Resveratrol: Its Path from Isolation to Therapeutic Action in Eye Diseases
Abstract
:1. Introduction
2. The Occurrence and Sources of Resveratrol
3. Biochemical Insight into Biosynthesis Pathways of Resveratrol and Its Derivatives
4. Methods of Extraction, Isolation and Analysis of Stilbenes
4.1. Extraction
4.2. Isolation
4.3. Analysis
5. The Stability of Resveratrol
6. The Bioavailability and Safety of Resveratrol
7. The Anti-Angiogenic Modulatory Effect of Resveratrol
8. The Antioxidant Role of Resveratrol in Eye Diseases
Resveratrol Concentration | Cell Line | Method/Target Molecule | Effect | Reference |
---|---|---|---|---|
10 mM—inside microcapsules | D407 RPE | ELISA | ↓ VEGF ↓ IL-6 | [160] |
25, 50, and 100 μM | D407 RPE | The activity of antioxidant enzymes | ↑ SOD ↑ Catalase ↑ Reduced glutathione | [153] |
12.5, 25, 50 and 100 mg/L | D407 RPE | ELISA Western Blotting qRT-PCR | ↑ SOD ↓ MDA ↑ Bcl-2 ↓ Caspase-3 | [161] |
10 μM | ARPE-19 | SIRT1 Activity Assay Kit DNMT Activity Quantification Kit qRT-PCR | ↑ SIRT1 levels ↑ DNA methyltransferases (DNMTs) | [162] |
10 μM | ARPE-19 | Flow cytometry Acridine orange staining | ↓ Cell death ↑ Autophagy | [163] |
10 μM | ARPE-19 | ELISA | ↓ VEGF | [164] |
10 μM | ARPE-19 | ELISA | ↓ VEGF ↓ IL-6 ↓ IL-8 | [142] |
10 μm 20 μM 50 μM | ARPE-19 | ELISA Western blotting | ↓ VEGF | [143] |
50 μM 100 μM | ARPE-19 | Western blotting | ↓ ERK 1/2 | [151] |
2–50 μM | ARPE-19 | ELISA RT-PCR | ↓ VEGF-A ↓ VEGF-C | [144] |
40 μM | E1A.NR3 retinal cells | Western blotting | ↑ SIRT-1 ↑ Ku70 ↓ Bax | [165] |
Pre-treatment for 24 h with 1 μM | HRECs | Carboxy-DCFDA | ↓ Intracellular ROS levels | [166] |
5 μM | HUVECs | Western blotting PCR | ↑ SIRT1 levels | [167] |
25 μM | Primary porcine trabecular meshwork cells | Carboxy-DCFDA RT-PCR Flow cytometry | ↓ Intracellular ROS levels ↓ IL1α, IL6, IL8, ↓ sa-β-gal and lipofuscin | [155] |
2.5 μM, 5 μM, 10 μM and 20 μM | Human lens epithelial cells (HLEB-3) | WST-1 Flow cytometry Western blotting | ↑ Cell viability after H2O2 damage ↓ p38 and JNK phosphorylation ↑ SOD, Catalase and HO-1 expression | [156] |
9. The Relationship between Resveratrol and Sirtuins
10. Resveratrol Delivery Systems
11. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Ginsburg, H.; Deharo, E. A call for using natural compounds in the development of new antimalarial treatments—An introduction. Malar. J. 2011, 10 (Suppl. 1), S1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; The International Natural Product Sciences Taskforce; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
- Samec, D.; Karalija, E.; Sola, I.; Vujcic Bok, V.; Salopek-Sondi, B. The Role of Polyphenols in Abiotic Stress Response: The Influence of Molecular Structure. Plants 2021, 10, 118. [Google Scholar] [CrossRef] [PubMed]
- Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxidative Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
- Leifer, A.; Barberio, D.M. Direct ingestion method for enhancing production and bioavailability of resveratrol and other phytoalexins in Vitis vinifera. Med. Hypotheses 2016, 88, 1–5. [Google Scholar] [CrossRef]
- Takaoka, M.J. Of the Phenolic Substances of White Hellebore (Veratrum grandiflorum Loe. fil.). J. Fac. Sci. Hokkaido Imp. Univ. 1940, 3, 1–16. [Google Scholar]
- Nonomura, S.; Kanagawa, H.; Makimoto, A. Chemical constituents of polygonaceous plants. I. Studies on the components of Ko-jo-kon (Polygonum cuspidatum Sieb. et Zucc.). Yakugaku Zasshi J. Pharm. Soc. Jpn. 1963, 83, 988–990. [Google Scholar] [CrossRef] [Green Version]
- Burr, M.L. Explaining the French paradox. J. R. Soc. Health 1995, 115, 217–219. [Google Scholar] [CrossRef]
- Burns, J.; Yokota, T.; Ashihara, H.; Lean, M.E.; Crozier, A. Plant foods and herbal sources of resveratrol. J. Agric. Food Chem. 2002, 50, 3337–3340. [Google Scholar] [CrossRef]
- Feijóo, O.; Moreno, A.; Falqué, E. Content of trans- and cis-resveratrol in Galician white and red wines. J. Food Compos. Anal. 2008, 21, 608–613. [Google Scholar] [CrossRef]
- Teka, T.; Zhang, L.; Ge, X.; Li, Y.; Han, L.; Yan, X. Stilbenes: Source plants, chemistry, biosynthesis, pharmacology, application and problems related to their clinical Application-A comprehensive review. Phytochemistry 2022, 197, 113128. [Google Scholar] [CrossRef]
- Hurst, W.J.; Glinski, J.A.; Miller, K.B.; Apgar, J.; Davey, M.H.; Stuart, D.A. Survey of the trans-resveratrol and trans-piceid content of cocoa-containing and chocolate products. J. Agric. Food Chem. 2008, 56, 8374–8378. [Google Scholar] [CrossRef] [PubMed]
- Ragab, A.S.; Van Fleet, J.; Jankowski, B.; Park, J.H.; Bobzin, S.C. Detection and quantitation of resveratrol in tomato fruit (Lycopersicon esculentum Mill.). J. Agric. Food Chem. 2006, 54, 7175–7179. [Google Scholar] [CrossRef] [PubMed]
- Sales, J.M.; Resurreccion, A.V. Resveratrol in peanuts. Crit. Rev. Food Sci. Nutr. 2014, 54, 734–770. [Google Scholar] [CrossRef] [PubMed]
- Lyons, M.M.; Yu, C.; Toma, R.B.; Cho, S.Y.; Reiboldt, W.; Lee, J.; van Breemen, R.B. Resveratrol in raw and baked blueberries and bilberries. J. Agric. Food Chem. 2003, 51, 5867–5870. [Google Scholar] [CrossRef] [PubMed]
- Chan, E.W.C.; Wong, C.W.; Tan, Y.H.; Foo, J.P.Y.; Wong, S.K.; Chan, H.T. Resveratrol and pterostilbene: A comparative overview of their chemistry, biosynthesis, plant sources and pharmacological properties. J. Appl. Pharm. Sci. 2019, 9, 124–129. [Google Scholar] [CrossRef] [Green Version]
- He, S.; Yan, X. From resveratrol to its derivatives: New sources of natural antioxidant. Curr. Med. Chem. 2013, 20, 1005–1017. [Google Scholar] [PubMed]
- Keylor, M.H.; Matsuura, B.S.; Stephenson, C.R. Chemistry and Biology of Resveratrol-Derived Natural Products. Chem. Rev. 2015, 115, 8976–9027. [Google Scholar] [CrossRef]
- Jeandet, P.; Delaunois, B.; Conreux, A.; Donnez, D.; Nuzzo, V.; Cordelier, S.; Clement, C.; Courot, E. Biosynthesis, metabolism, molecular engineering, and biological functions of stilbene phytoalexins in plants. Biofactors 2010, 36, 331–341. [Google Scholar] [CrossRef]
- Dubrovina, A.S.; Kiselev, K.V. Regulation of stilbene biosynthesis in plants. Planta 2017, 246, 597–623. [Google Scholar] [CrossRef]
- Shi, J.; He, M.; Cao, J.; Wang, H.; Ding, J.; Jiao, Y.; Li, R.; He, J.; Wang, D.; Wang, Y. The comparative analysis of the potential relationship between resveratrol and stilbene synthase gene family in the development stages of grapes (Vitis quinquangularis and Vitis vinifera). Plant Physiol. Biochem. 2014, 74, 24–32. [Google Scholar] [CrossRef] [PubMed]
- Ciaffi, M.; Paolacci, A.R.; Paolocci, M.; Alicandri, E.; Bigini, V.; Badiani, M.; Muganu, M. Transcriptional regulation of stilbene synthases in grapevine germplasm differentially susceptible to downy mildew. BMC Plant Biol. 2019, 19, 404. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.; Shi, J.; Yu, Y.; Shen, Y.; Tan, B.; Ye, X.; Li, J.; Feng, J. Exploration of Elite Stilbene Synthase Alleles for Resveratrol Concentration in Wild Chinese Vitis spp. and Vitis Cultivars. Front. Plant Sci. 2017, 8, 487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, W.; Tang, K.; Yang, H.R.; Wen, P.F.; Zhang, P.; Wang, H.L.; Huang, W.D. Distribution of resveratrol and stilbene synthase in young grape plants (Vitis vinifera L. cv. Cabernet Sauvignon) and the effect of UV-C on its accumulation. Plant Physiol. Biochem. 2010, 48, 142–152. [Google Scholar] [CrossRef]
- Tian, B.; Liu, J. Resveratrol: A review of plant sources, synthesis, stability, modification and food application. J. Sci. Food Agric. 2020, 100, 1392–1404. [Google Scholar] [CrossRef]
- Fan, E.; Zhang, K.; Zhu, M.; Wang, Q. Obtaining Resveratrol: From Chemical Synthesis to Biotechnological Production. Mini-Rev. Org. Chem. 2010, 7, 272–281. [Google Scholar] [CrossRef]
- Zielinska-Przyjemska, M.; Kaczmarek, M.; Krajka-Kuzniak, V.; Wierzchowski, M.; Baer-Dubowska, W. Effect of methoxy stilbenes-analogs of resveratrol-on the viability and induction of cell cycle arrest and apoptosis in human myeloid leukemia cells. Mol. Cell. Biochem. 2020, 474, 113–123. [Google Scholar] [CrossRef]
- Alosi, J.A.; McDonald, D.E.; Schneider, J.S.; Privette, A.R.; McFadden, D.W. Pterostilbene inhibits breast cancer in vitro through mitochondrial depolarization and induction of caspase-dependent apoptosis. J. Surg. Res. 2010, 161, 195–201. [Google Scholar] [CrossRef]
- Rossi, M.; Caruso, F.; Antonioletti, R.; Viglianti, A.; Traversi, G.; Leone, S.; Basso, E.; Cozzi, R. Scavenging of hydroxyl radical by resveratrol and related natural stilbenes after hydrogen peroxide attack on DNA. Chem. Biol. Interact. 2013, 206, 175–185. [Google Scholar] [CrossRef]
- Murias, M.; Handler, N.; Erker, T.; Pleban, K.; Ecker, G.; Saiko, P.; Szekeres, T.; Jäger, W. Resveratrol analogues as selective cyclooxygenase-2 inhibitors: Synthesis and structure-activity relationship. Bioorg. Med. Chem. 2004, 12, 5571–5578. [Google Scholar] [CrossRef]
- Cheng, J.C.; Fang, J.G.; Chen, W.F.; Zhou, B.; Yang, L.; Liu, Z.L. Structure-activity relationship studies of resveratrol and its analogues by the reaction kinetics of low density lipoprotein peroxidation. Bioorg. Chem. 2006, 34, 142–157. [Google Scholar] [CrossRef] [PubMed]
- Cai, Y.J.; Wei, Q.Y.; Fang, J.G.; Yang, L.; Liu, Z.L.; Wyche, J.H.; Han, Z. The 3,4-dihydroxyl groups are important for trans-resveratrol analogs to exhibit enhanced antioxidant and apoptotic activities. Anticancer Res. 2004, 24, 999–1002. [Google Scholar] [PubMed]
- Liu, L.; Li, J.; Kundu, J.K.; Surh, Y.J. Piceatannol inhibits phorbol ester-induced expression of COX-2 and iNOS in HR-1 hairless mouse skin by blocking the activation of NF-kappaB and AP-1. Inflamm. Res. 2014, 63, 1013–1021. [Google Scholar] [CrossRef] [PubMed]
- Peng, X.L.; Xu, J.; Sun, X.F.; Ying, C.J.; Hao, L.P. Analysis of trans-resveratrol and trans-piceid in vegetable foods using high-performance liquid chromatography. Int. J. Food Sci. Nutr. 2015, 66, 729–735. [Google Scholar] [CrossRef]
- Zamora-Ros, R.; Andres-Lacueva, C.; Lamuela-Raventos, R.M.; Berenguer, T.; Jakszyn, P.; Martinez, C.; Sanchez, M.J.; Navarro, C.; Chirlaque, M.D.; Tormo, M.J.; et al. Concentrations of resveratrol and derivatives in foods and estimation of dietary intake in a Spanish population: European Prospective Investigation into Cancer and Nutrition (EPIC)-Spain cohort. Br. J. Nutr. 2008, 100, 188–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Perrone, D.; Fuggetta, M.P.; Ardito, F.; Cottarelli, A.; De Filippis, A.; Ravagnan, G.; De Maria, S.; Lo Muzio, L. Resveratrol (3,5,4′-trihydroxystilbene) and its properties in oral diseases. Exp. Ther. Med. 2017, 14, 3–9. [Google Scholar] [CrossRef] [Green Version]
- Fabris, S.; Momo, F.; Ravagnan, G.; Stevanato, R. Antioxidant properties of resveratrol and piceid on lipid peroxidation in micelles and monolamellar liposomes. Biophys. Chem. 2008, 135, 76–83. [Google Scholar] [CrossRef]
- Nikfardjam, M.; Laszlo, G.; Dietrich, H. Resveratrol-derivatives and antioxidative capacity in wines made from grapes. Food Chem. 2006, 96, 74–79. [Google Scholar] [CrossRef]
- Liu, R.; Zhang, Y.; Yao, X.; Wu, Q.; Wei, M.; Yan, Z. Epsilon-Viniferin, a promising natural oligostilbene, ameliorates hyperglycemia and hyperlipidemia by activating AMPK in vivo. Food Funct. 2020, 11, 10084–10093. [Google Scholar] [CrossRef]
- Wu, C.W.; Nakamoto, Y.; Hisatome, T.; Yoshida, S.; Miyazaki, H. Resveratrol and its dimers epsilon-viniferin and delta-viniferin in red wine protect vascular endothelial cells by a similar mechanism with different potency and efficacy. Kaohsiung J. Med. Sci. 2020, 36, 535–542. [Google Scholar] [CrossRef] [Green Version]
- Zghonda, N.; Yoshida, S.; Ezaki, S.; Otake, Y.; Murakami, C.; Mliki, A.; Ghorbel, A.; Miyazaki, H. ε-Viniferin is more effective than its monomer resveratrol in improving the functions of vascular endothelial cells and the heart. Biosci. Biotechnol. Biochem. 2012, 76, 954–960. [Google Scholar] [CrossRef]
- Romero-Perez, A.I.; Lamuela-Raventos, R.M.; Andres-Lacueva, C.; de La Torre-Boronat, M.C. Method for the quantitative extraction of resveratrol and piceid isomers in grape berry skins. Effect of powdery mildew on the stilbene content. J. Agric. Food Chem. 2001, 49, 210–215. [Google Scholar] [CrossRef] [PubMed]
- Karacabey, E.; Mazza, G. Optimization of solid-liquid extraction of resveratrol and other phenolic compounds from milled grape canes (Vitis vinifera). J. Agric. Food Chem. 2008, 56, 6318–6325. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Jiang, X.; Yang, G.; Bi, Y.; Liu, W. Green and Efficient Extraction of Resveratrol from Peanut Roots Using Deep Eutectic Solvents. J. Chem. 2018, 2018, 4091930. [Google Scholar] [CrossRef]
- Kosovic, E.; Topiar, M.; Curinova, P.; Sajfrtova, M. Stability testing of resveratrol and viniferin obtained from Vitis vinifera L. by various extraction methods considering the industrial viewpoint. Sci. Rep. 2020, 10, 5564. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.; Wang, L.; Wang, J.; Wu, B.; Liu, W.; Fan, P.; Liang, Z.; Li, S. Resveratrols in Vitis berry skins and leaves: Their extraction and analysis by HPLC. Food Chem. 2013, 136, 643–649. [Google Scholar] [CrossRef]
- Brglez Mojzer, E.; Knez Hrncic, M.; Skerget, M.; Knez, Z.; Bren, U. Polyphenols: Extraction Methods, Antioxidative Action, Bioavailability and Anticarcinogenic Effects. Molecules 2016, 21, 901. [Google Scholar] [CrossRef]
- Averilla, J.N.; Oh, J.; Wu, Z.; Liu, K.H.; Jang, C.H.; Kim, H.J.; Kim, J.S.; Kim, J.S. Improved extraction of resveratrol and antioxidants from grape peel using heat and enzymatic treatments. J. Sci. Food Agric. 2019, 99, 4043–4053. [Google Scholar] [CrossRef]
- Chen, F.; Zhang, X.; Du, X.; Yang, L.; Zu, Y.; Yang, F. A new approach for obtaining trans -resveratrol from tree peony seed oil extracted residues using ionic liquid-based enzymatic hydrolysis in situ extraction. Sep. Purif. Technol. 2016, 170, 294–305. [Google Scholar] [CrossRef]
- La Torre, G.L.; Laganà, G.; Bellocco, E.; Vilasi, F.; Salvo, F.; Dugo, G. Improvement on enzymatic hydrolysis of resveratrol glucosides in wine. Food Chem. 2004, 85, 259–266. [Google Scholar] [CrossRef]
- Gligor, O.; Mocan, A.; Moldovan, C.; Locatelli, M.; Crișan, G.; Ferreira, I.C. Enzyme-assisted extractions of polyphenols—A comprehensive review. Trends Food Sci. Technol. 2019, 88, 302–315. [Google Scholar] [CrossRef]
- Mantell, C.; Rodríguez, M.; Martínez de la Ossa, E. A Screening Analysis of the High-Pressure Extraction of Anthocyanins from Red Grape Pomace with Carbon Dioxide and Cosolvent. Eng. Life Sci. 2003, 3, 38–42. [Google Scholar] [CrossRef]
- Ruan, N.; Jiao, Z.; Tang, L. Response Surface Methodology to Optimize Supercritical Carbon Dioxide Extraction of Polygonum cuspidatum. J. AOAC Int. 2022, 105, 272–281. [Google Scholar] [CrossRef]
- Beňová, B.; Adam, M.; Pavlíková, P.; Fischer, J. Supercritical fluid extraction of piceid, resveratrol and emodin from Japanese knotweed. J. Supercrit. Fluids 2010, 51, 325–330. [Google Scholar] [CrossRef]
- Casas, L.; Mantell, C.; Rodríguez, M.; Martínez de la Ossa, E.J.; Roldán, A.; De Ory, I.; Caro, I.; Blandino, A. Extraction of resveratrol from the pomace of Palomino fino grapes by supercritical carbon dioxide. J. Food Eng. 2010, 96, 304–308. [Google Scholar] [CrossRef]
- Babazadeh, A.; Taghvimi, A.; Hamishehkar, H.; Tabibiazar, M. Development of new ultrasonic–solvent assisted method for determination of trans-resveratrol from red grapes: Optimization, characterization, and antioxidant activity (ORAC assay). Food Biosci. 2017, 20, 36–42. [Google Scholar] [CrossRef]
- Piñeiro, Z.; Marrufo-Curtido, A.; Vela, C.; Palma, M. Microwave-assisted extraction of stilbenes from woody vine material. Food Bioprod. Process. 2017, 103, 18–26. [Google Scholar] [CrossRef]
- Wang, L.X.; Heredia, A.; Song, H.; Zhang, Z.; Yu, B.; Davis, C.; Redfield, R. Resveratrol glucuronides as the metabolites of resveratrol in humans: Characterization, synthesis, and anti-HIV activity. J. Pharm. Sci. 2004, 93, 2448–2457. [Google Scholar] [CrossRef]
- Ribeiro de Lima, M.T.; Waffo-Teguo, P.; Teissedre, P.L.; Pujolas, A.; Vercauteren, J.; Cabanis, J.C.; Merillon, J.M. Determination of stilbenes (trans-astringin, cis- and trans-piceid, and cis- and trans-resveratrol) in Portuguese wines. J. Agric. Food Chem. 1999, 47, 2666–2670. [Google Scholar] [CrossRef]
- Sun, H.; Lin, Q.; Wei, W.; Qin, G. Ultrasound-assisted extraction of resveratrol from grape leaves and its purification on mesoporous carbon. Food Sci. Biotechnol. 2018, 27, 1353–1359. [Google Scholar] [CrossRef]
- Abbott, J.A.; Medina-Bolivar, F.; Martin, E.M.; Engelberth, A.S.; Villagarcia, H.; Clausen, E.C.; Carrier, D.J. Purification of resveratrol, arachidin-1, and arachidin-3 from hairy root cultures of peanut (Arachis hypogaea) and determination of their antioxidant activity and cytotoxicity. Biotechnol. Prog. 2010, 26, 1344–1351. [Google Scholar] [CrossRef] [PubMed]
- Schwarz, L.J.; Danylec, B.; Yang, Y.; Harris, S.J.; Boysen, R.I.; Hearn, M.T. Enrichment of (E)-resveratrol from peanut byproduct with molecularly imprinted polymers. J. Agric. Food Chem. 2011, 59, 3539–3543. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Zhao, Y.; Chen, Z. Separation and purification of resveratrol by liquid extraction assistance macroporous adsorption resin mixed-bed technology. Sep. Sci. Technol. 2021, 56, 3106–3118. [Google Scholar] [CrossRef]
- Coskun, O. Separation techniques: Chromatography. North. Clin. Istanb. 2016, 3, 156–160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prasain, J.K. Tandem Mass Spectrometry—Applications and Principles; IntechOpen: London, UK, 2012. [Google Scholar] [CrossRef]
- Urpi-Sarda, M.; Zamora-Ros, R.; Lamuela-Raventos, R.; Cherubini, A.; Jauregui, O.; de la Torre, R.; Covas, M.I.; Estruch, R.; Jaeger, W.; Andres-Lacueva, C. HPLC-tandem mass spectrometric method to characterize resveratrol metabolism in humans. Clin. Chem. 2007, 53, 292–299. [Google Scholar] [CrossRef] [Green Version]
- Huang, X.; Mazza, G. Simultaneous analysis of serotonin, melatonin, piceid and resveratrol in fruits using liquid chromatography tandem mass spectrometry. J. Chromatogr. A 2011, 1218, 3890–3899. [Google Scholar] [CrossRef]
- Chen, W.; Yeo, S.C.; Elhennawy, M.G.; Xiang, X.; Lin, H.S. Determination of naturally occurring resveratrol analog trans-4,4′-dihydroxystilbene in rat plasma by liquid chromatography-tandem mass spectrometry: Application to a pharmacokinetic study. Anal. Bioanal. Chem. 2015, 407, 5793–5801. [Google Scholar] [CrossRef]
- Lu, S.C.; Liao, W.R.; Chen, S.F. Quantification of Trans-resveratrol in Red Wines Using QuEChERS Extraction Combined with Liquid Chromatography–Tandem Mass Spectrometry. Anal. Sci. 2018, 34, 439–444. [Google Scholar] [CrossRef] [Green Version]
- Sahebi, H.; Zandavar, H.; Pourmortazavi, S.M.; Mirsadeghi, S. Construction of Fe3O4/SiO2/chitosan-grafted-poly (N-vinylcaprolactam) magnetic nanocomposite and their application in simultaneous extraction of Trans-resveratrol and its metabolites from rat plasma. J. Chromatogr. B 2021, 1179, 122841. [Google Scholar] [CrossRef]
- Zhu, Z.; Zhong, B.; Yang, Z.; Zhao, W.; Shi, L.; Aziz, A.; Rauf, A.; Aljohani, A.S.M.; Alhumaydhi, F.A.; Suleria, H.A.R. LC-ESI-QTOF-MS/MS Characterization and Estimation of the Antioxidant Potential of Phenolic Compounds from Different Parts of the Lotus (Nelumbo nucifera) Seed and Rhizome. ACS Omega 2022, 7, 14630–14642. [Google Scholar] [CrossRef]
- Lachowicz, S.; Oszmiański, J.; Wojdyło, A.; Cebulak, T.; Hirnle, L.; Siewiński, M. UPLC-PDA-Q/TOF-MS identification of bioactive compounds and on-line UPLC-ABTS assay in Fallopia japonica Houtt and Fallopia sachalinensis (F.Schmidt) leaves and rhizomes grown in Poland. Eur. Food Res. Technol. 2019, 245, 691–706. [Google Scholar] [CrossRef] [Green Version]
- Moretón-Lamas, E.; Lago-Crespo, M.; Lage-Yusty, M.A.; López-Hernández, J. Comparison of methods for analysis of resveratrol in dietary vegetable supplements. Food Chem. 2017, 224, 219–223. [Google Scholar] [CrossRef] [PubMed]
- Fu, J.; Wang, M.; Guo, H.; Tian, Y.; Zhang, Z.; Song, R. Profiling of components of rhizoma et radix polygoni cuspidati by high-performance liquid chromatography with ultraviolet diode-array detector and ion trap/time-of-flight mass spectrometric detection. Pharmacogn. Mag. 2015, 11, 486–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Careri, M.; Corradini, C.; Elviri, L.; Nicoletti, I.; Zagnoni, I. Direct HPLC Analysis of Quercetin and trans-Resveratrol in Red Wine, Grape, and Winemaking Byproducts. J. Agric. Food Chem. 2003, 51, 5226–5231. [Google Scholar] [CrossRef] [PubMed]
- Guerrero, R.F.; Puertas, B.; Fernández, M.I.; Palma, M.; Cantos-Villar, E. Induction of stilbenes in grapes by UV-C: Comparison of different subspecies of Vitis. Innov. Food Sci. Emerg. Technol. 2010, 11, 231–238. [Google Scholar] [CrossRef]
- Vilanova, M.; Rodríguez, I.; Canosa, P.; Otero, I.; Gamero, E.; Moreno, D.; Talaverano, I.; Valdés, E. Variability in chemical composition of Vitis vinifera cv Mencı’a from different geographic areas and vintages in Ribeira Sacra (NW Spain). Food Chem. 2015, 169, 187–196. [Google Scholar] [CrossRef]
- Kawakami, S.; Kinoshita, Y.; Maruki-Uchida, H.; Yanae, K.; Sai, M.; Ito, T. Piceatannol and its metabolite, isorhapontigenin, induce SIRT1 expression in THP-1 human monocytic cell line. Nutrients 2014, 6, 4794–4804. [Google Scholar] [CrossRef] [Green Version]
- AL-Bukhaiti, W.Q.; Anwar Noman, A.; Qasim, A.S.; AL-Farga, A. Gas Chromatography: Principles, Advantages and Applications in Food Analysis. Int. J. Agric. Innov. Res. 2017, 6, 123–128. [Google Scholar]
- Rimando, A.M.; Cody, R. Determination of stilbenes in blueberries. LCGC N. Am. 2005, 23, 1192–1200. [Google Scholar]
- Viñas, P.; Martínez-Castillo, N.; Campillo, N.; Hernández-Córdoba, M. Directly suspended droplet microextraction with in injection-port derivatization coupled to gas chromatography–mass spectrometry for the analysis of polyphenols in herbal infusions, fruits and functional foods. J. Chromatogr. A 2011, 1218, 639–646. [Google Scholar] [CrossRef]
- Viñas, P.; Campillo, N.; Martínez-Castillo, N.; Hernández-Córdoba, M. Solid-phase microextraction on-fiber derivatization for the analysis of some polyphenols in wine and grapes using gas chromatography-mass spectrometry. J. Chromatogr. A 2009, 1216, 1279–1284. [Google Scholar] [CrossRef] [PubMed]
- Gu, X.; Chu, Q.; O’Dwyer, M.; Zeece, M. Analysis of resveratrol in wine by capillary electrophoresis. J. Chromatogr. A 2000, 881, 471–481. [Google Scholar] [CrossRef] [PubMed]
- Moze, S.; Polak, T.; Gasperlin, L.; Koron, D.; Vanzo, A.; Poklar Ulrih, N.; Abram, V. Phenolics in Slovenian bilberries (Vaccinium myrtillus L.) and blueberries (Vaccinium corymbosum L.). J. Agric. Food Chem. 2011, 59, 6998–7004. [Google Scholar] [CrossRef] [PubMed]
- Kiselev, K.V.; Aleynova, O.A.; Grigorchuk, V.P.; Dubrovina, A.S. Stilbene accumulation and expression of stilbene biosynthesis pathway genes in wild grapevine Vitis amurensis Rupr. Planta 2017, 245, 151–159. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; He, S.; Sun, C.; Pan, Y. Selective 1O2 quenchers, oligostilbenes, from Vitis wilsonae: Structural identification and biogenetic relationship. Phytochemistry 2012, 77, 294–303. [Google Scholar] [CrossRef] [PubMed]
- Counet, C.; Callemien, D.; Collin, S. Chocolate and cocoa: New sources of trans-resveratrol and trans-piceid. Food Chem. 2006, 98, 649–657. [Google Scholar] [CrossRef]
- Rosso, M.D.; Soligo, S.; Panighel, A.; Carraro, R.; Vedova, A.D.; Maoz, I.; Tomasi, D.; Flamini, R. Changes in grape polyphenols (V. vinifera L.) as a consequence of post-harvest withering by high-resolution mass spectrometry: Raboso Piave versus Corvina. J. Mass Spectrom. 2016, 51, 750–760. [Google Scholar] [CrossRef]
- Rimando, A.M.; Kalt, W.; Magee, J.B.; Dewey, J.; Ballington, J.R. Resveratrol, pterostilbene, and piceatannol in vaccinium berries. J. Agric. Food Chem. 2004, 52, 4713–4719. [Google Scholar] [CrossRef]
- Tokuşoǧlu, Ö.; Ünal, M.K.; Yemiş, F. Determination of the Phytoalexin Resveratrol (3,5,4‘-Trihydroxystilbene) in Peanuts and Pistachios by High-Performance Liquid Chromatographic Diode Array (HPLC-DAD) and Gas Chromatography−Mass Spectrometry (GC-MS). J. Agric. Food Chem. 2005, 53, 5003–5009. [Google Scholar] [CrossRef]
- Di Fabio, E.; Incocciati, A.; Palombarini, F.; Boffi, A.; Bonamore, A.; Macone, A. Ethylchloroformate Derivatization for GC–MS Analysis of Resveratrol Isomers in Red Wine. Molecules 2020, 25, 4603. [Google Scholar] [CrossRef]
- Rocha, S.; Araújo, A.M.; Almeida, A.; de Pinho, P.G.; Fernandes, E. Development and Validation of a GC-MS/MS Method for cis- and trans-Resveratrol Determination: Application to Portuguese Wines. Food Anal. Methods 2019, 12, 1536–1544. [Google Scholar] [CrossRef]
- Guo, J.; Zhao, J.; Zhang, M.; Sun, Z.; Liu, L. Optimization of the ultrasonic-assisted extraction of trans-resveratrol and its glucoside from grapes followed by UPLC-MS/MS using the response surface methodology. J. Food Meas. Charact. 2022, 16, 1124–1136. [Google Scholar] [CrossRef]
- Ji, M.; Li, Q.; Ji, H.; Lou, H. Investigation of the distribution and season regularity of resveratrol in Vitis amurensis via HPLC–DAD–MS/MS. Food Chem. 2014, 142, 61–65. [Google Scholar] [CrossRef] [PubMed]
- Kong, Q.J.; Ren, X.Y.; Hu, N.; Sun, C.R.; Pan, Y.J. Identification of isomers of resveratrol dimer and their analogues from wine grapes by HPLC/MSn and HPLC/DAD-UV. Food Chem. 2011, 127, 727–734. [Google Scholar] [CrossRef]
- Spanilá, M.; Pazourek, J.; Farková, M.; Havel, J. Optimization of solid-phase extraction using artificial neural networks in combination with experimental design for determination of resveratrol by capillary zone electrophoresis in wines. J. Chromatogr. A 2005, 1084, 180–185. [Google Scholar] [CrossRef]
- Clardy, J.; Walsh, C. Lessons from natural molecules. Nature 2004, 432, 829–837. [Google Scholar] [CrossRef]
- Nur Mursyida, S.; Mahendran, S.; Siew Hua, G.; Pei Teng, L.; Jaishree, V.; Subban, R. Resveratrol: Latest Scientific Evidences of its Chemical, Biological Activities and Therapeutic Potentials. Pharmacogn. J. 2020, 12, 1779–1791. [Google Scholar]
- Francioso, A.; Mastromarino, P.; Masci, A.; d’Erme, M.; Mosca, L. Chemistry, stability and bioavailability of resveratrol. Med. Chem. 2014, 10, 237–245. [Google Scholar] [CrossRef]
- Zupancic, S.; Lavric, Z.; Kristl, J. Stability and solubility of trans-resveratrol are strongly influenced by pH and temperature. Eur. J. Pharm. Biopharm. 2015, 93, 196–204. [Google Scholar] [CrossRef]
- Bancuta, O.R.; Chilian, A.; Bancuta, I.; Setnescu, R.; Setnescu, T.; Ion, R.M. Thermal characterization of resveratrol. Rev. Chim 2018, 69, 1346–1351. [Google Scholar] [CrossRef]
- Robinson, K.; Mock, C.; Liang, D. Pre-formulation studies of resveratrol. Drug Dev. Ind. Pharm. 2015, 41, 1464–1469. [Google Scholar] [CrossRef] [PubMed]
- Neves, A.R.; Lucio, M.; Lima, J.L.; Reis, S. Resveratrol in medicinal chemistry: A critical review of its pharmacokinetics, drug-delivery, and membrane interactions. Curr. Med. Chem. 2012, 19, 1663–1681. [Google Scholar] [CrossRef] [PubMed]
- Lu, Z.; Zhang, Y.; Liu, H.; Yuan, J.; Zheng, Z.; Zou, G. Transport of a cancer chemopreventive polyphenol, resveratrol: Interaction with serum albumin and hemoglobin. J. Fluoresc. 2007, 17, 580–587. [Google Scholar] [CrossRef]
- Vitaglione, P.; Sforza, S.; Galaverna, G.; Ghidini, C.; Caporaso, N.; Vescovi, P.P.; Fogliano, V.; Marchelli, R. Bioavailability of trans-resveratrol from red wine in humans. Mol. Nutr. Food Res. 2005, 49, 495–504. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Xu, Y.; Lv, H.; Pang, W.; Wang, J.; Ma, H.; Wang, S. Intestinal pharmacokinetics of resveratrol and regulatory effects of resveratrol metabolites on gut barrier and gut microbiota. Food Chem. 2021, 357, 129532. [Google Scholar] [CrossRef]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [Green Version]
- Goldberg, D.M.; Yan, J.; Soleas, G.J. Absorption of three wine-related polyphenols in three different matrices by healthy subjects. Clin. Biochem. 2003, 36, 79–87. [Google Scholar] [CrossRef]
- Boocock, D.J.; Faust, G.E.; Patel, K.R.; Schinas, A.M.; Brown, V.A.; Ducharme, M.P.; Booth, T.D.; Crowell, J.A.; Perloff, M.; Gescher, A.J.; et al. Phase I dose escalation pharmacokinetic study in healthy volunteers of resveratrol, a potential cancer chemopreventive agent. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1246–1252. [Google Scholar] [CrossRef] [Green Version]
- Vaz-da-Silva, M.; Loureiro, A.I.; Falcao, A.; Nunes, T.; Rocha, J.F.; Fernandes-Lopes, C.; Soares, E.; Wright, L.; Almeida, L.; Soares-da-Silva, P. Effect of food on the pharmacokinetic profile of trans-resveratrol. Int. J. Clin. Pharmacol. Ther. 2008, 46, 564–570. [Google Scholar] [CrossRef]
- la Porte, C.; Voduc, N.; Zhang, G.; Seguin, I.; Tardiff, D.; Singhal, N.; Cameron, D.W. Steady-State pharmacokinetics and tolerability of trans-resveratrol 2000 mg twice daily with food, quercetin and alcohol (ethanol) in healthy human subjects. Clin. Pharm. 2010, 49, 449–454. [Google Scholar] [CrossRef]
- Almeida, L.; Vaz-da-Silva, M.; Falcão, A.; Soares, E.; Costa, R.; Loureiro, A.I.; Fernandes-Lopes, C.; Rocha, J.F.; Nunes, T.; Wright, L.; et al. Pharmacokinetic and safety profile of trans-resveratrol in a rising multiple-dose study in healthy volunteers. Mol. Nutr. Food Res. 2009, 53 (Suppl. 1), S7–S15. [Google Scholar] [CrossRef] [PubMed]
- Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Penn, J.S.; Madan, A.; Caldwell, R.B.; Bartoli, M.; Caldwell, R.W.; Hartnett, M.E. Vascular endothelial growth factor in eye disease. Prog. Retin. Eye Res. 2008, 27, 331–371. [Google Scholar] [CrossRef] [Green Version]
- Ferrara, N. From the discovery of vascular endothelial growth factor to the introduction of avastin in clinical trials—An interview with Napoleone Ferrara by Domenico Ribatti. Int. J. Dev. Biol. 2011, 55, 383–388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frank, S.; Hübner, G.; Breier, G.; Longaker, M.T.; Greenhalgh, D.G.; Werner, S. Regulation of vascular endothelial growth factor expression in cultured keratinocytes. Implications for normal and impaired wound healing. J. Biol. Chem. 1995, 270, 12607–12613. [Google Scholar] [CrossRef] [Green Version]
- Gerber, H.P.; Vu, T.H.; Ryan, A.M.; Kowalski, J.; Werb, Z.; Ferrara, N. VEGF couples hypertrophic cartilage remodeling, ossification and angiogenesis during endochondral bone formation. Nat. Med. 1999, 5, 623–628. [Google Scholar] [CrossRef]
- Sunderkötter, C.; Steinbrink, K.; Goebeler, M.; Bhardwaj, R.; Sorg, C. Macrophages and angiogenesis. J. Leukoc. Biol. 1994, 55, 410–422. [Google Scholar] [CrossRef]
- Itakura, J.; Ishiwata, T.; Shen, B.; Kornmann, M.; Korc, M. Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int. J. Cancer 2000, 85, 27–34. [Google Scholar] [CrossRef]
- Verheul, H.M.; Hoekman, K.; Luykx-de Bakker, S.; Eekman, C.A.; Folman, C.C.; Broxterman, H.J.; Pinedo, H.M. Platelet: Transporter of vascular endothelial growth factor. Clin. Cancer Res. 1997, 3, 2187–2190. [Google Scholar]
- Ribatti, D. The discovery of the fundamental role of VEGF in the development of the vascular system. Mech. Dev. 2019, 160, 103579. [Google Scholar] [CrossRef]
- Flaxman, S.R.; Bourne, R.R.A.; Resnikoff, S.; Ackland, P.; Braithwaite, T.; Cicinelli, M.V.; Das, A.; Jonas, J.B.; Keeffe, J.; Kempen, J.H.; et al. Global causes of blindness and distance vision impairment 1990–2020: A systematic review and meta-analysis. Lancet Glob. Health 2017, 5, e1221–e1234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.Q.; Welchowski, T.; Schmid, M.; Letow, J.; Wolpers, C.; Pascual-Camps, I.; Holz, F.G.; Finger, R.P. Prevalence, incidence and future projection of diabetic eye disease in Europe: A systematic review and meta-analysis. Eur. J. Epidemiol. 2020, 35, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Jager, R.D.; Mieler, W.F.; Miller, J.W. Age-related macular degeneration. N. Engl. J. Med. 2008, 358, 2606–2617. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, A.; Jayesh Sodha, S.; Junnuthula, V.; Kolimi, P.; Dyawanapelly, S. Novel and investigational therapies for wet and dry age-related macular degeneration. Drug Discov. Today 2022, 27, 2322–2332. [Google Scholar] [CrossRef]
- Zafar, S.; Sachdeva, M.; Frankfort, B.J.; Channa, R. Retinal Neurodegeneration as an Early Manifestation of Diabetic Eye Disease and Potential Neuroprotective Therapies. Curr. Diabetes Rep. 2019, 19, 17. [Google Scholar] [CrossRef]
- Gündüz, K.; Bakri, S.J. Management of proliferative diabetic retinopathy. Compr. Ophthalmol. Update 2007, 8, 245–256. [Google Scholar]
- Ellis, D.; Burgess, P.I.; Kayange, P. Management of diabetic retinopathy. Malawi Med. J. 2013, 25, 116–120. [Google Scholar]
- Dulull, N.; Kwa, F.; Osman, N.; Rai, U.; Shaikh, B.; Thrimawithana, T.R. Recent advances in the management of diabetic retinopathy. Drug Discov. Today 2019, 24, 1499–1509. [Google Scholar] [CrossRef]
- Ferrara, N.; Damico, L.; Shams, N.; Lowman, H.; Kim, R. Development of ranibizumab, an anti-vascular endothelial growth factor antigen binding fragment, as therapy for neovascular age-related macular degeneration. Retina 2006, 26, 859–870. [Google Scholar] [CrossRef]
- Rodrigues, E.B.; Farah, M.E.; Maia, M.; Penha, F.M.; Regatieri, C.; Melo, G.B.; Pinheiro, M.M.; Zanetti, C.R. Therapeutic monoclonal antibodies in ophthalmology. Prog. Retin. Eye Res. 2009, 28, 117–144. [Google Scholar] [CrossRef] [PubMed]
- Bheemidi, A.B.; Valentim, C.C.; Singh, R.P. Aflibercept in Diabetic Retinopathy. Touchrev. Ophthalmol. 2021, 15, 18–22. [Google Scholar] [CrossRef]
- Mukherji, S.K. Bevacizumab (Avastin). Am. J. Neuroradiol. 2009, 31, 235–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, L.; Acon, D.; Berrocal, M.H.; Gallego-Pinazo, R.; Dolz-Marco, R.; Roca, J.A.; Maia, M.; Rojas, S.; Zas, M.; Arevalo, J.F.; et al. Five-year outcomes after intravitreal bevacizumab of treatment-naive eyes with macular edema secondary to CRVO in routine clinical practice: Results of the Pan-American Collaborative Retina Study (PACORES) group. Int. Ophthalmol. 2022, 42, 951–958. [Google Scholar] [CrossRef] [PubMed]
- Stewart, M.W. Aflibercept (VEGF-TRAP): The Next Anti-VEGF Drug. Inflamm. Allergy Drug Targets 2011, 10, 497–508. [Google Scholar] [CrossRef] [PubMed]
- Fogli, S.; Del Re, M.; Rofi, E.; Posarelli, C.; Figus, M.; Danesi, R. Clinical pharmacology of intravitreal anti-VEGF drugs. Eye 2018, 32, 1010–1020. [Google Scholar] [CrossRef] [PubMed]
- Cheung, N.; Mitchell, P.; Wong, T.Y. Diabetic retinopathy. Lancet 2010, 376, 124–136. [Google Scholar] [CrossRef]
- Al Rasheed, R.; Adhi, M.I.; Alowedi, S.A.; Albdah, B.; Aldebasi, T.; Hazzazi, M.A. Long-term peripheral retinal vascular behavior in retinopathy of prematurity patients treated with ranibizumab intravitreal injection as monotherapy using fluorescein angiography. Int. J. Retin. Vitr. 2022, 8, 53. [Google Scholar] [CrossRef]
- Lin, Y.L.; Chang, H.C.; Chen, T.L.; Chang, J.H.; Chiu, W.T.; Lin, J.W.; Chen, R.M. Resveratrol protects against oxidized LDL-induced breakage of the blood-brain barrier by lessening disruption of tight junctions and apoptotic insults to mouse cerebrovascular endothelial cells. J. Nutr. 2010, 140, 2187–2192. [Google Scholar] [CrossRef] [Green Version]
- Duarte, A.C.; Rosado, T.; Costa, A.R.; Santos, J.; Gallardo, E.; Quintela, T.; Ishikawa, H.; Schwerk, C.; Schroten, H.; Goncalves, I.; et al. The bitter taste receptor TAS2R14 regulates resveratrol transport across the human blood-cerebrospinal fluid barrier. Biochem. Pharmacol. 2020, 177, 113953. [Google Scholar] [CrossRef]
- Losso, J.N.; Truax, R.E.; Richard, G. Trans-resveratrol inhibits hyperglycemia-induced inflammation and connexin downregulation in retinal pigment epithelial cells. J. Agric. Food Chem. 2010, 58, 8246–8252. [Google Scholar] [CrossRef]
- Seong, H.; Ryu, J.; Jeong, J.Y.; Chung, I.Y.; Han, Y.S.; Hwang, S.H.; Park, J.M.; Kang, S.S.; Seo, S.W. Resveratrol suppresses vascular endothelial growth factor secretion via inhibition of CXC-chemokine receptor 4 expression in ARPE-19 cells. Mol. Med. Rep. 2015, 12, 1479–1484. [Google Scholar] [CrossRef] [Green Version]
- Nagineni, C.N.; Raju, R.; Nagineni, K.K.; Kommineni, V.K.; Cherukuri, A.; Kutty, R.K.; Hooks, J.J.; Detrick, B. Resveratrol Suppresses Expression of VEGF by Human Retinal Pigment Epithelial Cells: Potential Nutraceutical for Age-related Macular Degeneration. Aging Dis. 2014, 5, 88–100. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Jiang, D. Effect of Resveratrol on Bcl-2 and VEGF Expression in Oxygen-Induced Retinopathy of Prematurity. J. Pediatr. Ophthalmol. Strabismus 2012, 49, 230–235. [Google Scholar] [CrossRef] [PubMed]
- Yar, A.S.; Menevse, S.; Dogan, I.; Alp, E.; Ergin, V.; Cumaoglu, A.; Aricioglu, A.; Ekmekci, A.; Menevse, A. Investigation of Ocular Neovascularization–Related Genes and Oxidative Stress in Diabetic Rat Eye Tissues After Resveratrol Treatment. J. Med. Food 2011, 15, 391–398. [Google Scholar] [CrossRef] [PubMed]
- Kim, Y.H.; Kim, Y.S.; Roh, G.S.; Choi, W.S.; Cho, G.J. Resveratrol blocks diabetes-induced early vascular lesions and vascular endothelial growth factor induction in mouse retinas. Acta Ophthalmol. 2012, 90, e31–e37. [Google Scholar] [CrossRef]
- Chen, Y.; Meng, J.; Li, H.; Wei, H.; Bi, F.; Liu, S.; Tang, K.; Guo, H.; Liu, W. Resveratrol exhibits an effect on attenuating retina inflammatory condition and damage of diabetic retinopathy via PON1. Exp. Eye Res. 2019, 181, 356–366. [Google Scholar] [CrossRef]
- Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative Stress: Harms and Benefits for Human Health. Oxidative Med. Cell. Longev. 2017, 2017, 8416763. [Google Scholar] [CrossRef] [Green Version]
- Calderon, G.D.; Juarez, O.H.; Hernandez, G.E.; Punzo, S.M.; De la Cruz, Z.D. Oxidative stress and diabetic retinopathy: Development and treatment. Eye 2017, 31, 1122–1130. [Google Scholar] [CrossRef]
- King, R.E.; Kent, K.D.; Bomser, J.A. Resveratrol reduces oxidation and proliferation of human retinal pigment epithelial cells via extracellular signal-regulated kinase inhibition. Chem. Biol. Interact. 2005, 151, 143–149. [Google Scholar] [CrossRef]
- Li, J.; Yu, S.; Ying, J.; Shi, T.; Wang, P. Resveratrol Prevents ROS-Induced Apoptosis in High Glucose-Treated Retinal Capillary Endothelial Cells via the Activation of AMPK/Sirt1/PGC-1α Pathway. Oxidative Med. Cell. Longev. 2017, 2017, 7584691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pintea, A.; Rugină, D.; Pop, R.; Bunea, A.; Socaciu, C.; Diehl, H.A. Antioxidant effect of trans-resveratrol in cultured human retinal pigment epithelial cells. J. Ocul. Pharmacol. Ther. 2011, 27, 315–321. [Google Scholar] [CrossRef] [PubMed]
- Lançon, A.; Frazzi, R.; Latruffe, N. Anti-Oxidant, Anti-Inflammatory and Anti-Angiogenic Properties of Resveratrol in Ocular Diseases. Molecules 2016, 21, 304. [Google Scholar] [CrossRef] [PubMed]
- Luna, C.; Li, G.; Liton, P.B.; Qiu, J.; Epstein, D.L.; Challa, P.; Gonzalez, P. Resveratrol prevents the expression of glaucoma markers induced by chronic oxidative stress in trabecular meshwork cells. Food Chem. Toxicol. 2009, 47, 198–204. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Liu, Y.; Ge, J.; Wang, X.; Liu, L.; Bu, Z.; Liu, P. Resveratrol protects human lens epithelial cells against H2O2-induced oxidative stress by increasing catalase, SOD-1, and HO-1 expression. Mol. Vis. 2010, 16, 1467–1474. [Google Scholar]
- Soufi, F.G.; Mohammad-Nejad, D.; Ahmadieh, H. Resveratrol improves diabetic retinopathy possibly through oxidative stress—Nuclear factor κB—Apoptosis pathway. Pharmacol. Rep. 2012, 64, 1505–1514. [Google Scholar] [CrossRef]
- Soufi, F.G.; Vardyani, M.; Sheervalilou, R.; Mohammadi, M.; Somi, M.H. Long-term treatment with resveratrol attenuates oxidative stress pro-inflammatory mediators and apoptosis in streptozotocin-nicotinamide-induced diabetic rats. Gen. Physiol. Biophys. 2012, 31, 431–438. [Google Scholar] [CrossRef]
- Al-Hussaini, H.; Kittaneh, R.S.; Kilarkaje, N. Effects of trans-resveratrol on type 1 diabetes-induced up-regulation of apoptosis and mitogen-activated protein kinase signaling in retinal pigment epithelium of Dark Agouti rats. Eur. J. Pharmacol. 2021, 904, 174167. [Google Scholar] [CrossRef]
- Ruginǎ, D.; Ghiman, R.; Focșan, M.; Tăbăran, F.; Copaciu, F.; Suciu, M.; Pintea, A.; Aștilean, S. Resveratrol-delivery vehicle with anti-VEGF activity carried to human retinal pigmented epithelial cells exposed to high-glucose induced conditions. Colloids Surf. B Biointerfaces 2019, 181, 66–75. [Google Scholar] [CrossRef]
- Yang, Y.; Wu, Z.Z.; Cheng, Y.L.; Lin, W.; Qu, C. Resveratrol protects against oxidative damage of retinal pigment epithelium cells by modulating SOD/MDA activity and activating Bcl-2 expression. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 378–388. [Google Scholar] [CrossRef]
- Maugeri, A.; Barchitta, M.; Mazzone, M.G.; Giuliano, F.; Basile, G.; Agodi, A. Resveratrol Modulates SIRT1 and DNMT Functions and Restores LINE-1 Methylation Levels in ARPE-19 Cells under Oxidative Stress and Inflammation. Int. J. Mol. Sci. 2018, 19, 2118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Josifovska, N.; Albert, R.; Nagymihály, R.; Lytvynchuk, L.; Moe, M.C.; Kaarniranta, K.; Veréb, Z.J.; Petrovski, G. Resveratrol as Inducer of Autophagy, Pro-Survival, and Anti-Inflammatory Stimuli in Cultured Human RPE Cells. Int. J. Mol. Sci. 2020, 21, 813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhatt, P.; Fnu, G.; Bhatia, D.; Shahid, A.; Sutariya, V. Nanodelivery of Resveratrol-Loaded PLGA Nanoparticles for Age-Related Macular Degeneration. AAPS PharmSciTech 2020, 21, 291. [Google Scholar] [CrossRef] [PubMed]
- Anekonda, T.S.; Adamus, G. Resveratrol prevents antibody-induced apoptotic death of retinal cells through upregulation of Sirt1 and Ku70. BMC Res. Notes 2008, 1, 122. [Google Scholar] [CrossRef] [Green Version]
- Giordo, R.; Nasrallah, G.K.; Posadino, A.M.; Galimi, F.; Capobianco, G.; Eid, A.H.; Pintus, G. Resveratrol-Elicited PKC Inhibition Counteracts NOX-Mediated Endothelial to Mesenchymal Transition in Human Retinal Endothelial Cells Exposed to High Glucose. Antioxidants 2021, 10, 224. [Google Scholar] [CrossRef]
- Santini, S.J.; Cordone, V.; Mijit, M.; Bignotti, V.; Aimola, P.; Dolo, V.; Falone, S.; Amicarelli, F. SIRT1-Dependent Upregulation of Antiglycative Defense in HUVECs Is Essential for Resveratrol Protection against High Glucose Stress. Antioxidants 2019, 8, 346. [Google Scholar] [CrossRef] [Green Version]
- Grabowska, W.; Sikora, E.; Bielak-Zmijewska, A. Sirtuins, a promising target in slowing down the ageing process. Biogerontology 2017, 18, 447–476. [Google Scholar] [CrossRef]
- Michan, S.; Sinclair, D. Sirtuins in mammals: Insights into their biological function. Biochem. J. 2007, 404, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 Activation by Natural Phytochemicals: An Overview. Front. Pharmacol. 2020, 11, 1225. [Google Scholar] [CrossRef]
- Carafa, V.; Rotili, D.; Forgione, M.; Cuomo, F.; Serretiello, E.; Hailu, G.S.; Jarho, E.; Lahtela-Kakkonen, M.; Mai, A.; Altucci, L. Sirtuin functions and modulation: From chemistry to the clinic. Clin. Epigenet. 2016, 8, 61. [Google Scholar] [CrossRef]
- Alhazzazi, T.Y.; Kamarajan, P.; Verdin, E.; Kapila, Y.L. SIRT3 and cancer: Tumor promoter or suppressor? Biochim. Biophys. Acta BBA Rev. Cancer 2011, 1816, 80–88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Taurone, S.; De Ponte, C.; Rotili, D.; De Santis, E.; Mai, A.; Fiorentino, F.; Scarpa, S.; Artico, M.; Micera, A. Biochemical Functions and Clinical Characterizations of the Sirtuins in Diabetes-Induced Retinal Pathologies. Int. J. Mol. Sci. 2022, 23, 4048. [Google Scholar] [CrossRef] [PubMed]
- Bordone, L.; Motta, M.C.; Picard, F.; Robinson, A.; Jhala, U.S.; Apfeld, J.; McDonagh, T.; Lemieux, M.; McBurney, M.; Szilvasi, A.; et al. Sirt1 regulates insulin secretion by repressing UCP2 in pancreatic beta cells. PLoS Biol. 2006, 4, e31. [Google Scholar] [CrossRef] [Green Version]
- Qin, K.; Zhang, N.; Zhang, Z.; Nipper, M.; Zhu, Z.; Leighton, J.; Xu, K.; Musi, N.; Wang, P. SIRT6-mediated transcriptional suppression of Txnip is critical for pancreatic beta cell function and survival in mice. Diabetologia 2018, 61, 906–918. [Google Scholar] [CrossRef] [Green Version]
- Gertz, M.; Nguyen, G.T.; Fischer, F.; Suenkel, B.; Schlicker, C.; Franzel, B.; Tomaschewski, J.; Aladini, F.; Becker, C.; Wolters, D.; et al. A molecular mechanism for direct sirtuin activation by resveratrol. PLoS ONE 2012, 7, e49761. [Google Scholar] [CrossRef] [Green Version]
- Jiang, T.; Gu, J.; Chen, W.; Chang, Q. Resveratrol inhibits high-glucose-induced inflammatory “metabolic memory” in human retinal vascular endothelial cells through SIRT1-dependent signaling. Can. J. Physiol. Pharmacol. 2019, 97, 1141–1151. [Google Scholar] [CrossRef]
- Bagul, P.K.; Dinda, A.K.; Banerjee, S.K. Effect of resveratrol on sirtuins expression and cardiac complications in diabetes. Biochem. Biophys. Res. Commun. 2015, 468, 221–227. [Google Scholar] [CrossRef]
- Kubota, S.; Ozawa, Y.; Kurihara, T.; Sasaki, M.; Yuki, K.; Miyake, S.; Noda, K.; Ishida, S.; Tsubota, K. Roles of AMP-activated protein kinase in diabetes-induced retinal inflammation. Investig. Ophthalmol. Vis. Sci. 2011, 52, 9142–9148. [Google Scholar] [CrossRef] [Green Version]
- Kubota, S.; Kurihara, T.; Ebinuma, M.; Kubota, M.; Yuki, K.; Sasaki, M.; Noda, K.; Ozawa, Y.; Oike, Y.; Ishida, S.; et al. Resveratrol prevents light-induced retinal degeneration via suppressing activator protein-1 activation. Am. J. Pathol. 2010, 177, 1725–1731. [Google Scholar] [CrossRef]
- Singh, G. Resveratrol: Nanocarrier-based delivery systems to enhance its therapeutic potential. Nanomedicine 2020, 15, 2801–2817. [Google Scholar] [CrossRef]
- Manojlovic, V.; Rajic, N.; Djonlagic, J.; Obradovic, B.; Nedovic, V.; Bugarski, B. Application of Electrostatic Extrusion—Flavour Encapsulation and Controlled Release. Sensors 2008, 8, 1488–1496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davidov-Pardo, G.; McClements, D.J. Resveratrol encapsulation: Designing delivery systems to overcome solubility, stability and bioavailability issues. Trends Food Sci. Technol. 2014, 38, 88–103. [Google Scholar] [CrossRef]
- Tapeinos, C.; Battaglini, M.; Ciofani, G. Advances in the design of solid lipid nanoparticles and nanostructured lipid carriers for targeting brain diseases. J. Control. Release 2017, 264, 306–332. [Google Scholar] [CrossRef] [PubMed]
- Neves, A.R.; Lúcio, M.; Martins, S.; Lima, J.L.C.; Reis, S. Novel resveratrol nanodelivery systems based on lipid nanoparticles to enhance its oral bioavailability. Int. J. Nanomed. 2013, 8, 177–187. [Google Scholar] [CrossRef] [Green Version]
- Mohseni, R.; ArabSadeghabadi, Z.; Ziamajidi, N.; Abbasalipourkabir, R.; RezaeiFarimani, A. Oral Administration of Resveratrol-Loaded Solid Lipid Nanoparticle Improves Insulin Resistance Through Targeting Expression of SNARE Proteins in Adipose and Muscle Tissue in Rats with Type 2 Diabetes. Nanoscale Res. Lett. 2019, 14, 227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Z.; Tan, S.; Li, S.; Shen, Q.; Wang, K. Cancer drug delivery in the nano era: An overview and perspectives (Review). Oncol. Rep. 2017, 38, 611–624. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.N.; Cao, Y.N.; Sun, J.; Liang, Z.; Wu, Q.; Cui, S.H.; Zhi, D.F.; Guo, S.T.; Zhen, Y.H.; Zhang, S.B. Anti-breast cancer activity of resveratrol encapsulated in liposomes. J. Mater. Chem. B 2020, 8, 27–37. [Google Scholar] [CrossRef]
- Jhaveri, A.; Deshpande, P.; Pattni, B.; Torchilin, V. Transferrin-targeted, resveratrol-loaded liposomes for the treatment of glioblastoma. J. Control. Release 2018, 277, 89–101. [Google Scholar] [CrossRef]
- Jagwani, S.; Jalalpure, S.; Dhamecha, D.; Jadhav, K.; Bohara, R. Pharmacokinetic and Pharmacodynamic Evaluation of Resveratrol Loaded Cationic Liposomes for Targeting Hepatocellular Carcinoma. ACS Biomater. Sci. Eng. 2020, 6, 4969–4984. [Google Scholar] [CrossRef]
- Wang, M.; Li, L.; Zhang, X.; Liu, Y.; Zhu, R.; Liu, L.; Fang, Y.; Gao, Z.; Gao, D. Magnetic Resveratrol Liposomes as a New Theranostic Platform for Magnetic Resonance Imaging Guided Parkinson’s Disease Targeting Therapy. ACS Sustain. Chem. Eng. 2018, 6, 17124–17133. [Google Scholar] [CrossRef]
- Yücel, Ç.; Karatoprak, G.; Aktaş, Y. Nanoliposomal Resveratrol as a Novel Approach to Treatment of Diabetes Mellitus. J. Nanosci. Nanotechnol. 2018, 18, 3856–3864. [Google Scholar] [CrossRef] [PubMed]
- Machado, N.D.; Fernandez, M.A.; Diaz, D.D. Recent Strategies in Resveratrol Delivery Systems. Chempluschem 2019, 84, 951–973. [Google Scholar] [CrossRef] [PubMed]
- Muzzalupo, R.; Tavano, L. Niosomal drug delivery for transdermal targeting: Recent advances. Res. Rep. Transdermal Drug Deliv. 2015, 4, 23–33. [Google Scholar] [CrossRef] [Green Version]
- El-Haddad, M.E.; Hussien, A.A.; Saeed, H.M.; Farid, R.M. Down regulation of inflammatory cytokines by the bioactive resveratrol-loaded chitoniosomes in induced ocular inflammation model. J. Drug Deliv. Sci. Technol. 2021, 66, 102787. [Google Scholar] [CrossRef]
- Pircalabioru, G.G.; Bleotu, C.; Curutiu, C.; Mihaescu, G.; Chifiriuc, M.-C. Chapter 2—Nanodrug delivery systems in cancer. In Biomedical Applications of Nanoparticles; Grumezescu, A.M., Ed.; William Andrew Publishing: Norwich, NY, USA, 2019; pp. 31–62. [Google Scholar]
- Ahmadi, Z.; Mohammadinejad, R.; Ashrafizadeh, M. Drug delivery systems for resveratrol, a non-flavonoid polyphenol: Emerging evidence in last decades. J. Drug Deliv. Sci. Technol. 2019, 51, 591–604. [Google Scholar] [CrossRef]
- Devi, P.; Sharma, P.; Rathore, C.; Negi, P. Novel Drug Delivery Systems of Resveratrol to Bioavailability and Therapeutic Effects. In Resveratrol—Adding Life to Years, Not Adding Years to Life; IntechOpen: London, UK, 2019. [Google Scholar] [CrossRef] [Green Version]
- Li, M.; Zhang, L.; Li, R.; Yan, M. New resveratrol micelle formulation for ocular delivery: Characterization and in vitro/in vivo evaluation. Drug Dev. Ind. Pharm. 2020, 46, 1960–1970. [Google Scholar] [CrossRef]
- Selvarajan, V.; Obuobi, S.; Ee, P.L.R. Silica Nanoparticles—A Versatile Tool for the Treatment of Bacterial Infections. Front. Chem. 2020, 8, 602. [Google Scholar] [CrossRef]
- Kumar, A.; Ma, H.; Zhang, X.; Huang, K.; Jin, S.; Liu, J.; Wei, T.; Cao, W.; Zou, G.; Liang, X.-J. Gold nanoparticles functionalized with therapeutic and targeted peptides for cancer treatment. Biomaterials 2012, 33, 1180–1189. [Google Scholar] [CrossRef]
- Dong, Y.; Wan, G.; Yan, P.; Qian, C.; Li, F.; Peng, G. Fabrication of resveratrol coated gold nanoparticles and investigation of their effect on diabetic retinopathy in streptozotocin induced diabetic rats. J. Photochem. Photobiol. B Biol. 2019, 195, 51–57. [Google Scholar] [CrossRef]
- Sarma, S.; Agarwal, S.; Bhuyan, P.; Hazarika, J.; Ganguly, M. Resveratrol-loaded chitosan-pectin core-shell nanoparticles as novel drug delivery vehicle for sustained release and improved antioxidant activities. R. Soc. Open Sci. 2022, 9, 210784. [Google Scholar] [CrossRef]
- Soldati, P.P.; Polonini, H.C.; Paes, C.Q.; Restrepob, J.A.S.; Creczynksi-Pasa, T.B.; Chaves, M.G.A.M.; Brandão, M.A.F.; Pittella, F.; Raposo, N.R.B. Controlled release of resveratrol from lipid nanoparticles improves antioxidant effect. IFAC Pap. 2018, 51, 16–21. [Google Scholar] [CrossRef]
- Ha, E.-S.; Park, H.; Lee, S.-K.; Sim, W.-Y.; Jeong, J.-S.; Baek, I.-h.; Kim, M.-S. Pure Trans-Resveratrol Nanoparticles Prepared by a Supercritical Antisolvent Process Using Alcohol and Dichloromethane Mixtures: Effect of Particle Size on Dissolution and Bioavailability in Rats. Antioxidants 2020, 9, 342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Natesan, S.; Pandian, S.; Ponnusamy, C.; Palanichamy, R.; Muthusamy, S.; Kandasamy, R. Co-encapsulated resveratrol and quercetin in chitosan and peg modified chitosan nanoparticles: For efficient intra ocular pressure reduction. Int. J. Biol. Macromol. 2017, 104, 1837–1845. [Google Scholar] [CrossRef] [PubMed]
- Saha, M.; Saha, D.R.; Ulhosna, T.; Sharker, S.M.; Shohag, M.H.; Islam, M.S.; Ray, S.K.; Rahman, G.M.S.; Reza, H.M. QbD based development of resveratrol-loaded mucoadhesive lecithin/chitosan nanoparticles for prolonged ocular drug delivery. J. Drug Deliv. Sci. Technol. 2021, 63, 102480. [Google Scholar] [CrossRef]
- Razzacki, S.Z.; Thwar, P.K.; Yang, M.; Ugaz, V.M.; Burns, M.A. Integrated microsystems for controlled drug delivery. Adv. Drug Deliv. Rev. 2004, 56, 185–198. [Google Scholar] [CrossRef] [PubMed]
- Rabanel, J.M.; Banquy, X.; Zouaoui, H.; Mokhtar, M.; Hildgen, P. Progress technology in microencapsulation methods for cell therapy. Biotechnol. Prog. 2009, 25, 946–963. [Google Scholar] [CrossRef] [PubMed]
- Lengyel, M.; Kállai-Szabó, N.; Antal, V.; Laki, A.J.; Antal, I. Microparticles, Microspheres, and Microcapsules for Advanced Drug Delivery. Sci. Pharm. 2019, 87, 20. [Google Scholar] [CrossRef] [Green Version]
- Kalaycioglu, G.D.; Aydogan, N. Layer-by-layer coated microcapsules with lipid nanodomains for dual-drug delivery. Colloids Surf. A Physicochem. Eng. Asp. 2020, 584, 124037. [Google Scholar] [CrossRef]
- Sun, Z.; Wu, B.; Ren, Y.; Wang, Z.; Zhao, C.X.; Hai, M.; Weitz, D.A.; Chen, D. Diverse Particle Carriers Prepared by Co-Precipitation and Phase Separation: Formation and Applications. Chempluschem 2021, 86, 49–58. [Google Scholar] [CrossRef]
- Cano-Higuita, D.M.; Malacrida, C.R.; Telis, V.R.N. Stability of Curcumin Microencapsulated by Spray and Freeze Drying in Binary and Ternary Matrices of Maltodextrin, Gum Arabic and Modified Starch. J. Food Process. Preserv. 2015, 39, 2049–2060. [Google Scholar] [CrossRef]
- de Moura, S.C.S.R.; Berling, C.L.; Germer, S.P.M.; Alvim, I.D.; Hubinger, M.D. Encapsulating anthocyanins from Hibiscus sabdariffa L. calyces by ionic gelation: Pigment stability during storage of microparticles. Food Chem. 2018, 241, 317–327. [Google Scholar] [CrossRef]
- Ren, X.; Hou, T.; Liang, Q.; Zhang, X.; Hu, D.; Xu, B.; Chen, X.; Chalamaiah, M.; Ma, H. Effects of frequency ultrasound on the properties of zein-chitosan complex coacervation for resveratrol encapsulation. Food Chem. 2019, 279, 223–230. [Google Scholar] [CrossRef]
- Wang, L.; Lai, C.; Li, D.; Luo, Z.; Liu, L.; Jiang, Y.; Li, L. Lecithin-Polysaccharide Self-Assembled Microspheres for Resveratrol Delivery. Antioxidants 2022, 11, 1666. [Google Scholar] [CrossRef] [PubMed]
- Gartziandia, O.; Lasa, A.; Pedraz, J.L.; Miranda, J.; Portillo, M.P.; Igartua, M.; Hernandez, R.M. Preparation and Characterization of Resveratrol Loaded Pectin/Alginate Blend Gastro-Resistant Microparticles. Molecules 2018, 23, 1886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stoia, D.; Pop, R.; Campu, A.; Nistor, M.; Astilean, S.; Pintea, A.; Suciu, M.; Rugina, D.; Focsan, M. Hybrid polymeric therapeutic microcarriers for thermoplasmonic-triggered release of resveratrol. Colloids Surf. B Biointerfaces 2022, 220, 112915. [Google Scholar] [CrossRef] [PubMed]
- Harrison, I.P.; Spada, F. Hydrogels for Atopic Dermatitis and Wound Management: A Superior Drug Delivery Vehicle. Pharmaceutics 2018, 10, 71. [Google Scholar] [CrossRef]
- Ulijn, R.V.; Bibi, N.; Jayawarna, V.; Thornton, P.D.; Todd, S.J.; Mart, R.J.; Smith, A.M.; Gough, J.E. Bioresponsive hydrogels. Mater. Today 2007, 10, 40–48. [Google Scholar] [CrossRef] [Green Version]
- Buosi, F.S.; Alaimo, A.; Di Santo, M.C.; Elías, F.; García Liñares, G.; Acebedo, S.L.; Castañeda Cataña, M.A.; Spagnuolo, C.C.; Lizarraga, L.; Martínez, K.D.; et al. Resveratrol encapsulation in high molecular weight chitosan-based nanogels for applications in ocular treatments: Impact on human ARPE-19 culture cells. Int. J. Biol. Macromol. 2020, 165, 804–821. [Google Scholar] [CrossRef] [PubMed]
Analytical Method | Identified Stilbene | Source | Reference |
---|---|---|---|
HPLC-MS | Trans-resveratrol | Bilberries (Vaccinium myrtillus L.) Highbush blueberries (Vaccinium corymbosum L.) | [84] |
Trans-resveratrol | Red wines | [69] | |
Trans-piceid Cis-piceid | Red seedless grape Raspberry frozen Raspberry Peach Plum Different varieties of pears and apples | [67] | |
Trans-resveratrol | Red seedless grape Raspberry frozen Raspberry Tomato Plum | ||
Cis-piceid Trans-piceid Trans-resveratrol ε-viniferin | Grapes (Vitis amurensis) | [85] | |
ε-viniferin | Wild grape (Vitis wilsonae) | [86] | |
HPLC-MS/MS | Trans-resveratrol Trans-piceid | Dark chocolate and cocoa liquor | [87] |
UPLC-QTOF-MS | Resveratrol Resveratrol 3-O-glucoside | Lotus (Nelumbo nucifera) | [71] |
Trans-resveratrol Vis-resveratrol Trans-piceid Cis-piceid | Fallopia japonica Fallopia sachalinensis | [72] | |
Trans-resveratrol Resveratrol oligomers Viniferins Piceatannol | Grapes (Vitis vinifera) | [88] | |
GC-MS | Resveratrol Piceatannol | Berries (Vaccinium) | [89] |
Trans-resveratrol Cis-resveratrol Trans-piceid Cis-piceid | Tomato fruit (Lycopersicon esculentum Mill.) | [13] | |
Trans-resveratrol Cis-resveratrol | Black tea Green tea Red tea Chamomile | [82] | |
Trans-resveratrol Cis-resveratrol | Peanut (Arachis hypogaea L.) varieties Pistachio (Pistacia vera L.) varieties | [90] | |
Trans-resveratrol Cis-resveratrol | Red Wines | [91] | |
GC-MS/MS | Trans-resveratrol Cis-resveratrol | Red Wines | [92] |
UPLC-MS/MS | Trans-resveratrol 3-O-β-D-glucuronide 4′-O-β-D-glucuronide | Grape skins, grape seeds and grape stems | [93] |
HPLC-DAD/UV | Resveratrol | Tissues of Vitis amurensis | [94] |
Trans-resveratrol Cis-resveratrol | Peanut (Arachis hypogaea L.) varieties Pistachio (Pistacia vera L.) varieties | [90] | |
Trans-resveratrol | Nero d’Avola red grape variety | [75] | |
Trans-resveratrol | Grapes of a wide range of Vitis subspecies (sativa and sylvestris) | [76] | |
Resveratrol-O-sulfate Resveratrol-O-diglucoside | Rhizoma et Radix Polygoni Cuspidati | [74] | |
ε-Viniferins | Xinjiang wine grapes (Vitis Vinifera) | [95] | |
CE | Trans-resveratrol Cis-resveratrol | White wines Red wines | [96] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Pop, R.; Daescu, A.; Rugina, D.; Pintea, A. Resveratrol: Its Path from Isolation to Therapeutic Action in Eye Diseases. Antioxidants 2022, 11, 2447. https://doi.org/10.3390/antiox11122447
Pop R, Daescu A, Rugina D, Pintea A. Resveratrol: Its Path from Isolation to Therapeutic Action in Eye Diseases. Antioxidants. 2022; 11(12):2447. https://doi.org/10.3390/antiox11122447
Chicago/Turabian StylePop, Roxana, Adela Daescu, Dumitrita Rugina, and Adela Pintea. 2022. "Resveratrol: Its Path from Isolation to Therapeutic Action in Eye Diseases" Antioxidants 11, no. 12: 2447. https://doi.org/10.3390/antiox11122447
APA StylePop, R., Daescu, A., Rugina, D., & Pintea, A. (2022). Resveratrol: Its Path from Isolation to Therapeutic Action in Eye Diseases. Antioxidants, 11(12), 2447. https://doi.org/10.3390/antiox11122447