Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1588 word(s) 1588 2021-03-25 07:29:10 |
2 format correct -209 word(s) 1379 2021-04-06 12:04:35 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Shaabani, E. Chitosan-Coated Gold Nanoparticles. Encyclopedia. Available online: https://encyclopedia.pub/entry/8475 (accessed on 29 March 2024).
Shaabani E. Chitosan-Coated Gold Nanoparticles. Encyclopedia. Available at: https://encyclopedia.pub/entry/8475. Accessed March 29, 2024.
Shaabani, Elnaz. "Chitosan-Coated Gold Nanoparticles" Encyclopedia, https://encyclopedia.pub/entry/8475 (accessed March 29, 2024).
Shaabani, E. (2021, April 06). Chitosan-Coated Gold Nanoparticles. In Encyclopedia. https://encyclopedia.pub/entry/8475
Shaabani, Elnaz. "Chitosan-Coated Gold Nanoparticles." Encyclopedia. Web. 06 April, 2021.
Chitosan-Coated Gold Nanoparticles
Edit

Delivery of small interfering RNA (siRNA) provides one of the most powerful strategies for downregulation of therapeutic targets. Despite the widely explored capabilities of this strategy, intracellular delivery is hindered by a lack of carriers that have high stability, low toxicity and high transfection efficiency. Here we propose a layer by layer (LBL) self-assembly method to fabricate chitosan-coated gold nanoparticles (CS-AuNPs) as a more stable and efficient siRNA delivery system. 

gold nanoparticles Chitosan layer by layer assembly siRNA gene delivery endosomal escape

1. Introduction

Small interfering RNA (siRNA) is a powerful therapeutic technology which induces post-transcriptional gene silencing via translation inhibition or by cleavage of the target mRNA by incorporating into the RNA-induced silencing complex (RISC) in the cytoplasm [1][2]. Fast enzymatic digestion, limited cellular uptake, inefficient release from endosomes, and lack of cell-specific targeting are the major obstacles of using naked (free) siRNA [3][4][5]. Thus, the therapeutic application of siRNA molecules requires suitable carriers to allow them to be delivered inside target cells in a safe and effective manner. Many different delivery systems have been explored to date. These include viral [5] and non-viral vectors [6][7], each of which come with their own set of advantages and disadvantages. For example, although viral vectors are highly efficient, they are associated with safety concerns such as inherent immunogenicity [8], mutagenesis [9], oncogenic potential, and inflammation [10]. These concerns have triggered the development of non-viral vectors for siRNA delivery including cationic lipids, polymers, peptides and inorganic nanoparticles [7][11][12]. While non-viral vectors are considered to be safer with a reduced immunogenic response, they come with their own drawbacks. For instance, cationic liposomes can suffer from low colloidal stability, difficulties in controlled release, fast elimination in the body, and poor targeting [13]. Polypeptides for gene delivery have limited efficiency and are associated with toxic side effects [14]. In addition, cationic polymer-based carriers can suffer from low stability [15] as well as poor cell recognition and internalization [16]. In spite of those challenges, non-viral vectors remain of great current interest given their better safety profiles and ease of production at an affordable costs [17].

One of the most widely used polymers for the design of nanocarriers for siRNA delivery is chitosan [18][19][20]. Chitosan is a linear biopolymer consisting of randomly repeating D-glucosamine and N-acetyl-d-glucosamine units [21]. Chitosan is a weak polybase with a pKa around 6.5 which offers the advantage of being biodegradable and biocompatible while at the same time being highly positively charged at a pH below the pKa so that it can easily form electrostatic complexes with nucleic acids [22]. On the downside, it suffers from poor stability [23][24] and an undefined structural composition [25]. While improvements have been suggested by making blends with other polymers or modifying its chemical structure [23][26], there is still a need for increasing the stability of chitosan as a gene delivery vehicle. Inorganic nanoparticles like gold nanoparticles (AuNPs) have attracted great interest because of several advantages including their simple synthesis, tunable size and surface properties, good biocompatibility, and multifunctional capabilities [27][28][29]. These unique properties make AuNPs attractive stabilizing scaffolds for gene delivery vehicles. In particular, AuNPs can be used as scaffolds for layer-by-layer (LBL) self-assembly, which is a widely used technique to deposit multiple layers of positively and negatively charged polymers onto surfaces of films or nanoparticles [30][31][32]. Such hybrids of cationic polymers and inorganic nanoparticles combine the advantages of both systems to achieve increased gene delivery efficacy [33]. For instance, LBL assembled AuNP-siRNA have been prepared with polymers that can induce endosomal escape, such as polyethylenimine (PEI) [34][35][36][37][38]. However, release of siRNA in the cellular cytoplasm remains limited due to the high binding affinity between AuNPs-PEI and siRNA [39]. In addition, PEI is often associated with high cytotoxicity due to inducing membrane perturbations and chromosome aberrations [40].

Therefore, in this study we propose LBL assembled AuNP-siRNA in combination with chitosan as a biocompatible and biodegradable cationic polymer to obtain highly stable gene delivery carriers for efficient intracellular siRNA delivery. AuNPs capped with chitosan (CS-AuNPs) are synthesized by using chitosan as both the reducing and stabilizing agent [41][42]. Next, negatively charged siRNA is incorporated as the next layer on top of the positively charged CS-AuNPs. Finally, a third chitosan layer is applied to protect the siRNA and endow the particles with a net positive charge which allows them to easily adsorb to the negatively charged cell membrane, hence facilitating endocytic uptake.

Long term stability in storage buffer, stability in different media, and siRNA release are investigated. siRNA gene silencing is subsequently evaluated in H1299 cells stably expressing green fluorescent protein (H1299-eGFP) (as shown in Scheme 1) as a model that allows for an easy readout of siRNA silencing. Cellular uptake, endosomal escape transfection efficiency and cell viability are all studied. We find that the here presented AuNP-based carriers show better stability and efficacy than commercial transfection agents (Lipofectamine (cationic lipid mediated transfection) and jetPEI® (cationic polymer)) or nanocarriers prepared from chitosan alone (CSNPs).

Scheme 1. Schematic representation of LBL-CS-AuNPs for siRNA delivery. Under normal conditions, H1299-eGFP cells continuously express enhanced Green Fluorescent Protein (eGFP). When LBL-CS-AuNPs are taken up by cells via endocytosis, chitosan can induce endosomal escape. Then, the released siRNA (si-eGFP) is processed by the RNA-induced silencing complex (RISC), which targets and cleaves GFP-mRNA. The cleavage of GFP-mRNA leads to decreased GFP expression and a diminished green fluorescence intensity of H1299-eGFP cells.

2. Nanoparticle Formation, Characterization and Stability

CS-AuNPs have been synthetized in a one-step synthetic method which used chitosan (CS) both as the reducing agent and stabilizer to generate CS-capped AuNPs. The appearance of a LSPR peak at 524 nm in the UV-Visible spectrum and the absence of plasmonic bands associated with agglomeration of nanoparticles confirmed that the CS-AuNPs are stable and do not show aggregation. For the second layer of the LBL coating, different ratios of siRNA were evaluated for siRNA attachment on the surface of the CS-AuNPs. Then, a final CS layer was applied to protect the loaded siRNA from preventing fast release and ensuring efficient uptake by cells and efficient endosomal escape.

Evaluation of siRNA release profiles and colloidal stability indicated that LBL-CS-AuNPs were more stable than CSNPs which are composed of chitosan alone. We hypothesize that the macromolecular organization of the polymer layer on the surface of the gold nanoparticles confers to their high colloidal stability due to the high cationic charge and the steric effect of the chitosan.

3. Uptake and Transfection Efficiency of Nanoparticles

The biocompatibility of a vector for siRNA delivery is an important consideration. Investigation of metabolic activity and induction of apoptosis showed that CSNPs induced very little toxicity, even at the highest concentration, while for LBL-CS-AuNPs, the toxicity gradually increased with increasing concentration. Chitosan is known to be a biocompatible polymer so the low toxicity by CSNPs is not surprising. The fact that LBL-CS-AuNP induced more toxicity is very likely due to enhanced cellular uptake, as we indeed could observe by using fluorescently labeled siRNA. For CSNPs, the rMFI did not increase substantially with increasing NP concentration, indicating that under the studied concentrations, the uptake machinery was already saturated. Indeed, it has been previously suggested that endocytic uptake pathways may be different for particles of different sizes [43][44]. Therefore, it cannot be excluded that the larger CSNPs are taken up via a different endocytic pathway, which perhaps may saturate more quickly, than the smaller LBL-CS-AuNPs. Regardless of the underlying mechanism, enhanced uptake of LBL-CS-AuNPs resulted in a maximum gene silencing of 76% for LBL-CS-AuNP with 24 nM siRNA, while this remained limited to 49% for CSNPs with 50 nM siRNA.

When comparing transfection efficiencies with two commercial transfection reagents, jet-PEI and Lipofectamine, it was found that CSNPs showed similar effects as jetPEI®, while being much less effective than Lipofectamine. LBL-CS-AuNPs at 8 nM, on the other hand, performed even better than Lipofectamine, with similar knockdown efficiencies at higher siRNA concentrations. Importantly, unlike the other carriers (commercial transfection reagents or CSNPs), LBL-CS-AuNPs were still as functional 7 days after synthesis, which is yet another demonstration of superior stability of LBL-CS-AuNPs.

4. Endosomal Escape Efficiency

In order to explain the experimental observations of the silencing effect, we proceeded to determine and quantify the endosomal escape capacity of all the NPs evaluated. After image analysis (~500 cells for each sample), a direct correlation was found between the extent of endosomal escape and the transfection efficiencies, indicating that the escape from endosomes is one of the main factors in the effectiveness of the siRNA delivery carriers evaluated here.

References

  1. Burnett, J.C.; Rossi, J.J. RNA-based therapeutics: Current progress and future prospects. Chem. Biol. 2012, 19, 60–71.
  2. Rao, D.D.; Vorhies, J.S.; Senzer, N.; Nemunaitis, J. siRNA vs. shRNA: Similarities and differences. Adv. Drug Deliv. Rev. 2009, 61, 746–759.
  3. Oh, Y.-K.; Park, T.G. siRNA delivery systems for cancer treatment. Adv. Drug Deliv. Rev. 2009, 61, 850–862.
  4. Aagaard, L.; Rossi, J.J. RNAi therapeutics: Principles, prospects and challenges. Adv. Drug Deliv. Rev. 2007, 59, 75–86.
  5. Juliano, R.L. The delivery of therapeutic oligonucleotides. Nucleic Acids Res. 2016, 44, 6518–6548.
  6. Kanasty, R.; Dorkin, J.R.; Vegas, A.; Anderson, D. Delivery materials for siRNA therapeutics. Nat. Mater. 2013, 12, 967–977.
  7. Dong, Y.; Siegwart, D.J.; Anderson, D.G. Strategies, design, and chemistry in siRNA delivery systems. Adv. Drug Deliv. Rev. 2019, 144, 133–147.
  8. Verma, I.M. A tumultuous year for gene therapy. Mol. Ther. 2000, 2, 415–416.
  9. Couzin, J.; Kaiser, J. As Gelsinger Case Ends, Gene Therapy Suffers Another Blow; American Association for the Advancement of Science: Washington, DC, USA, 2005.
  10. Timmers, A.M.; Newmark, J.A.; Turunen, H.T.; Farivar, T.; Liu, J.; Song, C.; Ye, G.-j.; Pennock, S.; Gaskin, C.; Knop, D.R. Ocular inflammatory response to intravitreal injection of adeno-associated virus vector: Relative contribution of genome and capsid. Hum. Gene Ther. 2020, 31, 80–89.
  11. Wicki, A.; Witzigmann, D.; Balasubramanian, V.; Huwyler, J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J. Control. Release 2015, 200, 138–157.
  12. Kim, H.J.; Ishii, A.; Miyata, K.; Lee, Y.; Wu, S.; Oba, M.; Nishiyama, N.; Kataoka, K. Introduction of stearoyl moieties into a biocompatible cationic polyaspartamide derivative, PAsp (DET), with endosomal escaping function for enhanced siRNA-mediated gene knockdown. J. Control. Release 2010, 145, 141–148.
  13. Sriraman, S.K.; Torchilin, V.P. Recent advances with liposomes as drug carriers. Adv. Biomater. Biodevices 2014, 2, 79–119.
  14. Kang, Z.; Meng, Q.; Liu, K. Peptide-based gene delivery vectors. J. Mater. Chem. B 2019, 7, 1824–1841.
  15. Wang, Y.; Ye, M.; Xie, R.; Gong, S. Enhancing the in vitro and in vivo stabilities of polymeric nucleic acid delivery nanosystems. Bioconjugate Chem. 2018, 30, 325–337.
  16. Ullah, I.; Muhammad, K.; Akpanyung, M.; Nejjari, A.; Neve, A.L.; Guo, J.; Feng, Y.; Shi, C. Bioreducible, hydrolytically degradable and targeting polymers for gene delivery. J. Mater. Chem. B 2017, 5, 3253–3276.
  17. Bono, N.; Ponti, F.; Mantovani, D.; Candiani, G. Non-Viral in Vitro Gene Delivery: It is Now Time to Set the Bar! Pharmaceutics 2020, 12, 183.
  18. Chuan, D.; Jin, T.; Fan, R.; Zhou, L.; Guo, G. Chitosan for gene delivery: Methods for improvement and applications. Adv. Colloid Interface Sci. 2019, 268, 25–38.
  19. Ragelle, H.; Vanvarenberg, K.; Vandermeulen, G.; Préat, V. Chitosan nanoparticles for SiRNA delivery in vitro. In SiRNA Delivery Methods; Springer: Berlin/Heidelberg, Germany, 2016; pp. 143–150.
  20. Kang, S.H.; Revuri, V.; Lee, S.-J.; Cho, S.; Park, I.-K.; Cho, K.J.; Bae, W.K.; Lee, Y.-K. Oral siRNA delivery to treat colorectal liver metastases. ACS Nano 2017, 11, 10417–10429.
  21. Andrés, E.; Albesa-Jové, D.; Biarnés, X.; Moerschbacher, B.M.; Guerin, M.E.; Planas, A. Structural basis of chitin oligosaccharide deacetylation. Angew. Chem. 2014, 126, 7002–7007.
  22. Cao, Y.; Tan, Y.F.; Wong, Y.S.; Liew, M.W.J.; Venkatraman, S. Recent advances in chitosan-based carriers for gene delivery. Mar. Drugs 2019, 17, 381.
  23. Szymańska, E.; Winnicka, K. Stability of chitosan—A challenge for pharmaceutical and biomedical applications. Mar. Drugs 2015, 13, 1819–1846.
  24. Supper, S.; Anton, N.; Boisclair, J.; Seidel, N.; Riemenschnitter, M.; Curdy, C.; Vandamme, T. Chitosan/glucose 1-phosphate as new stable in situ forming depot system for controlled drug delivery. Eur. J. Pharm. Biopharm. 2014, 88, 361–373.
  25. Lv, H.; Zhang, S.; Wang, B.; Cui, S.; Yan, J. Toxicity of cationic lipids and cationic polymers in gene delivery. J. Control. Release 2006, 114, 100–109.
  26. Ragelle, H.; Riva, R.; Vandermeulen, G.; Naeye, B.; Pourcelle, V.; le Duff, C.S.; D’Haese, C.; Nysten, B.; Braeckmans, K.; de Smedt, S.C. Chitosan nanoparticles for siRNA delivery: Optimizing formulation to increase stability and efficiency. J. Control. Release 2014, 176, 54–63.
  27. Li, N.; Larson, T.; Nguyen, H.H.; Sokolov, K.V.; Ellington, A.D. Directed evolution of gold nanoparticle delivery to cells. Chem. Commun. 2010, 46, 392–394.
  28. Pissuwan, D.; Niidome, T.; Cortie, M.B. The forthcoming applications of gold nanoparticles in drug and gene delivery systems. J. Control. Release 2011, 149, 65–71.
  29. Singh, P.; Pandit, S.; Mokkapati, V.; Garg, A.; Ravikumar, V.; Mijakovic, I. Gold nanoparticles in diagnostics and therapeutics for human cancer. Int. J. Mol. Sci. 2018, 19, 1979.
  30. Bricaud, Q.; Fabre, R.M.; Brookins, R.N.; Schanze, K.S.; Reynolds, J.R. Energy transfer between conjugated polyelectrolytes in layer-by-layer assembled films. Langmuir 2011, 27, 5021–5028.
  31. Decher, G.; Hong, J.D. Buildup of ultrathin multilayer films by a self-assembly process, 1 consecutive adsorption of anionic and cationic bipolar amphiphiles on charged surfaces. In Makromolekulare Chemie. Macromolecular Symposia; Hüthig & Wepf Verlag: Basel, Switzerland, 1991; pp. 321–327.
  32. Fraire, J.C.; Houthaeve, G.; Liu, J.; Raes, L.; Vermeulen, L.; Stremersch, S.; Brans, T.; Barriga, G.G.-D.; de Keulenaer, S.; van Nieuwerburgh, F. Vapor nanobubble is the more reliable photothermal mechanism for inducing endosomal escape of siRNA without disturbing cell homeostasis. J. Control. Release 2020, 319, 262–275.
  33. Guo, K.; Zhao, X.; Dai, X.; Zhao, N.; Xu, F.J. Organic/inorganic nanohybrids as multifunctional gene delivery systems. J. Gene Med. 2019, 21, e3084.
  34. Akinc, A.; Thomas, M.; Klibanov, A.M.; Langer, R. Exploring polyethylenimine-mediated DNA transfection and the proton sponge hypothesis. J. Gene Med. Cross-Discip. J. Res. Sci. Gene Transf. Its Clin. Appl. 2005, 7, 657–663.
  35. Elbakry, A.; Zaky, A.; Liebl, R.; Rachel, R.; Goepferich, A.; Breunig, M. Layer-by-layer assembled gold nanoparticles for siRNA delivery. Nano Lett. 2009, 9, 2059–2064.
  36. Han, L.; Zhao, J.; Zhang, X.; Cao, W.; Hu, X.; Zou, G.; Duan, X.; Liang, X.-J. Enhanced siRNA delivery and silencing gold–chitosan nanosystem with surface charge-reversal polymer assembly and good biocompatibility. ACS Nano 2012, 6, 7340–7351.
  37. Chen, Z.; Zhang, L.; He, Y.; Shen, Y.; Li, Y. Enhanced shRNA Delivery and ABCG2 Silencing by Charge-Reversible Layered Nanocarriers. Small 2015, 11, 952–962.
  38. Labala, S.; Jose, A.; Venuganti, V.V.K. Transcutaneous iontophoretic delivery of STAT3 siRNA using layer-by-layer chitosan coated gold nanoparticles to treat melanoma. Colloids Surf. B Biointerfaces 2016, 146, 188–197.
  39. Guo, S.; Huang, Y.; Jiang, Q.; Sun, Y.; Deng, L.; Liang, Z.; Du, Q.; Xing, J.; Zhao, Y.; Wang, P.C. Enhanced gene delivery and siRNA silencing by gold nanoparticles coated with charge-reversal polyelectrolyte. ACS Nano 2010, 4, 5505–5511.
  40. Parhamifar, L.; Larsen, A.K.; Hunter, A.C.; Andresen, T.L.; Moghimi, S.M. Polycation cytotoxicity: A delicate matter for nucleic acid therapy—focus on polyethylenimine. Soft Matter 2010, 6, 4001–4009.
  41. Huang, H.; Yang, X. Synthesis of chitosan-stabilized gold nanoparticles in the absence/presence of tripolyphosphate. Biomacromolecules 2004, 5, 2340–2346.
  42. Simeonova, S.; Georgiev, P.; Exner, K.S.; Mihaylov, L.; Nihtianova, D.; Koynov, K.; Balashev, K. Kinetic study of gold nanoparticles synthesized in the presence of chitosan and citric acid A Physicochemical and engineering aspects. Colloids Surf. A Physicochem. Eng. Asp. 2018, 557, 106–115.
  43. Zhang, S.; Li, J.; Lykotrafitis, G.; Bao, G.; Suresh, S. Size-dependent endocytosis of nanoparticles. Adv. Mater. 2009, 21, 419–424.
  44. Rejman, J.; Oberle, V.; Zuhorn, I.S.; Hoekstra, D. Size-dependent internalization of particles via the pathways of clathrin-and caveolae-mediated endocytosis. Biochem. J. 2004, 377, 159–169.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 409
Revisions: 2 times (View History)
Update Date: 06 Apr 2021
1000/1000