Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco)
Abstract
:1. Introduction
2. Materials and Methods
2.1. Chemical Reagents
2.2. Cherry Samples
2.3. Preparation of Crude Cherry Extracts
Preparation of Phenolic Concentrated Cherry Extracts
2.4. Fourier Transform Infrared (FT-IR) Spectrometry Analysis
2.5. Cancer Cell Models
HepG2 Culture Conditions and Treatments
2.6. MTT Reduction
2.7. Membrane Integrity Assay
2.8. Cells’ Morphology
2.9. Determination of DNA Damage (Comet Assay)
2.10. Determination of NO Levels
2.11. Nitric Oxide (NO) Scavenging Assay
2.12. Measurement of Intracellular Reactive Oxygen Species
2.13. Preparation of Protein and Ligands
2.14. Receptor-Ligand Docking
2.15. Inhibition of Lipid Peroxidation
2.16. Ferric Reducing-Antioxidant Power Assay
2.17. Statistical Analysis of Results
3. Results and Discussion
3.1. FT-IR Spectroscopy Analysis
3.2. Effect of Sweet Cherry Fractions in HepG2 Cell Viability
3.3. Cell Culture Radical Scavenging Activity
3.4. NO Scavenging Activity
3.5. Rule of Five (Ro5)
3.6. Docking Results
3.7. Protective Effects of Sweet Cherry Fractions against Lipid Peroxidation and Ferric Species
4. Conclusions
Supplementary Materials
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- World Health Organization. Available online: https://www.who.int/news-room/fact-sheets/detail/cancer (accessed on 2 September 2021).
- Malaguarnera, G.; Giordano, M. Serum markers of hepatocellular carcinoma. Dig. Dis. Sci. 2010, 55, 2744–2755. [Google Scholar] [CrossRef]
- Shen, H.; Wang, H.; Wang, L.; Wang, L.; Zhu, M.; Ming, Y.; Zhao, S.; Fan, J.; Lai, E.Y. Ethanol extract of root of Prunus persica inhibited the growth of liver cancer cell HepG2 by inducing cell cycle arrest and migration suppression. Evid. Based Complement. Altern. Med. 2017, 8, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Eduardo, C.; De Souza, A.; Andrade, R.; Riverin, C.; Maria, R.; Maria, S.; Correia, S.; Acco, A. Antineoplastic activity of a novel ruthenium complex against human hepatocellular carcinoma (HepG2) and human cervical adenocarcinoma (HeLa) cells. Heliyon 2020, 6, e03862. [Google Scholar]
- Lin, L.; Yan, L.; Liu, Y.; Qu, C.; Ni, J.; Li, H. The burden and trends of primary liver cancer caused by specific etiologies from 1990 to 2017 at the global, regional, national, age, and sex level results from the global burden of disease study 2017. Liver Cancer 2020, 9, 563–582. [Google Scholar] [CrossRef]
- Frager, S.Z.; Schwartz, J.M. Hepatocellular carcinoma: Epidemiology, screening, and assessment of hepatic reserve. Curr. Oncol. 2020, 27, 138–143. [Google Scholar] [CrossRef]
- Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
- Probst, Y.C.; Guan, V.X.; Kent, K. Dietary phytochemical intake from foods and health outcomes: A systematic review protocol and preliminary scoping. BMJ Open 2017, 7, e013337. [Google Scholar] [CrossRef]
- Duthie, S.J.; Duthie, G.G.; Russell, W.R.; Kyle, J.A.M.; Macdiarmid, J.I.; Rungapamestry, V.; Stephen, S.; Megias, C.; Joanna, B.; Lindsey, J.K.; et al. Effect of increasing fruit and vegetable intake by dietary intervention on nutritional biomarkers and attitudes to dietary change: A randomised trial. Eur. J. Nutr. 2018, 57, 1855–1872. [Google Scholar] [CrossRef] [Green Version]
- Bamia, C.; Lagiou, P.; Jenab, M.; Aleksandrova, K.; Fedirko, V.; Trichopoulos, D.; Overvad, K.; Tjønneland, A.; Olsen, A.; Clavel-Chapelon, F.; et al. Fruit and vegetable consumption in relation to hepatocellular carcinoma in a multi-centre, European cohort study. Br. J. Cancer 2015, 112, 1273–1282. [Google Scholar] [CrossRef] [Green Version]
- Hafeez, B.B.; Siddiqui, I.A.; Asim, M.; Malik, A.; Afaq, F.; Adhami, V.M.; Saleem, M.; Din, M.; Mukhtar, H. A dietary anthocyanidin delphinidin induces apoptosis of human prostate cancer PC3 cells in vitro and in vivo: Involvement of nuclear factor-κB signaling. Cancer Res. 2008, 68, 8564–8572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gonçalves, A.C.; Rodrigues, M.; Santos, A.O.; Alves, G.; Silva, L.R. Antioxidant status, antidiabetic properties and effects on Caco-2 cells of colored and non-colored enriched extracts of sweet cherry fruits. Nutrients 2018, 10, 1688. [Google Scholar] [CrossRef] [Green Version]
- Briguglio, G.; Costa, C.; Pollicino, M.; Giambò, F.; Catania, S.; Fenga, C. Polyphenols in cancer prevention: New insights (Review). Int. J. Funct. Nutr. 2020, 1, 9–20. [Google Scholar] [CrossRef]
- Abu, H.; Khalil-ur, R.; Nazish, J.; Zara, J. The role of polyphenol (flavonoids) compounds in the treatment of cancer. Nutr. Cancer 2020, 72, 386–397. [Google Scholar]
- Acero, N.; Gradillas, A.; Beltran, M.; García, A.; Mingarro, D.M. Comparison of phenolic compounds profile and antioxidant properties of different sweet cherry (Prunus avium L.) varieties. Food Chem. 2019, 279, 260–271. [Google Scholar] [CrossRef] [PubMed]
- Hayaloglu, A.A.; Demir, N. Physicochemical characteristics, antioxidant activity, organic acid and sugar contents of 12 sweet cherry (Prunus avium L.) cultivars grown in Turkey. J. Food Sci. 2015, 80, C564–C570. [Google Scholar] [CrossRef]
- Serradilla, M.J.; Lozano, M.; Bernalte, M.J.; Ayuso, M.C.; López-Corrales, M.; González-Gómez, D. Physicochemical and bioactive properties evolution during ripening of “Ambrunés” sweet cherry cultivar. LWT Food Sci. Technol. 2011, 44, 199–205. [Google Scholar] [CrossRef]
- Skrzyński, J.; Leja, M.; Gonkiewicz, A.; Banach, P. Cultivar effect on the sweet cherry antioxidant and some chemical attributes. Folia Hortic. 2016, 28, 95–102. [Google Scholar] [CrossRef] [Green Version]
- Gonçalves, A.C.; Bento, C.; Silva, B.M.; Silva, L.R. Sweet cherries from Fundão possess antidiabetic potential and protect human erythrocytes against oxidative damage. Food Res. Int. 2017, 95, 91–100. [Google Scholar] [CrossRef] [Green Version]
- Nunes, A.R.; Gonçalves, A.C.; Alves, G.; Garcia-Viguera, C.; Moreno, D.A.; Silva, L.R. Valorisation of Prunus avium L. by-products: Phenolic composition and effect on Caco-2 cells viability. Foods 2021, 10, 1185. [Google Scholar] [CrossRef]
- Gonçalves, A.C.; Bento, C.; Silva, B.; Simões, M.; Silva, L.R. Nutrients, bioactive compounds and bioactivity: The health benefits of sweet cherries (Prunus avium L.). Curr. Nutr. Food Sci. 2019, 15, 208–227. [Google Scholar] [CrossRef]
- Serra, A.T.; Seabra, I.J.; Braga, M.E.M.; Bronze, M.R.; De Sousa, H.C.; Duarte, C.M.M. Processing cherries (Prunus avium) using supercritical fluid technology. Part 1: Recovery of extract fractions rich in bioactive compounds. J. Supercrit. Fluids 2010, 55, 184–191. [Google Scholar] [CrossRef]
- Serra, A.T.; Matias, A.A.; Almeida, A.P.C.; Bronze, M.R.; Alves, P.M.; De Sousa, H.C.; Duarte, C.M.M. Processing cherries (Prunus avium) using supercritical fluid technology. Part 2. Evaluation of SCF extracts as promising natural chemotherapeutical agents. J. Supercrit. Fluids 2011, 55, 1007–1013. [Google Scholar] [CrossRef]
- Serra, A.T.; Duarte, R.O.; Bronze, M.R.; Duarte, C.M.M. Identification of bioactive response in traditional cherries from Portugal. Food Chem. 2011, 125, 318–325. [Google Scholar] [CrossRef]
- Matias, A.A.; Rosado-Ramos, R.; Nunes, S.L.; Figueira, I.; Serra, A.T.; Bronze, M.R.; Santos, C.N.; Duarte, C.M.M. Protective effect of a (poly)phenol-rich extract derived from sweet cherries culls against oxidative cell damage. Molecules 2016, 21, 406. [Google Scholar] [CrossRef]
- Gonçalves, B.; Landbo, A.K.; Let, M.; Silva, A.P.; Rosa, E.; Meyer, A.S. Storage affects the phenolic profiles and antioxidant activities of cherries (Prunus avium L.) on human low-density lipoproteins. J. Sci. Food Agric. 2004, 84, 1013–1020. [Google Scholar] [CrossRef]
- Gonçalves, A.C.; Campos, G.; Alves, G.; Garcia-Viguera, C.; Moreno, D.A.; Silva, L.R. Physical and phytochemical composition of 23 Portuguese sweet cherries as conditioned by variety (or genotype). Food Chem. 2021, 335, 127637. [Google Scholar] [CrossRef]
- Forbes-Hernández, T.Y.; Gasparrini, M.; Afrin, S.; Cianciosi, D.; González-Paramás, A.M.; Santos-Buelga, C.; Mezzetti, B.; Quiles, J.L.; Battino, M.; Giampieri, F.; et al. Strawberry (cv. Romina) methanolic extract and anthocyanin-enriched fraction improve lipid profile and antioxidant status in HepG2 cells. Int. J. Mol. Sci. 2017, 18, 1149. [Google Scholar] [CrossRef] [Green Version]
- Zhou, L.; Zhang, H.; Wu, J. Effects of nitric oxide on the biological behavior of HepG2 human hepatocellular carcinoma cells. Exp. Ther. Med. 2016, 11, 1875–1880. [Google Scholar] [CrossRef] [Green Version]
- Gonçalves, A.C.; Bento, C.; Nunes, A.R.; Simões, M.; Alves, G.; Silva, L.R. Multitarget protection of Pterospartum tridentatum phenolic-rich extracts against a wide range of free radical species, antidiabetic activity and effects on human colon carcinoma (Caco-2) cells. J. Food Sci. 2020, 85, 4377–4388. [Google Scholar] [CrossRef]
- Costa, A.R.; Marcelino, H.; Gonçalves, I.; Quintela, T.; Tomás, J.; Duarte, A.C.; Fonseca, A.M.; Santos, C.R.A. Sex hormones protect against amyloid-β induced oxidative stress in the choroid plexus cell Line Z310. J. Neuroendocrinol. 2016, 28. [Google Scholar] [CrossRef]
- Tulipani, S.; Armeni, T.; Giampieri, F.; Alvarez-Suarez, J.M.; Gonzalez-Paramás, A.M.; Santos-Buelga, C.; Busco, F.; Principato, G.; Bompadre, S.; Quiles, J.L.; et al. Strawberry intake increases blood fluid, erythrocyte and mononuclear cell defenses against oxidative challenge. Food Chem. 2014, 156, 87–93. [Google Scholar] [CrossRef] [PubMed]
- Chiu, Y.; Lo, H.; Huang, H.; Chao, P.; Hwang, J.-M.; Huang, P.-Y.; Huang, S.-J.; Liu, J.-Y.; Lai, T.-J. The antioxidant and cytoprotective activity of Ocimum gratissimum extracts against hydrogen peroxide-induced toxicity in human HepG2 cells. J. Food Drug Anal. 2013, 21, 253–260. [Google Scholar] [CrossRef] [Green Version]
- Jesus, F.; Gonçalves, A.C.; Alves, G.; Silva, L.R. Exploring the phenolic profile, antioxidant, antidiabetic and anti-hemolytic potential of Prunus avium vegetal parts. Food Res. Int. 2018, 116, 600–610. [Google Scholar] [CrossRef]
- Valdés-Tresanco, M.S.; Valdés-Tresanco, M.E.; Valiente, P.A.; Moreno, E. AMDock: A versatile graphical tool for assisting molecular docking with Autodock Vina and Autodock4. Biol. Direct 2020, 15, 1–12. [Google Scholar] [CrossRef]
- Trott, O.; Olson, A.J. AutoDock Vina: Improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J. Comput. Chem. 2010, 31, 455–461. [Google Scholar] [CrossRef] [Green Version]
- Seeliger, D.; de Groot, B.L. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J. Comput. Aided. Mol. Des. 2010, 24, 417–422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bastos, C.; Barros, L.; Dueñas, M.; Calhelha, R.C.; Queiroz, M.J.R.P.; Santos-Buelga, C.; Ferreira, I.C.F.R. Chemical characterisation and bioactive properties of Prunus avium L.: The widely studied fruits and the unexplored stems. Food Chem. 2015, 173, 1045–1053. [Google Scholar] [CrossRef] [Green Version]
- Balanč, B.; Kalušević, A.; Drvenica, I.; Coelho, M.T.; Djordjević, V.; Alves, V.D.; Sousa, I.; Moldão-Martins, M.; Rakić, V.; Nedović, V.; et al. Calcium-alginate-inulin microbeads as carriers for aqueous carqueja extract. J. Food Sci. 2016, 81, E65–E75. [Google Scholar] [CrossRef]
- Benzie, I.F.; Strain, J.J. The ferric reducing ability of plasma (FRAP) as a measure of “antioxidant power”: The FRAP assay. Anal. Biochem. 1996, 239, 70–76. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucarini, M.; Durazzo, A.; Kiefer, J.; Santini, A.; Lombardi-Boccia, G.; Souto, E.B.; Romani, A.; Lampe, A.; Nicoli, S.F.; Gabrielli, P.; et al. Grape seeds: Chromatographic profile of fatty acids and phenolic compounds and qualitative analysis by FTIR-ATR Spectroscopy. Foods 2020, 9, 10. [Google Scholar] [CrossRef] [Green Version]
- Qi, Y.; Liu, J.; Liu, Y.; Yan, D.; Wu, H.; Li, R.; Jiang, Z.; Yang, Y.; Ren, X. Polyphenol oxidase plays a critical role in melanin formation in the fruit skin of persimmon (Diospyros kaki cv. ‘Heishi’). Food Chem. 2020, 330, 127253. [Google Scholar] [CrossRef] [PubMed]
- Pop, R.M.; Bocsan, I.C.; Buzoianu, A.D.; Pecoraro, M.; Popolo, A. Evaluation of the antioxidant activity of Nigella sativa L. and Allium ursinum extracts in a cellular model of doxorubicin-induced cardiotoxicity. Molecules 2020, 25, 5259. [Google Scholar] [CrossRef]
- Okur, İ.; Baltacıoğlu, C.; Ağçam, E.; Baltacıoğlu, H.; Alpas, H. Evaluation of the effect of different extraction techniques on sour cherry pomace phenolic content and antioxidant activity and determination of phenolic compounds by FTIR and HPLC. Waste Biomass Valorization 2019, 10, 3545–3555. [Google Scholar] [CrossRef]
- Umar, A.H.; Ratnadewi, D.; Rafi, M. Untargeted metabolomics analysis using FTIR and UHPLC-Q-Orbitrap HRMS of two Curculigo species and evaluation of their antioxidant and α-glucosidase inhibitory activities. Metabolites 2021, 11, 42. [Google Scholar] [CrossRef]
- Golubtsova, J. Study of fruit raw material by Fourier Transform Infrared spectroscopy. J. Pharm. Sci. Res. 2017, 9, 1081–1090. [Google Scholar]
- Oliveira, R.N.; Mancini, M.C.; Cabral, F.; De Oliveira, S.; Passos, T.M.; Quilty, B.; Thiré, R.M.D.S.M.; McGuinness, G.B. FTIR analysis and quantification of phenols and flavonoids of five commercially available plants extracts used in wound healing. Rev. Matéria 2016, 21, 767–779. [Google Scholar] [CrossRef] [Green Version]
- Kozłowicz, K.; Różyło, R.; Gładyszewska, B.; Matwijczuk, A.; Gładyszewski, G.; Chocyk, D.; Samborska, K.; Piekut6, J.; Smolewska, M. Identification of sugars and phenolic compounds in honey powders with the use of GC–MS, FTIR spectroscopy, and X-ray diffraction. Sci. Rep. 2020, 10, 16269. [Google Scholar] [CrossRef]
- Ayed, L.; Abid, S.B.; Hamdi, M. Development of a beverage from red grape juice fermented with the Kombucha consortium. Ann. Microbiol. 2017, 67, 111–121. [Google Scholar] [CrossRef]
- Cui, Y.; Chang, J.-M.; Wang, W.-L.; Li, B.; Re, X.-Y. Preparation of activated carbon using bio-oil phenol-formaldehyde resin. BioResources 2015, 10, 3865–3873. [Google Scholar] [CrossRef]
- Mohammed, W.H.; Hussein, A.A.; Saleem, M.M.N.M. Influence of antibioticsand stick sweet cherry (Prunus avium) on pathogenic bacteria and evaluation of tissues bioavailability, bioactive phytochemical compound and functional properties. Plant. Arch. 2020, 20, 298–308. [Google Scholar]
- Møller, P.; Loft, S.; Alfthan, G.; Freese, R. Oxidative DNA damage in circulating mononuclear blood cells after ingestion of blackcurrant juice or anthocyanin-rich drink. Fundam. Mol. Mech. Mutagen. 2004, 551, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Ficus, S.; Muhammad, H.; Omar, M.H.; Nurafnie, E.; Rasid, I.; Mohkiar, F.H.; Siu, L.M.; Awang, N. Phytochemical and in vitro genotoxicity studies of standardized Ficus deltoidea var. kunstleri aqueous extract. Plants 2021, 10, 343. [Google Scholar]
- Jung, H.; Kwak, H.-K.; Hwang, K.T. Antioxidant and antiinflammatory activities of cyanidin-3-glucoside and cyanidin-3-rutinoside in hydrogen peroxide and lipopolysaccharide-treated RAW 264.7 cells. Food Sci. Biotechnol. 2014, 23, 2053–2062. [Google Scholar] [CrossRef]
- Xiao, T.; Luo, Z.; Guo, Z.; Wang, X.; Ding, M.; Wang, W.; Shen, X.; Zhao, Y. Multiple roles of black raspberry anthocyanins protecting against alcoholic liver disease. Molecules 2021, 26, 2313. [Google Scholar] [CrossRef]
- Kim, M.J.; Paramanantham, A.; Lee, W.S.; Yun, J.W.; Chang, S.H.; Kim, D.C.; Park, H.S.; Choi, Y.H.; Kim, G.S.; Ryu, C.H.; et al. Anthocyanins derived from Vitis coignetiae Pulliat contributes anti-cancer effects by suppressing NF-κB pathways in Hep3B human hepatocellular carcinoma cells and in vivo. Molecules 2020, 25, 5445. [Google Scholar] [CrossRef]
- Zhou, L.; Wang, H.; Yi, J.; Yang, B.; Li, M.; He, D.; Yang, W.; Zhang, Y.; Ni, H. Anti-tumor properties of anthocyanins from Lonicera caerulea ‘Beilei’ fruit on human hepatocellular carcinoma: In vitro and in vivo study. Biomed. Pharmacother. 2018, 104, 520–529. [Google Scholar] [CrossRef]
- Gonçalves, A.C.; Nunes, A.R.; Falcão, A.; Alves, G.; Silva, L.R. Dietary effects of anthocyanins in human health: A comprehensive review. Pharmaceuticals 2021, 14, 690. [Google Scholar] [CrossRef]
- Hui, C.; Bin, Y.; Xiaoping, Y.; Long, Y.; Chunye, C.; Mantian, M.; Wenhua, L. Anticancer activities of an anthocyanin-rich extract from black rice against breast cancer cells in vitro and in vivo. Nutr. Cancer 2010, 62, 1128–1136. [Google Scholar] [CrossRef]
- Lazzè, M.C.; Savio, M.; Pizzala, R.; Cazzalini, O.; Perucca, P.; Scovassi, A.I.; Stivala, L.A.; Bianchi, L. Anthocyanins induce cell cycle perturbations and apoptosis in different human cell lines. Carcinogenesis 2004, 25, 1427–1433. [Google Scholar] [CrossRef] [PubMed]
- Sousa, C.; Moita, E.; Valentão, P.; Fernandes, F.; Monteiro, P.; Andrade, P.B. Effects of colored and noncolored phenolics of Echium plantagineum L. bee pollen in Caco-2 cells under oxidative stress induced by tert -butyl hydroperoxide. J. Agric. Food Chem. 2015, 63, 2083–2091. [Google Scholar] [CrossRef] [PubMed]
- Fukumoto, L.R.; Mazza, G. Assessing antioxidant and prooxidant activities of phenolic compounds. J. Agric. Food Chem. 2000, 48, 3597–3604. [Google Scholar] [CrossRef] [PubMed]
- Galati, G.; Sabzevari, O.; Wilson, J.X.; O’Brien, P.J. Prooxidant activity and cellular effects of the phenoxyl radicals of dietary flavonoids and other polyphenolics. Toxicology 2002, 177, 91–104. [Google Scholar] [CrossRef]
- Graft-Johnson, J.; Nowak, D. Effect of selected plant phenolics on Fe2+-EDTA-H2O2 system mediated deoxyribose oxidation: Molecular structure-derived relationships of anti- and pro-oxidant actions. Molecules 2017, 22, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cavalluzzi, M.M.; Mangiatordi, G.F.; Nicolotti, O.; Lentini, G. Ligand efficiency metrics in drug discovery: The pros and cons from a practical perspective. Expert Opin. Drug Discov. 2017, 12, 1087–1104. [Google Scholar] [CrossRef]
- Kenny, P.W. The nature of ligand efficiency. J. Cheminformatics 2019, 11, 1–18. [Google Scholar] [CrossRef]
- Lipinski, C.A. Rule of five in 2015 and beyond: Target and ligand structural limitations, ligand chemistry structure and drug discovery project decisions. Adv. Drug Deliv. Rev. 2016, 101, 34–41. [Google Scholar] [CrossRef]
- Krämer, S.D.; Aschmann, H.E.; Hatibovic, M.; Hermann, K.F.; Neuhaus, C.S.; Brunner, C.; Belli, S. When barriers ignore the “rule-of-five”. Adv. Drug Deliv. Rev. 2016, 101, 62–74. [Google Scholar] [CrossRef]
- Hollman, P.C.H.; Cassidy, A.; Comte, B.; Heinonen, M.; Ric, M. The biological relevance of direct antioxidant effects of polyphenols for cardiovascular health in humans is not established. J. Nutr. 2011, 141, 989S–1009S. [Google Scholar] [CrossRef] [Green Version]
- Martini, S.; Conte, A.; Tagliazucchi, D. Bioactivity and cell metabolism of in vitro digested sweet cherry (Prunus avium) phenolic compounds. Int. J. Food Sci. Nutr. 2018, 70, 335–348. [Google Scholar] [CrossRef] [Green Version]
- Bresciani, L.; Martini, D.; Mena, P.; Tassotti, M.; Calani, L.; Brigati, G.; Brighenti, F.; Holasek, S.; Malliga, D.E.; Lamprecht, M.; et al. Absorption profile of (poly)phenolic compounds after consumption of three food supplements containing 36 different fruits, vegetables, and berries. Nutrients 2017, 9, 194. [Google Scholar] [CrossRef]
- Loza-Mejía, M.A.; Salazar, J.R. Sterols and triterpenoids as potential anti-inflammatories: Molecular docking studies for binding to some enzymes involved in inflammatory pathways. J. Mol. Graph. Model. 2015, 62, 18–25. [Google Scholar] [CrossRef] [PubMed]
- Gou, K.J.; Zeng, R.; Ren, X.D.; Dou, Q.L.; Yang, Q.B.; Dong, Y.; Qu, Y. Anti-rheumatoid arthritis effects in adjuvant-induced arthritis in rats and molecular docking studies of Polygonum orientale L. extracts. Immunol. Lett. 2018, 201, 59–69. [Google Scholar] [CrossRef] [PubMed]
- Heim, K.E.; Tagliaferro, A.R.; Bobilya, D.J. Flavonoid antioxidants: Chemistry, metabolism and structure-activity relationships. J. Nutr. Biochem. 2002, 13, 572–584. [Google Scholar] [CrossRef]
- Lecci, R.M.; D’Antuono, I.; Cardinali, A.; Garbetta, A.; Linsalata, V.; Logrieco, A.F.; Leone, A. Antioxidant and pro-oxidant capacities as mechanisms of photoprotection of olive polyphenols on uva-damaged human keratinocytes. Molecules 2021, 26, 2153. [Google Scholar] [CrossRef] [PubMed]
- Parcheta, M.; Świsłocka, R.; Orzechowska, S.; Akimowicz, M.; Choińska, R.; Lewandowski, W. Recent developments in effective antioxidants: The structure and antioxidant properties. Materials 2021, 14, 1984. [Google Scholar] [CrossRef] [PubMed]
- Tural, S.; Koca, I. Physico-chemical and nutritional properties of cornelian cherry fruits (Cornus mas L.) grown in Turkey. Sci. Hortic. 2008, 116, 362–366. [Google Scholar] [CrossRef]
- Martini, S.; Conte, A.; Tagliazucchi, D. Phenolic compounds profile and antioxidant properties of six sweet cherry (Prunus avium) cultivars. Food Res. Int. 2017, 97, 15–26. [Google Scholar] [CrossRef]
Compound | LogP (<5) | H Bond Donor (<5) | H Bond Acceptor (<10) | Violations | Meet Ro5 Criteria | Binding Energy Value ΔG (−kcal/mol) | Estimated Ki (uM) | Ligand Efficiency |
---|---|---|---|---|---|---|---|---|
3-O-Caffeoylquinic acid | −0.83 | 6 | 9 | 1 | Yes | −9.1 | 0.215 | −0.36 |
Caffeic acid | 0.25 | 3 | 4 | 0 | Yes | −6.8 | 10.800 | −0.52 |
Cyanidin 3-O-glucoside | −2.28 | 8 | 11 | 2 | No | −10.1 | 0.038 | −0.32 |
Cyanidin 3-O-rutinoside | −3.24 | 10 | 15 | 3 | No | −11.4 | 0.005 | −0.27 |
Kaempferol 3-O-rutinoside | −0.79 | 9 | 15 | 3 | No | −10.8 | 0.012 | −0.26 |
p-Coumaric acid | 1.02 | 2 | 3 | 0 | Yes | −7.1 | 0.006 | −0.59 |
Quercetin | 0.72 | 5 | 7 | 0 | Yes | −9.4 | 0.128 | −0.43 |
Quercetin 3-O-glucoside | −2.30 | 8 | 12 | 2 | No | −9.4 | 0.128 | −0.28 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Gonçalves, A.C.; Flores-Félix, J.D.; Costa, A.R.; Falcão, A.; Alves, G.; Silva, L.R. Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco). Foods 2021, 10, 2623. https://doi.org/10.3390/foods10112623
Gonçalves AC, Flores-Félix JD, Costa AR, Falcão A, Alves G, Silva LR. Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco). Foods. 2021; 10(11):2623. https://doi.org/10.3390/foods10112623
Chicago/Turabian StyleGonçalves, Ana C., José D. Flores-Félix, Ana R. Costa, Amílcar Falcão, Gilberto Alves, and Luís R. Silva. 2021. "Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco)" Foods 10, no. 11: 2623. https://doi.org/10.3390/foods10112623
APA StyleGonçalves, A. C., Flores-Félix, J. D., Costa, A. R., Falcão, A., Alves, G., & Silva, L. R. (2021). Hepatoprotective Effects of Sweet Cherry Extracts (cv. Saco). Foods, 10(11), 2623. https://doi.org/10.3390/foods10112623