Document Type : Research Article

Authors

1 Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran

2 Research Institute of Food Science and Technology (RIFST), Mashhad, Iran

Abstract

Introduction
 Gelatin is one of the most widely used colloidal proteins, which has unique hydrocolloidal property. Gelatin is derived from collagen by changing the thermal nature. This product is widely used in food, pharmaceutical, biomedical, cosmetic and photography industries. Global gelatin demand for food and non-food products is increasing. Two important properties of nanoparticles are: Increasing the surface-to-volume ratio of nanoparticles causes the atoms on the surface to have a much greater effect on their properties than the atoms within the particle volume. The effects of quantum size, which is the second feature. Methods for preparing nanoparticles from natural macromolecules: In general, two major methods for making protein nanoparticles have been reported Emulsion-solvent evaporation method and sedimentation or phase separation method in aqueous medium. Numerous methods have been reported for the preparation of nanoparticles from natural macromolecules. The first method is based on emulsification and the second method is based on phase separation in aqueous medium. In the first method, due to the instability of the emulsion, it is not possible to prepare nanoparticles smaller than 500 nm with a narrow particle size distribution. Therefore, coagulation method or anti-solvent method which is based on phase separation was proposed to prepare nanoparticles from natural macromolecules.
 
Materials and Methods
 Type B (cow) gelatin was purchased from processing company with Bloom 260-240 food and pharmaceutical Iran solvent gelatin solution of 25% aqueous acetate glutaraldehyde from Iran Neutron Company. Two-stage anti-solvent method was used to produce gelatin nanoparticles. Then, to form nanoparticles, acetone was added dropwise while stirring until the dissolved acetone begins to change color and eventually turns white, which indicates the formation of nanoparticles. Finally, glutaraldehyde solution was added for cross-linking and finally centrifuged.
 
 Results and Discussion
 The results showed that with increasing gelatin concentration, nanoparticle size and PDI increased significantly. According to the announced results, the solvent has a direct effect on the size. Therefore, the best mixing speed is determined to achieve the smallest particle size. Zeta potential is the best indicator for determining the electrical status of the particle surface and a factor for the stability of the potential of the colloidal system because it indicates the amount of charge accumulation in the immobile layer and the intensity of adsorption of opposite ions on the particle surface. If all the particles in the suspension are negatively or positively charged, the particles tend to repel each other and do not tend to accumulate. The tendency of co-particles to repel each other is directly related to the zeta potential. Fabricated gelatin nanoparticles have a stable structure, and are heat resistant. These nanoparticles are ready to be used to accept a variety of aromatic substances, compounds with high antioxidant properties, a variety of vitamins and heat-sensitive substances.
 
Conclusion
The results of this study showed that the optimal conditions for the production of a particle of 88.6 nm at 40 ° C, the volume of acetone consumption was 15 ml, concentration 200 mg and speed 1000 rpm, and the morphology of gelatin nanoparticles have resistant, spherical polymer structure and mesh with a smooth surface that can be clearly seen under an electron microscope.   

Keywords

Main Subjects

©2023 The author(s). This is an open access article distributed under Creative Commons Attribution 4.0 International License (CC BY 4.0), which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source.

  1. Alavijeh, R., Shokrollahi, P., & Barzin, J. (2017). A thermally and water activated shape memory gelatin physical hydrogel, with a gel point above the physiological temperature, for biomedical applications. Journal of Materials Chemistry B, 5)12(, 2302-2314. https://doi.org/10.1039/C7TB00014F
  2. , S., Abrams, D., Huang, Y., McQuarrie, S., Roa, W., Chen, H., Löbenberg, R., Miller, G.G., & Finlay, W.H. (2006). Optimization of a twostep desolvation method for preparing gelatin nanoparticles and cell uptake studies in 143B osteosarcoma cancer cells. Journal Pharmacy Science, 9(1), 124–132. https://doi.org/10.7939/R3J96097M
  3. Devika, R.B., & Varsha, B.P. (2006). Studies on effect of pH on cross-linking of chitosan with sodium tripolyphosphate: a technical note, AAPS Pharm. Science Technology, 7, 1–6. https://doi.org/10.1208/pt070250
  4. Elzoghby, O. (2013). Gelatin-based nanoparticles as drug and gene delivery systems: Reviewing three decades of research, Journal Control Release, 172(3), 1075–1091.
  5. , Z., & Jafarpour, A. (2020). A comparative study on physio-chemical properties of recovered gelatin from beluga (Huso huso) fish skin by enzymatic and chemical methods, Iranian Food Science and Technology Research Journal, 16, 157-170. https://doi.org/10.22067/ifstrj.v16i1.76991
  6. , S.Y., Yang, H., & Chen, P. (2007). Formation of colloidal suspension of hydrophobic compounds with an amphiphilic self-assembling peptide, Colloids and Surfaces B: Biointerfaces, 55(2), 200-211. https://doi.org/10.1016/j.colsurfb.2006.12.002
  7. GMIA, G.H. (2012). Gelatin Manufacturers Institute of America, New York.
  8. Gomathi, T., Prasad, P.S., Sudha, P.N., & Anil, S. (2017). Size optimization and in vitro biocompatibility studies of chitosan nanoparticles, International Journal of Biological Macromolecules, https://doi.org/10.1016/j.ijbiomac.2017.08.057
  9. Gupta, A.K., Gupta, M.S., Yarwood, J., & Curtis, A.S.G. (2004). Effect of cellular uptake of gelatin nanoparticles on adhesion, morphology and cytoskeleton organisation of human fibroblasts, Journal of Controlled Release, 95(2), 197-207. https://doi.org/10.1016/j.jconrel.2003.11.006
  10. Hosseini, S.M., Emam-Djomeh, H.Z., Razavi, S.H., Saboury, A.A., Moosavi-Movahedi, A.A., & Atri, Meeren, M.S. (2013). Food Hydrocolloids b -Lactoglobulin e sodium alginate interaction as affected by polysaccharide depolymerization using high intensity ultrasound, FOOHYD, 32(2), 235–244. https://doi.org/10.1016/j.foodhyd.2013.01.002
  11. Ishikawa, H., Nakamura, Y., Jo, J., & Tabata, Y. (2012). Gelatin nanospheres incorporating siRNA for controlled intracellular release, Biomaterials, 33(35), 9097-104. https://doi.org/10.1016/j.biomaterials.2012.08.032
  12. Iida, H., Takayanagi, K., Nakanishi, T., & Osaka, T. (2007). Synthesis of Fe3O4 nanoparticles with various sizes and magnetic properties by controlled hydrolysis, Journal of Colloid and Interface Science, 314(1), 274-280. https://doi.org/10.1016/j.jcis.2007.05.047
  13. Jahanshahi, M., Sanati, M.H., Minuchehr, Z., Hajizadeh, S., & Babaei, Z. (2008). Controlled fabrication of gelatin nanoparticles as drug carriers, Nanotechnol. Its Appl. First Sharjah Int. Conf., 228, pp. 228–232. https://doi.org/10.1063/1.2776720
  14. Jalaja, K., Naskar, D., Kundu, S.C., & James, N.R. (2015). Fabrication of cationized gelatin nanofibers by electrospinning for tissue regeneration. Rsc Advances, 5(109), 89521-89530. https://doi.org/10.1039/C5RA10384C
  15. Jones, O., Decker, E.A., & McClements, D.J. (2010). Thermal analysis of ß-lactoglobulin complexes with pectins. https://doi.org/10.1016/j.foodhyd.2009.10.001
  16. Kirby, B.J., & Hasselbrink, E.F. (2004). Zeta potential of microfluidic substrates: 1. Theory, experimental techniques, and effects on separations, Electrophoresis, 25(2), 187-202. https://doi.org/1002/elps.200305754
  17. Kaintura, R., Sharma, P., Singh, S., Rawat, K., & Solanki, P. (2015). Gelatin nanoparticles as a drug delivery system for proteins, Journal of Nanomedicine Research, 2, 00018. or carrageenan for production of stable biopolymer particles, Food Hydrocolloids, 24(2), 239-248. https://doi.org/1007/s10853-015-9287-3
  18. Khan, S.A., & Schneider, M. (2013). Improvement of nanoprecipitation technique for preparation of gelatin nanoparticles and potential macromolecular drug loading, Macromolecular Bioscience, 13(4), 455-463. http://doi.org//10.1002/mabi.201200382
  19. Kommareddy,S., Shenoy, D.B., & Amiji, M.M. (2007). Gelatin nanoparticles and their biofunctionalization, Nanotechnologies Lifesci no. september. https://doi.org/10.1002/9783527610419.ntls0011
  20. Lohcharoenkal, W., Wang, L., Wang, Y., Chen, C., & Rojanasakul, Y. (2014). Protein nanoparticles as drug delivery carriers for cancer therap Wang,y, BioMed research international. https://doi.org/10.1155/2014/180549
  21. Li, J., Li, K., Wang, N., & Wu, X.S. (1997). A novel biodegradable system based on gelatin nanoparticles and poly (lactic‐co‐glycolic acid) microspheres for protein and peptide drug delivery, Journal of Pharmaceutical Sciences, 86(8), 891-895.
  22. Luo, Y., Teng, Z., & Wang, Q. (2012). Development of zein nanoparticles coated with carboxymethyl chitosan for encapsulation and controlled release of vitamin D3, Journal of Agricultural and Food Chemistry.
  23. Ma, Y., Zheng, Y., Zeng, X., Jiang, L., Chen, H., Liu, R., Huang, L., & Mei., L. (2011). Novel docetaxel-loaded nanoparticles based on PCL-Tween 80 copolymer for cancer treatment, International Journal of Nanomedicine, 6(2679), e88. https://doi.org/10.2147/IJN.S25251
  24. Nejat, H., Rabiee, M., Tahriri, M., Jazayeri, H.E., Rajadas, J., Ye, H., Cui, Z., & Tayebi, L. (2017). Preparation and characterization of cardamom extract-loaded gelatin nanoparticles as effective targeted drug delivery system to treat glioblastoma. Reactive and Functional Polymers, 120, 46–56. https://doi.org/10.1016/j.reactfunctpolym.2017.09.008
  25. Narayanan, D., Koyakutty, G.M.G, L. H, M., Nair, S., & Menon, D. (2013). Poly-(ethylene glycol) modified gelatin nanoparticles for sustained delivery of the anti-inflammatory drug Ibuprofen-Sodium: An in vitro and in vivo analysis, Nanomedicine: Nanotechnology, Biology and Medicine, 9(6), 818-828. https://doi.org/1016/j.nano.2013.02.001
  26. Nguyen, T.H., & Lee, B.T. (2010). Fabrication and characterization of cross-linked gelatin electro-spun nano-fibers, Journal of Biomedical Science and Engineering, 3(12), 11-17. https://doi.org/4236/jbise.2010.312145
  27. Rejinold, N.S., Muthunarayanan, M., Muthuchelian, K., Chennazhi, K.P., Nair., S.V., & Jayakumar, R. (2011). Saponin-loaded chitosan nanoparticles and their cytotoxicity to cancer cell lines in vitro. Carbohydrate Polymers, 84, 407–416.
  28. Shutava, T.G., Balkundi, S.S., Vangala, P., Steffan, J.J., Bigelow, R.L., Cardelli, J.A., O’Neal, D.P., & Lvov, Y.M. (2009). Layer-by-layer-coated gelatin nanoparticles as a vehicle for delivery of natural polyphenols, Journal American Chemical Society, 3(7), 1877–1885. https://doi.org/1021/nn900451a
  29. Yang, L., Jian, W., Qiu, L., Jian, X., Zuo, D., Wang, D., & Yang, L. (2015). One pot synthesis of highly luminescent polyethylene glycol anchored carbon dots functionalized with a nuclear localization signal peptide for cell nucleus imaging, Nanoscale, 7(14), 6104-6113. https://doi.org/1039/C5NR01080B
  30. Y, W., W, Z., L, Y., G.X, C. (2000). Preparation and characterization of gelatin gel with a gradient structure. Polymer International, 49, 1600-1603.

https://doi.org/10.1002/1097-0126(200012)49:12<1600::AID-PI554>3.0.CO;2-K

 

CAPTCHA Image