Synthesis and biological activity of siRNA and Etoposide with magnetic nanoparticles on drug resistance model MCF-7 Cells: Molecular docking study with MRP1 enzyme

Document Type : Research Paper

Authors

1 Department of Molecular Biology and Genetics, Kırsehir Ahi Evran University, Kırsehir, Turkey

2 Department of Biology, Middle East Technical University, Ankara,Turkey

Abstract

Objective(s): In this work, MRP-1 (Multidrug resistance-associated protein 1) gene expression levels and anticancer activity of siRNA and Etoposide loaded Poly-hydroxybutyrate (PHB) coated magnetic nanoparticles (MNPs) was studied on MCF-7/Sensitive and MCF-7/1000Etoposide resistance cells. For this purpose, PHB covered iron oxide-based magnetic nanoparticles (PHB-MNPs) were prepared by coprecipitation. We used magnetic nanoparticles because they include highly targeted to tumors in vivo cancer therapy.
Materials and Methods: Etoposide, anti-cancer drug, was loaded onto the PHB-MNPs. The in vitro cytotoxicity analysis of siRNA and Etoposide-loaded PHB-MNPs was applied on cancer cells. The expression levels of MRP1 related to drug resistance were shown using qRT-PCR. In the present study, we also investigated whether nanoparticle system could be a potential anticancer drug target with molecular docking analyses.
Results: The IC50 values of Etoposide on MCF-7/sensitive and MCF-7/1000Eto resistance cells were identified as 50,6 μM and 135,7 μM, respectively. IC50 values of siRNA and Etoposide loaded PHB coated magnetic nanoparticles were determined as 10,18 μM and 39,21 μM on MCF-7 and MCF-7/1000 Eto cells, respectively. According to the gene expression results, MRP1 expression was 4 fold upregulated in MCF-7/1000Eto cells. However, it was about 3 fold downregulated due to the application of siRNA-Etoposide loaded magnetic nanoparticles.
Conclusion: According to the docking results, nanoparticle system may be a drug active substance with obtained results. The results of this study demonstrated that siRNA and Etoposide loaded PHB covered iron oxide based magnetic nanoparticles can be a potential targeted therapeutic agent to overcome drug resistance.

Keywords


1.Clark PI, Slevin ML. The clinical pharmacology of etoposide and teniposide. Clin Pharmacokinet. 1987; 12(4): 223–252.
2.Chamberlain M. Recurrent brainstem gliomas treated with oral VP-16. J Neurooncol. 1993; 15(2): 133–139.
3.Ashley DM, Meier L, Kerby T. Response of recurrent medulloblastoma to low-dose oral etoposide. J Clin Oncol. 1996; 14(6): 1922–1927.
4.Shah JC, Chen JR, Chow D. Preformulation study of etoposide: identification of physicochemical characteristics responsible for the low and erratic oral bioavailability of etoposide. Pharm Res. 1989; 6(5): 408–412.
5.Zhang T, Chen J, Zhang Y. Characterization and evaluation of nanostructured lipid carrier asa vehicle for oral delivery of etoposide. Eur J Pharm Sci. 2011;43(3): 174–179.
6.Lamprecht A, Benoit JP. Etoposide nanocarriers suppress glioma cell growth by intracellular drug delivery and simultaneous P-glycoprotein inhibition. J Control Release. 2006; 112(2): 208–213.
7.Masquelier M, Zhou QF, Gruber A. Relationship between daunorubicin concentration and apoptosis induction in leukemic cells. Biochem Pharmacol. 2004; 67(6): 1047–1056.
8.Varshosaz J, Hassanzadeh F, Sadeghi AH. Uptake of etoposide in CT-26 cells of colorectal cancer using folate targeted dextran stearate polymericmicelles. Biomed Res Int. 2014; 708593.
9.Varthya M, Pawar H, Singh C. Development of Novel Polymer-Lipid Hybrid Nanoparticles of Tamoxifen: in vitro and in vivo Evaluation. J Nanosci Nanotechnol. 2016; 16(1): 253–260.
10.Khodadust R, Unsoy G, Yalcın S. PAMAM dendrimer-coated iron oxide nanoparticles: synthesis and characterization of different generations. J Nanopart Res. 2013; 15: 1488.
11.Gabizon AA. in: V. Torchilin (Ed.), Nanoparticulates as Drug Carriers, Imperial College Press, London. 2011.
12.Lawrence MJ, Warisnoicharoen W. in: V. Torchilin (Ed.), Nanoparticulates as Drug Carriers, Imperial College Press, London. 2006.
13.Mäder K. in: V. Torchilin (Ed.) Nanoparticulates as Drug Carriers, Imperial College Press, London. 2006.
14.Yordanov G, Skrobanska R, Evangelatov A. Colloidal formulations of etoposide based on poly(butyl cyanoacrylate) nanoparticles: preparation,physicochemical properties and cytotoxicity. Colloids Surf B Biointerfaces. 2013; 101: 215–22.
15.Callewaert M, Dukic S, Gulick L. Etoposide encapsulation in surface-modified poly(lactide-co-glycolide) nanoparticles strongly enhances glioma antitumor efficiency. J Biomed Mater Res A. 2012; 101(5): 1319–1327.
16.Gaucher G, Poreba M, Ravenelle F. Poly(N-vinyl-pyrrolidone)- block-poly(D,L-lactide) as polymeric emulsifier for the preparation of biodegradable nanoparticles. J Pharm Sci. 2007;96(7): 1763–1775.
17.Kilicay E, Demirbilek M, Turk M, et al. Preparation and characterization of poly(3-hydroxybutyrate-co-3- hydroxyhexanoate) based nanoparticles for targeted cancer therapy. Eur J Pharm Sci. 2011; 44(3): 310–320.
18.Poreba R, Gac P, Poreba M. Environmental and occupational exposure to lead as a potential risk factor for cardiovascular disease. Environ Toxicol Pharmacol 2011; 31(2): 267–277.
19.Yadav KS, Jacob S, Sachdeva G, et al. Intracellular delivery of etoposide loaded biodegradable nanoparticles: cytotoxicity and cellular uptake studies. J Nanosci Nanotechnol .2011; 11(8): 6657–6667.
20.Khajavinia A, Varshosaz J, Dehkordi AJ. Targeting etoposide to acute myelogenous leukaemia cells using nanostructured lipid carriers coated with transferrin. Nanotechnology. 2012; 23(40): 405101.
21.Jinturkar KA, Anish C, Kumar MK. Liposomal formulations of Etoposide and Docetaxel for p53 mediated enhanced cytotoxicity in lung cancer cell lines. Biomaterials. 2012; 33(8): 2492–2507.
22.Varshosaz J, Hasanzadeh F, Eslamdoost M. Optimization of self-assembling properties of fatty acids grafted to methoxy poly(ethylene glycol) as nanocarriers for etoposide. Acta Pharm. 2012; 62(1): 31–44.
23.Xiong YC, Yao YC, Zhan XYl. Application of polyhydroxyalkanoates nanoparticles as intracellular sustained drug-release vectors. Biomater Sci Polym Ed. 2010; 21(1): 127–140.
24.Yalçın S, Khodadust R, Unsoy G, et al. Synthesis and characterization of Poly-hydroxybutyrate (PHB) coated magnetic nanoparticles: toxicity analyses on different cell lines. Inorganic and Nano-Metal Chemistry. 2015; 45(5): 700–708.
25.Su Z, Liu G, Fang T. Silencing MRP1-4 genes by RNA interference enhances sensitivity of human hepatoma cells to chemotherapy. Am J Transl Res. 2016; 8(6): 2790–2802.
26.Kaplan E, Gunduz U. Expression analysis of TOP2A, MSH2 and MLH1 genes in MCF7 cells at different levels of etoposide resistance. Biomed Pharmacother. 2012; 66(1): 29–35.
27.Livak KJ, Schmittgen TD. Analysis of Relative Gene Expression Data Using Real-Time Quantitative PCR and the 2−ΔΔCT Method. Methods. 2001; 25(4): 402–408.
28.Morris GM, Goodsell DS, Halliday RS, Huey R, Hart WE, Belew RK, Olson AJ (1998). Automated docking using a Lamarckian genetic algorithm and an empirical binding free energy function. J Comput Chem. 19(14): 1639-1662.
29.Trott O, Olson AJ(2010). AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem. 31 (2): 455e461.
30.Ramaen O, Leulliot N, Sizun C, Ulryck N, Pamlard O, Lallemand JY, Tilbeurgh Hv, Jacquet E (2006). Structure of the human multidrug resistance protein 1 nucleotide binding domain 1 bound to Mg2+/ATP reveals a non-productive catalytic site. J Mol Biol. 359(4):940-9.
31.Frisch, M. J., Trucks, G. W., Schlegel, H. B., Scuseria, G. E., Robb, M. A., Cheeseman, J. R., ... & Nakatsuji, H. (2009). Gaussian09 Revision D. 01, Gaussian Inc. Wallingford CT.
32.Thomsen R, Christensen, MH (2006). MolDock: a new technique for high-accuracy molecular docking. J Med Chem. 49(11):3315-3321.
33.Nomura T, Saikawa A, Morita S. 1998. Pharmacokinetic characteristics and therapeutic effects of mitomycin C-dextran conjugates after intratumoral injection. J Control Release. 1998; 52(3): 239–252.
34.Maeda H, Sawa T, Konno T. Mechanism of tumor-targeted delivery of macromolecular drugs, including the EPR effect in solid tumor and clinical overview of prototype polymeric drug SMANCS. J Control Release. 2001; 74(1–3): 47–61.
35.Yalcin S, Unsoy G, Mutlu P, et al. Polyhydroxybutyrate coated magnetic nanoparticles for Doxorubicin delivery: Cytotoxic effect against Doxorubicin-resistant breast cancer cell line. Am J Ther. 2014; 21(6): 453–461.
36.Marin A, Sun H, Husseini GA. Drug delivery in pluronic micelles: effect of high-frequency ultrasound on drug release from micelles and intracellular uptake. J Control Release. 2002;84(1–2): 39–47.
37.Williams J, Lansdown R, Sweitzer R. Nanoparticle drug delivery system for intravenous delivery of topoisomerase inhibitors. J Control Release. 2003; 91(1–2): 167–172.
38.Harivardhan RL, Sharma RK, Chuttani K, et al. Influence of administration route on tumor uptake and biodistribution of etoposide loaded solid lipidnanoparticles in Dalton’s lymphoma tumor bearing mice. J Control Release. 2005; 105(3): 185–98.
39.Xia F, Hou W, Zhang Cl. pH-responsive gold nanoclusters-based nanoprobes for lung cancer targeted near-infrared fluorescence imaging and chemo-photodynamic therapy. Acta biomaterialia. 2018; 68: 308–319.
40.Hao Y, Zheng C, Wang L et al. 2017. Tumor acidity-activatable manganese phosphate nanoplatform for amplification of photodynamic cancer therapy and magnetic resonance imaging. Acta Biomater. 2017; 62: 293–305.
41.Shi XX, Ma X, Hou M, et al. pH-responsive unimolecular micelles based on amphiphilic star-like copolymers with high drug loading for effective drug delivery and cellular imaging. J Mater Chem B. 2017; 5: 6847–6859.
42.Lin WJ, Yao N, Qian L. pH-responsive unimolecular micelle-gold nanoparticles-drug nanohybrid system for cancer theranostics. Acta Biomater. 2017; 58: 455–465.
43.Fang S, Lin J, Li C et al. Dual-stimuli responsive Nanotheranostics for multimodal imaging guided Trimodal synergistic therapy. Small. 2017;13(6).
44.Du J, Lane LA, Nie S. Stimuli-responsive nanoparticles for targeting the tumor microenvironment. J Control Release. 2015;219: 205–214.
45.Chen Q, Feng LZ, Liu JJ. 2016. Intelligent albumin-MnO2 nanoparticles as pH-/H2O2-responsive dissociable Nanocarriers to modulate tumor hypoxia for effective combination therapy. Adv Mater. 2016; 28(33): 7129–7136.
46.Feng LL, Gai S, He F. Controllable generation of free radicals from multifunctional heat-responsive Nanoplatform for targeted cancer therapy. Chem Mater. 2018; 30(2): 526–539.
47.Yang GB, Zhang R, Liang C. Manganese dioxide coated WS2@Fe3O4/sSiO(2) Nanocomposites for pH-responsive MR imaging and oxygen-elevated synergetic therapy. Small. 2018; 14(2).
48.Cho MH, Choi ES, Kim S, Goh SH, Choi Y. Redox-responsive manganese dioxide nanoparticles for enhanced MR imaging and radiotherapy of lung cancer. Front Chem. 2017; 5: 109.
49.Moreira AF, Dias DR, Correia IJ. Stimuli-responsive mesoporous silica nanoparticles for cancer therapy: a review. Micropor Mesopor Mat. 2016; 236: 141–157.
50.Hu QY, Katti PS, Gu Z. Enzyme-responsive nanomaterials for controlled drug delivery. Nanoscale. 2014; 6(21): 12273–12286.
51.Li JM, Liu F, Shao Q, Min YZ, et al. Enzyme-responsive cell-penetrating peptide conjugated Mesoporous silica quantum dot Nanocarriers for controlled release of nucleus-targeted drug molecules and real-time intracellular fluorescence imaging of tumor cells. Adv Healthc Mater. 2014; 3(8): 1230–1239.
52.de la Rica R, Aili D, Stevens MM. Enzyme-responsive nanoparticles for drug release and diagnostics. Adv Drug Deliver Rev. 2012; 64(11): 967–978.
53.John JV, Uthaman S, Augustine R, et al. pH/redox dual stimuli-responsive sheddable nanodaisies for efficient intracellular tumour-triggered drug delivery. J Mater Chem B. 2017;5(25): 5027–5036.
54.Uthaman S, Huh K, Park IK. Tumor microenvironment responsive nanoparticles for cancer theragnostic applications. Biomater Res. 2018; 22:22.
55.Hande KR. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur J Cancer. 1998; 34(10): 1514–1521
56.Lundstrom K. Cancer therapy applying viral nanoparticles. In: Khudyakov Y, Pumpens P, editors. Viral Nanotechnology. Boca Raton: Taylor & Francis 2015; 455–466.
57.Baker JR. Dendrimer-based nanoparticles for cancer therapy. Hematology Am Soc Hematol Educ Program. 2009; 708–719.
58.Lee SJ, Huh MS, Lee SY, Min S, Lee S, Koo H, Chu JU, Lee KE, Jeon H, Choi Y, Choi K, Byun Y, Jeong SY, Park K, Kim K, Kwon IC. Tumor-homing poly-siRNA/glycol chitosan self-cross-linked nanoparticles for systemic siRNA delivery in cancer treatment. Angew Chem Int Ed Engl. 2012 Jul 16; 51(29): 7203-7207.
59.Predescu AM, Matei E, Berbecaru AC, Pantilimon C, Drăgan C, Vidu R, Predescu C, Kuncser V.Synthesis and characterization of dextran-coated iron oxide nanoparticles. R Soc Open Sci. 2018; 5(3): 171525.
60.Dung DTK, Hai TH, Phuc LH, Long BD, Vinh LK, Truc PN. Preparation and characterization of magnetic nanoparticles with chitosan coating, APCTP–ASEAN Workshop on Advanced Materials Science and Nanotechnology (AMSN08) IOP Publishing Journal of Physics: Conference Series. 2009; 187: 012036
61.Zhang XL, Niu HY, Zhang SX, Cai YQ. Preparation of a chitosan-coated C18-functionalized magnetite nanoparticle sorbent for extraction of phthalate ester compounds from environmental water samples. Anal Bioanal Chem. 2010b; 397: 791–798.