The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications
Abstract
:1. Introduction
2. Stabilizers for Phytochemical-Based NPs Preparation and Their Effects
2.1. CS and Its Derivatives
2.1.1. Stabilizing Mechanisms
2.1.2. Applications
2.2. CN
2.2.1. Stabilizing Mechanisms
2.2.2. Applications
2.3. Saponins
2.3.1. Stabilizing Mechanisms
2.3.2. Applications
2.4. Cyclodextrin
2.4.1. Stabilizing Mechanisms
2.4.2. Applications
2.5. PEGs
2.5.1. Stabilizing Mechanisms
2.5.2. Applications
2.6. Other Stabilizers for NPs
2.6.1. Lentinan: Stabilizing Mechanisms and Applications
2.6.2. Glycosylated Lactoferrin: Stabilizing Mechanisms and Applications
2.6.3. Trehalose: Stabilizing Mechanisms and Applications
2.6.4. Didodecyldimethyl Ammonium Bromide and Polyvinyl Alcohol: Stabilizing Mechanisms and Applications
3. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Mohanty, C.; Das, M.; Sahoo, S.K. Emerging role of nanocarriers to increase the solubility and bioavailability of curcumin. Expert Opin. Drug Deliv. 2012, 9, 1347–1364. [Google Scholar] [CrossRef] [PubMed]
- Wang, W.; Sun, C.; Mao, L.; Ma, P.; Liu, F.; Yang, J.; Gao, Y. The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci. Technol. 2016, 56, 21–38. [Google Scholar] [CrossRef]
- Patel, R.V.; Mistry, B.M.; Shinde, S.K.; Syed, R.; Singh, V.; Shin, H.-S. Therapeutic potential of quercetin as a cardiovascular agent. Eur. J. Med. Chem. 2018, 155, 889–904. [Google Scholar] [CrossRef] [PubMed]
- Khonkarn, R.; Mankhetkorn, S.; Hennink, W.E.; Okonogi, S. PEG-OCL micelles for quercetin solubilization and inhibition of cancer cell growth. Eur. J. Pharm. Biopharm. 2011, 79, 268–275. [Google Scholar] [CrossRef]
- Ndiaye, M.; Kumar, R.; Ahmad, N. Resveratrol in cancer management: Where are we and where we go from here? Ann. N. Y. Acad. Sci. 2011, 1215, 144–149. [Google Scholar] [CrossRef]
- Li, X.; He, L.; Li, N.; He, D. Curcumin loaded hydrogel with anti-inflammatory activity to promote cartilage regeneration in immunocompetent animals. J. Biomater. Sci. Polym. Ed. 2023, 34, 200–216. [Google Scholar] [CrossRef]
- Khan, I.; Saeed, K.; Khan, I. Nanoparticles: Properties, applications and toxicities. Arab. J. Chem. 2019, 12, 908–931. [Google Scholar] [CrossRef]
- Kansom, T.; Sajomsang, W.; Saeeng, R.; Rojanarata, T.; Ngawhirunpat, T.; Patrojanasophon, P.; Opanasopit, P. Fabrication and characterization of andrographolide analogue (3A.1) nanosuspensions stabilized by amphiphilic chitosan derivatives for colorectal cancer therapy. J. Drug Deliv. Sci. Technol. 2019, 54, 101287. [Google Scholar] [CrossRef]
- Li, J.; Yang, M.; Xu, W.R. Enhanced oral bioavailability of fluvastatin by using nanosuspensions containing cyclodextrin. Drug Des. Dev. Ther. 2018, 12, 3491–3499. [Google Scholar]
- Hossin, A.Y.; Inafuku, M.; Oku, H. Dihydropyranocoumarins exerted anti-obesity activity in vivo and its activity was enhanced by nanoparticulation with polylactic-co-glycolic acid. Nutrients 2019, 11, 3053. [Google Scholar] [CrossRef]
- Gao, L.; Liu, G.; Ma, J.; Wang, X.; Zhou, L.; Li, X.; Wang, F. Application of drug nanocrystal technologies on oral drug delivery of poorly soluble drugs. Pharm. Res. 2013, 30, 307–324. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, I.; Bose, S.; Vippagunta, R.; Harmon, F. Nanosuspension for improving the bioavailability of a poorly soluble drug and screening of stabilizing agents to inhibit crystal growth. Int. J. Pharm. 2011, 409, 260–268. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Zheng, Y.; Zhang, L.; Wang, Q.; Zhang, D. Stability of nanosuspensions in drug delivery. J. Control. Release 2013, 172, 1126–1141. [Google Scholar] [CrossRef]
- Wu, L.; Zhang, J.; Watanabe, W. Physical and chemical stability of drug nanoparticles. Adv. Drug Deliv. Rev. 2011, 63, 456–469. [Google Scholar] [CrossRef] [PubMed]
- Yue, P.F.; Li, Y.; Wan, J.; Yang, M.; Zhu, W.F.; Wang, C.H. Study on formability of solid nanosuspensions during nanodispersion and solidification: I. Novel role of stabilizer/drug property. Int. J. Pharm. 2013, 454, 269–277. [Google Scholar] [CrossRef] [PubMed]
- Fang, C.; Bhattarai, N.; Sun, C.; Zhang, M. Functionalized nanoparticles with long-term stability in biological media. Small 2009, 5, 1637–1641. [Google Scholar] [CrossRef]
- Qiao, H.; Chen, L.; Rui, T.; Wang, J.; Chen, T.; Fu, T.; Li, J.; Di, L. Fabrication and in vitro/in vivo evaluation of amorphous andrographolide nanosuspensions stabilized by d-alpha-tocopheryl polyethylene glycol 1000 succinate/sodium lauryl sulfate. Int. J. Nanomed. 2017, 12, 1033–1046. [Google Scholar] [CrossRef]
- Lu, Y.; Li, Y.; Wu, W. Injected nanocrystals for targeted drug delivery. Acta Pharm. Sin. B 2016, 6, 106–113. [Google Scholar] [CrossRef]
- Gao, L.; Zhang, D.; Chen, M. Drug nanocrystals for the formulation of poorly soluble drugs and its application as a potential drug delivery system. J. Nanoparticle Res. 2008, 10, 845–862. [Google Scholar] [CrossRef]
- Pitkowski, A.; Durand, D.; Nicolai, T. Structure and dynamical mechanical properties of suspensions of sodium caseinate. J. Colloid Interface Sci. 2008, 326, 96–102. [Google Scholar] [CrossRef]
- Zhang, H.; Sun, X.; Wang, J.; Dong, M.; Li, L.; Bai, F.; Xu, K.; Wang, L. Sodium caseinate decorating on shellac nanoparticles as a stabilizer for the encapsulation of quercetin. Food Chem. 2022, 395, 133580. [Google Scholar] [CrossRef]
- Muller, R.H.; Keck, C.M. Challenges and solutions for the delivery of biotech drugs—A review of drug nanocrystal technology and lipid nanoparticles. J. Biotechnol. 2004, 113, 151–170. [Google Scholar] [CrossRef]
- Heydenreich, A. Preparation and purification of cationic solid lipid nanospheres—Effects on particle size, physical stability and cell toxicity. Int. J. Pharm. 2003, 254, 83–87. [Google Scholar] [CrossRef] [PubMed]
- Phan, T.T.V.; Hoang, G.; Nguyen, V.T.; Nguyen, T.P.; Kim, H.H.; Mondal, S.; Manivasagan, P.; Moorthy, M.S.; Lee, K.D.; Junghwan, O. Chitosan as a stabilizer and size-control agent for synthesis of porous flower-shaped palladium nanoparticles and their applications on photo-based therapies. Carbohydr. Polym. 2019, 205, 340–352. [Google Scholar] [CrossRef] [PubMed]
- Atamer, Z.; Post, A.E.; Schubert, T.; Holder, A.; Boom, R.M.; Hinrichs, J. Bovine β-casein: Isolation, properties and functionality. A review. Int. Dairy J. 2017, 66, 115–125. [Google Scholar] [CrossRef]
- Long, J.; Song, J.; Zhang, X.; Deng, M.; Xie, L.; Zhang, L.; Li, X. Tea saponins as natural stabilizers for the production of hesperidin nanosuspensions. Int. J. Pharm. 2020, 583, 119406. [Google Scholar] [CrossRef]
- Ravindran, J.; Nair, H.B.; Sung, B.; Prasad, S.; Tekmal, R.R.; Aggarwal, B.B. Thymoquinone poly (lactide-co-glycolide) nanoparticles exhibit enhanced anti-proliferative, anti-inflammatory, and chemosensitization potential. Biochem. Pharmacol. 2010, 79, 1640–1647. [Google Scholar] [CrossRef]
- Suo, Z.; Sun, Q.; Peng, X.; Zhang, S.; Gan, N.; Zhao, L.; Yuan, N.; Zhang, Y.; Li, H. Lentinan as a natural stabilizer with bioactivities for preparation of drug-drug nanosuspensions. Int. J. Biol. Macromol. 2021, 184, 101–108. [Google Scholar] [CrossRef]
- Chen, Y.; Gao, X.; Liu, S.; Cai, Q.; Wu, L.; Sun, Y.; Xia, G.; Wang, Y. Establishment and Characterization of Stable Zein/Glycosylated Lactoferrin Nanoparticles to Enhance the Storage Stability and in vitro Bioaccessibility of 7,8-Dihydroxyflavone. Front. Nutr. 2021, 8, 806623. [Google Scholar] [CrossRef]
- Almalik, A.; Alradwan, I.; Kalam, M.A.; Alshamsan, A. Effect of cryoprotection on particle size stability and preservation of chitosan nanoparticles with and without hyaluronate or alginate coating. Saudi Pharm. J. 2017, 25, 861–867. [Google Scholar] [CrossRef]
- Sonaje, K.; Italia, J.; Sharma, G.; Bhardwaj, V.; Tikoo, K.; Kumar, M.R. Development of biodegradable nanoparticles for oral delivery of ellagic acid and evaluation of their antioxidant efficacy against cyclosporine A-induced nephrotoxicity in rats. Pharm. Res. 2007, 24, 899–908. [Google Scholar] [CrossRef] [PubMed]
- Benhabiles, M.S.; Tazdait, D.; Abdi, N.; Lounici, H.; Drouiche, N.; Goosen, M.F.A.; Mameri, N. Assessment of coating tomato fruit with shrimp shell chitosan and N,O-carboxymethyl chitosan on postharvest preservation. J. Food Meas. Charact. 2013, 7, 66–74. [Google Scholar] [CrossRef]
- Rashmi, S.H.; Biradar, B.; Maladkar, K.; Kittur, A.A. Extraction of chitin from prawn shell and preparation of chitosan. Res. J. Chem. Environ. Sci. 2016, 4, 70–73. [Google Scholar]
- Smith, A.; Perelman, M.; Hinchcliffe, M. Chitosan a promising safe and immune-enhancing adjuvant for intranasal vaccines. Hum. Vaccines Immunother. 2014, 10, 797–807. [Google Scholar] [CrossRef] [PubMed]
- Collado-Gonzalez, M.; Montalban, M.G.; Pena-Garcia, J.; Perez-Sanchez, H.; Villora, G.; Diaz Banos, F.G. Chitosan as stabilizing agent for negatively charged nanoparticles. Carbohydr. Polym. 2017, 161, 63–70. [Google Scholar] [CrossRef]
- Raftery, R.M.; Tierney, E.G.; Curtin, C.M.; Cryan, S.A.; O’Brien, F.J. Development of a gene-activated scaffold platform for tissue engineering applications using chitosan-pDNA nanoparticles on collagen-based scaffolds. J. Control. Release 2015, 210, 84–94. [Google Scholar] [CrossRef]
- Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B.A.; Thiessen, P.A.; Yu, B.; et al. PubChem 2023 update. Nucleic Acids Res. 2023, 51, D1373–D1380. [Google Scholar] [CrossRef]
- Gadkari, R.R.; Suwalka, S.; Yogi, M.R.; Ali, W.; Das, A.; Alagirusamy, R. Green synthesis of chitosan-cinnamaldehyde cross-linked nanoparticles: Characterization and antibacterial activity. Carbohydr. Polym. 2019, 226, 115298. [Google Scholar] [CrossRef]
- Yusof, N.A.A.; Zain, N.M.; Pauzi, N. Synthesis of ZnO nanoparticles with chitosan as stabilizing agent and their antibacterial properties against Gram-positive and Gram-negative bacteria. Int. J. Biol. Macromol. 2019, 124, 1132–1136. [Google Scholar] [CrossRef]
- Mohamed, A.A.R.; Khater, S.I.; Hamed Arisha, A.; Metwally, M.M.M.; Mostafa-Hedeab, G.; El-Shetry, E.S. Chitosan-stabilized selenium nanoparticles alleviate cardio-hepatic damage in type 2 diabetes mellitus model via regulation of caspase, Bax/Bcl-2, and Fas/FasL-pathway. Gene 2021, 768, 145288. [Google Scholar] [CrossRef]
- Kim, D.G.; Jeong, Y.I.; Choi, C.; Roh, S.H.; Kang, S.K.; Jang, M.K.; Nah, J.W. Retinol-encapsulated low molecular water-soluble chitosan nanoparticles. Int. J. Pharm. 2006, 319, 130–138. [Google Scholar] [CrossRef] [PubMed]
- Sogias, I.A.; Khutoryanskiy, V.V.; Williams, A.C. Exploring the factors affecting the solubility of chitosan in water. Macromol. Chem. Phys. 2010, 211, 426–433. [Google Scholar] [CrossRef]
- Phan, T.T.V.; Moorthy, M.S.; Kang, H.W.; Nam, S.Y.; Lee, Y.W.; Oh, J. Coating Chitosan Thin Shells: A Facile Technique to Improve Dispersion Stability of Magnetoliposomes. J. Nanosci. Nanotechnol. 2018, 18, 583–590. [Google Scholar] [CrossRef] [PubMed]
- Shao, Z.; Vollrath, F. Surprising strength of silkworm silk. Nature 2002, 418, 741. [Google Scholar] [CrossRef]
- Omenetto, F.G.; Kaplan, D.L. New opportunities for an ancient material. Science 2010, 329, 528–531. [Google Scholar] [CrossRef]
- Bonferoni, M.C.; Sandri, G.; Rossi, S.; Usai, D.; Liakos, I.; Garzoni, A.; Fiamma, M.; Zanetti, S.; Athanassiou, A.; Caramella, C.; et al. A novel ionic amphiphilic chitosan derivative as a stabilizer of nanoemulsions: Improvement of antimicrobial activity of Cymbopogon citratus essential oil. Colloids Surf. B Biointerfaces 2017, 152, 385–392. [Google Scholar] [CrossRef]
- Dai, Y.; Chen, S.R.; Chai, L.; Zhao, J.; Wang, Y.; Wang, Y. Overview of pharmacological activities of Andrographis paniculata and its major compound andrographolide. Crit. Rev. Food Sci. Nutr. 2019, 59, S17–S29. [Google Scholar] [CrossRef]
- Woraphatphadung, T.; Sajomsang, W.; Gonil, P.; Saesoo, S.; Opanasopit, P. Synthesis and characterization of pH-responsive N-naphthyl-N,O-succinyl chitosan micelles for oral meloxicam delivery. Carbohydr. Polym. 2015, 121, 99–106. [Google Scholar] [CrossRef]
- Sanna, V.; Roggio, A.M.; Siliani, S.; Piccinini, M.; Marceddu, S.; Mariani, A.; Sechi, M. Development of novel cationic chitosan-and anionic alginate-coated poly(D,L-lactide-co-glycolide) nanoparticles for controlled release and light protection of resveratrol. Int. J. Nanomed. 2012, 7, 5501–5516. [Google Scholar]
- Guo, C.; Gemeinhart, R.A. Understanding the adsorption mechanism of chitosan onto poly(lactide-co-glycolide) particles. Eur. J. Pharm. Biopharm. 2008, 70, 597–604. [Google Scholar] [CrossRef]
- Chamcheu, J.C.; Siddiqui, I.A.; Adhami, V.M.; Esnault, S.; Bharali, D.J.; Babatunde, A.S.; Adame, S.; Massey, R.J.; Wood, G.S.; Longley, B.J.; et al. Chitosan-based nanoformulated (-)-epigallocatechin-3-gallate (EGCG) modulates human keratinocyte-induced responses and alleviates imiquimod-induced murine psoriasiform dermatitis. Int. J. Nanomed. 2018, 13, 4189–4206. [Google Scholar] [CrossRef] [PubMed]
- Chethan, P.D.; Vishalakshi, B.; Sathish, L.; Ananda, K.; Poojary, B. Preparation of substituted quaternized arylfuran chitosan derivatives and their antimicrobial activity. Int. J. Biol. Macromol. 2013, 59, 158–164. [Google Scholar] [CrossRef] [PubMed]
- Hassan, M.A.; Tamer, T.M.; Omer, A.M.; Baset, W.M.A.; Abbas, E.; Mohy-Eldin, M.S. Therapeutic potential of two formulated novel chitosan derivatives with prominent antimicrobial activities against virulent microorganisms and safe profiles toward fibroblast cells. Int. J. Pharm. 2023, 634, 122649. [Google Scholar] [CrossRef] [PubMed]
- Hu, Q.; Zhou, F.; Ly, N.K.; Ordyna, J.; Peterson, T.; Fan, Z.; Wang, S. Development of Multifunctional Nanoencapsulated trans-Resveratrol/Chitosan Nutraceutical Edible Coating for Strawberry Preservation. ACS Nano 2023, 17, 8586–8597. [Google Scholar] [CrossRef] [PubMed]
- Sondi, I.; Salopek-Sondi, B. Silver nanoparticles as antimicrobial agent: A case study on E. coli as a model for Gram-negative bacteria. J. Colloid Interface Sci. 2004, 275, 177–182. [Google Scholar] [CrossRef]
- Chithrani, D.B. Optimization of bio-nano interface using gold nanostructures as a model nanoparticle system. Insciences J. 2011, 1, 115–135. [Google Scholar] [CrossRef]
- Zhang, J.; Nie, S.; Wang, S. Nanoencapsulation enhances epigallocatechin-3-gallate stability and its antiatherogenic bioactivities in macrophages. J. Agric. Food Chem. 2013, 61, 9200–9209. [Google Scholar] [CrossRef]
- Zu, Y.; Zhao, L.; Hao, L.; Mechref, Y.; Zabet-Moghaddam, M.; Keyel, P.A.; Abbasi, M.; Wu, D.; Dawson, J.A.; Zhang, R.; et al. Browning white adipose tissue using adipose stromal cell-targeted resveratrol-loaded nanoparticles for combating obesity. J. Control. Release 2021, 333, 339–351. [Google Scholar] [CrossRef]
- Zu, Y.; Overby, H.; Ren, G.; Fan, Z.; Zhao, L.; Wang, S. Resveratrol liposomes and lipid nanocarriers: Comparison of characteristics and inducing browning of white adipocytes. Colloids Surf. B Biointerfaces 2018, 164, 414–423. [Google Scholar] [CrossRef]
- Phan, T.T.V.; Phan, D.T.; Cao, X.T.; Huynh, T.C.; Oh, J. Roles of Chitosan in Green Synthesis of Metal Nanoparticles for Biomedical Applications. Nanomaterials 2021, 11, 273. [Google Scholar] [CrossRef]
- de Figueiredo Furtado, G.; Mantovani, R.A.; Consoli, L.; Hubinger, M.D.; da Cunha, R.L. Structural and emulsifying properties of sodium caseinate and lactoferrin influenced by ultrasound process. Food Hydrocoll. 2017, 63, 178–188. [Google Scholar] [CrossRef]
- Chakraborty, A.; Basak, S. Effect of surfactants on casein structure: A spectroscopic study. Colloids Surf. B Biointerfaces 2008, 63, 83–90. [Google Scholar] [CrossRef] [PubMed]
- Liang, Y.; Gillies, G.; Matia-Merino, L.; Ye, A.; Patel, H.; Golding, M. Structure and stability of sodium-caseinate-stabilized oil-in-water emulsions as influenced by heat treatment. Food Hydrocoll. 2017, 66, 307–317. [Google Scholar] [CrossRef]
- Park, E.K.; Kim, S.Y.; Lee, S.B.; Lee, Y.M. Folate-conjugated methoxy poly(ethylene glycol)/poly(epsilon-caprolactone) amphiphilic block copolymeric micelles for tumor-targeted drug delivery. J. Control. Release 2005, 109, 158–168. [Google Scholar] [CrossRef] [PubMed]
- Guo, M.; Fox, P.; Flynn, A.; Kindstedt, P. Heat-induced modifications of the functional properties of sodium caseinate. Int. Dairy J. 1996, 6, 473–483. [Google Scholar] [CrossRef]
- Patel, A.R.; Heussen, P.C.; Hazekamp, J.; Drost, E.; Velikov, K.P. Quercetin loaded biopolymeric colloidal particles prepared by simultaneous precipitation of quercetin with hydrophobic protein in aqueous medium. Food Chem. 2012, 133, 423–429. [Google Scholar] [CrossRef]
- Patel, A.; Hu, Y.; Tiwari, J.K.; Velikov, K.P. Synthesis and characterisation of zein–curcumin colloidal particles. Soft Matter 2010, 6, 6192–6199. [Google Scholar] [CrossRef]
- Corradini, E.; Curti, P.S.; Meniqueti, A.B.; Martins, A.F.; Rubira, A.F.; Muniz, E.C. Recent advances in food-packing, pharmaceutical and biomedical applications of zein and zein-based materials. Int. J. Mol. Sci. 2014, 15, 22438–22470. [Google Scholar] [CrossRef]
- Zhou, J.-F.; Zheng, G.-D.; Wang, W.-J.; Yin, Z.-P.; Chen, J.-G.; Li, J.-E.; Zhang, Q.-F. Physicochemical properties and bioavailability comparison of two quercetin loading zein nanoparticles with outer shell of caseinate and chitosan. Food Hydrocoll. 2021, 120, 106959. [Google Scholar] [CrossRef]
- Li, S.; You, J.; Wang, Z.; Liu, Y.; Wang, B.; Du, M.; Zou, T. Curcumin alleviates high-fat diet-induced hepatic steatosis and obesity in association with modulation of gut microbiota in mice. Food Res. Int. 2021, 143, 110270. [Google Scholar] [CrossRef]
- Guclu-Ustundag, O.; Mazza, G. Saponins: Properties, applications and processing. Crit. Rev. Food Sci. Nutr. 2007, 47, 231–258. [Google Scholar] [CrossRef] [PubMed]
- Kharat, M.; McClements, D.J. Fabrication and characterization of nanostructured lipid carriers (NLC) using a plant-based emulsifier: Quillaja saponin. Food Res. Int. 2019, 126, 108601. [Google Scholar] [CrossRef] [PubMed]
- Golemanov, K.; Tcholakova, S.; Denkov, N.; Pelan, E.; Stoyanov, S.D. The role of the hydrophobic phase in the unique rheological properties of saponin adsorption layers. Soft Matter 2014, 10, 7034–7044. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.R.; Fan, C.L.; Chen, Y.; Quan, J.Y.; Shi, L.Z.; Tian, C.Y.; Shang, X.; Xu, N.S.; Ye, W.C.; Yu, L.Z.; et al. Anti-inflammatory, anti-angiogenetic and antiviral activities of dammarane-type triterpenoid saponins from the roots of Panax notoginseng. Food Funct. 2022, 13, 3590–3602. [Google Scholar] [CrossRef]
- He, F.; Ding, Y.; Liang, C.; Song, S.B.; Dou, D.Q.; Song, G.Y.; Kim, Y.H. Antitumor effects of dammarane-type saponins from steamed Notoginseng. Pharmacogn. Mag. 2014, 10, 314–317. [Google Scholar] [CrossRef]
- Cui, C.; Zong, J.; Sun, Y.; Zhang, L.; Ho, C.T.; Wan, X.; Hou, R. Triterpenoid saponins from the genus Camellia: Structures, biological activities, and molecular simulation for structure-activity relationship. Food Funct. 2018, 9, 3069–3091. [Google Scholar] [CrossRef]
- Yu, X.L.; He, Y. Tea saponins: Effective natural surfactants beneficial for soil remediation, from preparation to application. RSC Adv. 2018, 8, 24312–24321. [Google Scholar] [CrossRef]
- Penfold, J.; Thomas, R.; Tucker, I.; Petkov, J.; Stoyanov, S.; Denkov, N.; Golemanov, K.; Tcholakova, S.; Webster, J. Saponin adsorption at the air–water interface—Neutron reflectivity and surface tension study. Langmuir 2018, 34, 9540–9547. [Google Scholar] [CrossRef]
- Chen, Y.; Liu, Y.; Xu, J.; Xie, Y.; Zheng, Q.; Yue, P.; Yang, M. A Natural Triterpenoid Saponin as Multifunctional Stabilizer for Drug Nanosuspension Powder. AAPS PharmSciTech 2017, 18, 2744–2753. [Google Scholar] [CrossRef]
- Wang, T.; Liu, B.; Zhang, W.; Wilson, B.; Hong, J.S. Andrographolide reduces inflammation-mediated dopaminergic neurodegeneration in mesencephalic neuron-glia cultures by inhibiting microglial activation. J. Pharmacol. Exp. Ther. 2004, 308, 975–983. [Google Scholar] [CrossRef]
- Coon, J.T.; Ernst, E. Andrographis paniculata in the treatment of upper respiratory tract infections: A systematic review of safety and efficacy. Planta Med. 2004, 70, 293–298. [Google Scholar]
- Chou, C.; Pan, S.; Teng, C.; Guh, J. Pharmacological evaluation of several major ingredients of Chinese herbal medicines in human hepatoma Hep3B cells. Eur. J. Pharm. Sci. 2003, 19, 403–412. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Lin, G.; Zuo, Z. Involvement of UDP-glucuronosyltransferases in the extensive liver and intestinal first-pass metabolism of flavonoid baicalein. Pharm. Res. 2007, 24, 81–89. [Google Scholar] [CrossRef] [PubMed]
- Xie, Y.; Ma, Y.; Xu, J.; Liu, Y.; Yue, P.; Zheng, Q.; Hu, P.; Yang, M. Panax Notoginseng Saponins as a Novel Nature Stabilizer for Poorly Soluble Drug Nanocrystals: A Case Study with Baicalein. Molecules 2016, 21, 1149. [Google Scholar] [CrossRef] [PubMed]
- Salminen, H.; Gommel, C.; Leuenberger, B.H.; Weiss, J. Influence of encapsulated functional lipids on crystal structure and chemical stability in solid lipid nanoparticles: Towards bioactive-based design of delivery systems. Food Chem. 2016, 190, 928–937. [Google Scholar] [CrossRef]
- Makhlof, A.; Miyazaki, Y.; Tozuka, Y.; Takeuchi, H. Cyclodextrins as stabilizers for the preparation of drug nanocrystals by the emulsion solvent diffusion method. Int. J. Pharm. 2008, 357, 280–285. [Google Scholar] [CrossRef]
- Davis, M.E.; Brewster, M.E. Cyclodextrin-based pharmaceutics: Past, present and future. Nat. Rev. Drug Discov. 2004, 3, 1023–1035. [Google Scholar] [CrossRef]
- Hassan, A.S.; Soliman, G.M. Rutin Nanocrystals with Enhanced Anti-Inflammatory Activity: Preparation and Ex Vivo/In Vivo Evaluation in an Inflammatory Rat Model. Pharmaceutics 2022, 14, 2727. [Google Scholar] [CrossRef]
- Negahdari, R.; Bohlouli, S.; Sharifi, S.; Maleki Dizaj, S.; Rahbar Saadat, Y.; Khezri, K.; Jafari, S.; Ahmadian, E.; Gorbani Jahandizi, N.; Raeesi, S. Therapeutic benefits of rutin and its nanoformulations. Phytother. Res. 2021, 35, 1719–1738. [Google Scholar] [CrossRef]
- Wang, H.; Xiao, Y.; Wang, H.; Sang, Z.; Han, X.; Ren, S.; Du, R.; Shi, X.; Xie, Y. Development of daidzein nanosuspensions: Preparation, characterization, in vitro evaluation, and pharmacokinetic analysis. Int. J. Pharm. 2019, 566, 67–76. [Google Scholar] [CrossRef]
- Onodera, R.; Hayashi, T.; Motoyama, K.; Tahara, K.; Takeuchi, H. Hydroxypropyl-beta-cyclodextrin Enhances Oral Absorption of Silymarin Nanoparticles Prepared Using PureNano Continuous Crystallizer. Pharmaceutics 2022, 14, 394. [Google Scholar] [CrossRef] [PubMed]
- Das, S.; Roy, P.; Auddy, R.G.; Mukherjee, A. Silymarin nanoparticle prevents paracetamol-induced hepatotoxicity. Int. J. Nanomed. 2011, 6, 1291–1301. [Google Scholar]
- Javed, S.; Kohli, K.; Ali, M. Reassessing bioavailability of silymarin. Altern. Med. Rev. 2011, 16, 239–249. [Google Scholar] [PubMed]
- Zheng, K.; Huang, Z.; Huang, J.; Liu, X.; Ban, J.; Huang, X.; Luo, H.; Chen, Z.; Xie, Q.; Chen, Y.; et al. Effect of a 2-HP-beta-Cyclodextrin Formulation on the Biological Transport and Delivery of Chemotherapeutic PLGA Nanoparticles. Drug Des. Dev. Ther. 2021, 15, 2605–2618. [Google Scholar] [CrossRef]
- Lan, H.; Zhang, W.; Jin, K.; Liu, Y.; Wang, Z. Modulating barriers of tumor microenvironment through nanocarrier systems for improved cancer immunotherapy: A review of current status and future perspective. Drug Deliv. 2020, 27, 1248–1262. [Google Scholar] [CrossRef]
- Ghadi, M.; Hosseinimehr, S.J.; Talebpour Amiri, F.; Mardanshahi, A.; Noaparast, Z. Data on the in vitro and in vivo anti-tumor effects of itraconazole, paclitaxel, and the two in combination in HT-29 and YM-1 cancer cell line and HT-29 colon cancer xenograft models. Data Brief 2021, 35, 106862. [Google Scholar] [CrossRef]
- Chen, X.M.; Chen, Y.; Hou, X.F.; Wu, X.; Gu, B.H.; Liu, Y. Sulfonato-beta-Cyclodextrin Mediated Supramolecular Nanoparticle for Controlled Release of Berberine. ACS Appl. Mater. Interfaces 2018, 10, 24987–24992. [Google Scholar] [CrossRef]
- Shan, W.-J.; Huang, L.; Zhou, Q.; Meng, F.-C.; Li, X.-S. Synthesis, biological evaluation of 9-N-substituted berberine derivatives as multi-functional agents of antioxidant, inhibitors of acetylcholinesterase, butyrylcholinesterase and amyloid-β aggregation. Eur. J. Med. Chem. 2011, 46, 5885–5893. [Google Scholar] [CrossRef]
- Hong, J.; Li, Y.; Xiao, Y.; Li, Y.; Guo, Y.; Kuang, H.; Wang, X. Annonaceous acetogenins (ACGs) nanosuspensions based on a self-assembly stabilizer and the significantly improved anti-tumor efficacy. Colloids Surf. B Biointerfaces 2016, 145, 319–327. [Google Scholar] [CrossRef]
- Oberlies, N.H.; Chang, C.J.; McLaughlin, J.L. Structure-activity relationships of diverse Annonaceous acetogenins against multidrug resistant human mammary adenocarcinoma (MCF-7/Adr) cells. J. Med. Chem. 1997, 40, 2102–2106. [Google Scholar] [CrossRef]
- Tormo, J.R.; DePedro, N.; Royo, I.; Barrachina, I.; Zafra-Polo, M.C.; Cuadrillero, C.; Hernandez, P.; Cortes, D.; Pelaez, F. In vitro antitumor structure-activity relationships of threo/trans/threo/trans/erythro bis-tetrahydrofuranic acetogenins: Correlations with their inhibition of mitochondrial complex I. Oncol. Res. 2005, 15, 129–138. [Google Scholar] [CrossRef]
- Loftsson, T.; Duchene, D. Cyclodextrins and their pharmaceutical applications. Int. J. Pharm. 2007, 329, 1–11. [Google Scholar] [CrossRef] [PubMed]
- Shende, P.; Deshmukh, K.; Trotta, F.; Caldera, F. Novel cyclodextrin nanosponges for delivery of calcium in hyperphosphatemia. Int. J. Pharm. 2013, 456, 95–100. [Google Scholar] [CrossRef] [PubMed]
- Ferreira, M.; Gomes, D.; Neto, M.; Passarinha, L.A.; Costa, D.; Sousa, A. Development and Characterization of Quercetin-Loaded Delivery Systems for Increasing Its Bioavailability in Cervical Cancer Cells. Pharmaceutics 2023, 15, 936. [Google Scholar] [CrossRef] [PubMed]
- D’Souza, A.A.; Shegokar, R. Polyethylene glycol (PEG): A versatile polymer for pharmaceutical applications. Expert Opin. Drug Deliv. 2016, 13, 1257–1275. [Google Scholar] [CrossRef] [PubMed]
- Harris, J.M.; Chess, R.B. Effect of pegylation on pharmaceuticals. Nat. Rev. Drug Discov. 2003, 2, 214–221. [Google Scholar] [CrossRef]
- Hatakeyama, H.; Akita, H.; Harashima, H. A multifunctional envelope type nano device (MEND) for gene delivery to tumours based on the EPR effect: A strategy for overcoming the PEG dilemma. Adv. Drug Deliv. Rev. 2011, 63, 152–160. [Google Scholar] [CrossRef]
- Patravale, V.B.; Date, A.A.; Kulkarni, R.M. Nanosuspensions: A promising drug delivery strategy. J. Pharm. Pharmacol. 2004, 56, 827–840. [Google Scholar] [CrossRef]
- Junghanns, J.U.; Muller, R.H. Nanocrystal technology, drug delivery and clinical applications. Int. J. Nanomed. 2008, 3, 295–309. [Google Scholar]
- Wu, J.; Zhao, C.; Lin, W.; Hu, R.; Wang, Q.; Chen, H.; Li, L.; Chen, S.; Zheng, J. Binding characteristics between polyethylene glycol (PEG) and proteins in aqueous solution. J. Mater. Chem. B 2014, 2, 2983–2992. [Google Scholar] [CrossRef]
- Shi, L.; Zhang, J.; Zhao, M.; Tang, S.; Cheng, X.; Zhang, W.; Li, W.; Liu, X.; Peng, H.; Wang, Q. Effects of polyethylene glycol on the surface of nanoparticles for targeted drug delivery. Nanoscale 2021, 13, 10748–10764. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Cano, C.; Carril, M. Recent Developments in the Design of Non-Biofouling Coatings for Nanoparticles and Surfaces. Int. J. Mol. Sci. 2020, 21, 1007. [Google Scholar] [CrossRef]
- Nogueira, C.d.C.; de Araújo Padilha, C.E.; de Souza Filho, P.F.; dos Santos, E.S. Effects of the addition of poly (ethylene glycol) and non-ionic surfactants on pretreatment, enzymatic hydrolysis, and ethanol fermentation. BioEnergy Res. 2022, 15, 889–904. [Google Scholar] [CrossRef]
- Amin, F.U.; Shah, S.A.; Badshah, H.; Khan, M.; Kim, M.O. Anthocyanins encapsulated by PLGA@PEG nanoparticles potentially improved its free radical scavenging capabilities via p38/JNK pathway against Abeta(1-42)-induced oxidative stress. J. Nanobiotechnology 2017, 15, 12. [Google Scholar] [CrossRef] [PubMed]
- Badary, O.A.; Taha, R.A.; Gamal El-Din, A.M.; Abdel-Wahab, M.H. Thymoquinone is a potent superoxide anion scavenger. Drug Chem. Toxicol. 2003, 26, 87–98. [Google Scholar] [CrossRef]
- Al-Ali, A.; Alkhawajah, A.A.; Randhawa, M.A.; Shaikh, N.A. Oral and intraperitoneal LD50 of thymoquinone, an active principle of Nigella sativa, in mice and rats. J. Ayub Med. Coll. Abbottabad 2008, 20, 25–27. [Google Scholar]
- Anand, P.; Nair, H.B.; Sung, B.; Kunnumakkara, A.B.; Yadav, V.R.; Tekmal, R.R.; Aggarwal, B.B. Design of curcumin-loaded PLGA nanoparticles formulation with enhanced cellular uptake, and increased bioactivity in vitro and superior bioavailability in vivo. Biochem. Pharmacol. 2010, 79, 330–338. [Google Scholar] [CrossRef]
- Kunnumakkara, A.B.; Anand, P.; Aggarwal, B.B. Curcumin inhibits proliferation, invasion, angiogenesis and metastasis of different cancers through interaction with multiple cell signaling proteins. Cancer Lett. 2008, 269, 199–225. [Google Scholar] [CrossRef]
- Coomaraswamy, J.; Kilger, E.; Wolfing, H.; Schafer, C.; Kaeser, S.A.; Wegenast-Braun, B.M.; Hefendehl, J.K.; Wolburg, H.; Mazzella, M.; Ghiso, J.; et al. Modeling familial Danish dementia in mice supports the concept of the amyloid hypothesis of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2010, 107, 7969–7974. [Google Scholar] [CrossRef]
- Zheng, Z.; Zhang, Y.; Liu, Y.; Wang, J.; Cui, Z.; Pan, X.; Liu, Y.; Tang, W.; Wang, K. Metabolic degradation of lentinan in liver mediated by CYP450 enzymes and epoxide hydrolase. Carbohydr. Polym. 2021, 253, 117255. [Google Scholar] [CrossRef]
- Zhang, Y.; Li, S.; Wang, X.; Zhang, L.; Cheung, P.C.K. Advances in lentinan: Isolation, structure, chain conformation and bioactivities. Food Hydrocoll. 2011, 25, 196–206. [Google Scholar] [CrossRef]
- Wasser, S.P. Medicinal mushrooms as a source of antitumor and immunomodulating polysaccharides. Appl. Microbiol. Biotechnol. 2002, 60, 258–274. [Google Scholar] [PubMed]
- Li, W.; Wang, J.; Hu, H.; Li, Q.; Liu, Y.; Wang, K. Functional polysaccharide Lentinan suppresses human breast cancer growth via inducing autophagy and caspase-7-mediated apoptosis. J. Funct. Foods 2018, 45, 75–85. [Google Scholar] [CrossRef]
- Li, R.; Dong, L.; Liang, Y.; Cui, Y.; Ji, X.; Xiao, H.; Gao, S.; Wang, L. Palladium Nanoparticles Stabilized by Lentinan with Enhanced Peroxidase-like Activity for Sensitive Detection of H2O2. ChemistrySelect 2022, 7, e202200247. [Google Scholar] [CrossRef]
- Chan, C.B.; Tse, M.C.; Liu, X.; Zhang, S.; Schmidt, R.; Otten, R.; Liu, L.; Ye, K. Activation of muscular TrkB by its small molecular agonist 7,8-dihydroxyflavone sex-dependently regulates energy metabolism in diet-induced obese mice. Chem. Biol. 2015, 22, 355–368. [Google Scholar] [CrossRef]
- Chen, C.; Wang, Z.; Zhang, Z.; Liu, X.; Kang, S.S.; Zhang, Y.; Ye, K. The prodrug of 7,8-dihydroxyflavone development and therapeutic efficacy for treating Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2018, 115, 578–583. [Google Scholar] [CrossRef]
- Liu, C.; Chan, C.B.; Ye, K. 7,8-dihydroxyflavone, a small molecular TrkB agonist, is useful for treating various BDNF-implicated human disorders. Transl. Neurodegener. 2016, 5, 2. [Google Scholar] [CrossRef]
- Rao, W.; Huang, H.; Wang, H.; Zhao, S.; Dumbleton, J.; Zhao, G.; He, X. Nanoparticle-mediated intracellular delivery enables cryopreservation of human adipose-derived stem cells using trehalose as the sole cryoprotectant. ACS Appl. Mater. Interfaces 2015, 7, 5017–5028. [Google Scholar] [CrossRef]
- Rothschild, L.J.; Mancinelli, R.L. Life in extreme environments. Nature 2001, 409, 1092–1101. [Google Scholar] [CrossRef]
- Crowe, J.H.; Crowe, L.M. Preservation of mammalian cells-learning nature’s tricks. Nat. Biotechnol. 2000, 18, 145–146. [Google Scholar] [CrossRef]
- Mandal, S.; Debnath, K.; Jana, N.R.; Jana, N.R. Trehalose-Conjugated, Catechin-Loaded Polylactide Nanoparticles for Improved Neuroprotection against Intracellular Polyglutamine Aggregates. Biomacromolecules 2020, 21, 1578–1586. [Google Scholar] [CrossRef] [PubMed]
- Pradhan, N.; Debnath, K.; Mandal, S.; Jana, N.R.; Jana, N.R. Antiamyloidogenic chemical/biochemical-based designed nanoparticle as artificial chaperone for efficient inhibition of protein aggregation. Biomacromolecules 2018, 19, 1721–1731. [Google Scholar] [CrossRef] [PubMed]
- Jana, N.R.; Nukina, N. Recent advances in understanding the pathogenesis of polyglutamine diseases: Involvement of molecular chaperones and ubiquitin-proteasome pathway. J. Chem. Neuroanat. 2003, 26, 95–101. [Google Scholar] [CrossRef] [PubMed]
- Ross, C.A.; Aylward, E.H.; Wild, E.J.; Langbehn, D.R.; Long, J.D.; Warner, J.H.; Scahill, R.I.; Leavitt, B.R.; Stout, J.C.; Paulsen, J.S.; et al. Huntington disease: Natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol. 2014, 10, 204–216. [Google Scholar] [CrossRef] [PubMed]
- Ratnam, D.V.; Ankola, D.D.; Bhardwaj, V.; Sahana, D.K.; Kumar, M.N. Role of antioxidants in prophylaxis and therapy: A pharmaceutical perspective. J. Control. Release 2006, 113, 189–207. [Google Scholar] [CrossRef]
- Liu, X.; Huang, N.; Wang, H.; Li, H.; Jin, Q.; Ji, J. The effect of ligand composition on the in vivo fate of multidentate poly(ethylene glycol) modified gold nanoparticles. Biomaterials 2013, 34, 8370–8381. [Google Scholar] [CrossRef]
- Wang, L.; Du, J.; Zhou, Y.; Wang, Y. Safety of nanosuspensions in drug delivery. Nanomedicine 2017, 13, 455–469. [Google Scholar] [CrossRef]
- Wolfram, J.; Zhu, M.; Yang, Y.; Shen, J.; Gentile, E.; Paolino, D.; Fresta, M.; Nie, G.; Chen, C.; Shen, H.; et al. Safety of Nanoparticles in Medicine. Curr. Drug Targets 2015, 16, 1671–1681. [Google Scholar] [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Zhou, F.; Peterson, T.; Fan, Z.; Wang, S. The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications. Nutrients 2023, 15, 3881. https://doi.org/10.3390/nu15183881
Zhou F, Peterson T, Fan Z, Wang S. The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications. Nutrients. 2023; 15(18):3881. https://doi.org/10.3390/nu15183881
Chicago/Turabian StyleZhou, Fang, Tiffany Peterson, Zhaoyang Fan, and Shu Wang. 2023. "The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications" Nutrients 15, no. 18: 3881. https://doi.org/10.3390/nu15183881
APA StyleZhou, F., Peterson, T., Fan, Z., & Wang, S. (2023). The Commonly Used Stabilizers for Phytochemical-Based Nanoparticles: Stabilization Effects, Mechanisms, and Applications. Nutrients, 15(18), 3881. https://doi.org/10.3390/nu15183881