ferreira AM. Isatin-Schiff base copper(II) complexes and their influence on cellular viability. J Inorg Biochem 2005; 99:1433-1440.
26. Filomeni G, Cerchiaro G, Da Costa Ferreira AM, De Martino A, Pedersen JZ, Rotilio G, Ciriolo MR. Pro-apoptotic activity of novel Isatin-Schiff base copper(II) complexes depends on oxidative stress induction and organelle-selective damage. J Biol Chem 2007;282:12010-12021.
27. Ali AQ, Teoh SG, Salhin A, Eltayeb NE, Khadeer Ahamed MB, Abdul Majid AM. Synthesis of isatin thiosemicarbazones derivatives: In vitro anti-cancer, DNA binding and cleavage activities. Spectrochimica Acta A: Mol Biomol Spectrosc 2014;125:440-448.
28. Borges BE, Teixeira VR, Appel MH, Steclan CA, Rigo F, Filipak Neto F, da Costa, Ferreira AM, Chammas R, Zanata SM, Nakao LS. De novo galectin-3 expression influences the response of melanoma cells to isatin-Schiff base copper (II) complex-induced oxidative stimulus. Chem Biol Interact 2013;206:37-46.
29. Davidovich P, Aksenova V, Petrova V, Tentler D, Orlova D, Smirnov S, Gurzhiy V, Okorokov AL, Garabadzhiu A, Melino G, Barlev N, Tribulovich V. Discovery of Novel Isatin-Based p53 Inducers. ACS Med Chem Lett 2015;6:856-860.
30. Bulatov E, Sayarova R, Mingaleeva R, Miftakhova R, Gomzikova M, Ignatyev Y, Petukhov A, Davidovich P, Rizvanov A, Barlev NA. Isatin-Schiff base-copper (II) complex induces cell death in p53-positive tumors. Cell Death Discov 2018;4:103.
31. Fedorova O, Daks A, Petrova V, Petukhov A, Lezina L, Shuvalov O, Davidovich P, Kriger D, Lomert E, Tentler D, Kartsev V, Uyanik B, Tribulovich V, Demidov O, Melino G, Barlev NA. Novel isatin-derived molecules activate p53 via interference with Mdm2 to promote apoptosis. Cell Cycle 2018;17:1917-1930.
32. Smirnov AS, Martins LMDRS, Nikolaev DN, Manzhos RA, Gurzhiy VV, Krivenko AG, Nikolaenko KO, Belyakov AV, Garabadzhiu AV, Davidovich PB. Structure and catalytic properties of novel copper isatin schiff base complexes. New J Chem 2019;43:188-198.
33. Joerger AC, Fersht AR. Structure–function–rescue: the diverse nature of common p53 cancer mutants. Oncogene 2007;26:2226-2242.
34. Brown CJ, Lain S, Verma CS, Fersht AR, Lane DP. Awakening guardian angels: drugging the p53 pathway. Nat Rev Cancer 2009;9: 862-873.
35. Yu J, Zhang L. PUMA, a potent killer with or without p53. Oncogene 2008;27 Suppl 1:S71-83.
36. Komeda S, Casini A. Next-generation anticancer metallodrugs. Curr Top Med Chem 2012; 12:219-235.
37. Liu C, Zhou J, Li Q, Wang L, Liao Z, Xu H. DNA damage by copper(II) complexes: coordination-structural dependence of reactivities. J Inorg Biochem 1999;75: 233-240.
38. Ostrakhovitch EA, Cherian MG. Role of p53 and reactive oxygen species in apoptotic response to copper and zinc in epithelial breast cancer cells. Apoptosis 2005;10:111-121.
39. Loh SN. The missing zinc: p53 misfolding and cancer. Metallomics 2010;2:442-449.
40. Formigari A, Gregianin E, Irato P. The effect of zinc and the role of p53 in copper-induced cellular stress responses. J Appl Toxicol 2013;33:527-536.