ANTIOXIDANT EFFECT OF CATECHIN LOADED POLYMERIC NANOPARTICLE

Flavonoids are polyphenols compounds a structural class of mainly natural and a products of secondary metabolism of plants. Flavonoids, which can be found in herbs and trees, act as natural chemopreventives and anticancer agents [1]. As natural products, they have major importance for human life and also have a widespread effect such as antioxidant, antinflammatory, anticarcinogen, antidiabetic [2, 3]. However, bioavailability of this polyphenolic antioxidant have limited because of their high water solubility [4], low absorption, permeability, stability, slow dissolution rate, and light-induced decomposition over time in the physiological medium. In recent years, various methods have been tried to overcome the limitations of flavonoids, including its incorporation into micels and nanoparticles.

___

  • [1] Papademetrio, D.L., et al., The catechin flavonoid reduces proliferation and induces apoptosis of murine lymphoma cells LB02 through modulation of antiapoptotic proteins. Revista Brasileira de Farmacognosia, 2013. 23(3): p. 455-463.
  • [2] Middleton, E., C. Kandaswami, and T.C. Theoharides, The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacological reviews, 2000. 52(4): p. 673-751.
  • [3] Sporn, M.B. and N. Suh, Chemoprevention: an essential approach to controlling cancer. Nature Reviews Cancer, 2002. 2(7): p. 537-543.
  • [4] Aditya, N., et al., Co-delivery of hydrophobic curcumin and hydrophilic catechin by a water-in-oil-in-water double emulsion. Food chemistry, 2015. 173: p. 7-13.
  • [5] Srivastava, A.K., et al., Synthesis of PLGA nanoparticles of tea polyphenols and their strong in vivo protective effect against chemically induced DNA damage. International journal of nanomedicine, 2013. 8: p. 1451.
  • [6] Bhardwaj, P. and D. Khanna, Green tea catechins: defensive role in cardiovascular disorders. Chinese journal of natural medicines, 2013. 11(4): p. 345-353.
  • [7] Zanwar, A., et al., Antioxidant role of catechin in health and disease. Polyphenols in Human Health and Disease; Watson, RR, Preedy, VR, Eds, 2014: p. 267-271.
  • [8] Velayutham, P., A. Babu, and D. Liu, Green tea catechins and cardiovascular health: an update. Current medicinal chemistry, 2008. 15(18): p. 1840.
  • [9] Dufresne, C.J. and E.R. Farnworth, A review of latest research findings on the health promotion properties of tea. The Journal of nutritional biochemistry, 2001. 12(7): p. 404-421.
  • [10] How, C.W., et al., Effects of a synthetic antitumoral catechin and its tyrosinase-processed product on the structural properties of phosphatidylcholine membranes. Biochimica et Biophysica Acta (BBA)-Biomembranes, 2014. 1838(5): p. 1215-1224.
  • [11] Cabrera, C., R. Artacho, and R. Giménez, Beneficial effects of green tea—a review. Journal of the American College of Nutrition, 2006. 25(2): p. 79-99.
  • [12] Crespy, V. and G. Williamson, A review of the health effects of green tea catechins in in vivo animal models. The Journal of nutrition, 2004. 134(12): p. 3431S-3440S.
  • [13] Cabrera, C., R. Giménez, and M.C. López, Determination of tea components with antioxidant activity. Journal of agricultural and food chemistry, 2003. 51(15): p. 4427-4435.
  • [14] Horžić, D., et al., The composition of polyphenols and methylxanthines in teas and herbal infusions. Food chemistry, 2009. 115(2): p. 441-448.
  • [15] Goodin, M., et al., Estrogen receptor-mediated actions of polyphenolic catechins in vivo and in vitro. Toxicological Sciences, 2002. 69(2): p. 354-361.
  • [16] Zaveri, N.T., Green tea and its polyphenolic catechins: medicinal uses in cancer and noncancer applications. Life sciences, 2006. 78(18): p. 2073-2080.
  • [17] Chiu, F.-L. and J.-K. Lin, HPLC analysis of naturally occurring methylated catechins, 3''-and 4''-methyl-epigallocatechin gallate, in various fresh tea leaves and commercial teas and their potent inhibitory effects on inducible nitric oxide synthase in macrophages. Journal of agricultural and food chemistry, 2005. 53(18): p. 7035-7042.
  • [18] Chou, C.W., et al., (−)‐Epigallocatechin gallate, the most active polyphenolic catechin in green tea, presynaptically facilitates Ca2+‐dependent glutamate release via activation of protein kinase C in rat cerebral cortex. Synapse, 2007. 61(11): p. 889-902.
  • [19] Mukhtar, H., S.K. Katiyar, and R. Agarwal, Green tea and skin--anticarcinogenic effects. Journal of investigative dermatology, 1994. 102(1): p. 3-7.
  • [20] Mukhtar, H., et al., Tea components: antimutagenic and anticarcinogenic effects. Preventive Medicine, 1992. 21(3): p. 351-360.
  • [21] Higdon, J.V. and B. Frei, Tea catechins and polyphenols: health effects, metabolism, and antioxidant functions. 2003.
  • [22] Matsuzaki, T. and Y. Hara, Antioxidative activity of tea leaf catechins. Journal of the Agricultural Chemical Society of Japan (Japan), 1985.
  • [23] Ruch, R.J., S.-j. Cheng, and J.E. Klaunig, Prevention of cytotoxicity and inhibition of intercellular communication by antioxidant catechins isolated from Chinese green tea. Carcinogenesis, 1989. 10(6): p. 1003-1008.
  • [24] Cao, J., et al., Chemopreventive effects of green and black tea on pulmonary and hepatic carcinogenesis. Toxicological Sciences, 1996. 29(2): p. 244-250.
  • [25] El-Beshbishy, H.A., Hepatoprotective effect of green tea (Camellia sinensis) extract against tamoxifen-induced liver injury in rats. BMB Reports, 2005. 38(5): p. 563-570.
  • [26] Raj, P.V., et al., Protective role of catechin on d-galactosamine induced hepatotoxicity through a p53 dependent pathway. Indian Journal of Clinical Biochemistry, 2010. 25(4): p. 349-356.
  • [27] Yang, B., et al., Relationship of electrochemical oxidation of catechins on their antioxidant activity in microsomal lipid peroxidation. Chemical and pharmaceutical bulletin, 2001. 49(6): p. 747-751.
  • [28] Nanjo, F., et al., Scavenging effects of tea catechins and their derivatives on 1, 1-diphenyl-2-picrylhydrazyl radical. Free Radical Biology and Medicine, 1996. 21(6): p. 895-902.
  • [29] Nash, K.M. and S. Ahmed, Nanomedicine in the ROS-mediated pathophysiology: Applications and clinical advances. Nanomedicine: Nanotechnology, Biology and Medicine, 2015. 11(8): p. 2033-2040.
  • [30] Embuscado, M.E., Spices and herbs: natural sources of antioxidants–a mini review. Journal of Functional Foods, 2015. 18: p. 811-819.
  • [31] Frei, B. and J.V. Higdon, Antioxidant activity of tea polyphenols in vivo: evidence from animal studies. The Journal of nutrition, 2003. 133(10): p. 3275S-3284S.
  • [32] Fang, J.-Y., et al., Effect of liposome encapsulation of tea catechins on their accumulation in basal cell carcinomas. Journal of dermatological science, 2006. 42(2): p. 101-109.
  • [33] Paquay, J.B., et al., Protection against nitric oxide toxicity by tea. Journal of Agricultural and Food Chemistry, 2000. 48(11): p. 5768-5772.
  • [34] Haenen, G. and A. Bast, Nitric oxide radical scavenging of flavonoids. Methods in enzymology, 1999. 301: p. 490.
  • [35] Haenen, G.R., et al., Peroxynitrite scavenging by flavonoids. Biochemical and biophysical research communications, 1997. 236(3): p. 591-593.
  • [36] Agarwal, R., et al., Protection against ultraviolet B radiation‐induced effects in the skin of SKH‐1 hairless mice by a polyphenolic fraction isolated from green tea. Photochemistry and photobiology, 1993. 58(5): p. 695-700.
  • [37] Park, H.J., et al., Green tea extract attenuates hepatic steatosis by decreasing adipose lipogenesis and enhancing hepatic antioxidant defenses in ob/ob mice. The Journal of nutritional biochemistry, 2011. 22(4): p. 393-400.
  • [38] Bruno, R., J.A. Bomser, and M.G. Ferruzzi. Antioxidant Capacity of Green Tea (Camellia sinensis). in Elsevier Inc. 2014.
  • [39] Morris, J.D., et al., Selenium-or quercetin-induced retardation of DNA synthesis in primary prostate cells occurs in the presence of a concomitant reduction in androgen-receptor activity. Cancer letters, 2006. 239(1): p. 111-122.
  • [40] Renold, W., et al., An investigation of the tea aroma. Part I. New volatile black tea constituents. Helvetica Chimica Acta, 1974. 57(5): p. 1301-1308.
  • [41] Chaudhuri, T., et al., Phytochemical investigation of the roots of Camellia sinensis L.(O. Kuntze). Journal of the Indian Chemical Society, 1997. 74(2).
  • [42] Sabu, M., K. Smitha, and R. Kuttan, Anti-diabetic activity of green tea polyphenols and their role in reducing oxidative stress in experimental diabetes. Journal of ethnopharmacology, 2002. 83(1): p. 109-116.
  • [43] Riso, P., et al., Effect of green tea extract on DNA repair and oxidative damage due to H 2 O 2 in Jurkat T cells. Nutrition Research, 2002. 22(10): p. 1143-1150.
  • [44] Geetha, T., et al., Delineation of antimutagenic activity of catechin, epicatechin and green tea extract. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis, 2004. 556(1): p. 65- 74.
  • [45] Finger, A., U.H. Engelhardt, and V. Wray, Flavonol triglycosides containing galactose in tea. Phytochemistry, 1991. 30(6): p. 2057-2060.
  • [46] Mikaberidze, K. and I. Moniava, Umbelliferone from tea leaves. Chemistry of Natural Compounds, 1974. 10(1): p. 81-81.
  • [47] Sandhir, R., et al., Nano-antioxidants: An emerging strategy for intervention against neurodegenerative conditions. Neurochemistry international, 2015. 89: p. 209-226.
  • [48] Aditya, N., et al., Curcumin and catechin co-loaded water-in-oil-in-water emulsion and its beverage application. Journal of Functional Foods, 2015. 15: p. 35-43.
  • [49] Tolochko, N., History of nanotechnology. Nanoscience and nanotechnologies. Encyclopaedia of Life Support Systems (EOLSS), Developed under the auspices of the UNESCO, Eolss Publishers, Oxford. http:// www. eolss. net, 2009.
  • [50] Dangi, R. and S. Shakya, Preparation, optimization and characterization of PLGA nanoparticle. International Journal of Pharmacy & Life Sciences, 2013. 4(7).
  • [51] Li, Z., et al., A review: Using nanoparticles to enhance absorption and bioavailability of phenolic phytochemicals. Food Hydrocolloids, 2015. 43: p. 153-164.
  • [52] Gao, S. and M. Hu, Bioavailability challenges associated with development of anti-cancer phenolics. Mini reviews in medicinal chemistry, 2010. 10(6): p. 550.
  • [53] Gentile, P., et al., An overview of poly (lactic-co-glycolic) acid (PLGA)-based biomaterials for bone tissue engineering. International journal of molecular sciences, 2014. 15(3): p. 3640-3659.
  • [54] McCarron, P.A., R.F. Donnelly, and W. Marouf, Celecoxib-loaded poly (D, L-lactide-co-glycolide) nanoparticles prepared using a novel and controllable combination of diffusion and emulsification steps as part of the salting-out procedure. Journal of microencapsulation, 2006. 23(5): p. 480-498.
  • [55] Reis, C.P., et al., Nanoencapsulation I. Methods for preparation of drug-loaded polymeric nanoparticles. Nanomedicine: Nanotechnology, Biology and Medicine, 2006. 2(1): p. 8-21.
  • [56] Rao, J.P. and K.E. Geckeler, Polymer nanoparticles: preparation techniques and size-control parameters. Progress in Polymer Science, 2011. 36(7): p. 887-913.
  • [57] Nagavarma, B., et al., Different techniques for preparation of polymeric nanoparticles—a review. Asian J. Pharm. Clin. Res, 2012. 5(3): p. 16-23.
  • [58] Nordström, P., Formation of polymeric nanoparticles encapsulating and releasing a new hydrophobic cancer drug. 2011.
  • [59] Bennet, D. and S. Kim, Polymer Nanoparticles for Smart Drug Delivery. 2014.
  • [60] Ramazani, F., et al., Strategies for encapsulation of small hydrophilic and amphiphilic drugs in PLGA microspheres: State-of-the-art and challenges. International Journal of Pharmaceutics, 2016. 499(1): p. 358-367.
  • [61] Kulhari, H., et al., Peptide conjugated polymeric nanoparticles as a carrier for targeted delivery of docetaxel. Colloids and Surfaces B: Biointerfaces, 2014. 117: p. 166-173.
  • [62] Mohanraj, V. and Y. Chen, Nanoparticles-a review. Tropical Journal of Pharmaceutical Research, 2007. 5(1): p. 561-573.
  • [63] Mali, A.D. and R.S. Bathe, UPDATED REVIEW ON NANOPARTICLES AS DRUG DELIVERY SYSTEMS.
  • [64] Hirsjärvi, S., Preparation and characterization of Poly (lactic acid) nanoparticles for pharmaceutical use. 2008.
  • [65] Song, X., et al., Dual agents loaded PLGA nanoparticles: systematic study of particle size and drug entrapment efficiency. European journal of pharmaceutics and biopharmaceutics, 2008. 69(2): p. 445- 453.
  • [66] Koppel, D.E., Analysis of macromolecular polydispersity in intensity correlation spectroscopy: the method of cumulants. The Journal of Chemical Physics, 1972. 57(11): p. 4814-4820.
  • [67] Bououdina, M., Handbook of Research on Nanoscience, Nanotechnology, and Advanced Materials. 2014: IGI Global.
  • [68] Varma, J.R., et al., Formulation and characterization of pyrazinamide polymeric nanoparticles for pulmonary tuberculosis: Efficiency for alveolar macrophage targeting. Indian journal of pharmaceutical sciences, 2015. 77(3): p. 258.
  • [69] Xu, R., C. Wu, and H. Xu, Particle size and zeta potential of carbon black in liquid media. Carbon, 2007. 45(14): p. 2806-2809.
  • [70] He, C., et al., Effects of particle size and surface charge on cellular uptake and biodistribution of polymeric nanoparticles. Biomaterials, 2010. 31(13): p. 3657-3666.
  • [71] Delgado, Á.V., et al., Measurement and interpretation of electrokinetic phenomena. Journal of colloid and interface science, 2007. 309(2): p. 194-224.
  • [72] Xu, R. and G. Smart, Electrophoretic Mobility Study of Dodecyltrimethylammonium Bromide in Aqueous Solution and Adsorption on Microspheres1. Langmuir, 1996. 12(17): p. 4125-4133.
  • [73] Ware, B.R., & Haas, D. D., Electrophoretic light scattering, in Fast methods in physical biochemistry and cell biology, I.R. I. and S.a.S.M. Fernandez, Editors. 1983, Elsevier: New York. p. 173–220.
  • [74] Xu, R., Progress in nanoparticles characterization: Sizing and zeta potential measurement. Particuology, 2008. 6(2): p. 112-115.
  • [75] Javidparvar, A., B. Ramezanzadeh, and E. Ghasemi, Effects of surface morphology and treatment of iron oxide nanoparticles on the mechanical properties of an epoxy coating. Progress in Organic Coatings, 2016. 90: p. 10-20.
  • [76] Chan, H.-K., What is the role of particle morphology in pharmaceutical powder aerosols? Expert opinion on drug delivery, 2008. 5(8): p. 909-914.
  • [77] Todokoro, H. and M. Ezumi, Scanning electron microscope. 1999, Google Patents.
  • [78] Yue, P.-F., et al., The study on the entrapment efficiency and in vitro release of puerarin submicron emulsion. Aaps Pharmscitech, 2009. 10(2): p. 376-383.
  • [79] Bader, R.A. and D.A. Putnam, Engineering polymer systems for improved drug delivery. 2014: John Wiley & Sons.
  • [80] Fonseca, C., S. Simoes, and R. Gaspar, Paclitaxel-loaded PLGA nanoparticles: preparation, physicochemical characterization and in vitro anti-tumoral activity. Journal of Controlled Release, 2002. 83(2): p. 273-286.
  • [81] Peltonen, L., et al., Improved entrapment efficiency of hydrophilic drug substance during nanoprecipitation of poly (I) lactide nanoparticles. Aaps Pharmscitech, 2004. 5(1): p. 115-120.
  • [82] Zhang, Z. and S.-S. Feng, The drug encapsulation efficiency, in vitro drug release, cellular uptake and cytotoxicity of paclitaxel-loaded poly (lactide)–tocopheryl polyethylene glycol succinate nanoparticles. Biomaterials, 2006. 27(21): p. 4025-4033.
  • [83] Mudshinge, S.R., et al., Nanoparticles: emerging carriers for drug delivery. Saudi pharmaceutical journal, 2011. 19(3): p. 129-141.
  • [84] Magenheim, B., M. Levy, and S. Benita, A new in vitro technique for the evaluation of drug release profile from colloidal carriers-ultrafiltration technique at low pressure. International journal of pharmaceutics, 1993. 94(1): p. 115-123.
  • [85] Fang, Z. and B. Bhandari, Encapsulation of polyphenols–a review. Trends in Food Science & Technology, 2010. 21(10): p. 510-523.
  • [86] Liu, J., et al., A modified double-emulsion method for the preparation of daunorubicin-loaded polymeric nanoparticle with enhanced in vitro anti-tumor activity. Biomedical Materials, 2010. 5(6): p. 065002.
  • [87] Tice, T.R. and R.M. Gilley, Preparation of injectable controlled-release microcapsules by a solvent-evaporation process. Journal of Controlled Release, 1985. 2: p. 343-352.
  • [88] Anand, P., et al., RETRACTED: Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo. Biochemical pharmacology, 2010. 79(3): p. 330-338.
  • [89] Lambert, G., E. Fattal, and P. Couvreur, Nanoparticulate systems for the delivery of antisense oligonucleotides. Advanced drug delivery reviews, 2001. 47(1): p. 99-112.
  • [90] Quintanar-Guerrero, D., et al., Preparation techniques and mechanisms of formation of biodegradable nanoparticles from preformed polymers. Drug development and industrial pharmacy, 1998. 24(12): p. 1113-1128.
  • [91] Jacobson, G.B., et al., Biodegradable nanoparticles with sustained release of functional siRNA in skin. Journal of pharmaceutical sciences, 2010. 99(10): p. 4261-4266.