Effects of self-assembled cell-penetrating peptides and their nano-complexes on ABCB1 expression and activity

Document Type : Original Article

Authors

1 Student Research Committee, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran

2 Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

3 Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

4 Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran

5 Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran

6 College of Pharmacy, Al Ain University of Science and Technology, Al Ain 64141, UAE

7 Faculty of Pharmacy and Pharmaceutical Sciences, Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, Alberta, T6G 2H5, Canada

8 Biotechnology Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran

Abstract

Objective(s): Doxorubicin (Dox) is one of the most well-known chemotherapeutics that are commonly applied for a wide range of cancer treatments. However, in most cases, efflux pumps like P-glycoprotein (P-gp), expel the taken drugs out of the cell and decrease the Dox bioavailability. Expression of P-gp is associated with elevated mRNA expression of the ATP-binding cassette B1 (ABCB1) gene.
Materials and Methods: In the current study, different sequences of cell-penetrating peptides (CPPs) containing tryptophan, lysine, and arginine and their nano-complexes were synthesized and their impact on the expression and activity of the ABCB1 gene was evaluated in the A549 lung carcinoma cell line. Furthermore, the cellular uptake of designed CPPs in the A549 cell line was assessed.
Results: The designed peptides, including [W4K4], [WR]3-QGR, R10, and K10 increased Dox cytotoxicity after 48 hr. Furthermore, arginine-rich peptides showed higher cellular uptake. Rhodamin123 accumulation studies illustrated that all the obtained peptides could successfully inhibit the P-gp pump. The designed peptides inhibited the ABCB1 gene expression, of which, [W4K4] resulted in the lowest expression ratio.
Conclusion: [W4K4], [WR]3-QGR, R10, and K10 could successfully increase the Dox cytotoxicity by decreasing the efflux pump gene expression.

Keywords


1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin 2017;67:7-30.
2. Wang Y, Dou L, He H, Zhang Y, Shen Q. Multifunctional nanoparticles as nanocarrier for vincristine sulfate delivery to overcome tumor multidrug resistance. Mol Pharm 2014;11:885-894.
3. Dawar S, Singh N, Kanwar RK, Kennedy RL, Veedu RN, Zhou SF, et al. Multifunctional and multitargeted nanoparticles for drug delivery to overcome barriers of drug resistance in human cancers. Drug Discov Today 2013;18:1292-1300.
4.    Chang A. Chemotherapy, chemoresistance and the changing treatment landscape for NSCLC. Lung Cancer 2011;71:3-10.
5.    Wang Y, Dou L, He H, Zhang Y, Shen Q. Multifunctional nanoparticles as nanocarrier for vincristine sulfate delivery to overcome tumor multidrug resistance. Mol Pharm 2014;11:885-894.
6.    Li W, Zhang H, Assaraf YG, Zhao K, Xu X, Xie J, et al. Overcoming ABC transporter-mediated multidrug resistance: molecular mechanisms and novel therapeutic drug strategies. Drug Resist Updat 2016;27:14-29.
7.    Niazi M, Zakeri-Milani P, Najafi Hajivar S, Soleymani Goloujeh M, Ghobakhlou N, Shahbazi Mojarrad J, et al. Nano-based strategies to overcome p-glycoprotein-mediated drug resistance. Expert Opin Drug Metab Toxicol 2016;12:1021-1033.
8. Dong X, Mumper RJ. Nanomedicinal strategies to treat multidrug-resistant tumors: current progress. Nanomedicine 2010;5:597-615.
9.    Yardley DA. Drug resistance and the role of combination chemotherapy in improving patient outcomes. Int J Breast Cancer 2013;2013:15-??.
10.    Cukierman E, Khan DR. The benefits and challenges associated with the use of drug delivery systems in cancer therapy. Biochem Pharmacol 2010;80:762-770.
11.    Patel NR, Pattni BS, Abouzeid AH, Torchilin VP. Nanopreparations to overcome multidrug resistance in cancer. Adv Drug Deliv Rev 2013;65:1748-1762.
12.    Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev 2013;65:1866-1879.
13.    Livney YD, Assaraf YG. Rationally designed nanovehicles to overcome cancer chemoresistance. Adv Drug Deliv Rev 2013;65:1716-1730.
14.    Fodale V, Pierobon M, Liotta L, Petricoin E. Mechanism of cell adaptation: when and how do cancer cells develop chemoresistance? Cancer J 2011;17:89-95.
15.    Gillet JP, Gottesman MM. Mechanisms of multidrug resistance in cancer. Methods Mol Biol 2010;596:47-76.
16.    Mesgari Abbasi M, Valizadeh H, Hamishekar H, Mohammadnejad L, Zakeri-Milani P. The effects of cetirizine on P-glycoprotein expression and function in vitro and in situ. Adv Pharm Bull 2016;6:111-118.
17.    Callaghan R, Luk F, Bebawy M. Inhibition of the multidrug resistance P-glycoprotein: time for a change of strategy? Drug Metab Dispos 2014;42:623-631.
18.    Abd Ellah NH, Taylor L, Ayres N, Elmahdy MM, Fetih GN, Jones HN, et al. NF-[kappa]B decoy polyplexes decrease P-glycoprotein-mediated multidrug resistance in colorectal cancer cells. Cancer Gene Ther 2016;23:149-155.
19.    Mohammadzadeh R, Baradaran B, Valizadeh H, Yousefi B, Zakeri-Milani P. Reduced ABCB1 expression and activity in the presence of acrylic copolymers. Adv Pharm Bull 2014;4:219-224.
20.    Hanke U, May K, Rozehnal V, Nagel S, Siegmund W, Weitschies W. Commonly used nonionic surfactants interact differently with the human efflux transporters ABCB1 (p-glycoprotein) and ABCC2 (MRP2). Eur J Pharm Biopharm 2010;76:260-268.
21.    Sodani K, Patel A, Kathawala RJ, Chen Z-S. Multidrug resistance associated proteins in multidrug resistance. Chin J Cancer 2012;31:58-72.
22.    Cavaco MC, Pereira C, Kreutzer B, Gouveia LF, Silva-Lima B, Brito AM, et al. Evading P-glycoprotein mediated-efflux chemoresistance using solid lipid nanoparticles. Eur J Pharm Biopharm 2017;110:76-84.
23.    Sharom FJ. ABC multidrug transporters: structure, function and role in chemoresistance. Pharmacogenomics 2008;9:105-127.
24.    Zakeri-Milani P, Valizadeh H. Intestinal transporters: enhanced absorption through P-glycoprotein-related drug interactions. Expert Opin Drug Metab Toxicol 2014;10:859-871.
25.    Kunjachan S, Rychlik B, Storm G, Kiessling F, Lammers T. Multidrug resistance: physiological principles and nanomedical solutions. Adv Drug Deliv Rev 2013;65:1852-1865.
26.    Esser L, Zhou F, Pluchino KM, Shiloach J, Ma J, Tang W-k, et al. Structures of the multidrug transporter P-glycoprotein reveal asymmetric ATP binding and the mechanism of polyspecificity. J Biol Chem 2017;292:446-461.
27.    Thorn CF, Oshiro C, Marsh S, Hernandez-Boussard T, McLeod H, Klein TE, et al. Doxorubicin pathways: pharmacodynamics and adverse effects. Pharmacogenet Genomics 2011;21:440-446.
28.    Ren F, Shen J, Shi H, Hornicek FJ, Kan Q, Duan Z. Novel mechanisms and approaches to overcome multidrug resistance in the treatment of ovarian cancer. Biochim Biophys Acta Rev Cancer 2016;1866:266-275.
29.    Markman JL, Rekechenetskiy A, Holler E, Ljubimova JY. Nanomedicine therapeutic approaches to overcome cancer drug resistance. Adv Drug Deliv Rev 2013;65:1866-1879.
30.    Omote H, Al-Shawi MK. Interaction of transported drugs with the lipid bilayer and P-glycoprotein through a solvation exchange mechanism. Biophys J 2006;90:4046-4059.
31.    Najafi-Hajivar S, Zakeri-Milani P, Mohammadi H, Niazi M, Soleymani-Goloujeh M, Baradaran B, et al. Overview on experimental models of interactions between nanoparticles and the immune system. Biomed Pharmacother 2016;83:1365-1378.
32.    Vargas JR, Stanzl EG, Teng NN, Wender PA. Cell-penetrating, guanidinium-rich molecular transporters for overcoming efflux-mediated multidrug resistance. Mol Pharm 2014;11:2553-2565.
33.    Copolovici DM, Langel K, Eriste E, Langel Ü. Cell-penetrating peptides: design, synthesis, and applications. ACS Nano 2014;8:1972-1994.
34.    Ru Q, Shang B-y, Miao Q-f, Li L, Wu S-y, Gao R-j, et al. A cell penetrating peptide-integrated and enediyne-energized fusion protein shows potent antitumor activity. Eur J Pharm Sci 2012;47:781-789.
35.    Farkhani SM, Johari-Ahar M, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. Enhanced cellular internalization of CdTe quantum dots mediated by arginine- and tryptophan-rich cell-penetrating peptides as efficient carriers. Artif Cells Nanomed Biotechnol 2016;44:1424-1428.
36.    Mohammadi S, Zakeri-Milani P, Golkar N, Farkhani SM, Shirani A, Shahbazi Mojarrad J, et al. Synthesis and cellular characterization of various nano-assemblies of cell penetrating peptide-epirubicin-polyglutamate conjugates for the enhancement of antitumor activity. Artif Cells Nanomed Biotechnol 2018;46:1572-1585.
37.    Farshbaf M, Salehi R, Annabi N, Khalilov R, Akbarzadeh A, Davaran S. pH- and thermo-sensitive MTX-loaded magnetic nanocomposites: synthesis, characterization, and in vitro studies on A549 lung cancer cell and MR imaging. Drug Dev Ind Pharm 2018;44:452-462.
38.    Meuzelaar H, Vreede J, Woutersen S. Influence of Glu/Arg, Asp/Arg, and Glu/Lys salt bridges on α-helical stability and folding kinetics. Biophys J 2016;110:2328-2341.
39.    Madani F, Lindberg S, Langel, #220, lo, Futaki S, et al. Mechanisms of cellular uptake of cell-penetrating peptides. J Biophys 2011;2011:414729.
40.    Trabulo S, Cardoso AL, Mano M, de Lima MCP. Cell-penetrating peptides—mechanisms of cellular uptake and generation of delivery systems. Pharmaceuticals 2010;3:961-993.
41.    Åmand HL, Rydberg HA, Fornander LH, Lincoln P, Nordén B, Esbjörner EK. Cell surface binding and uptake of arginine- and lysine-rich penetratin peptides in absence and presence of proteoglycans. Biochim Biophys Acta Biomembr 2012;1818:2669-2678.
42.    Bechara C, Sagan S. Cell-penetrating peptides: 20 years later, where do we stand? FEBS Lett 2013;587:1693-702.
43.    Schmidt N, Mishra A, Lai GH, Wong GCL. Arginine-rich cell-penetrating peptides. FEBS Lett 2010;584:1806-1813.
44.    Takeuchi T, Futaki S. Current understanding of direct translocation of arginine-rich cell-penetrating peptides and Its internalization mechanisms. Chem Pharml Bull 2016;64:1431-1437.
45.    Mohammadi S, Shahbazi Mojarrad J, Zakeri-Milani P, Shirani A, Mussa Farkhani S, Samadi N, et al. Synthesis and in vitro evaluation of amphiphilic peptides and their nanostructured conjugates. Adv Pharm Bull 2015;5:41-49.
46.    Mussa Farkhani S, Asoudeh Fard A, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. Enhancing antitumor activity of silver nanoparticles by modification with cell-penetrating peptides. Artif Cells Nanomed Biotechnol 2017;45:1029-1035.
47.    Soleymani-Goloujeh M, Nokhodchi A, Niazi M, Najafi-Hajivar S, Shahbazi-Mojarrad J, Zarghami N, et al. Effects of N-terminal and C-terminal modification on cytotoxicity and cellular uptake of amphiphilic cell penetrating peptides. Artif Cells Nanomed Biotechnol 2018;46:91-103.
48.    Zakeri-Milani P, Mussa Farkhani S, Shirani A, Mohammadi S, Shahbazi Mojarrad J, Akbari J, et al. Cellular uptake and anti-tumor activity of gemcitabine conjugated with new amphiphilic cell penetrating peptides. EXCLI J 2017;16:650-662.
49.    Farkhani SM, Shirani A, Mohammadi S, Zakeri-Milani P, Shahbazi Mojarrad J, Valizadeh H. Effect of poly-glutamate on uptake efficiency and cytotoxicity of cell penetrating peptides. IET Nanobiotechnol 2016;10:87-95.
50.    Mitchell DJ, Kim DT, Steinman L, Fathman CG, Rothbard JB. Polyarginine enters cells more efficiently than other polycationic homopolymers. J Pept Res 2000;56:318-325.
51.    Morris MC, Depollier J, Mery J, Heitz F, Divita G. A peptide carrier for the delivery of biologically active proteins into mammalian cells. Nat Biotechnol 2001;19:1173-1176.
52.    Banković J, Andrä J, Todorović N, Podolski-Renić A, Milošević Z, Miljković Đ, et al. The elimination of P-glycoprotein over-expressing cancer cells by antimicrobial cationic peptide NK-2: The unique way of multi-drug resistance modulation. Exp Cell Res 2013;319:1013-1027.
53.    Nakase I, Konishi Y, Ueda M, Saji H, Futaki S. Accumulation of arginine-rich cell-penetrating peptides in tumors and the potential for anticancer drug delivery in vivo. J Control Release 2012;159:181-188.
54.    Duan Z, Chen C, Qin J, Liu Q, Wang Q, Xu X, et al. Cell-penetrating peptide conjugates to enhance the antitumor effect of paclitaxel on drug-resistant lung cancer. Drug Deliv 2017;24:752-64.
55.    Zheng Z, Aojula H, Clarke D. Reduction of doxorubicin resistance in P-glycoprotein overexpressing cells by hybrid cell-penetrating and drug-binding peptide. J Drug Target 2010;18:477-487.