EFFECTS OF SQUALANE ON THE SPERMINE-BASED CATIONIC NIOSOMES FOR GENE DELIVERY

Authors

  • Supusson Pengnam Faculty of Pharmacy, Silpakorn University, Sanamchandra Palace Campus, Nakhon Pathom
  • Prasopchai Tonglairoum Faculty of Pharmacy, Silpakorn University, Sanamchandra Palace Campus, Nakhon Pathom
  • Nattisa Ni-yomtham Faculty of Science, Ramkhamhaeng University, Bangkok
  • Boon-ek Yingyongnarongkul Faculty of Science, Ramkhamhaeng University, Bangkok
  • Praneet Opanasopit Faculty of Pharmacy, Silpakorn University, Sanamchandra Palace Campus, Nakhon Pathom
  • Samarwadee Plianwong Faculty of Pharmaceutical Sciences, Burapha University, Chonburi

DOI:

https://doi.org/10.14456/tbps.2018.12

Keywords:

gene delivery, cationic niosomes, lipid nanoparticles, squalane, helper lipids

Abstract

Gene therapy is a delivery of a defined therapeutic gene into specific cells of a patient to replace a defective gene. Lipid nanoparticles (LNPs) have been widely used as a carrier to improve the delivery efficiency. Of the LNPs delivery systems, niosomes which are formulated from non-ionic surfactants are generally cheaper and potentially more stable than liposomes which are formulated from phospholipids. However, it still has a storage stability problem. The aim of this study was therefore to investigate the effect of a helper lipid, namely squalane (Sq), on the physical stability, particle size, zeta potential, transfection efficiency and cytotoxicity of cationic niosomes. The cationic niosomes were composed of Span 20, cholesterol and spermine-based cationic lipid at a fixed molar ratio of 2.5:2.5:1, while the molar ratio of Sq was varied from 0.25 to 1. The zeta potential and the particle size of niosomes and niosome/DNA complexes were characterized. The results showed that the addition of Sq to the spermine-based niosomes reduced the particle size of the niosomes from 162.3 to 119.5 nm and increased the physical stability after kept at 4 °C for at least 4 weeks. In vitro transfection efficiency tested in HeLa cell revealed that niosome containing Sq at molar ratio of 1 (Sq1) exhibited comparable transfection efficiency to the niosome without Sq; however, lower amount of the Sq1 niosomes was required to form the complexes with DNA. None of the niosome formulations were toxic to cells at the niosome to DNA weight ratio which gave the highest transfection efficiency. These findings suggested that Sq may be used as a potential helper lipid in cationic niosomes for gene delivery.

References

1. Jin L, Zeng X, Liu M, Deng Y, He N. Current Progress in Gene Delivery Technology Based on Chemical Methods and Nano-carriers. Theranostics. 2014;4(3):240-55.

2. Yin H, Kanasty RL, Eltoukhy AA, Vegas AJ, Dorkin JR, Anderson DG. Non-viral vectors for gene-based therapy. Nat Rev Genet. 2014;15(8):541.

3. Loh XJ, Lee T-C, Dou Q, Deen GR. Utilising inorganic nanocarriers for gene delivery. Biomat sci. 2016;4(1):70-86.

4. Rezaee M, Oskuee RK, Nassirli H, Malaekeh-Nikouei B. Progress in the development of lipopolyplexes as efficient non-viral gene delivery systems. J Control Release. 2016;236:1-14.

5. Bishop CJ, Tzeng SY, Kozielski KL, Quinones-Hinojosa A, Green J. Polymeric nanoparticle systems for non-viral gene delivery. Front Bioeng Biotechnol.

6. Puras G, Mashal M, Zárate J, Agirre M, Ojeda E, Grijalvo S, et al. A novel cationic niosome formulation for gene delivery to the retina. J Control Release. 2014;174:27-36.

7. Li W, Szoka FC. Lipid-based Nanoparticles for Nucleic Acid Delivery. Pharm Res. 2007;24(3):438-49.

8. Vyas SP, Singh RP, Jain S, Mishra V, Mahor S, Singh P, et al. Non-ionic surfactant based vesicles (niosomes) for non-invasive topical genetic immunization against hepatitis B. Int J Pharm. 2005;296(1):80-6.

9. Zhdanov RI, Podobed OV, Vlassov VV. Cationic lipid–DNA complexes—lipoplexes—for gene transfer and therapy. Bioelectrochemistry. 2002;58(1):53-64.

10. Zuhorn IS, Engberts JBFN, Hoekstra D. Gene delivery by cationic lipid vectors: overcoming cellular barriers. Eur Biophys J. 2007;36(4):349-62.

11. Niyomtham N, Apiratikul N, Suksen K, Opanasopit P, Yingyongnarongkul B-e. Synthesis and in vitro transfection efficiency of spermine-based cationic lipids with different central core structures and lipophilic tails. Bioorg Med Chem Lett. 2015;25(3):496-503.

12. Kerner M, Meyuhas O, Hirsch-Lerner D, Rosen LJ, Min Z, Barenholz Y. Interplay in lipoplexes between type of pDNA promoter and lipid composition determines transfection efficiency of human growth hormone in NIH3T3 cells in culture. Biochim Biophys Acta. 2001;1532(1):128-36.

13. Hui SW, Langner M, Zhao YL, Ross P, Hurley E, Chan K. The role of helper lipids in cationic liposome-mediated gene transfer. Biophys J. 1996;71(2):590-9.

14. Dabkowska AP, Barlow DJ, Hughes AV, Campbell RA, Quinn PJ, Lawrence MJ. The effect of neutral helper lipids on the structure of cationic lipid monolayers. J R Soc Interface. 2012;9(68):548-61.

15. Hirsch-Lerner D, Zhang M, Eliyahu H, Ferrari ME, Wheeler CJ, Barenholz Y. Effect of “helper lipid” on lipoplex electrostatics. Biochim Biophys Acta. 2005;1714(2):71-84.

16. Köster F, Finas D, Schulz C, Hauser C, Diedrich K, Felberbaum R. Additive effect of steroids and cholesterol on the liposomal transfection of the breast cancer cell line T-47D. Int J Mol Med. 2004;14(4):769-841.

17. Zuidam NJ, Barenholz Y. Electrostatic and structural properties of complexes involving plasmid DNA and cationic lipids commonly used for gene delivery1A preliminary report of this study was presented at the 3rd annual conference: Artificial Self-Assembling Systems for Gene Delivery, organized by Cambridge Healthtec Institute, November 17–18, 1996, Coronado, CA.1. Biochim Biophys Acta. 1998;1368(1):115-28.

18. Simberg D, Danino D, Talmon Y, Minsky A, Ferrari ME, Wheeler CJ, et al. Phase behavior, DNA ordering, and size instability of cationic lipoplexes Relevance to optimal transfection activity. J Biol Chem. 2001;276(50):47453-9.

19. Du Z, Munye MM, Tagalakis AD, Manunta MD, Hart SL. The role of the helper lipid on the DNA transfection efficiency of lipopolyplex formulations. Sci Rep. 2014;4:7107.

20. Ojeda E, Puras G, Agirre M, Zarate J, Grijalvo S, Eritja R, et al. The role of helper lipids in the intracellular disposition and transfection efficiency of niosome formulations for gene delivery to retinal pigment epithelial cells. Int J Pharm. 2016;503(1):115-26.

21. Sakurai F, Nishioka T, Yamashita F, Takakura Y, Hashida M. Effects of erythrocytes and serum proteins on lung accumulation of lipoplexes containing cholesterol or DOPE as a helper lipid in the single-pass rat lung perfusion system. Eur J Pharm Biopharm. 2001;52(2):165-72.

22. Cheng X, Lee RJ. The role of helper lipids in lipid nanoparticles (LNPs) designed for oligonucleotide delivery. Adv Drug Del Rev. 2016;99:129-37.

23. Kircheis R, Wightman L, Wagner E. Design and gene delivery activity of modified polyethylenimines. Adv Drug Del Rev. 2001;53(3):341-58.

24. Motta S, Brocca P, Favero ED, Rondelli V, Cantù L, Amici A, et al. Nanoscale structure of protamine/DNA complexes for gene delivery. Appl Phys Lett. 2013;102(5):053703.

25. Inoh Y, Nagai M, Matsushita K, Nakanishi M, Furuno T. Gene transfection efficiency into dendritic cells is influenced by the size of cationic liposomes/DNA complexes. Eur J Pharm Sci. 2017;102:230-6.

26. Kneuer C, Ehrhardt C, Bakowsky H, Ravi Kumar MNV, Oberle V, Lehr CM, et al. The Influence of Physicochemical Parameters on the Efficacy of Non-Viral DNA Transfection Complexes: A Comparative Study. J Nanosci Nanotechnol. 2006;6(9-10):2776-82.

27. Prabha S, Zhou W-Z, Panyam J, Labhasetwar V. Size-dependency of nanoparticle-mediated gene transfection: studies with fractionated nanoparticles. Int J Pharm. 2002;244(1):105-15.

28. Mansouri S, Cuie Y, Winnik F, Shi Q, Lavigne P, Benderdour M, et al. Characterization of folate-chitosan-DNA nanoparticles for gene therapy. Biomaterials. 2006;27(9):2060-5.

29. Opanasopit P, Leksantikul L, Niyomtham N, Rojanarata T, Ngawhirunpat T, Yingyongnarongkul B-E. Cationic niosomes an effective gene carrier composed of novel spermine-derivative cationic lipids: effect of central core structures. Pharm Dev Technol. 2017;22(3):350-9.

30. Paecharoenchai O, Niyomtham N, Ngawhirunpat T, Rojanarata T, Yingyongnarongkul B-e, Opanasopit P. Cationic niosomes composed of spermine-based cationic lipids mediate high gene transfection efficiency. J Drug Target. 2012;20(9):783-92.

31. Dash PR, Read ML, Barrett LB, Wolfert MA, Seymour LW. Factors affecting blood clearance and in vivo distribution of polyelectrolyte complexes for gene delivery. Gene Ther. 1999;6:643.

32. Fröhlich E. The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. Int J Nanomed. 2012;7:5577-91.

Downloads

Published

2018-06-27

Issue

Section

Original Research Articles