Next Article in Journal
Treatment with Angiotensin-(1-7) Prevents Development of Oral Papilloma Induced in K-ras Transgenic Mice
Previous Article in Journal
CRISPR/Cas9-Mediated Knockout of miR-130b Affects Mono- and Polyunsaturated Fatty Acid Content via PPARG-PGC1α Axis in Goat Mammary Epithelial Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant-Derived Nanoscale-Encapsulated Antioxidants for Oral and Topical Uses: A Brief Review

Department of BioNano Technology, Gachon University, Seongnam-daero 1342, Sujeong-gu, Seongnam-si 13120, Korea
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(7), 3638; https://doi.org/10.3390/ijms23073638
Submission received: 11 February 2022 / Revised: 23 March 2022 / Accepted: 24 March 2022 / Published: 26 March 2022
(This article belongs to the Topic Oxidative Stress and Inflammation)

Abstract

:
Several plant-based nanoscale-encapsulated antioxidant compounds (rutin, myricetin, β-carotene, fisetin, lycopene, quercetin, genkwanin, lutein, resveratrol, eucalyptol, kaempferol, glabridin, pinene, and whole-plant bio-active compounds) are briefly introduced in this paper, along with their characteristics. Antioxidants’ bioavailability has become one of the main research topics in bio-nanomedicine. Two low patient compliance drug delivery pathways (namely, the oral and topical delivery routes), are described in detail in this paper, for nanoscale colloidal systems and gel formulations. Both routes and/or formulations seek to improve bioavailability and maximize the drug agents’ efficiency. Some well-known compounds have been robustly studied, but many remain elusive. The objective of this review is to discuss recent studies and advantages of nanoscale formulations of plant-derived antioxidant compounds.

1. Introduction

Plant-derived antioxidants have many beneficial effects on humans; one of these is the reduction of the oxidative stress [1] that has been linked to the development of degenerative diseases and cancers [2,3,4]. In addition, many antioxidants exhibit anti-microbial effects [5], anti-inflammatory effects [6], and skin protection effects [7]. The present review focuses on the properties of carotenoids, polyphenols, terpenes, and xanthophylls (Table 1).
In the present review, we consider carotenoids such as β-carotene and lycopene. Carotenoids are synthesized not only by plants, but also by bacteria, fungi, and algae [56]. For example, carotenoids are present in salmon, tomato, and watermelon, explaining the colors of these foods [57,58]. In addition, carotenoids have been implicated in the mitigation of cardiovascular diseases, cancers [59], and osteoporosis [60,61]. Polyphenols, namely rutin, myricetin, fisetin, quercetin, genkwanin, resveratrol, kaempferol, and glabridin are discussed as well. It is well-known that, for example, chocolate [62] and green tea [63] are enriched in polyphenols. Polyphenols have been shown to mitigate carcinogenesis [64], cancers [65], and chronic diseases [66]. Terpenes, eucalyptol and pinene, are introduced in this review as penetration enhancers. Terpenes are commonly present in conifers [67], and act as skin-penetration enhancers by disrupting the lipid bilayer [68]. Terpenes have been reported to exhibit radical-scavenging effects [69] and anti-inflammatory effects [70,71,72]. Xanthophylls such as lutein are discussed as well. Xanthophylls are abundant in green vegetables and corn [73,74].
The effective delivery of drugs requires proper formulations. Drug properties (such as release kinetics, effectiveness, and retention time) are affected by specific formulations. Nanoscale miniaturization has been shown to improve the drugs’ bioavailability [75,76,77]. Owing to small particle sizes and high surface ratios, nanoscale formulations have become very desirable [78,79]. With the adoption of nanoscale formulations, the importance of nanoscale delivery systems has also increased [80,81]. The physical properties of nanoscale formulations (such as size, porosity, geometry, surface charge, and hydrophobicity) impart them with unique characteristics [82]. Nanoscale formulations can be used for effective targeting and delivery of drug agents, owing to the nanoparticles’ unique properties, which make them resilient to metabolic processes and immune response [78]. Several nanocolloid and nanogel formulations are discussed in this review. Colloidal systems are promising for various formulations, including ointments, emulsions, and aerosols. The synthesis of nanoparticles in colloidal systems has garnered significant attention. Gel (or nanogel) formulations have also been widely considered for synthesizing nanoparticles. Owing to the swelling effect of gels, they can be used for effectively delivering and/or controlling molecules [83,84].
The delivery route is one of the most addressed research issues in the nanomedicine field. Determining appropriate delivery routes is important for improving the drugs’ efficiency, which is often hindered owing to the barriers imposed by the human body. The oral and topical delivery routes are discussed in the present review. The oral delivery route is one of the more convenient routes but suffers from low patient compliance. Topical delivery, also among the more convenient drug delivery routes, allows the drugs to bypass the acidic environment of the stomach and the gastro-intestinal tract, but topically administered drugs have to overcome several hurdles in the skin layer [87]. Both routes (topical and oral) should ensure safe and effective drug delivery to the target. In this review, we discuss both the oral and topical routes for delivery of nanoscale colloidal and gel formulations (Figure 1).

2. The Oral Route

2.1. Gels

Mujtaba et al. studied nanogels containing rutin-loaded chitosan-alginate nanoparticles [88]. Rutin is present in passionflower, apples, and buckwheat [89]. Rutin exhibits anti-diabetic [8] and neuroprotective [9,10] effects. The absorption rate of rutin by the human body is poor, owing to the low water solubility of rutin [90]. The objective of the study by Mujtaba et al. was to formulate rutin-loaded nanoparticles with enhanced bioavailability and better water solubility and dissolution. Rutin-loaded chitosan-alginate nanoparticles were formulated using the coacervation method with slight modifications, and gels were formed using the ionotropic pre-gelation technique. Using the novel formulation, rutin was entrapped successfully by the nanogel particles. Rutin was released rapidly and sustainably from the rutin-loaded chitosan-alginate nanogel particles, whose positively charged surface enabled easy delivery of the payload to negatively charged cellular membrane channels [91].
Yao et al. studied chitosan-based nanogels of myricetin, for oral delivery [92]. Myricetin is abundantly found in berries and vegetables [93]. Myricetin was reported to inhibit cancers [16,17] and reduce hyperglycemia [18,19,20]. The nanogel formulation was prepared by following a typical procedure [92]. Nanogels with three-dimensional physical structures exhibited sustainable release properties [94], as well as good solubility and cellular uptake properties [95]. Myricetin-loaded nanogels exhibited the sustained and controlled release of myricetin, with good bioavailability.
Liu et al. studied β-carotene-encapsulated internal-phase emulsion gels, for edible applications, for improving the retention of β-carotene; for their studies, these authors simulated the gastro-intestinal tract conditions [96]. β-carotene is among the more well-known antioxidant components [21]. β-carotene-encapsulated internal-phase emulsion gels were formulated using a slightly modified cold gelation method [22]. Increasing the gel network density and antioxidant capacity improved the stability of β-carotene-loaded high-internal-phase emulsion gels. In addition, the improved bio-accessibility of β-carotene was demonstrated for β-carotene-loaded gels and was attributed to their gel network structure.
Whole flavonoids of Satsuma mandarin (Citrus unshiu) peel extracts with pectin nanoparticles were considered by Hu et al. for effective delivery [97]. Before loading with pectin nanoparticles, Satsuma mandarin peels were extracted, and whole components of Satsuma mandarin were used in the studies. Then, the Satsuma mandarin peel extract was loaded with pectin nanoparticles, using the gelation technique [98]. Owing to the novel formulation, Satsuma mandarin peel gels exhibited improved bio-accessibility and controlled release of bioactive compounds.
Mahmoudi et al. formulated gels with chitosan nanoparticles containing the Chinese lantern (Physalis alkekengi L.) extract [99]. The Chinese lantern extract contains bioactive compounds, including carotenoids [100] and flavonoids [101]. This extract has been used in traditional medicine [102]. The seeds of the Chinese lantern were dried, ground, and then extracted using the percolation method. Next, the Chinese lantern extract was entrapped in chitosan nanoparticles using the ionic gelation method [103]. Polycationic chitosan presents high bio-affinity to a negative charged cell surface, including a negative charged cancer cell or bacteria membrane [104,105]. Using this formulation, high-biocompatibility oral consumption became possible. Chitosan nanoparticles containing the Chinese lantern extract exhibited good antioxidant effects and stability, enabling the protection of bioactive compounds using the novel formulation.

2.2. Colloids

Sechi et al. studied the encapsulation of fisetin with polymeric nanoparticles, for controlled oral delivery and release (Figure 2) [106]. Polymers in this work were biodegradable and biocompatible, of the type that is typically used for the encapsulation of drugs to enhance oral bioavailability. Fisetin, commonly found in fruits and vegetables, such as onions and strawberries [107], was reported as an antioxidant flavonoid [108,109] with anti-cancer effects [23,24]. Fisetin was loaded into nanoparticles, using a modified nanoprecipitation method [110]. With polymeric nanoparticles, fisetin-loaded nanoparticles exhibited an effective fisetin-loading capacity and controlled release of fisetin, in simulated gastro-intestinal conditions.
Regiellen et al. studied rutin-loaded bovine serum albumin nanoparticles for addressing the low oral bioavailability of rutin [11]; the study was performed using nanoscale spray drying methods [12]. Rutin-loaded bovine serum albumin nanoparticles exhibited improved radical scavenging after 72 h, implying a sustained release of rutin. Rutin penetrated into the tissue, owing to the fine particle size of rutin-loaded nanoparticles, yielding better performance than original drugs for similar in vivo conditions.
Singh et al. considered a lipid-based lycopene nanoemulsion system for improving oral bioavailability [111]. Lycopene, a carotenoid, is found in tomatoes [112] and olives [113]. It is well-known that lycopene inhibits and prevents prostate cancer [25,26]. Singh et al. formulated lycopene-loaded nano-lipid carriers, using the ultra-sonication method. Lipid nanoparticles participated in lipophilicity of the drug, so that the drug entered easily to the central nerve system (CNS) [114]. Lycopene-loaded nano-lipid carriers exhibited no precipitates or phase separation, yielding high dispersibility. Lipid nanoparticles delivered the drug inside of the cells for nucleoside transporters so the drug could access rapidly with lycopene into the cells. High stability of this formulation, good gut permeation, and good cytotoxicity against human breast tumor cells were shown for lycopene-loaded nano-lipid carriers.
Hao et al. considered quercetin-loaded polymeric nanocapsules with soybean lecithin [115]. Quercetin is a flavonoid that is abundantly present in onions, berries, and apples [116]. Quercetin was reported to inhibit the progression of the cancer cell cycle [28,29,30], and to downregulate cardiovascular diseases [31]. Hao et al. used the electrostatic deposition method for encapsulating quercetin with chitosan-coated liposomes. Quercetin-loaded nanoparticles in an aqueous solution exhibited higher stability, reducing power, and cytotoxicity with respect to the human liver hepatocellular carcinoma cells compared with free quercetin. Trypan blue staining in the MTT assay revealed cytotoxicity to the human liver hepatocellular carcinoma cells by observation of blue spots inside of the cells.
Li. et al. considered a nanosuspension system with genkwanin [117]. Genkwanin is found in the seeds of black alder (Alnus glutinosa) [117] and laurel-leaf cistus (Cistus laurifolius) [118]. Genkwanin has been reported to inhibit breast cancer [34,119]. A nanosuspension system containing genkwanin was synthesized using the anti-solvent precipitation method [35]. Genkwanin nanosuspensions demonstrated a stronger anti-tumor effect and tolerance, enabling the oral delivery route.
Zhou et al. studied whey protein isolate-based nanoemulsions for the bio-accessibility of edible β-carotene [120]. Interestingly, Zhou et al. used whole edible ingredients in their formulation. The bio-accessibility and stability of β-carotene increased owing to the smaller particle size, suggesting a suitable formulation of this oil-based bioactive compound.
For management of osteoporosis, Gera et al. developed a rutin-nanoparticle colloidal system [121] using the anti-solvent precipitation technique [13]. The rutin nanosuspension exhibited a high drug absorption rate, good solubility, and good intestinal permeability. Enhanced bioavailability, dose reduction, and long-term stability were also demonstrated by this novel formulation.
Kumar et al. formulated isradipine-loaded solid lipid nanoparticles with rutin [122]. Solid lipid nanoparticles were also formulated using a method developed by Gardouh et al. [123]. Isradipine was shown to treat stroke and heart attacks by blocking calcium channels [124]. Isradipine released from the formulated solid lipid nanoparticles exhibited enhanced sustainability and a higher absorption rate than original isradipine, owing to rutin.
For improving the oral bioavailability of lycopene, Mishra and Kumari developed lycopene nanosuspensions [75]. The nanoprecipitation method was used for formulating a lycopene nanosuspension system [27]. Lycopene nanosuspensions decreased the level of triglycerides; at the same time, they improved the effect of insulin. It was also shown that lycopene nanosuspensions increased the amount of the released drug and reduced the blood glucose level.

3. Topical Use

3.1. Gels

To develop topical delivery through the eye, Bodoki et al. considered a thermosensitive lutein nanogel system for treating selenite-induced cataracts [125]. Lutein is present in kale [126] and spinach [127,128]. Lutein has been widely shown to exert beneficial effects on eyes [36,37]. Lutein-loaded poly(lactic-co-glycolic acid) (PLGA) nanogels were formulated using the emulsion/evaporation method, while lutein-loaded zein nanogels were formulated using a liquid-liquid dispersion. Lutein-loaded nanogels demonstrated better stability and more efficient delivery to the lens, compared with the similar amounts of free lutein. These nanogels were shown to reduce selenite-induced cataracts, with better bioavailability and longer antioxidant retention.
Andleeb et al. developed yarrow (Achillea millefolium) extract-loaded nanoethosomes for topical delivery through skin (Figure 3) [129]. Yarrow is well-known as a traditional medical plant [130]. The yarrow extract reportedly exerts choleretic effects [130,131]. Andleeb et al. used a simple cold method for loading the yarrow extract into nanoethosomes. In the case of dermal delivery, ethosomes cause skin disruption. More and deeper ethosomes could permeate inside of the skin [132]. By loading the yarrow extract into nanoethosomes, ethanol helped to penetrate easily through the skin. In addition, with a narrow distribution of particle sizes, small-size yarrow extract-loaded nanoethosomes successfully delivered bioactive compounds into deep skin layers. These unique properties imparted the nanoethosomes with better skin penetration characteristics than common gels [133,134], demonstrating a higher efficiency of the yarrow extract.
Imran et al. studied a nanostructured lipid carrier gel for loading two drugs, quercetin and resveratrol, to improve the disposition for their topical delivery [135]. Resveratrol is a polyphenol compound found in grapes, peanuts [136], mulberry fruit, and Jamun seed [137] and was shown to exhibit anti-cancer effects [38,39,40] and the reduction of cardiovascular risk factors [41,42], which explains the beneficial effects of the wine [138]. Nanostructured lipid carriers were prepared by melt emulsification using the ultra-sonication method and formulated to the gel form using the methods of Naz et al. [139]. The nanostructured lipid carrier gel exhibited better skin hydration, owing to its nanoscale particles with the novel formulation, leading to better permeation of drugs encapsulated in the gel particles. The nanostructured lipid carrier gel demonstrated lower IC50 and better drug permeation than the conventional gel, implying the better topical bioavailability of the former. In addition, the nanostructured lipid carrier gel demonstrated the inhibition of migration in a bidirectional wound-healing assay, owing to its anti-metastatic effect.
Gokhale et al. developed a gel based on a quercetin-loaded nanoemulsion [140]. Quercetin was used as a drug agent for treating rheumatism, while the nanoemulsion formulation was used for effective topical delivery. The formulation was made using the spontaneous emulsification technique. Then, gels were synthesized based on the prepared nanoemulsion [141]. Quercetin-loaded gels exhibited good solubility, diffusion rate, skin permeability, and physicochemical stability.
Rutin nanocrystal gels were studied by Li et al. for improving the bioavailability and efficiency of rutin [142]. Rutin nanosuspensions were formulated using high-speed shearing and the high-pressure homogenization technique [14,15]. After preparing rutin nanosuspensions, they were freeze-dried [143] and dispersed to form a nanocrystal gel. There was a remarkable improvement of the saturation solubility, release behavior, transdermal bioavailability of the drug, and antioxidant activity of the rutin nanocrystal gel. In addition, the nanocrystal gel inhibited the oxidative damage associated with the skin photoaging.

3.2. Colloids

Hatahet et al. formulated a quercetin smartCrystals® into a nanosuspension system [144]. As mentioned above, quercetin has some beneficial health effects; on the other hand, its bioavailability is poor. By formulating quercetin into nanocrystals, its bioavailability can be improved, owing to the nanocrystals’ high performance when delivered through the dermal route [145]. Hatahet et al. synthesized quercetin nanosuspensions, using the smartCrystals® technology [32]. Using this novel formulation, quercetin nanosuspensions exhibited higher saturation solubility, better antioxidant activity, and better physical stability. In addition, the protective activity on Vero cells with respect to hydrogen peroxide toxicity was demonstrated and the MTT assay revealed that quercetin smartCrystals® do not show cellular cytotoxicity at a higher concentration than crude quercetin.
To improve the delivery of drugs and large-molecular-weight compounds into deep skin layers, Kahraman et al. formulated nanomicelles with a combination of terpenes and tacrolimus, obtaining an aqueous formulation for topical delivery [146]. Tacrolimus is an immunosuppressive drug that is used in transplant medicine [147,148]. Comprehensive terpenes were used for enhancing the tacrolimus penetration for topical delivery. Terpenes are powerful skin-penetration enhancers [149,150] and anti-inflammation agents [151,152]. Tacrolimus monohydrate-loaded polymeric micelles were prepared using the thin film hydration method [153]. Nanomicelles enhanced the drug delivery through skin [154,155]. Owing to this novel formulation, tacrolimus-loaded nanomicelles exhibited improved colloidal stability, higher drug-loading efficiency, and higher accumulation of large molecules in the viable epidermis and dermis.
Chao et al. studied a kaempferol-loaded nanoemulsion system for topical delivery [156]. Kaempferol has poor water solubility, which implies a low bio-absorption rate [157]; thus, a kaempferol-loaded nanoemulsion system has been considered for overcoming this problem. Kaempferol is abundantly found in broccoli, spinach, beans, and kale [48]. Importantly, it was reported that kaempferol reduces the risk of Alzheimer’s disease [48,49,50]. The kaempferol-loaded nanoemulsion system exhibited enhanced permeation capacity, a higher drug accumulation over the period of 12 h, a higher deposition amount in skin, and a lower lagging time.
Using glabridin, Wang et al. designed a nanosuspension system for topical delivery [158]. Glabridin, a flavonoid, is typically found in licorice [159,160], and was reported to inhibit the tyrosinase activity [51]. Wang et al. formulated a glabridin nanosuspension system using the anti-solvent precipitation-homogenization method [52]. Using this method, Wang et al. demonstrated that the glabridin nanosuspension system is promising for topical use. Glabridin nanosuspension has higher solubility due to its reduced size, so that several advantages are demonstrated. A specially formulated glabridin nanosuspension system demonstrated enhanced stability for short-term storage, with no significant particle aggregation; it also enhanced skin permeation, thanks to enhanced skin penetration due to an enhanced concentration gradient, both in vitro and in vivo.
Nikolic et al. studied curcumin-loaded nanoemulsions containing eucalyptol and pinene, for evaluating a curcumin-loaded low-energy nanoemulsion containing terpenes, for curcumin delivery [161]. Curcumin, a plant-derived polyphenol, is used in skin disease treatments [162,163]. Terpenes, eucalyptol, and pinene were used as penetration enhancers. Eucalyptol was reported to exhibit anti-inflammatory [43,44] and lung-protective effects [45,46,47]. Pinene is a major component of the essential oil in sage [163] and was reported to exhibit anti-tumor activity [53,54,55]. The spontaneous emulsification method was used for formulating the nanoemulsion systems. Both eucalyptol and pinene acted as surfactants, imparting the nanoemulsions with a low interface energy, and reduced the amount of the surfactant needed for maintaining a stable formulation, which resulted in higher safety with enhanced penetration effects of the nanoemulsions.
In another study, solid lipid nanoparticles were formulated with a mixture of surfactants [164]. Bose et al. developed a solvent-free solid lipid-based nanoscale system for topical delivery. Quercetin was used as a drug agent, and quercetin nanoparticles were prepared using the probe ultra-sonication method [33]. Owing to the drug enrichment in the outer shell, the diffusion path of the active component was shorter. Remarkable physical stability and a high initial burst-like release, as well as a prolonged release, were observed for quercetin-encapsulated solid lipid nanoparticles.

4. Conclusions

In this article, we reviewed some recent studies on plant-derived antioxidants. Applications of plant-derived antioxidants have been widely discussed. Several plant-derived antioxidants were reviewed in this paper, but the discussion is by no means exhaustive, implying additional discoveries. In addition, while some components have been well-characterized, others remain elusive.
Much remains to be discovered in the field of nanomedicine. The unique properties of nanoscale compounds are notable but remain elusive, implying the need for additional research and optimization [165,166]. At the same time, nanomedicine approaches are very promising for improving the efficiency of drug delivery systems, which in turn is likely to improve the quality of life.

Author Contributions

Y.-C.L. conceptualized and revised the study. S.-H.K. wrote the manuscript mainly. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education (2021R1F1A1047906), and by the Basic Science Research Capacity Enhancement Project through Korea Basic Science Institute (National Research Facilities and Equipment Center) grant funded by the Ministry of Education (2019R1A6C1010016), and by the Gachon University research fund of 2020 (GCU-202008480014).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative Stress and Antioxidant Defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Satue, M.; del Mar Arriero, M.; Monjo, M.; Ramis, J.M. Quercitrin and Taxifolin Stimulate Osteoblast Differentiation in Mc3t3-E1 Cells and Inhibit Osteoclastogenesis in Raw 264.7 Cells. Biochem. Pharmacol. 2013, 86, 1476–1486. [Google Scholar] [CrossRef] [PubMed]
  3. Ren, W.; Qiao, Z.; Wang, H.; Zhu, L.; Zhang, L. Flavonoids: Promising Anticancer Agents. Med. Res. Rev. 2003, 23, 519–534. [Google Scholar] [CrossRef] [PubMed]
  4. Willcox, J.K.; Ash, S.L.; Catignani, G.L. Antioxidants and Prevention of Chronic Disease. Crit. Rev. Food Sci. Nutr. 2004, 44, 275–295. [Google Scholar] [CrossRef] [PubMed]
  5. Cushnie, T.P.; Lamb, A.J. Antimicrobial Activity of Flavonoids. Int. J. Antimicrob. Agents 2005, 26, 343–356. [Google Scholar] [CrossRef]
  6. Garcia-Lafuente, A.; Guillamon, E.; Villares, A.; Rostagno, M.A.; Martinez, J.A. Flavonoids as Anti-Inflammatory Agents: Implications in Cancer and Cardiovascular Disease. Inflamm. Res. 2009, 58, 537–552. [Google Scholar] [CrossRef]
  7. Ebrahimzadeh, M.A.; Enayatifard, R.; Khalili, M.; Ghaffarloo, M.; Saeedi, M.; Charati, J.Y. Correlation between Sun Protection Factor and Antioxidant Activity, Phenol and Flavonoid Contents of Some Medicinal Plants. Iran. J. Pharm. Res. IJPR 2014, 13, 1041–1047. [Google Scholar] [PubMed]
  8. Tongjaroenbuangam, W.; Ruksee, N.; Chantiratikul, P.; Pakdeenarong, N.; Kongbuntad, W.; Govitrapong, P. Neuroprotective Effects of Quercetin, Rutin and Okra (Abelmoschus Esculentus Linn.) in Dexamethasone-Treated Mice. Neurochem. Int. 2011, 59, 677–685. [Google Scholar] [CrossRef]
  9. Miyake, K.; Arima, H.; Hirayama, F.; Yamamoto, M.; Horikawa, T.; Sumiyoshi, H.; Noda, S.; Uekama, K. Improvement of Solubility and Oral Bioavailability of Rutin by Complexation with 2-Hydroxypropyl-Β-Cyclodextrin. Pharm. Dev. Technol. 2000, 5, 399–407. [Google Scholar] [CrossRef] [PubMed]
  10. Katuwavila, N.P.; Chandani Perera, A.D.L.; Samarakoon, S.R.; Soysa, P.; Karunaratne, V.; Amaratunga, G.A.J.; Karunaratne, D.N.; Samarakoon, S.R.; Soysa, P.; Karunaratne, V.; et al. Chitosan-Alginate Nanoparticle System Efficiently Delivers Doxorubicin to Mcf-7 Cells. J. Nanomater. 2016, 2016, 1–12. [Google Scholar] [CrossRef] [Green Version]
  11. Singh, A.; Neupane, Y.R.; Panda, B.P.; Kohli, K. Lipid Based Nanoformulation of Lycopene Improves Oral Delivery: Formulation Optimization, Ex Vivo Assessment and Its Efficacy against Breast Cancer. J. Microencapsul. 2017, 34, 416–429. [Google Scholar] [CrossRef]
  12. Rao, A.V.; Waseem, Z.; Agarwal, S. Lycopene Content of Tomatoes and Tomato Products and Their Contribution to Dietary Lycopene. Food Res. Int. 1998, 31, 737–741. [Google Scholar] [CrossRef]
  13. Gardouh, A.R.; Gad, S.; Ghonaim, H.M.; Ghorab, M.M. Design and Characterization of Glyceryl Monostearate Solid Lipid Nanoparticles Prepared by High Shear Homogenization. J. Pharm. Res. Int. 2013, 3, 326–346. [Google Scholar] [CrossRef]
  14. Yang, H.; Teng, F.; Wang, P.; Tian, B.; Lin, X.; Hu, X.; Zhang, L.; Zhang, K.; Zhang, Y.; Tang, X. Investigation of a Nanosuspension Stabilized by Soluplus(R) to Improve Bioavailability. Int. J. Pharm. 2014, 477, 88–95. [Google Scholar] [CrossRef] [PubMed]
  15. Hatahet, T.; Morille, M.; Hommoss, A.; Dorandeu, C.; Muller, R.H.; Begu, S. Dermal Quercetin Smartcrystals(R): Formulation Development, Antioxidant Activity and Cellular Safety. Eur. J. Pharm. Biopharm. 2016, 102, 51–63. [Google Scholar] [CrossRef] [PubMed]
  16. Ong, K.C.; Khoo, H.-E. Effects of Myricetin on Glycemia and Glycogen Metabolism in Diabetic Rats. Life Sci. 2000, 67, 1695–1705. [Google Scholar] [CrossRef]
  17. Kandasamy, N.; Ashokkumar, N. Myricetin, a Natural Flavonoid, Normalizes Hyperglycemia in Streptozotocin-Cadmium-Induced Experimental Diabetic Nephrotoxic Rats. Biomed. Prev. Nutr. 2012, 2, 246–251. [Google Scholar] [CrossRef]
  18. Kang, S.-J.; Park, J.-H.Y.; Choi, H.-N.; Kim, J.-I. A-Glucosidase Inhibitory Activities of Myricetin in Animal Models of Diabetes Mellitus. Food Sci. Biotechnol. 2015, 24, 1897–1900. [Google Scholar] [CrossRef]
  19. Oh, J.K.; Drumright, R.; Siegwart, D.J.; Matyjaszewski, K. The Development of Microgels/Nanogels for Drug Delivery Applications. Prog. Polym. Sci. 2008, 33, 448–477. [Google Scholar] [CrossRef]
  20. Li, N.; Wang, J.; Yang, X.; Li, L. Novel Nanogels as Drug Delivery Systems for Poorly Soluble Anticancer Drugs. Colloids Surf. B Biointerfaces 2011, 83, 237–244. [Google Scholar] [CrossRef] [PubMed]
  21. Hu, Y.; Zhang, W.; Ke, Z.; Li, Y.; Zhou, Z. In Vitro Release and Antioxidant Activity of Satsuma Mandarin (Citrus reticulata Blanco Cv. Unshiu) Peel Flavonoids Encapsulated by Pectin Nanoparticles. Int. J. Food Sci. Technol. 2017, 52, 2362–2373. [Google Scholar] [CrossRef]
  22. Zhang, Y.; Chen, T.; Yuan, P.; Tian, R.; Hu, W.; Tang, Y.; Jia, Y.; Zhang, L. Encapsulation of Honokiol into Self-Assembled Pectin Nanoparticles for Drug Delivery to Hepg2 Cells. Carbohydr. Polym. 2015, 133, 31–38. [Google Scholar] [CrossRef] [PubMed]
  23. Sanna, V.; Siddiqui, I.A.; Sechi, M.; Mukhtar, H. Resveratrol-Loaded Nanoparticles Based on Poly (Epsilon-Caprolactone) and Poly (D,L-Lactic-Co-Glycolic Acid)–Poly (Ethylene Glycol) Blend for Prostate Cancer Treatment. Mol. Pharm. 2013, 10, 3871–3881. [Google Scholar] [CrossRef] [Green Version]
  24. Pedrozo, R.C.; Antônio, E.; Khalil, N.M.; Mainardes, R.M. Bovine Serum Albumin-Based Nanoparticles Containing the Flavonoid Rutin Produced by Nano Spray Drying. Braz. J. Pharm. Sci. 2020, 56. [Google Scholar] [CrossRef] [Green Version]
  25. Kaur, I.P.; Bhandari, R.; Bhandari, S.; Kakkar, V. Potential of Solid Lipid Nanoparticles in Brain Targeting. J. Control. Release 2008, 127, 97–109. [Google Scholar] [CrossRef] [PubMed]
  26. Hao, J.; Guo, B.; Yu, S.; Zhang, W.; Zhang, D.; Wang, J.; Wang, Y. Encapsulation of the Flavonoid Quercetin with Chitosan-Coated Nano-Liposomes. LWT—Food Sci. Technol. 2017, 85, 37–44. [Google Scholar] [CrossRef]
  27. Becerra-Moreno, A.; Alanís-Garza, P.A.; Mora-Nieves, J.L.; Mora-Mora, J.P.; Jacobo-Velázquez, D.A. Kale: An Excellent Source of Vitamin C, Pro-Vitamin a, Lutein and Glucosinolates. CyTA—J. Food 2013, 12, 298–303. [Google Scholar] [CrossRef]
  28. Yoshida, M.; Sakai, T.; Hosokawa, N.; Marui, N.; Matsumoto, K.; Fujioka, A.; Nishino, H.; Aoike, A. The Effect of Quercetin on Cell Cycle Progression and Growth of Human Gastric Cancer Cells. FEBS Lett. 1990, 260, 10–13. [Google Scholar] [CrossRef] [Green Version]
  29. Edwards, R.L.; Lyon, T.; Litwin, S.E.; Rabovsky, A.; Symons, J.D.; Jalili, T. Quercetin Reduces Blood Pressure in Hypertensive Subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef] [PubMed]
  30. Li, Y.; Hong, J.; Li, H.; Qi, X.; Guo, Y.; Han, M.; Wang, X. Genkwanin Nanosuspensions: A Novel and Potential Antitumor Drug in Breast Carcinoma Therapy. Drug Deliv. 2017, 24, 1491–1500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. O’Rourke, C.; Byres, M.; Delazar, A.; Kumarasamy, Y.; Nahar, L.; Stewart, F.; Sarker, S.D. Hirsutanonol, Oregonin and Genkwanin from the Seeds of Alnus Glutinosa (Betulaceae). Biochem. Syst. Ecol. 2005, 33, 749–752. [Google Scholar] [CrossRef]
  32. Peters, D.H.; Fitton, A.; Plosker, G.L.; Faulds, D. Tacrolimus. Drugs 1993, 46, 746–794. [Google Scholar] [CrossRef] [PubMed]
  33. Maurer-Jones, M.A.; Gunsolus, I.L.; Murphy, C.J.; Haynes, C.L. Toxicity of Engineered Nanoparticles in the Environment. Anal. Chem. 2013, 85, 3036–3049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Das, S.; Suresh, P.K. Nanosuspension: A New Vehicle for the Improvement of the Delivery of Drugs to the Ocular Surface. Application to Amphotericin B. Nanomedicine 2011, 7, 242–247. [Google Scholar] [CrossRef] [PubMed]
  35. Gera, S.; Pooladanda, V.; Godugu, C.; Challa, V.S.; Wankar, J.; Dodoala, S.; Sampathi, S. Rutin Nanosuspension for Potential Management of Osteoporosis: Effect of Particle Size Reduction on Oral Bioavailability, in vitro and in vivo Activity. Pharm. Dev. Technol. 2020, 25, 971–988. [Google Scholar] [CrossRef]
  36. Andleeb, M.; Shoaib Khan, H.M.; Daniyal, M. Development, Characterization and Stability Evaluation of Topical Gel Loaded with Ethosomes Containing Achillea Millefolium L. Extract. Front. Pharmacol. 2021, 12, 603227. [Google Scholar] [CrossRef] [PubMed]
  37. Benedek, B.; Kopp, B. Achillea Millefolium, L.S.L. Revisited: Recent Findings Confirm the Traditional Use. Wien. Med. Wochenschr. 2007, 157, 312–314. [Google Scholar] [CrossRef]
  38. Bishayee, A. Cancer Prevention and Treatment with Resveratrol: From Rodent Studies to Clinical Trials. Cancer Prev. Res. 2009, 2, 409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Zortea, K.; Franco, V.C.; Francesconi, L.P.; Cereser, K.M.; Lobato, M.I.; Belmonte-de-Abreu, P.S. Resveratrol Supplementation in Schizophrenia Patients: A Randomized Clinical Trial Evaluating Serum Glucose and Cardiovascular Risk Factors. Nutrients 2016, 8, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Bradamante, S.; Barenghi, L.; Villa, A. Cardiovascular Protective Effects of Resveratrol. Cardiovasc. Drug Rev. 2004, 22, 169–188. [Google Scholar] [CrossRef] [PubMed]
  41. Frémont, L. Biological Effects of Resveratrol. Life Sci. 2000, 66, 663–673. [Google Scholar] [CrossRef]
  42. Naz, Z.; Ahmad, F.J. Curcumin-Loaded Colloidal Carrier System: Formulation Optimization, Mechanistic Insight, Ex Vivo and in Vivo Evaluation. Int. J. Nanomed. 2015, 10, 4293–4307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Kennedy-Feitosa, E.; Cattani-Cavalieri, I.; Barroso, M.V.; Romana-Souza, B.; Brito-Gitirana, L.; Valenca, S.S. Eucalyptol Promotes Lung Repair in Mice Following Cigarette Smoke-Induced Emphysema. Phytomedicine 2019, 55, 70–79. [Google Scholar] [CrossRef] [PubMed]
  44. Yu, N.; Sun, Y.-T.; Su, X.-M.; He, M.; Dai, B.; Kang, J. Eucalyptol Protects Lungs against Bacterial Invasion through Attenuating Ciliated Cell Damage and Suppressing Muc5ac Expression. J. Cell. Physiol. 2019, 234, 5842–5850. [Google Scholar] [CrossRef] [PubMed]
  45. De Lima Gondim, F.; Serra, D.S.; Cavalcante, F.S.Á. Effects of Eucalyptol in Respiratory System Mechanics on Acute Lung Injury after Exposure to Short-Term Cigarette Smoke. Respir. Physiol. Neurobiol. 2019, 266, 33–38. [Google Scholar] [CrossRef]
  46. Chen, W.; Liu, Y.; Li, M.; Mao, J.; Zhang, L.; Huang, R.; Jin, X.; Ye, L. Anti-Tumor Effect of A-Pinene on Human Hepatoma Cell Lines through Inducing G2/M Cell Cycle Arrest. J. Pharmacol. Sci. 2015, 127, 332–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zhao, Y.; Chen, R.; Wang, Y.; Yang, Y. A-Pinene Inhibits Human Prostate Cancer Growth in a Mouse Xenograft Model. Chemotherapy 2018, 63, 1–7. [Google Scholar] [CrossRef] [PubMed]
  48. Kim, J.K.; Choi, S.J.; Cho, H.Y.; Hwang, H.-J.; Kim, Y.J.; Lim, S.T.; Kim, C.-J.; Kim, H.K.; Peterson, S.; Shin, D.-H. Protective Effects of Kaempferol (3,4′,5,7-Tetrahydroxyflavone) against Amyloid Beta Peptide (Aβ)-Induced Neurotoxicity in Icr Mice. Biosci. Biotechnol. Biochem. 2010, 239, 091226180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Wang, W.P.; Hul, J.; Sui, H.; Zhao, Y.S.; Feng, J.; Liu, C. Glabridin Nanosuspension for Enhanced Skin Penetration: Formulation Optimization, in Vitro and in Vivo Evaluation. Pharmazie 2016, 71, 252–257. [Google Scholar] [PubMed]
  50. Martinez, R.M.; Pinho-Ribeiro, F.A.; Steffen, V.S.; Caviglione, C.V.; Pala, D.; Baracat, M.M.; Georgetti, S.R.; Verri, W.A.; Casagrande, R. Topical Formulation Containing Hesperidin Methyl Chalcone Inhibits Skin Oxidative Stress and Inflammation Induced by Ultraviolet B Irradiation. Photochem. Photobiol. Sci. 2016, 15, 554–563. [Google Scholar] [CrossRef] [PubMed]
  51. Nikolic, I.; Mitsou, E.; Pantelic, I.; Randjelovic, D.; Markovic, B.; Papadimitriou, V.; Xenakis, A.; Lunter, D.J.; Zugic, A.; Savic, S. Microstructure and Biopharmaceutical Performances of Curcumin-Loaded Low-Energy Nanoemulsions Containing Eucalyptol and Pinene: Terpenes’ Role Overcome Penetration Enhancement Effect? Eur. J. Pharm. Sci. 2020, 142, 105135. [Google Scholar] [CrossRef]
  52. Vollono, L.; Falconi, M.; Gaziano, R.; Iacovelli, F.; Dika, E.; Terracciano, C.; Bianchi, L.; Campione, E. Potential of Curcumin in Skin Disorders. Nutrients 2019, 11, 2169. [Google Scholar]
  53. Hou, J.; Zhang, Y.; Zhu, Y.; Zhou, B.; Ren, C.; Liang, S.; Guo, Y. A-Pinene Induces Apoptotic Cell Death Via Caspase Activation in Human Ovarian Cancer Cells. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2019, 25, 6631–6638. [Google Scholar] [CrossRef] [PubMed]
  54. Bose, S.; Du, Y.; Takhistov, P.; Michniak-Kohn, B. Formulation Optimization and Topical Delivery of Quercetin from Solid Lipid Based Nanosystems. Int. J. Pharm. 2013, 441, 56–66. [Google Scholar] [CrossRef] [PubMed]
  55. Delmas, T.; Couffin, A.C.; Bayle, P.A.; de Crecy, F.; Neumann, E.; Vinet, F.; Bardet, M.; Bibette, J.; Texier, I. Preparation and Characterization of Highly Stable Lipid Nanoparticles with Amorphous Core of Tuneable Viscosity. J. Colloid Interface Sci. 2011, 360, 471–481. [Google Scholar] [CrossRef] [PubMed]
  56. Stahl, W.; Sies, H. Antioxidant Activity of Carotenoids. Mol. Asp. Med. 2003, 24, 345–351. [Google Scholar] [CrossRef]
  57. Maoka, T. Carotenoids as Natural Functional Pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Lewinsohn, E.; Sitrit, Y.; Bar, E.; Azulay, Y.; Meir, A.; Zamir, D.; Tadmor, Y. Carotenoid Pigmentation Affects the Volatile Composition of Tomato and Watermelon Fruits, as Revealed by Comparative Genetic Analyses. J. Agric. Food Chem. 2005, 53, 3142–3148. [Google Scholar] [CrossRef] [PubMed]
  59. Paiva, S.A.R.; Russell, R.M. Β-Carotene and Other Carotenoids as Antioxidants. J. Am. Coll. Nutr. 1999, 18, 426–433. [Google Scholar] [CrossRef] [PubMed]
  60. Rao, L.G.; Mackinnon, E.S.; Josse, R.G.; Murray, T.M.; Strauss, A.; Rao, A.V. Lycopene Consumption Decreases Oxidative Stress and Bone Resorption Markers in Postmenopausal Women. Osteoporos. Int. 2007, 18, 109–115. [Google Scholar] [CrossRef]
  61. Park, C.-K.; Ishimi, Y.; Ohmura, M.; Yamaguchi, M.; Ikegami, S. Vitamin a and Carotenoids Stimulate Differentiation of Mouse Osteoblastic Cells. J. Nutr. Sci. Vitaminol. 1997, 43, 281–296. [Google Scholar] [CrossRef] [PubMed]
  62. Wollgast, J.; Anklam, E. Review on Polyphenols in Theobroma Cacao: Changes in Composition during the Manufacture of Chocolate and Methodology for Identification and Quantification. Food Res. Int. 2000, 33, 423–447. [Google Scholar] [CrossRef]
  63. Tachibana, H.; Koga, K.; Fujimura, Y.; Yamada, K. A Receptor for Green Tea Polyphenol Egcg. Nat. Struct. Mol. Biol. 2004, 11, 380–381. [Google Scholar] [CrossRef] [PubMed]
  64. Yang, C.S.; Landau, J.M.; Huang, M.-T.; Newmark, H.L. Inhibition of Carcinogenesis by Dietary Polyphenolic Compounds. Annu. Rev. Nutr. 2001, 21, 381–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Zaveri, N.T. Green Tea and Its Polyphenolic Catechins: Medicinal Uses in Cancer and Noncancer Applications. Life Sci. 2006, 78, 2073–2080. [Google Scholar] [CrossRef] [PubMed]
  66. Ramassamy, C. Emerging Role of Polyphenolic Compounds in the Treatment of Neurodegenerative Diseases: A Review of Their Intracellular Targets. Eur. J. Pharmacol. 2006, 545, 51–64. [Google Scholar] [CrossRef] [PubMed]
  67. Phillips, M.A.; Croteau, R.B. Resin-Based Defenses in Conifers. Trends Plant Sci. 1999, 4, 184–190. [Google Scholar] [CrossRef]
  68. Cornwell, P.A.; Barry, B.W.; Bouwstra, J.A.; Gooris, G.S. Modes of Action of Terpene Penetration Enhancers in Human Skin; Differential Scanning Calorimetry, Small-Angle X-Ray Diffraction and Enhancer Uptake Studies. Int. J. Pharm. 1996, 127, 9–26. [Google Scholar] [CrossRef]
  69. Zengin, H.; Baysal, A.H. Antibacterial and Antioxidant Activity of Essential Oil Terpenes against Pathogenic and Spoilage-Forming Bacteria and Cell Structure-Activity Relationships Evaluated by Sem Microscopy. Molecules 2014, 19, 17773–17798. [Google Scholar] [CrossRef] [Green Version]
  70. Ciaglia, E.; Malfitano, A.M.; Laezza, C.; Fontana, A.; Nuzzo, G.; Cutignano, A.; Abate, M.; Pelin, M.; Sosa, S.; Bifulco, M.; et al. Immuno-Modulatory and Anti-Inflammatory Effects of Dihydrogracilin a, a Terpene Derived from the Marine Sponge Dendrilla Membranosa. Int. J. Mol. Sci. 2017, 18, 1643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Rufino, A.T.; Ribeiro, M.; Judas, F.; Salgueiro, L.; Lopes, M.C.; Cavaleiro, C.; Mendes, A.F. Anti-Inflammatory and Chondroprotective Activity of (+)-A-Pinene: Structural and Enantiomeric Selectivity. J. Nat. Prod. 2014, 77, 264–269. [Google Scholar] [CrossRef] [PubMed]
  72. Da Franca Rodrigues, K.A.; Amorim, L.V.; Dias, C.N.; Moraes, D.F.C.; Carneiro, S.M.P.; de Amorim Carvalho, F.A. Syzygium cumini (L.) Skeels Essential Oil and Its Major Constituent A-Pinene Exhibit Anti-Leishmania Activity through Immunomodulation in Vitro. J. Ethnopharmacol. 2015, 160, 32–40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Thomas, S.E.; Johnson, E.J. Xanthophylls. Adv. Nutr. 2018, 9, 160–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Moros, E.E.; Darnoko, D.; Cheryan, M.; Perkins, E.G.; Jerrell, J. Analysis of Xanthophylls in Corn by Hplc. J. Agric. Food Chem. 2002, 50, 5787–5790. [Google Scholar] [CrossRef] [PubMed]
  75. Mishra, S.B.; Kumari, N. Engineering of Crystalline Nano-Suspension of Lycopene for Potential Management of Oxidative Stress–Linked Diabetes in Experimental Animals. BioNanoScience 2021, 11, 345–354. [Google Scholar] [CrossRef]
  76. Yao, M.; McClements, D.J.; Xiao, H. Improving Oral Bioavailability of Nutraceuticals by Engineered Nanoparticle-Based Delivery Systems. Curr. Opin. Food Sci. 2015, 2, 14–19. [Google Scholar] [CrossRef]
  77. Shaikh, J.; Ankola, D.D.; Beniwal, V.; Singh, D.; Kumar, M.N.V.R. Nanoparticle Encapsulation Improves Oral Bioavailability of Curcumin by at Least 9-Fold When Compared to Curcumin Administered with Piperine as Absorption Enhancer. Eur. J. Pharm. Sci. 2009, 37, 223–230. [Google Scholar] [CrossRef] [PubMed]
  78. Orive, G.; Hernandez, R.M.; Gascon, A.R.; Dominguez-Gil, A.; Pedraz, J.L. Drug Delivery in Biotechnology: Present and Future. Curr. Opin. Biotechnol. 2003, 14, 659–664. [Google Scholar] [CrossRef] [PubMed]
  79. Verma, A.; Stellacci, F. Effect of Surface Properties on Nanoparticle–Cell Interactions. Small 2010, 6, 12–21. [Google Scholar] [CrossRef]
  80. Sandhiya, S.; Dkhar, S.A.; Surendiran, A. Emerging Trends of Nanomedicine—an Overview. Fundam. Clin. Pharmacol. 2009, 23, 263–269. [Google Scholar] [CrossRef] [PubMed]
  81. Lammers, T.; Aime, S.; Hennink, W.E.; Storm, G.; Kiessling, F. Theranostic Nanomedicine. Acc. Chem. Res. 2011, 44, 1029–1038. [Google Scholar] [CrossRef] [PubMed]
  82. Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer Nanomedicine: Progress, Challenges and Opportunities. Nat. Rev. Cancer 2017, 17, 20–37. [Google Scholar] [CrossRef]
  83. Colombo, P.; Bettini, R.; Santi, P.; DeAscentiis, A.; Peppas, N.A. Analysis of the Swelling and Release Mechanisms from Drug Delivery Systems with Emphasis on Drug Solubility and Water Transport. J. Control. Release 1996, 39, 231–237. [Google Scholar] [CrossRef]
  84. Gutowska, A.; Bark, J.S.; Kwon, I.C.; Bae, Y.H.; Cha, Y.; Kim, S.W. Squeezing Hydrogels for Controlled Oral Drug Delivery. J. Control. Release 1997, 48, 141–148. [Google Scholar] [CrossRef]
  85. Jepps, O.G.; Dancik, Y.; Anissimov, Y.G.; Roberts, M.S. Modeling the Human Skin Barrier—Towards a Better Understanding of Dermal Absorption. Adv. Drug Deliv. Rev. 2013, 65, 152–168. [Google Scholar] [CrossRef] [PubMed]
  86. Mujtaba, A.; Hassan, K.; Ansari, M. Chitosan-Alginate Nanoparticles as a Novel Drug Delivery System for Rutin. Int. J. Adv. Biotechnol. Res. 2018, 9, 1895–1903. [Google Scholar]
  87. Ganeshpurkar, A.; Saluja, A.K. The Pharmacological Potential of Rutin. Saudi Pharm. J. 2017, 25, 149–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Li, Y.Q.; Zhou, F.C.; Gao, F.; Bian, J.S.; Shan, F. Comparative Evaluation of Quercetin, Isoquercetin and Rutin as Inhibitors of A-Glucosidase. J. Agric. Food Chem. 2009, 57, 11463–11468. [Google Scholar] [CrossRef] [PubMed]
  89. Pu, F.; Mishima, K.; Irie, K.; Motohashi, K.; Tanaka, Y.; Orito, K.; Egawa, T.; Kitamura, Y.; Egashira, N.; Iwasaki, K.; et al. Neuroprotective Effects of Quercetin and Rutin on Spatial Memory Impairment in an 8-Arm Radial Maze Task and Neuronal Death Induced by Repeated Cerebral Ischemia in Rats. J. Pharmacol. Sci. 2007, 104, 0707310004-04. [Google Scholar] [CrossRef] [Green Version]
  90. Yao, Y.; Xia, M.; Wang, H.; Li, G.; Shen, H.; Ji, G.; Meng, Q.; Xie, Y. Preparation and Evaluation of Chitosan-Based Nanogels/Gels for Oral Delivery of Myricetin. Eur. J. Pharm. Sci. 2016, 91, 144–153. [Google Scholar] [CrossRef] [PubMed]
  91. Semwal, D.K.; Semwal, R.B.; Combrinck, S.; Viljoen, A. Myricetin: A Dietary Molecule with Diverse Biological Activities. Nutrients 2016, 8, 90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Devi, K.P.; Rajavel, T.; Habtemariam, S.; Nabavi, S.F.; Nabavi, S.M. Molecular Mechanisms Underlying Anticancer Effects of Myricetin. Life Sci. 2015, 142, 19–25. [Google Scholar] [CrossRef] [PubMed]
  93. Phillips, P.A.; Sangwan, V.; Borja-Cacho, D.; Dudeja, V.; Vickers, S.M.; Saluja, A.K. Myricetin Induces Pancreatic Cancer Cell Death Via the Induction of Apoptosis and Inhibition of the Phosphatidylinositol 3-Kinase (Pi3k) Signaling Pathway. Cancer Lett. 2011, 308, 181–188. [Google Scholar] [CrossRef] [Green Version]
  94. Liu, W.; Gao, H.; McClements, D.J.; Zhou, L.; Wu, J.; Zou, L. Stability, Rheology, and Β-Carotene Bioaccessibility of High Internal Phase Emulsion Gels. Food Hydrocoll. 2019, 88, 210–217. [Google Scholar] [CrossRef]
  95. Miller, N.J.; Sampson, J.; Candeias, L.P.; Bramley, P.M.; Rice-Evans, C.A. Antioxidant Activities of Carotenes and Xanthophylls. FEBS Lett. 1996, 384, 240–242. [Google Scholar] [CrossRef] [Green Version]
  96. Mao, L.; Roos, Y.H.; Miao, S. Study on the Rheological Properties and Volatile Release of Cold-Set Emulsion-Filled Protein Gels. J. Agric. Food Chem. 2014, 62, 11420–11428. [Google Scholar] [CrossRef] [PubMed]
  97. Mahmoudi, R.; Tajali Ardakani, M.; Hajipour Verdom, B.; Bagheri, A.; Mohammad-Beigi, H.; Aliakbari, F.; Salehpour, Z.; Alipour, M.; Afrouz, S.; Bardania, H. Chitosan Nanoparticles Containing Physalis Alkekengi-L Extract: Preparation, Optimization and Their Antioxidant Activity. Bull. Mater. Sci. 2019, 42, 1–6. [Google Scholar] [CrossRef] [Green Version]
  98. Pintea, A.; Varga, A.; Stepnowski, P.; Socaciu, C.; Culea, M.; Diehl, H.A. Chromatographic Analysis of Carotenol Fatty Acid Esters in Physalis Alkekengi and Hippophae Rhamnoides. Phytochem. Anal. 2005, 16, 188–195. [Google Scholar] [CrossRef] [PubMed]
  99. Kranjc, E.; Albreht, A.; Vovk, I.; Makuc, D.; Plavec, J. Non-Targeted Chromatographic Analyses of Cuticular Wax Flavonoids from Physalis Alkekengi L. J. Chromatogr. A 2016, 1437, 95–106. [Google Scholar] [CrossRef] [PubMed]
  100. Helvaci, S.; Kokdil, G.; Kawai, M.; Duran, N.; Duran, G.; Guvenc, A. Antimicrobial Activity of the Extracts and Physalin D from Physalis Alkekengi and Evaluation of Antioxidant Potential of Physalin D. Pharm. Biol. 2010, 48, 142–150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Lee, J.S.; Kim, G.H.; Lee, H.G. Characteristics and Antioxidant Activity of Elsholtzia Splendens Extract-Loaded Nanoparticles. J. Agric. Food Chem. 2010, 58, 3316–3321. [Google Scholar] [CrossRef] [PubMed]
  102. Chung, Y.-C.; Su, Y.-P.; Chen, C.-C.; Jia, G.; Wang, H.-l.; Wu, J.C.G.; Lin, J.-G. Relationship between Antibacterial Activity of Chitosan and Surface Characteristics of Cell Wall. J. Acta Pharmacol. Sin. 2004, 25, 932–936. [Google Scholar]
  103. Adhikari, H.S.; Yadav, P.N. Anticancer Activity of Chitosan, Chitosan Derivatives, and Their Mechanism of Action. Int. J. Biomater. 2018, 2018, 2952085. [Google Scholar] [CrossRef] [Green Version]
  104. Sechi, M.; Syed, D.N.; Pala, N.; Mariani, A.; Marceddu, S.; Brunetti, A.; Mukhtar, H.; Sanna, V. Nanoencapsulation of Dietary Flavonoid Fisetin: Formulation and in Vitro Antioxidant and Alpha-Glucosidase Inhibition Activities. Mater. Sci. Eng C Mater. Biol. Appl. 2016, 68, 594–602. [Google Scholar] [CrossRef] [PubMed]
  105. Kim, H.J.; Kim, S.H.; Yun, J.-M. Fisetin Inhibits Hyperglycemia-Induced Proinflammatory Cytokine Production by Epigenetic Mechanisms. Evid.-Based Complementary Altern. Med. 2012, 2012, 639469. [Google Scholar] [CrossRef] [PubMed]
  106. Maher, P. Modulation of Multiple Pathways Involved in the Maintenance of Neuronal Function During Aging by Fisetin. Genes Nutr. 2009, 4, 297–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Sahu, B.D.; Kalvala, A.K.; Koneru, M.; Mahesh Kumar, J.; Kuncha, M.; Rachamalla, S.S.; Sistla, R. Ameliorative Effect of Fisetin on Cisplatin-Induced Nephrotoxicity in Rats Via Modulation of Nf-Kappab Activation and Antioxidant Defence. PLoS ONE 2014, 9, e105070. [Google Scholar] [CrossRef] [PubMed]
  108. Khan, N.; Afaq, F.; Syed, D.N.; Mukhtar, H. Fisetin, a Novel Dietary Flavonoid, Causes Apoptosis and Cell Cycle Arrest in Human Prostate Cancer Lncap Cells. Carcinogenesis 2008, 29, 1049–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Suh, Y.; Afaq, F.; Johnson, J.J.; Mukhtar, H. A Plant Flavonoid Fisetin Induces Apoptosis in Colon Cancer Cells by Inhibition of Cox2 and Wnt/Egfr/Nf-Κb-Signaling Pathways. Carcinogenesis 2009, 30, 300–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Baba, K.; Nishida, K. Steroid Nanocrystals Prepared Using the Nano Spray Dryer B-90. Pharmaceutics 2013, 5, 107–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Garrido, M.; Gonzalez-Flores, D.; Marchena, A.M.; Prior, E.; Garcia-Parra, J.; Barriga, C.; Rodriguez Moratinos, A.B. A Lycopene-Enriched Virgin Olive Oil Enhances Antioxidant Status in Humans. J. Sci. Food Agric. 2013, 93, 1820–1826. [Google Scholar] [CrossRef] [PubMed]
  112. Mirahmadi, M.; Azimi-Hashemi, S.; Saburi, E.; Kamali, H.; Pishbin, M.; Hadizadeh, F. Potential Inhibitory Effect of Lycopene on Prostate Cancer. Biomed. Pharmacother. 2020, 129, 110459. [Google Scholar] [CrossRef] [PubMed]
  113. Seren, S.; Lieberman, R.; Bayraktar, U.D.; Heath, E.; Sahin, K.; Andic, F.; Kucuk, O. Lycopene in Cancer Prevention and Treatment. Am. J. Ther. 2008, 15, 66–81. [Google Scholar] [CrossRef] [PubMed]
  114. Bhagwat, S.; Haytowitz, D.B.; Holden, J.M. Usda Database for the Flavonoid Content of Selected Foods, Release 3.1; US Department of Agriculture: Beltsville, MD, USA, 2014. [Google Scholar]
  115. Jeong, J.H.; An, J.Y.; Kwon, Y.T.; Rhee, J.G.; Lee, Y.J. Effects of Low Dose Quercetin: Cancer Cell-Specific Inhibition of Cell Cycle Progression. J. Cell. Biochem. 2009, 106, 73–82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Choi, J.-A.; Kim, J.-Y.; Lee, J.-Y.; Kang, C.-M.; Kwon, H.-J.; Yoo, Y.-D.; Kim, T.-W.; Lee, Y.-S.; Lee, S.-J. Induction of Cell Cycle Arrest and Apoptosis in Human Breast Cancer Cells by Quercetin. Int. J. Oncol. 2001, 19, 837–844. [Google Scholar] [CrossRef]
  117. Sadhu, S.K.; Okuyama, E.; Fujimoto, H.; Ishibashi, M.; Yesilada, E. Prostaglandin Inhibitory and Antioxidant Components of Cistus Laurifolius, a Turkish Medicinal Plant. J. Ethnopharmacol. 2006, 108, 371–378. [Google Scholar] [CrossRef] [PubMed]
  118. Androutsopoulos, V.P.; Ruparelia, K.; Arroo, R.R.J.; Tsatsakis, A.M.; Spandidos, D.A. Cyp1-Mediated Antiproliferative Activity of Dietary Flavonoids in Mda-Mb-468 Breast Cancer Cells. Toxicology 2009, 264, 162–170. [Google Scholar] [CrossRef] [PubMed]
  119. Zhou, X.; Wang, H.; Wang, C.; Zhao, C.; Peng, Q.; Zhang, T.; Zhao, C. Stability and in Vitro Digestibility of Beta-Carotene in Nanoemulsions Fabricated with Different Carrier Oils. Food Sci. Nutr. 2018, 6, 2537–2544. [Google Scholar] [CrossRef]
  120. Gera, S.; Talluri, S.; Rangaraj, N.; Sampathi, S. Formulation and Evaluation of Naringenin Nanosuspensions for Bioavailability Enhancement. AAPS PharmSciTech 2017, 18, 3151–3162. [Google Scholar] [CrossRef]
  121. Kumar, V.; Kharb, R.; Chaudhary, H. Optimization & Design of Isradipine Loaded Solid Lipid Nanobioparticles Using Rutin by Taguchi Methodology. Int. J. Biol. Macromol. 2016, 92, 338–346. [Google Scholar]
  122. Berjukow, S.; Marksteiner, R.; Gapp, F.; Sinnegger, M.J.; Hering, S. Molecular Mechanism of Calcium Channel Block by Isradipine. Role of a Drug-Induced Inactivated Channel Conformation. J. Biol. Chem. 2000, 275, 22114–22120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Singh, A.K.; Pandey, H.; Ramteke, P.W.; Mishra, S.B. Nano-Suspension of Ursolic Acid for Improving Oral Bioavailability and Attenuation of Type Ii Diabetes: A Histopathological Investigation. Biocatal. Agric. Biotechnol. 2019, 22, 101433. [Google Scholar] [CrossRef]
  124. Bodoki, E.; Vostinaru, O.; Samoila, O.; Dinte, E.; Bodoki, A.E.; Swetledge, S.; Astete, C.E.; Sabliov, C.M. Topical Nanodelivery System of Lutein for the Prevention of Selenite-Induced Cataract. Nanomedicine 2019, 15, 188–197. [Google Scholar] [CrossRef] [PubMed]
  125. Simonovska, B.; Vovk, I.; Glavnik, V.; Cernelic, K. Effects of Extraction and High-Performance Liquid Chromatographic Conditions on the Determination of Lutein in Spinach. J. Chromatogr. A 2013, 1276, 95–101. [Google Scholar] [CrossRef] [PubMed]
  126. Aman, R.; Schieber, A.; Carle, R. Effects of Heating and Illumination on Trans−Cis Isomerization and Degradation of Β-Carotene and Lutein in Isolated Spinach Chloroplasts. J. Agric. Food Chem. 2005, 53, 9512–9518. [Google Scholar] [CrossRef] [PubMed]
  127. Krinsky, N.I.; Landrum, J.T.; Bone, R.A. Biologic Mechanisms of the Protective Role of Lutein and Zeaxanthin in the Eye. Annu. Rev. Nutr. 2003, 23, 171–201. [Google Scholar] [CrossRef] [Green Version]
  128. Buscemi, S.; Corleo, D.; Di Pace, F.; Petroni, M.L.; Satriano, A.; Marchesini, G. The Effect of Lutein on Eye and Extra-Eye Health. Nutrients 2018, 10, 1321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Benedek, B.; Geisz, N.; Jäger, W.; Thalhammer, T.; Kopp, B. Choleretic Effects of Yarrow (Achillea Millefolium S.L.) in the Isolated Perfused Rat Liver. Phytomedicine 2006, 13, 702–706. [Google Scholar] [CrossRef] [PubMed]
  130. Satyam, G.; Shivani, S.; Garima, G. Ethosomes: A Novel Tool for Drug Delivery through the Skin. J. Pharm. Res. 2010, 3, 688–691. [Google Scholar]
  131. Baek, J.S.; Pham, C.V.; Myung, C.S.; Cho, C.W. Tadalafil-Loaded Nanostructured Lipid Carriers Using Permeation Enhancers. Int. J. Pharm. 2015, 495, 701–709. [Google Scholar] [CrossRef]
  132. Panchagnula, R.; Salve, P.S.; Thomas, N.S.; Jain, A.K.; Ramarao, P. Transdermal Delivery of Naloxone: Effect of Water, Propylene Glycol, Ethanol and Their Binary Combinations on Permeation through Rat Skin. Int. J. Pharm. 2001, 219, 95–105. [Google Scholar] [CrossRef]
  133. Imran, M.; Iqubal, M.K.; Imtiyaz, K.; Saleem, S.; Mittal, S.; Rizvi, M.M.A.; Ali, J.; Baboota, S. Topical Nanostructured Lipid Carrier Gel of Quercetin and Resveratrol: Formulation, Optimization, in Vitro and Ex Vivo Study for the Treatment of Skin Cancer. Int. J. Pharm. 2020, 587, 119705. [Google Scholar] [CrossRef]
  134. Chen, R.S.; Wu, P.L.; Chiou, R.Y.Y. Peanut Roots as a Source of Resveratrol. J. Agric. Food Chem. 2002, 50, 1665–1667. [Google Scholar] [CrossRef] [PubMed]
  135. Shrikanta, A.; Kumar, A.; Govindaswamy, V. Resveratrol Content and Antioxidant Properties of Underutilized Fruits. J. Food Sci. Technol. 2015, 52, 383–390. [Google Scholar] [CrossRef] [Green Version]
  136. Aggarwal, B.B.; Bhardwaj, A.; Aggarwal, R.S.; Seeram, N.P.; Shishodia, S.; Takada, Y. Role of Resveratrol in Prevention and Therapy of Cancer: Preclinical and Clinical Studies. Anticancer. Res. 2004, 24, 2783. [Google Scholar] [PubMed]
  137. Carter, L.G.; D’Orazio, J.A.; Pearson, K.J. Resveratrol and Cancer: Focus on in Vivo Evidence. Endocr.-Relat. Cancer 2014, 21, R209–R225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Gokhale, J.P.; Mahajan, H.S.; Surana, S.J. Quercetin Loaded Nanoemulsion-Based Gel for Rheumatoid Arthritis: In Vivo and in Vitro Studies. Biomed. Pharmacother. 2019, 112, 108622. [Google Scholar] [CrossRef] [PubMed]
  139. Rachmawati, H.; Budiputra, D.K.; Mauludin, R. Curcumin Nanoemulsion for Transdermal Application: Formulation and Evaluation. Drug Dev. Ind. Pharm. 2015, 41, 560–566. [Google Scholar] [CrossRef] [PubMed]
  140. Li, J.; Ni, W.; Aisha, M.; Zhang, J.; Sun, M. A Rutin Nanocrystal Gel as an Effective Dermal Delivery System for Enhanced Anti-Photoaging Application. Drug Dev. Ind. Pharm. 2021, 47, 429–439. [Google Scholar] [CrossRef]
  141. Kakran, M.; Shegokar, R.; Sahoo, N.G.; Shaal, L.A.; Li, L.; Muller, R.H. Fabrication of Quercetin Nanocrystals: Comparison of Different Methods. Eur. J. Pharm. Biopharm. 2012, 80, 113–121. [Google Scholar] [CrossRef] [PubMed]
  142. Jacobs, C.; Müller, R.H. Production and Characterization of a Budesonide Nanosuspension for Pulmonary Administration. Pharm. Res. 2002, 19, 189–194. [Google Scholar] [CrossRef] [PubMed]
  143. Shegokar, R.; Muller, R.H. Nanocrystals: Industrially Feasible Multifunctional Formulation Technology for Poorly Soluble Actives. Int. J. Pharm. 2010, 399, 129–139. [Google Scholar] [CrossRef] [PubMed]
  144. Müller, R.H.; Keck, C.M. Second Generation of Drug Nanocrystals for Delivery of Poorly Soluble Drugs: Smartcrystal Technology. Eur. J. Pharm. Sci. 2008, 34, S20–S21. [Google Scholar] [CrossRef]
  145. Kahraman, E.; Nesetoglu, N.; Gungor, S.; Unal, D.S.; Ozsoy, Y. The Combination of Nanomicelles with Terpenes for Enhancement of Skin Drug Delivery. Int. J. Pharm. 2018, 551, 133–140. [Google Scholar] [CrossRef] [PubMed]
  146. Venkataramanan, R.; Swaminathan, A.; Prasad, T.; Jain, A.; Zuckerman, S.; Warty, V.; McMichael, J.; Lever, J.; Burckart, G.; Starzl, T. Clinical Pharmacokinetics of Tacrolimus. Clin. Pharmacokinet. 1995, 29, 404–430. [Google Scholar] [CrossRef] [PubMed]
  147. Nokhodchi, A.; Sharabiani, K.; Rashidi, M.R.; Ghafourian, T. The Effect of Terpene Concentrations on the Skin Penetration of Diclofenac Sodium. Int. J. Pharm. 2007, 335, 97–105. [Google Scholar] [CrossRef] [PubMed]
  148. Williams, A.C.; Barry, B.W. Penetration Enhancers. Adv. Drug Deliv. Rev. 2012, 64, 128–137. [Google Scholar] [CrossRef]
  149. Marques, F.M.; Figueira, M.M.; Schmitt, E.F.P.; Kondratyuk, T.P.; Endringer, D.C.; Scherer, R.; Fronza, M. In Vitro Anti-Inflammatory Activity of Terpenes Via Suppression of Superoxide and Nitric Oxide Generation and the Nf-Kappab Signalling Pathway. Inflammopharmacology 2019, 27, 281–289. [Google Scholar] [CrossRef] [PubMed]
  150. Han, Y. Ginkgo Terpene Component Has an Anti-Inflammatory Effect on Candida Albicans-Caused Arthritic Inflammation. Int. Immunopharmacol. 2005, 5, 1049–1056. [Google Scholar] [CrossRef] [PubMed]
  151. Chen, J.; Jiang, Q.D.; Chai, Y.P.; Zhang, H.; Peng, P.; Yang, X.X. Natural Terpenes as Penetration Enhancers for Transdermal Drug Delivery. Molecules 2016, 21, 1709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Bachhav, Y.G.; Mondon, K.; Kalia, Y.N.; Gurny, R.; Moller, M. Novel Micelle Formulations to Increase Cutaneous Bioavailability of Azole Antifungals. J. Control. Release 2011, 153, 126–132. [Google Scholar] [CrossRef] [PubMed]
  153. Lapteva, M.; Mondon, K.; Moller, M.; Gurny, R.; Kalia, Y.N. Polymeric Micelle Nanocarriers for the Cutaneous Delivery of Tacrolimus: A Targeted Approach for the Treatment of Psoriasis. Mol. Pharm. 2014, 11, 2989–3001. [Google Scholar] [CrossRef] [PubMed]
  154. Chao, Y.; Huang, C.-T.; Fu, L.-T.; Huang, Y.-B.; Tsai, Y.-H.; Wu, P.-C. The Effect of Submicron Emulsion Systems on Transdermal Delivery of Kaempferol. Chem. Pharm. Bull. 2012, 60, 1171–1175. [Google Scholar] [CrossRef] [Green Version]
  155. Chen, A.Y.; Chen, Y.C. A Review of the Dietary Flavonoid, Kaempferol on Human Health and Cancer Chemoprevention. Food Chem. 2013, 138, 2099–2107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Holland, T.M.; Agarwal, P.; Wang, Y.; Leurgans, S.E.; Bennett, D.A.; Booth, S.L.; Morris, M.C. Dietary Flavonols and Risk of Alzheimer Dementia. Neurology 2020, 94, e1749–e1756. [Google Scholar] [CrossRef] [PubMed]
  157. Roth, A.; Schaffner, W.; Hertel, C. Phytoestrogen Kaempferol (3,4′,5,7-Tetrahydroxyflavone) Protects Pc12 and T47d Cells from Β-Amyloid–Induced Toxicity. J. Neurosci. Res. 1999, 57, 399–404. [Google Scholar] [CrossRef]
  158. Yu, X.-Q.; Xue, C.C.; Zhou, Z.-W.; Li, C.-G.; Du, Y.-M.; Liang, J.; Zhou, S.-F. In Vitro and in Vivo Neuroprotective Effect and Mechanisms of Glabridin, a Major Active Isoflavan from Glycyrrhiza Glabra (Licorice). Life Sci. 2008, 82, 68–78. [Google Scholar] [CrossRef] [PubMed]
  159. Yokota, T.; Nishio, H.; Kubota, Y.; Mizoguchi, M. The Inhibitory Effect of Glabridin from Licorice Extracts on Melanogenesis and Inflammation. Pigment Cell Res. 1998, 11, 355–361. [Google Scholar] [CrossRef] [PubMed]
  160. Pireddu, R.; Sinico, C.; Ennas, G.; Marongiu, F.; Muzzalupo, R.; Lai, F.; Fadda, A.M. Novel Nanosized Formulations of Two Diclofenac Acid Polymorphs to Improve Topical Bioavailability. Eur. J. Pharm. Sci. 2015, 77, 208–215. [Google Scholar] [CrossRef] [PubMed]
  161. Phan, T.-T.; See, P.; Lee, S.-T.; Chan, S.-Y. Protective Effects of Curcumin against Oxidative Damage on Skin Cells in Vitro: Its Implication for Wound Healing. J. Trauma Acute Care Surg. 2001, 51, 927–931. [Google Scholar] [CrossRef]
  162. Khan, A.; Vaibhav, K.; Javed, H.; Tabassum, R.; Ahmed, M.E.; Khan, M.M.; Khan, M.B.; Shrivastava, P.; Islam, F.; Siddiqui, M.S.; et al. 1,8-Cineole (Eucalyptol) Mitigates Inflammation in Amyloid Beta Toxicated Pc12 Cells: Relevance to Alzheimer’s Disease. Neurochem. Res. 2014, 39, 344–352. [Google Scholar] [CrossRef] [PubMed]
  163. Juergens, U.R.; Dethlefsen, U.; Steinkamp, G.; Gillissen, A.; Repges, R.; Vetter, H. Anti-Inflammatory Activity of 1.8-Cineol (Eucalyptol) in Bronchial Asthma: A Double-Blind Placebo-Controlled Trial. Respir. Med. 2003, 97, 250–256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. El-Ansary, A.; Al-Daihan, S. On the Toxicity of Therapeutically Used Nanoparticles: An Overview. J. Toxicol. 2009, 2009, 754810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Hua, S.; Marks, E.; Schneider, J.J.; Keely, S. Advances in Oral Nano-Delivery Systems for Colon Targeted Drug Delivery in Inflammatory Bowel Disease: Selective Targeting to Diseased Versus Healthy Tissue. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 1117–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Palmer, B.C.; DeLouise, L.A. Nanoparticle-Enabled Transdermal Drug Delivery Systems for Enhanced Dose Control and Tissue Targeting. Molecules 2016, 21, 1719. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Skin penetration delivery route and physiological factors affecting the oral delivery route in the gastrointestinal tract. Reprinted from Nanomedicine: Nanotechnology, Biology and Medicine, 11./5, Susan Hua et al., Advances in oral nano-delivery systems for colon targeted drug delivery in inflammatory bowel disease: Selective targeting to diseased versus healthy tissue, 1117–1132. Copyright (2015), with permission from Elsevier [85] and MDPI [86].
Figure 1. Skin penetration delivery route and physiological factors affecting the oral delivery route in the gastrointestinal tract. Reprinted from Nanomedicine: Nanotechnology, Biology and Medicine, 11./5, Susan Hua et al., Advances in oral nano-delivery systems for colon targeted drug delivery in inflammatory bowel disease: Selective targeting to diseased versus healthy tissue, 1117–1132. Copyright (2015), with permission from Elsevier [85] and MDPI [86].
Ijms 23 03638 g001aIjms 23 03638 g001b
Figure 2. (AC) SEM images of fisetin nanoparticles [106]. Adopted from Sechi, Mario, et al., “Nanoencapsulation of dietary flavonoid fisetin: Formulation and in vitro antioxidant and α-glucosidase inhibition activities”. Materials Science and Engineering: C 68 (2016): 594–602, with permission of Elsevier.
Figure 2. (AC) SEM images of fisetin nanoparticles [106]. Adopted from Sechi, Mario, et al., “Nanoencapsulation of dietary flavonoid fisetin: Formulation and in vitro antioxidant and α-glucosidase inhibition activities”. Materials Science and Engineering: C 68 (2016): 594–602, with permission of Elsevier.
Ijms 23 03638 g002
Figure 3. SEM image of the herbal nanoethosomes [129]. Adapted from Andleeb et al., “Development, Characterization and Stability Evaluation of Topical Gel Loaded with Ethosomes Containing Achillea millefolium L. Extract”. Frontiers in pharmacology 12 (2021) 336, with permission of Frontiers.
Figure 3. SEM image of the herbal nanoethosomes [129]. Adapted from Andleeb et al., “Development, Characterization and Stability Evaluation of Topical Gel Loaded with Ethosomes Containing Achillea millefolium L. Extract”. Frontiers in pharmacology 12 (2021) 336, with permission of Frontiers.
Ijms 23 03638 g003
Table 1. Classification, effects, and nanonization strategies for plant-derived antioxidants considered in this paper.
Table 1. Classification, effects, and nanonization strategies for plant-derived antioxidants considered in this paper.
NameStructureClassificationEffectNanonization Strategy
Rutin Ijms 23 03638 i001PolyphenolAnti-diabetic effects [8]
Neuroprotective effects [9,10]
Coacervation method
Nano-spray drying methods [11,12]
Anti-solvent precipitation technique [13]
High-speed shearing and high-pressure homogenization technique [14,15]
Myricetin Ijms 23 03638 i002PolyphenolAnti-cancer effects [16,17]
Hyperglycemia reduction [18,19,20]
Not considered in this review
β-carotene Ijms 23 03638 i003CarotenoidRadical scavengers [21]Cold gelation method [22]
Fisetin Ijms 23 03638 i004PolyphenolAnti-cancer effects [23,24]Nanoprecipitation method
Lycopene Ijms 23 03638 i005CarotenoidInhibition and prevention of prostate cancer [25,26]Ultrasonication method
Nanoprecipitation technique [27]
Quercetin Ijms 23 03638 i006PolyphenolInhibition of the cancer cell cycle progression [28,29,30]
Regulation of cardiovascular disease [31]
Electrostatic deposition
Spontaneous emulsification technique
SmartCrystals® technology [32]
Probe ultra-sonication method [33]
Genkwanin Ijms 23 03638 i007PolyphenolInhibition of breast cancer [34]Anti-solvent precipitation method [35]
Lutein Ijms 23 03638 i008XanthophyllBeneficial eye effects [36,37]Emulsion/evaporation method
Liquid-liquid dispersion
Resveratrol Ijms 23 03638 i009PolyphenolAnti-cancer effects [38,39,40]
Reduction of cardiovascular risk [41,42]
Melt emulsification with ultra-sonication method
Eucalyptol Ijms 23 03638 i010TerpeneAnti-inflammatory effects [43,44]
Lung protective effects [45,46,47]
Spontaneous emulsification method
Kaempferol Ijms 23 03638 i011PolyphenolReduction of Alzheimer’s disease risk [48,49,50]Not considered in this review
Glabridin Ijms 23 03638 i012PolyphenolInhibition of tyrosinase activity [51]Anti-solvent precipitation-homogenization method [52]
Pinene Ijms 23 03638 i013TerpeneAnti-cancer activity [53,54,55]Spontaneous emulsification method
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kim, S.-H.; Lee, Y.-C. Plant-Derived Nanoscale-Encapsulated Antioxidants for Oral and Topical Uses: A Brief Review. Int. J. Mol. Sci. 2022, 23, 3638. https://doi.org/10.3390/ijms23073638

AMA Style

Kim S-H, Lee Y-C. Plant-Derived Nanoscale-Encapsulated Antioxidants for Oral and Topical Uses: A Brief Review. International Journal of Molecular Sciences. 2022; 23(7):3638. https://doi.org/10.3390/ijms23073638

Chicago/Turabian Style

Kim, Seong-Hyeon, and Young-Chul Lee. 2022. "Plant-Derived Nanoscale-Encapsulated Antioxidants for Oral and Topical Uses: A Brief Review" International Journal of Molecular Sciences 23, no. 7: 3638. https://doi.org/10.3390/ijms23073638

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop