Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review
Abstract
:1. Introduction
2. Classification, Biosynthesis, and Metabolism of Polyphenols
2.1. Classification and Biosynthesis of Polyphenols
2.2. Metabolism of Polyphenols
3. Stilbenes and CVDs
3.1. Mechanism of Action
3.2. Pre-Clinical and Human Clinical Trials with RES Related to CVDs
3.2.1. Pre-Clinical Trials
Vascular Function, Hypertension
Cardiac Function, Remodeling
Other Conditions, Risk Factors
3.2.2. Human Clinical Trials
Hypertension
Vascular Protection
Heart Failure
4. Effect of Relevant Flavonoid Compounds on CVDs
4.1. Mechanism of Action
4.2. Pre-Clinical and Human Clinical Trials with Selected Flavonoids Related to CVDs
4.2.1. Quercetin
Pre-Clinical Trials
- Cardiac function, remodeling
- Vascular function
Human Clinical Trials
- Hypertension
- Myocardial ischemia
4.2.2. Catechins
Pre-Clinical Trials
- Vascular function, hypertension
- Cardiac function, remodeling
Human Clinical Trials
- Hypertension
- Atherosclerosis and myocardial ischemia
- Heart failure
4.2.3. Anthocyanins
Pre-Clinical Trials
- Cardiac function, remodeling
- Vascular function, hypertension
- Atherosclerosis and risk factors
Human Clinical Trials
- Myocardial ischemia
- Hypertension
5. The Association between CV Mortality and Polyphenol Intake
6. Red Wine Consumption and Its Association with CVDs
7. Adverse Effects of the Most Relevant Polyphenols
8. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
References
- Laslett, L.J.; Alagona, P., Jr.; Clark, B.A.; Drozda, J.P., Jr.; Saldivar, F.; Wilson, S.R.; Poe, C.; Hart, M. The worldwide environment of cardiovascular disease: Prevalence, diagnosis, therapy, and policy issues: A report from the American College of Cardiology. J. Am. Coll. Cardiol. 2012, 60, S1–S49. [Google Scholar] [CrossRef] [PubMed]
- Yu, C.; Moore, B.M.; Kotchetkova, I.; Cordina, R.L.; Celermajer, D.S. Causes of death in a contemporary adult congenital heart disease cohort. Heart 2018, 104, 1678–1682. [Google Scholar] [CrossRef] [PubMed]
- Cardiovascular Diseases (CVDs)—World Health Organization. Available online: https://www.who.int/news-room/fact-sheets/detail/cardiovascular-diseases-(cvds) (accessed on 4 September 2023).
- Chen, J.; Zhang, X.; Millican, R.; Sherwood, J.; Martin, S.; Jo, H.; Yoon, Y.-S.; Brott, B.C.; Jun, H.-W. Recent advances in nanomaterials for therapy and diagnosis for atherosclerosis. Adv. Drug Deliv. Rev. 2021, 170, 142–199. [Google Scholar] [CrossRef] [PubMed]
- Banez, M.J.; Geluz, M.I.; Chandra, A.; Hamdan, T.; Biswas, O.S.; Bryan, N.S.; Von Schwarz, E.R. A systemic review on the antioxidant and anti-inflammatory effects of resveratrol, curcumin, and dietary nitric oxide supplementation on human cardiovascular health. Nutr. Res. 2020, 78, 11–26. [Google Scholar] [CrossRef] [PubMed]
- Steven, S.; Frenis, K.; Oelze, M.; Kalinovic, S.; Kuntic, M.; Jimenez, M.T.B.; Vujacic-Mirski, K.; Helmstädter, J.; Kröller-Schön, S.; Münzel, T.; et al. Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. Oxid. Med. Cell. Longev. 2019, 2019, 7092151. [Google Scholar] [CrossRef] [PubMed]
- Finicelli, M.; Di Salle, A.; Galderisi, U.; Peluso, G. The Mediterranean Diet: An Update of the Clinical Trials. Nutrients 2022, 14, 2956. [Google Scholar] [CrossRef] [PubMed]
- Martínez-González, M.A.; Gea, A.; Ruiz-Canela, M. The Mediterranean Diet and Cardiovascular Health. Circ. Res. 2019, 124, 779–798. [Google Scholar] [CrossRef] [PubMed]
- Mozaffarian, D. Dietary and policy priorities for cardiovascular disease, diabetes, and obesity: A comprehensive review. Circulation 2016, 133, 187–225. [Google Scholar] [CrossRef]
- Silveira, B.K.S.; Oliveira, T.M.S.; Andrade, P.A.; Hermsdorff, H.H.M.; Rosa, C.O.B.; Franceschini, S.D.C.C. Dietary pattern and macronutrients profile on the variation of inflammatory biomarkers: Scientific Update. Cardiol. Res. Pract. 2018, 2018, 4762575. [Google Scholar]
- Visseren, F.L.J.; Mach, F.; Smulders, Y.M.; Carballo, D.; Koskinas, K.C. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef]
- Willett, W.C.; Sacks, F.; Trichopoulou, A.; Drescher, G.; Ferro-Luzzi, A.; Helsing, E.; Trichopoulos, D. Mediterranean diet pyramid: A cultural model for healthy eating. Am. J. Clin. Nutr. 1995, 61, 1402S–1406S. [Google Scholar] [CrossRef] [PubMed]
- Renaud, S.; De Lorgeril, M. Wine, alcohol, platelets, and the French paradox for coronary heart disease. Lancet 1992, 339, 1523–1526. [Google Scholar] [CrossRef] [PubMed]
- Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem. Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef] [PubMed]
- Hooper, L.; Kroon, P.A.; Rimm, E.B.; Cohn, J.S.; Harvey, I.; Le Cornu, K.A.; Ryder, J.J.; Hall, W.L.; Cassidy, A. Flavonoids, flavonoid-rich foods, and cardiovascular risk: A meta-analysis of randomized controlled trials. Am. J. Clin. Nutr. 2008, 88, 38–50. [Google Scholar] [CrossRef] [PubMed]
- Tungmunnithum, D.; Thongboonyou, A.; Pholboon, A.; Yangsabai, A. Flavonoids and Other Phenolic Compounds from Medicinal Plants for Pharmaceutical and Medical Aspects: An Overview. Medicines 2018, 5, 93. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.; Daniel, K.G.; Kuhn, D.J.; Kazi, A.; Bhuiyan, M.; Li, L.; Dou, Q.P. Green tea and tea polyphenols in cancer prevention. Front. Biosci. 2004, 9, 2618. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.L.; Fan, M.X.; Wu, J.L.; Li, N.; Guo, M.Q. Antioxidant and anti-inflammatory properties of flavonoids from lotus plumule. Food Chem. 2019, 277, 706–712. [Google Scholar] [CrossRef] [PubMed]
- Sarker, U.; Oba, S. Polyphenol and flavonoid profiles and radical scavenging activity in leafy vegetable Amaranthus gangeticus. BMC Plant Biol. 2020, 20, 499. [Google Scholar] [CrossRef]
- Martillanes, S.; Rocha-Pimienta, J.; Cabrera-Bañegil, M.; Martín-Vertedor, D.; Delgado-Adámez, J. Application of Phenolic Compounds for Food Preservation: Food Additive and Active Packaging. In Phenolic Compounds-Biological Activity; BoD–Books on Demand: London, UK, 2017. [Google Scholar] [CrossRef]
- Maqsood, S.; Benjakul, S.; Shahidi, F. Emerging Role of Phenolic Compounds as Natural Food Additives in Fish and Fish Products. Crit. Rev. Food Sci. Nutr. 2013, 53, 162–179. [Google Scholar] [CrossRef]
- Andriantsitohaina, R.; Auger, C.; Chataigneau, T.; Étenne-Selloum, N.; Li, H.; Martínez, M.C.; Schini-Kerth, V.B.; Laher, I. Molecular Mechanisms of the Cardiovascular Protective Effects of Polyphenols. Br. J. Nutr. 2012, 108, 1532–1549. [Google Scholar] [CrossRef]
- Gal, R.; Praksch, D.; Kenyeres, P.; Rabai, M.; Toth, K.; Halmosi, R.; Habon, T. Hemorheological Alterations in Patients with Heart Failure with Reduced Ejection Fraction Treated by Resveratrol. Cardiovasc. Ther. 2020, 2020, 7262474. [Google Scholar] [CrossRef] [PubMed]
- Castaldo, L.; Narváez, A.; Izzo, L.; Graziani, G.; Gaspari, A.; Minno, G.; Ritieni, A. Red Wine Consumption and Cardiovascular Health. Molecules 2019, 24, 3626. [Google Scholar] [CrossRef] [PubMed]
- Fraga, C.G.; Croft, K.D.; Kennedye, D.O.; Tomás-Barberán, F.A. The effects of polyphenols and other bioactives on human health. Food Funct. 2019, 10, 514–528. [Google Scholar] [CrossRef] [PubMed]
- Recio, M.; Andujar, I.; Rios, J. Anti-Inflammatory Agents from Plants: Progress and Potential. Curr. Med. Chem. 2012, 19, 2088–2103. [Google Scholar] [CrossRef] [PubMed]
- Magyar, K.; Halmosi, R.; Palfi, A.; Feher, G.; Czopf, L.; Fulop, A.; Battyany, I.; Sumegi, B.; Toth, K.; Szabados, E. Cardioprotection by resveratrol: A human clinical trial in patients with stable coronary artery disease. Clin. Hemorheol. Microcirc. 2012, 50, 179–187. [Google Scholar] [CrossRef] [PubMed]
- Riba, A.; Deres, L.; Sumegi, B.; Toth, K.; Szabados, E.; Halmosi, R. Cardioprotective Effect of Resveratrol in a Postinfarction Heart Failure Model. Oxid. Med. Cell. Longev. 2017, 2017, 6819281. [Google Scholar] [CrossRef] [PubMed]
- Li, A.N.; Li, S.; Zhang, Y.J.; Xu, X.R.; Chen, Y.M.; Li, H. Resources and biological activities of natural polyphenols. Nutrients 2014, 6, 6020–6047. [Google Scholar] [CrossRef]
- Rana, A.; Samtiya, M.; Dhewa, T.; Mishra, V.; Aluko, R.E. Health benefits of polyphenols: A concise review. J. Food Biochem. 2022, 46, e14264. [Google Scholar] [CrossRef]
- Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Remesy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230s–242s. [Google Scholar] [CrossRef]
- Bešlo, D.; Došlić, G.; Agić, D.; Rastija, V.; Šperanda, M.; Gantner, V.; Lučić, B. Polyphenols in Ruminant Nutrition and Their Effects on Reproduction. Antioxidants 2022, 11, 970. [Google Scholar] [CrossRef]
- Zhang, Y.; Butelli, E.; Alseekh, S.; Tohge, T.; Rallapalli, G.; Luo, J.; Kawar, P.G. Multi-level engineering facilitates the production of phenylpropanoid compounds in tomato. Nat. Commun. 2015, 6, 8635. [Google Scholar] [CrossRef] [PubMed]
- Dubrovina, A.S.; Kiselev, K.V. Regulation of stilbene biosynthesis in plants. Planta 2017, 246, 597–623. [Google Scholar] [CrossRef] [PubMed]
- El Khawand, T.; Courtois, A.; Valls, J.; Richard, T.; Krisa, S. A review of dietary stilbenes: Sources and bioavailability. Phytochem. Rev. 2018, 17, 1007–1029. [Google Scholar] [CrossRef]
- Pandey, K.B.; Rizv, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
- Dretcanu, G.; Stirbu, I.; Leopold, N.; Cruceriu, D.; Danciu, C.; Stănilă, A. Chemical Structure, Sources and Role of Bioactive Flavonoids in Cancer Prevention: A Review. Plants 2022, 11, 1117. [Google Scholar] [CrossRef] [PubMed]
- Santos, E.; Maia, B.; Ferriani, A.; Teixeira, S. Flavonoids: Classification, Biosynthesis and Chemical Ecology; InTechOpen: London, UK, 2017. [Google Scholar] [CrossRef]
- Tsao, R.; Deng, D. Separation procedures for naturally occurring antioxidant hytochemicals. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2004, 812, 85–99. [Google Scholar] [CrossRef] [PubMed]
- Sevgi, K.; Tepe, B.; Sarikurkcu, C. Antioxidant and DNA damage protection potentials of selected phenolic acids. Food Chem. Toxicol. 2015, 77, 12–21. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.H.; Tsao, R.; Yang, R.; Cui, S.W. Phenolic acid profiles and antioxidant activities of wheat bran extracts and the effect of hydrolysis conditions. Food Chem. 2006, 95, 466–473. [Google Scholar] [CrossRef]
- Durazzo, A. Lignans. In Phenolic Compounds in Food: Characterization and Analysis (Food Analysis and Properties); Leo, M.L.N., Ed.; CRC Press: Boca Raton, FL, USA, 2018. [Google Scholar]
- Lewis, N.G.; Davin, L.B. Lignans: Biosynthesis and function. In Comprehensive Natural Products Chemistry; Barton, D., Nakanishi, K., Meth-Cohn, O., Eds.; Elsevier: Amsterdam, The Netherlands, 1999; pp. 639–712. [Google Scholar]
- D’Archivio, M.; Filesi, C.; Di Benedetto, R.; Gargiulo, R.; Giovannini, C.; Masella, R. Polyphenols, dietary sources and bioavailability. Ann. dell’Istituto Super. Sanita 2007, 43, 348–361. [Google Scholar]
- Gambini, J.; Inglés, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Abdelaziz, K.M. Properties of resveratrol: In vitro and in vivo studies about metabolism, bioavailability, and biological effects in animal models and humans. Oxid. Med. Cell. Longev. 2015, 2015, 837042. [Google Scholar] [CrossRef]
- Scalbert, A.; Williamson, G. Dietary Intake and Bioavailability of Polyphenols. J. Nutr. 2000, 130, 2073S–2085S. [Google Scholar] [CrossRef] [PubMed]
- De-Souza, J.E.; Casanova, L.M.; Costa, S.S. Bioavailability of phenolic compounds: A major challenge for drug development? Rev. Fitos 2015, 9, 55–67. [Google Scholar]
- Lippolis, T.; Cofano, M.; Caponio, G.R.; De Nunzio, V.; Notarnicola, M. Bioaccessibility and bioavailability of diet polyphenols and their modulation of gut microbiota. Int. J. Mol. Sci. 2023, 24, 3813. [Google Scholar] [CrossRef] [PubMed]
- Rein, M.J.; Renouf, M.; Cruz-Hernandez, C.; Actis-Goretta, L.; Thakkar, S.K.; da Silva Pinto, M. Bioavailability of bioactive food compounds: A challenging journey to bioefficacy. Br. J. Clin. Pharmacol. 2013, 75, 588–602. [Google Scholar] [CrossRef] [PubMed]
- Stevens, J.F.; Maier, C.S. The chemistry of gut microbial metabolism of polyphenols. Phytochem. Rev. 2016, 15, 425–444. [Google Scholar] [CrossRef] [PubMed]
- Montané, X.; Kowalczyk, O.; Reig-Vano, B.; Bajek, A.; Roszkowski, K.; Tomczyk, R. Current perspectives of the applications of polyphenols and flavonoids in cancer therapy. Molecules 2020, 25, 3342. [Google Scholar] [CrossRef] [PubMed]
- Kim, D.-H.; Jung, E.-A.; Sohng, I.-S.; Han, J.-A.; Kim, T.-H.; Han, M.J. Intestinal bacterial metabolism of flavonoids and its relation to some biological activities. Arch. Pharmacol. Res. 1998, 21, 17. [Google Scholar] [CrossRef] [PubMed]
- Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
- Patel, K.R.; Brown, V.A.; Jones, D.J.; Britton, R.G.; Hemingway, D.; Miller, A.S.; West, K.P.; Booth, T.D.; Perloff, M.; Crowell, J.A.; et al. Clinical pharmacology of resveratrol and its metabolites in colorectal cancer patients. Cancer Res. 2010, 70, 7392–7399. [Google Scholar] [CrossRef]
- Walle, T. Methylation of Dietary Flavones Increases Their Metabolic Stability and Chemopreventive Effects. Int. J. Mol. Sci. 2009, 10, 5002–5019. [Google Scholar] [CrossRef]
- Zeng, X.; Su, W.; Bai, Y.; Chen, T.; Yan, Z.; Wang, J.; Su, M.; Zheng, Y.; Peng, W.; Hao, H. Urinary metabolite profiling of flavonoids in Chinese volunteers after consumption of orange juice by UFLC-Q-TOF-MS/MS. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2017, 1061–1062, 79–88. [Google Scholar] [CrossRef] [PubMed]
- Borges, G.; Ottaviani, J.I.; van der Hooft, J.J.J.; Schroeter, H.; Crozier, A. Absorption, metabolism, distribution and excretion of (−)-epicatechin: A review of recent findings. Mol. Asp. Med. 2018, 61, 18–30. [Google Scholar] [CrossRef] [PubMed]
- Tung, W.C.; Rizzo, B.; Dabbagh, Y.; Saraswat, S.; Romanczyk, M.; Codorniu-Hernández, E.; Rebollido-Rios, R. Polyphenols bind to low density lipoprotein at biologically relevant concentrations that are protective for heart disease. Arch. Biochem. Biophys. 2020, 694, 108589. [Google Scholar] [CrossRef]
- Sergides, C.; Chirilă, M.; Silvestro, L.; Pitta, D.; Pittas, A. Bioavailability and safety study of resveratrol 500 mg tablets in healthy male and female volunteers. Exp. Ther. Med. 2016, 11, 164–170. [Google Scholar] [CrossRef] [PubMed]
- Alldritt, I.; Whitham-Agut, B.; Sipin, M.; Studholme, J.; Trentacoste, A.; Tripp, J.A. Metabolomics reveals diet-derived plant polyphenols accumulate in physiological bone. Sci. Rep. 2019, 9, 8047. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Shen, X.; Mi, L.; Jing, J.; Gai, S. Simultaneous determinations of four major bioactive components in Acacia catechu (L.f.) Willd and Scutellaria baicalensis Georgi extracts by LC-MS/MS: Application to its herb-herb interactions based on pharmacokinetic, tissue distribution and excretion studies in rats. Phytomedicine 2019, 56, 64–73. [Google Scholar] [PubMed]
- Böhmdorfer, M.; Szakmary, A.; Schiestl, R.H.; Vaquero, J.; Riha, J.; Brenner, S.; Thalhammer, T.; Szekeres, T.; Jäger, W. Involvement of UDP-glucuronyltransferases and Sulfotransferases in the Excretion and Tissue Distribution of Resveratrol in Mice. Nutrients 2017, 9, 1347. [Google Scholar] [CrossRef] [PubMed]
- Leonard, S.S.; Xia, C.; Jiang, B.; Stinefelt, B.; Klandorf, H.; Harris, G.K.; Shi, X. Resveratrol scavenges reactive oxygen species and effects radical-induced cellular responses. Biochem. Biophys. Res. Commun. 2003, 309, 1017–1026. [Google Scholar] [CrossRef]
- Zamora-Ros, R.; Andres-Lacueva, C.; Lamuela-Raventós, R.; Berenguer, T.; Jakszyn, P.; Martínez, C.; Sánchez, M.J. 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]
- Li, H.; Horke, S.; Forstermann, U. Vascular oxidative stress, nitric oxide and atherosclerosis. Atherosclerosis 2014, 237, 208–219. [Google Scholar] [CrossRef]
- Gal, R.; Deres, L.; Horvath, O.; Eros, K.; Sandor, B.; Urban, P.; Soos, S.; Marton, Z.; Sumegi, B.; Toth, K.; et al. Resveratrol Improves Heart Function by Moderating Inflammatory Processes in Patients with Systolic Heart Failure. Antioxidants 2020, 9, 1108. [Google Scholar] [CrossRef] [PubMed]
- Su, M.; Dong, D.; Wan, J.; Zhou, M. Pharmacokinetics, tissue distribution and excretion study of trans-resveratrol-3-O-glucoside and its two metabolites in rats. Phytomedicine 2019, 58, 152882. [Google Scholar] [CrossRef] [PubMed]
- Jia, Z.; Zhu, H.; Misra, B.R.; Mahaney, J.E.; Li, Y.; Misra, H.P. EPR studies on the superoxide-scavenging capacity of the nutraceutical resveratrol. Mol. Cell. Biochem. 2008, 313, 187–194. [Google Scholar] [CrossRef]
- Holthoff, J.H.; Woodling, K.A.; Doerge, D.R.; Burns, S.T.; Hinson, J.A.; Mayeux, P.R. Resveratrol, a dietary polyphenolic phytoalexin, is a functional scavenger of peroxynitrite. Biochem. Pharmacol. 2010, 80, 1260–1265. [Google Scholar] [CrossRef] [PubMed]
- Mahal, H.S.; Mukherjee, T. Scavenging of reactive oxygen radicals by Resveratrol: Antioxidant effect. Res. Chem. Intermed. 2006, 32, 59–71. [Google Scholar] [CrossRef]
- Wang, J.; He, D.; Zhang, Q.; Han, Y.; Jin, S.; Qi, F. Resveratrol protects against Cisplatin-induced cardiotoxicity by alleviating oxidative damage. Cancer Biother. Radiopharm. 2009, 24, 675–680. [Google Scholar] [CrossRef] [PubMed]
- Ungvari, Z.; Orosz, Z.; Rivera, A.; Labinskyy, N.; Xiangmin, Z.; Olson, S.; Podlutsky, A.; Csiszar, A. Resveratrol increases vascular oxidative stress resistance. Am. J. Physiol. Heart Circ. Physiol. 2007, 292, H2417–H2424. [Google Scholar] [CrossRef] [PubMed]
- Hosoda, R.; Hamada, H.; Uesugi, D.; Iwahara, N.; Nojima, I.; Horio, Y.; Kuno, A. Different Antioxidative and Antiapoptotic Effects of Piceatannol and Resveratrol. J. Pharmacol. Exp. Ther. 2021, 376, 385–396. [Google Scholar] [CrossRef]
- Les, F.; Deleruyelle, S.; Cassagnes, L.-E.; Boutin, J.A.; Balogh, B.; Arbones-Mainar, J.M.; Biron, S.; Marceau, P.; Richard, D.; Nepveu, F.; et al. Piceatannol and resveratrol share inhibitory effects on hydrogen peroxide release, monoamine oxidase and lipogenic activities in adipose tissue, but differ in their antilipolytic properties. Chem. Biol. Interact. 2016, 258, 115–125. [Google Scholar] [CrossRef]
- Murias, M.; Jager, W.; Handler, N.; Erker, T.; Horvath, Z.; Szekeres, Z.; Nohl, H.; Gille, L. Antioxidant, prooxidant and cytotoxic activity of hydroxylated resveratrol analogues: Structure-activity relationship. Biochem. Pharmacol. 2005, 69, 903–912. [Google Scholar] [CrossRef]
- Yokozawa, T.; Kim, Y.J. Piceatannol inhibits melanogenesis by its antioxidative actions. Biol. Pharm. Bull. 2007, 30, 2007–2011. [Google Scholar] [CrossRef] [PubMed]
- Xia, N.; Forstermann, U.; Li, H. Resveratrol as a gene regulator in the vasculature. Curr. Pharm. Biotechnol. 2014, 15, 401–408. [Google Scholar] [CrossRef] [PubMed]
- Ungvari, Z.; Labinskyy, N.; Mukhopadhyay, P.; Pinto, J.T.; Bagi, Z.; Ballabh, P.; Zhang, C.; Pacher, P.; Csiszar, A.; Tarantini, S.; et al. Resveratrol attenuates mitochondrial oxidative stress in coronary arterial endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H1876–H1881. [Google Scholar] [CrossRef] [PubMed]
- Xia, N.; Daiber, A.; Förstermann, U.; Li, H. Antioxidant effects of resveratrol in the cardiovascular system. Br. J. Pharmacol. 2017, 174, 1633–1646. [Google Scholar] [CrossRef] [PubMed]
- Bagul, B.K.; Deepthi, N.; Sultana, R.; Banerjee, S.K. Resveratrol ameliorates cardiac oxidative stress in diabetes through deacetylation of NFkB-p65 and histone 3. J. Nutr. Biochem. 2015, 26, 1298–1307. [Google Scholar] [CrossRef] [PubMed]
- Spanier, G.; Xu, H.; Xia, N.; Tobias, S.; Deng, S.; Wojnowski, L.; Forstermann, U.; Li, H. Resveratrol reduces endothelial oxidative stress by modulating the gene expression of superoxide dismutase 1 (SOD1), glutathione peroxidase 1 (GPx1) and NADPH oxidase subunit (Nox4). J. Physiol. Pharmacol. 2009, 60, 111–116. [Google Scholar] [PubMed]
- Hsu, C.P.; Zhai, P.; Yamamoto, T.; Maejima, Y.; Matsushima, S.; Hariharan, N.; Shao, D.; Takagi, H.; Oka, S.; Sadoshimaet, J. Silent information regulator 1 protects the heart from ischemia/reperfusion. Circulation 2010, 122, 2170–2182. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.M.; Chen, H.; Wan, X.; Tania, M.; Xu, A.; Chen, F.; Zhang, D. Regulatory effects of resveratrol on antioxidant enzymes: A mechanism of growth inhibition and apoptosis induction in cancer cells. Mol. Cells 2013, 35, 219–225. [Google Scholar] [CrossRef]
- Li, H.; Forstermann, U. Pharmacological Prevention of eNOS Uncoupling. Curr. Pharm. Des. 2014, 20, 3595–3606. [Google Scholar] [CrossRef]
- Xia, N.; Daiber, A.; Habermeier, A.; Closs, E.I.; Thum, T.; Spanier, G.; Lu, Q.; Oelze, M.; Torzewski, M.; Lackner, K.J.; et al. Resveratrol reverses endothelial nitric-oxide synthase uncoupling in apolipoprotein E knockout mice. J. Pharmacol. Exp. Ther. 2010, 335, 149–154. [Google Scholar] [CrossRef]
- Li, S.; Zlia, G.; Chen, L.; Ding, Y.; Lian, J.; Hong, G.; Lu, Z. Resveratrol protects mice from paraquat-induced lung injury: The important role of SIRT1 and NRF2 antioxidant pathways. Mol. Med. Rep. 2016, 13, 1833–1838. [Google Scholar] [CrossRef] [PubMed]
- Meng, Q.; Guo, T.; Li, G.; Sun, S.; He, S.; Cheng, B.; Shi, B.; Shan, A. Dietary resveratrol improves antioxidant status of sows and piglets and regulates antioxidant gene expression in placenta by Keap1-Nrf2 pathway and Sirt1. J. Anim. Sci. Biotechnol. 2018, 9, 34. [Google Scholar] [CrossRef] [PubMed]
- Malaguarnera, L. Influence of resveratrol on the immune response. Nutrients 2019, 11, 946. [Google Scholar] [CrossRef] [PubMed]
- Dvorakova, M.; Landa, P. Anti-inflammatory activity of natural stilbenoids: A review. Pharmacol. Res. 2017, 124, 126–145. [Google Scholar] [CrossRef] [PubMed]
- Marko, L.; Vigolo, E.; Hinze, C.; Park, J.K.; Roel, G.; Balogh, A.; Choi, M.; Wubken, A.; Cording, J.; Blasig, I.E.; et al. Tubular Epithelial NF-kappaB Activity Regulates Ischemic AKI. J. Am. Soc. Nephrol. 2016, 27, 2658–2669. [Google Scholar] [CrossRef] [PubMed]
- Severgnini, M.; Takahashi, S.; Rozo, L.M.; Homer, R.J.; Kuhn, C.; Jhung, J.W.; Perides, G.; Steer, M.; Hassoun, P.M.; Fanburg, B.L.; et al. Activation of the STAT pathway in acute lung injury. Am. J. Physiol. Lung Cell. Mol. Physiol. 2004, 286, L1282–L1292. [Google Scholar] [CrossRef] [PubMed]
- Csiszar, A. Anti-inflammatory effects of resveratrol: Possible role in prevention of age-related cardiovascular disease. Ann. N. Y. Acad. Sci. 2011, 1215, 117–122. [Google Scholar] [CrossRef] [PubMed]
- Wang, M.; Weng, X.; Chen, H.; Chen, Z.; Liu, X. Resveratrol inhibits TNF-α-induced inflammation to protect against renal ischemia/reperfusion injury in diabetic rats. Acta Cir. Bras. 2020, 35, e202000506. [Google Scholar] [CrossRef]
- Ma, C.; Wang, Y.; Dong, L.; Li, M.; Cai, W. Anti-inflammatory effect of resveratrol through the suppression of NF-kB and JAK/STAT signaling pathways. Acta Bioch. Bioph. Sin. 2015, 47, 207–213. [Google Scholar] [CrossRef]
- Pinheiro, D.; de Oliveira, A.; Coutinho, L.G.; Fontes, F.L.; de Medeiros, O.R.; Oliveira, T.T.; Faustino, A.; Lira, D.S.V.; de Melo, C.J.; Lajus, T.; et al. Resveratrol decreases the expression of genes involved in inflammation through transcriptional regulation. Free Radic. Biol. Med. 2019, 130, 8–22. [Google Scholar] [CrossRef]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-κB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [PubMed]
- Jin, C.Y.; Moon, D.O.; Lee, K.J.; Kim, M.O.; Lee, J.D.; Choi, Y.H.; Park, Y.M.; Kim, G.Y. Piceatannol attenuates lipopolysaccharide-induced NF-κB activation and NF-κB -related proinflammatory mediators in BV2 microglia. Pharmacol. Res. 2006, 54, 461–467. [Google Scholar] [CrossRef] [PubMed]
- Su, L.; David, M. Distinct mechanisms of STAT phosphorylation via the interferon-a/b receptor. Selective inhibition of STAT3 and STAT5 by piceatannol. J. Biol. Chem. 2000, 275, 12661–12666. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Li, S.; Yang, Q.; Shi, Y.; Zheng, M.; Liu, Y.; Chen, F.; Song, G.; Xu, H.; Wan, T.; et al. Resveratrol reduces the proinflammatory effects and lipopolysaccharide- induced expression of HMGB1 and TLR4 in RAW264.7 cells. Cell. Physiol. Biochem. 2014, 33, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
- Narayanankutty, A. Toll like receptors as a novel therapeutic target for natural products against chronic diseases. Curr. Drug Target. 2019, 20, 1068–1080. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, H.; Tang, W.; Qiu, Q.; Peng, J. Resveratrol prevents TNF- α-induced VCAM-1 and ICAM-1 upregulation in endothelial progenitor cells via reduction of NF- κB activation. Int. Med. Res. 2020, 48, 300060520945131. [Google Scholar] [CrossRef] [PubMed]
- Seo, Y.; Park, J.; Choi, W.; Ju Son, D.; Sung Kim, Y.; Kim, M.; Yoon, B.; Pyee, J.; Tae Hong, J.; Go, Y.; et al. Antiatherogenic effect of resveratrol attributed to decreased expression of ICAM-1 (intercellular adhesion molecule-1). Arterioscler. Thromb. Vasc. Biol. 2019, 39, 675–684. [Google Scholar] [CrossRef] [PubMed]
- Palacz-Wrobel, M.; Borkowska, P.; Paul-Samojedny, M.; Kowalczyk, M.; Fila-Danilow, A.; Suchanek-Raif, R.; Kowalski, J. Effect of apigenin, kaempferol and resveratrol on the gene expression and protein secretion of tumor necrosis factor alpha (TNF-α) and interleukin-10 (IL-10) in RAW-264.7 macrophages. Biomed. Pharmacother. 2017, 93, 1205–1212. [Google Scholar] [CrossRef]
- Son, Y.; Chung, H.; Pae, H. Differential effects of resveratrol and its natural analogues, piceatannol and 3,5,40-trans-trimethoxystilbene, on anti-inflammatory heme oxigenase-1 expression in RAW264.7 macrophages. BioFactors 2014, 40, 138–145. [Google Scholar] [CrossRef]
- Jeong, S.O.; Son, Y.; Lee, J.H.; Cheong, Y.K.; Park, S.H.; Chung, T.H.; Pae, H.O. Resveratrol analogue piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. Mol. Med. Rep. 2015, 12, 937–944. [Google Scholar] [CrossRef]
- Szewczuk, L.M.; Forti, L.; Stivala, L.A.; Penning, T.M. Resveratrol is a peroxidase mediated inactivator of COX-1 but not COX-2: A mechanistic approach to the design of COX-1 selective agents. J. Biol. Chem. 2004, 279, 22727–22737. [Google Scholar] [CrossRef] [PubMed]
- Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef] [PubMed]
- Yang, C.M.; Chen, Y.W.; Chi, P.L.; Lin, C.C.; Hsiao, L.D. Resveratrol inhibits BK-induced COX-2 transcription by suppressing acetylation of AP-1 and NF-κB in human rheumatoid arthritis synovial fibroblasts. Biochem. Pharmacol. 2017, 132, 77–91. [Google Scholar] [CrossRef] [PubMed]
- Matsui, T.; Tao, J.; del Monte, F.; Lee, K.H.; Li, L.; Picard, M.; Force, T.L.; Franke, T.F.; Hajjar, R.J.; Rosenzweig, A. Akt activation preserves cardiac function and prevents injury after transient cardiac ischemia in vivo. Circulation 2001, 104, 330–335. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, S.; Murphy, A.N.; Brown, J.H. Akt mediated mitochondrial protection in the heart: Metabolic and survival pathways to the rescue. J. Bioenerg. Biomemb. 2009, 41, 169–180. [Google Scholar] [CrossRef] [PubMed]
- Muslin, A.J. MAPK signalling in cardiovascular health and disease: Molecular mechanisms and therapeutic targets. Clin. Sci. (Lond.) 2008, 115, 203–218. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.K.; Vinayak, M. Resveratrol alleviates inflammatory hyperalgesia by modulation of reactive oxygen species (ROS), antioxidant enzymes and ERK activation. Inflamm. Res. 2017, 66, 911–921. [Google Scholar] [CrossRef] [PubMed]
- Kondohand, K.; Nishida, E. Regulation of MAPkinases by MAP kinase phosphatases. Biochim. Biophys. Acta Mol. Cell Res. 2007, 1773, 1227–1237. [Google Scholar] [CrossRef]
- Ma, E.; Wu, C.; Chen, J.; Wo, D.; Ren, D.; Yan, H.; Peng, L.; Zhu, W. Resveratrol prevents Ang II-induced cardiac hypertrophy by inhibition of NF-κB signaling. Biomed. Pharmacother. 2023, 165, 115275. [Google Scholar] [CrossRef]
- Song, Y.; Huang, W.; Zheng, J.; Yan, M.; Cheng, R.; Zhao, L.; Chen, C.; Hu, C.; Jia, W. Resveratrol reduces intracellular reactive oxygen species levels by inducing autophagy through the AMPK-mTOR pathway. Front. Med. 2018, 12, 697–706. [Google Scholar] [CrossRef]
- Sanches-Silva, A.; Testai, L.; Nabavi, S.F.; Battino, M.; Pandima Devi, K.P.; Tejada, S.; Sureda, A.; Xu, S.; Yousefi, B.; Majidinia, M.; et al. Therapeutic potential of polyphenols in cardiovascular diseases: Regulation of mTOR signaling pathway. Pharmacol. Res. 2020, 152, 104626. [Google Scholar] [CrossRef] [PubMed]
- Hosoda, R.; Nakashima, R.; Yano, M.; Iwahara, N.; Asakura, S.; Nojima, I.; Saga, Y.; Kunimoto, R.; Horio, Y.; Kuno, A. Resveratrol, a SIRT1 activator, attenuates aging-associated alterations in skeletal muscle and heart in mice. J. Pharmacol. Sci. 2023, 152, 112–122. [Google Scholar] [CrossRef] [PubMed]
- Travers, J.G.; Kamal, F.A.; Robbins, J.; Yutzey, K.E.; Blaxall, B.C. Cardiac Fibrosis: The Fibroblast Awakens. Circ. Res. 2016, 118, 1021–1040. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.H.; Zhang, Y.; Wang, X.; Fan, X.F.; Zhang, Y.; Li, X.; Gong, Y.S.; Han, L.P. SIRT1 activation attenuates cardiac fibrosis by endothelial-to-mesenchymal transition. Biomed. Pharmacother. 2019, 118, 109227. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Lu, Y.; Ong’achwa, M.J.; Ge, L.; Qian, Y.; Chen, L.; Hu, X.; Li, F.; Wei, H.; Zhang, C.; et al. Resveratrol Inhibits the TGF-β1-Induced Proliferation of Cardiac Fibroblasts and Collagen Secretion by Downregulating miR-17 in Rat. BioMed. Res. Int. 2018, 2018, 8730593. [Google Scholar] [CrossRef] [PubMed]
- Dolinsky, V.W.; Soltys, C.L.M.; Rogan, K.R.; Chan, A.Y.M.; Nagendran, J.; Wang, S.; Dyck, J.R.B. Resveratrol prevents pathological but not physiological cardiac hypertrophy. J. Mol. Med. (Berl.) 2015, 93, 413–425. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Vanhoutte, P.M.; Leung, S.W. Vascular nitric oxide: Beyond eNOS. J. Pharmacol. Sci. 2015, 129, 83–94. [Google Scholar] [CrossRef] [PubMed]
- Forstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef]
- Parsamanesh, N.; Asghari, A.; Sardari, S.; Tasbandi, A.; Jamialahmadi, T.; Xu, S.; Sahebkar, A. Resveratrol and endothelial function: A literature review. Pharmacol. Res. 2021, 170, 105725. [Google Scholar] [CrossRef]
- Xia, N.; Forstermann, U.; Li, H. Resveratrol and endothelial nitric oxide. Molecules 2014, 19, 16102–16121. [Google Scholar] [CrossRef]
- Carrizzo, A.; Puca, A.; Damato, A.; Marino, M.; Franco, E.; Pompeo, F.; Traficante, A.; Civitillo, F.; Santini, L.; Trimarco, V.; et al. Resveratrol improves vascular function in patients with hypertension and dyslipidemia by modulating NO metabolism. Hypertension 2013, 62, 359–366. [Google Scholar] [CrossRef] [PubMed]
- Hostenbach, S.; D’Haeseleer, M.; Kooijman, R.; De Keyser, J. Modulation of Cytokine-Induced Astrocytic Endothelin-1 Production as a Possible New Approach to the Treatment of Multiple Sclerosis. Front. Pharmacol. 2020, 10, 1491. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wang, Y.; Yang, W.; Hou, X.; Zou, J.; Cao, K. Resveratrol inhibits angiotensin II-induced ERK1/2 activation by downregulating quinone reductase 2 in rat vascular smooth muscle cells. J. Biomed. Res. 2012, 26, 103–109. [Google Scholar] [CrossRef] [PubMed]
- Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef] [PubMed]
- Steinberg, G.R.; Hardie, D.G. New insights into activation and function of the AMPK. Nat. Rev. Mol. Cell Biol. 2023, 24, 255–272. [Google Scholar] [CrossRef] [PubMed]
- Guo, S.; Yao, Q.; Ke, Z.; Chen, H.; Wu, J.; Liu, C. Resveratrol attenuates high glucose-induced oxidative stress and cardiomyocyte apoptosis through AMPK. Mol. Cell. Endocrinol. 2015, 412, 85–94. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.; Lu, J.; Zhan, L.; Wang, M.; Shi, R.; Yuan, X.; Gao, X.; Liu, X.; Zang, J.; Liu, W.; et al. Resveratrol-induced Sirt1 phosphorylation by LKB1 mediates mitochondrial metabolism. J. Biol. Chem. 2021, 297, 100929. [Google Scholar] [CrossRef] [PubMed]
- Ma, S.; Feng, J.; Zhang, R.; Chen, J.; Han, D.; Li, X.; Yang, B.; Li, X.; Fan, M.; Li, C.; et al. SIRT1 Activation by Resveratrol Alleviates Cardiac Dysfunction via Mitochondrial Regulation in Diabetic Cardiomyopathy Mice. Oxid. Med. Cell. Longev. 2017, 2017, 4602715. [Google Scholar] [CrossRef]
- Ren, X.; Chen, L.; Xie, J.; Zhang, Z.; Dong, G.; Liang, J.; Liu, L.; Zhou, H.; Luo, P. Resveratrol Ameliorates Mitochondrial Elongation via Drp1/Parkin/PINK1 Signaling in Senescent-Like Cardiomyocytes. Oxid. Med. Cell. Longev. 2017, 2017, 4175353. [Google Scholar] [CrossRef]
- Zheng, M.; Bai, Y.; Sun, X.; Fu, R.; Liu, L.; Liu, M.; Li, Z.; Huang, X. Resveratrol Reestablishes Mitochondrial Quality Control in Myocardial Ischemia/Reperfusion Injury through Sirt1/Sirt3-Mfn2-Parkin-PGC-1 Pathway. Molecules 2022, 27, 5545. [Google Scholar] [CrossRef]
- Boa, S.; Ponzoa, V.; Cicconeb, G.; Evangelistab, A.; Sabaa, F.; Goitrea, I.; Procopioa, M.; PaganoaM, G.F.; Cassadera, M.; Gambinoa, R. Six months of resveratrol supplementation has no measurable effect in type 2 diabetic patients. A randomized, double blind, placebo-controlled trial. Pharmacol. Res. 2016, 111, 896–905. [Google Scholar] [CrossRef] [PubMed]
- Asgary, S.; Karimi, R.; Momtaz, S.; Naseri, R.; Farzaei, M.H. Effect of resveratrol on metabolic syndrome components: A systematic review and meta-analysis. Rev. Endocr. Metab. Disord. 2019, 20, 173–186. [Google Scholar] [CrossRef] [PubMed]
- Hoseini, A.; Namazi, G.; Farrokhian, A.; Reiner, Z.; Aghadavod, E.; Bahmani, F.; Asemi, Z. The effects of resveratrol on metabolic status in patients with type 2 diabetes mellitus and coronary heart disease. Food Funct. 2019, 10, 6042–6051. [Google Scholar] [CrossRef] [PubMed]
- Simental-Mendía, L.E.; Guerrero-Romero, F. Effect of resveratrol supplementation on lipid profile in subjects with dyslipidemia: A randomized double-blind, placebo-controlled trial. Nutrition 2019, 58, 7–10. [Google Scholar] [CrossRef] [PubMed]
- Fogacci, F.; Tocci, G.; Presta, V.; Fratter, A.; Borghi, C.; Cicero, A.F.G. Effect of resveratrol on blood pressure: A systematic review and meta-analysis of randomized, controlled, clinical trials. Crit. Rev. Food Sci. Nutr. 2019, 59, 1605–1618. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Ma, W.; Zhang, P.; He, S.; Huang, D. Effect of resveratrol on blood pressure: A meta-analysis of randomized controlled trials. Clin. Nutr. 2015, 34, 27–34. [Google Scholar] [CrossRef] [PubMed]
- Bhatt, J.K.; Thomas, S.; Nanjan, M.J. Resveratrol supplementation improves glycemic control in type 2 diabetes mellitus. Nutr. Res. 2012, 32, 537–541. [Google Scholar] [CrossRef] [PubMed]
- Imamura, H.; Yamaguchi, T.; Nagayama, D.; Saiki, A.; Shirai, K.; Tatsuno, I. Resveratrol ameliorates arterial stiffness assessed by cardio-ankle vascular index in patients with type 2 diabetes mellitus. Int. Heart J. 2017, 58, 577–583. [Google Scholar] [CrossRef]
- Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R.H.; van de Weijer, T.; Goossens, G.H.; Hoeks, J.; van der Krieken, S.; Ryu, D.; Kersten, S.; et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011, 14, 612–622. [Google Scholar] [CrossRef]
- Theodotou, M.; Fokianos, K.; Mouzouridou, A.; Konstantinou, C.; Aristotelous, A.; Prodromou, D.; Chrysikou, A. The effect of resveratrol on hypertension: A clinical trial. Exp. Ther. Med. 2017, 13, 295–301. [Google Scholar] [CrossRef]
- Walker, J.M.; Eckardt, P.; Aleman, J.O.; Da Rosa, J.C.; Liang, Y.; Iizumi, T.; Etheve, S.; Blaser, M.J.; Breslow, J.L.; Holt, P.R. The effects of trans-resveratrol on insulin resistance, inflammation, and microbiota in men with the metabolic syndrome: A pilot randomized, placebo-controlled clinical trial. J. Clin. Transl. Res. 2019, 4, 122–135. [Google Scholar] [PubMed]
- Wong, R.H.X.; Howe, P.R.C.; Buckley, J.D.; Coates, A.M.; Kunz, I.; Berry, N.M. Acute resveratrol supplementation improves flow-mediated dilatation in overweight/obese individuals with mildly elevated blood pressure. Nutr. Metab. Cardiovasc. Dis. 2011, 21, 851–856. [Google Scholar] [CrossRef] [PubMed]
- Marques, B.C.A.A.; Trindade, M.; Aquino, J.C.F.; Cunha, A.R.; Gismondi, R.O.; Neves, M.F.; Oigman, W. Beneficial effects of acute trans-resveratrol supplementation in treated hypertensive patients with endothelial dysfunction. Clin. Exp. Hypertens. 2018, 40, 218–223. [Google Scholar] [CrossRef] [PubMed]
- Edwards, R.; Lyon, T.; Litwin, S.; Rabovsky, A.; Symons, J. Quercetin Reduces Blood Pressure in Hypertensive Subjects. J. Nutr. 2007, 137, 2405–2411. [Google Scholar] [CrossRef] [PubMed]
- Zahedi, M.; Ghiasvand, R.; Feizi, A.; Asgari, G.; Darvish, L. Does quercetin improve cardiovascular risk factors and inflammatory biomarkers in women with type 2 diabetes: A double-blind randomized controlled clinical trial. Int. J. Prev. Med. 2013, 4, 777–785. [Google Scholar] [PubMed]
- Brüll, V.; Burak, C.; Stoffel-Wagner, B.; Wolffram, S.; Nickenig, G.; Müller, C.; Langguth, P.; Alteheld, B.; Fimmers, R.; Naaf, S.; et al. Effects of a quercetin-rich onion skin extract on 24 h ambulatory blood pressure and endothelial function in overweight-to-obese patients with pre-hypertension: A randomised double-blinded placebo-controlled cross-over trial. Br. J. Nutr. 2015, 114, 1263–1277. [Google Scholar] [CrossRef] [PubMed]
- Dower, J.I.; Geleijnse, J.M.; Gijsbers, L.; Zock, P.L.; Kromhout, D.; Hollman, P.C.H. Effects of the pure flavonoids epicatechin and quercetin on vascular function and cardiometabolic health: A randomized, double-blind, placebo-controlled, crossover trial. Am. J. Clin. Nutr. 2015, 101, 914–921. [Google Scholar] [CrossRef]
- Serban, M.; Sahebkar, A.; Zanchetti, A.; Mikhailidis, D.; Howard, G.; Antal, D.; Andrica, F.; Ahmed, A.; Aronow, W.S.; Muntner, P.; et al. Effects of Quercetin on Blood Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Am. Heart Assoc. 2016, 5, e002713. [Google Scholar] [CrossRef]
- Larson, A.; Witman, M.; Guo, Y.; Ives, S.; Richardson, R.; Bruno, R.; Jalili, T.; Symons, J.D. Acute, quercetin-induced reductions in blood pressure in hypertensive individuals are not secondary to lower plasma angiotensin-converting enzyme activity or endothelin-1: Nitric oxide. Nutr. Res. 2012, 32, 557–564. [Google Scholar] [CrossRef]
- Bondonno, N.; Bondonno, C.; Rich, L.; Mas, E.; Shinde, S.; Ward, N.; Hodgson, J.M.; Croft, K.D. Acute effects of quercetin-3-O-glucoside on endothelial function and blood pressure: A randomized dose-response study. Am. J. Clin. Nutr. 2016, 104, 97–103. [Google Scholar] [CrossRef]
- Chekalina, N.I.; Shut, S.V.; Trybrat, T.A.; Burmak, Y.H.; Petrov, Y.Y.; Manusha, Y.I.; Kazakov, Y.M. Effect of quercetin on parameters of central hemodynamics and myocardial ischemia in patients with stable coronary heart disease. Wiadomości Lek. 2017, 70, 707–711. [Google Scholar]
- Kondratiuk, V.E.; Synytsia, Y.P. Effect of quercetin on the echocardiographic parameters of left ventricular diastolic function in patients with gout and essential hypertension. Wiadomości Lek. 2018, 71, 1554–1559. [Google Scholar]
- Bogdanski, P.; Suliburska, J.; Szulinska, M.; Stepien, M.; Pupek-Musialik, D.; Jablecka, A. Green tea extract reduces blood pressure, inflammatory biomarkers, and oxidative stress and improves parameters associated with insulin resistance in obese, hypertensive patients. Nutr. Res. 2012, 32, 421–427. [Google Scholar] [CrossRef] [PubMed]
- Rostami, A.; Khalili, M.; Haghighat, N.; Eghtesadi, S.; Shidfar, F.; Heidari, I.; Ebrahimpour-Koujan, S.; Eghtesadi, M. High-cocoa polyphenol-rich chocolate improves blood pressure in patients with diabetes and hypertension. ARYA Atheroscler. 2015, 11, 21–29. [Google Scholar] [PubMed]
- Chatree, S.; Sitticharoon, C.; Maikaew, P.; Pongwattanapakin, K.; Keadkraichaiwat, I.; Churintaraphan, M.; Sripong, C.; Sririwichitchai, R.; Tapechum, S. Epigallocatechin gallate decreases plasma triglyceride, blood pressure, and serum kisspeptin in obese human subjects. Exp. Biol. Med. (Maywood) 2021, 246, 163–176. [Google Scholar] [CrossRef] [PubMed]
- Ellinger, S.; Reusch, A.; Stehle, P.; Helfrich, H.-P. Epicatechin ingested via cocoa products reduces blood pressure in humans: A nonlinear regression model with a Bayesian approach. Am. J. Clin. Nutr. 2012, 95, 1365–1377. [Google Scholar] [CrossRef] [PubMed]
- Oyama, J.-I.; Maeda, T.; Kouzuma, K.; Ochiai, R.; Tokimitsu, I.; Higuchi, Y.; Sugano, M.; Makino, N. Green tea catechins improve human forearm endothelial dysfunction and have antiatherosclerotic effects in smokers. Circ. J. 2010, 74, 578–588. [Google Scholar] [CrossRef] [PubMed]
- Alañón, M.E.; Castle, S.M.; Serra, G.; Lévèques, A.; Poquet, L.; Actis-Goretta, L.; Spencer, J.P.L. Acute study of dose-dependent effects of (−)-epicatechin on vascular function in healthy male volunteers: A randomized controlled trial. Clin. Nutr. 2020, 39, 746–754. [Google Scholar] [CrossRef]
- Samavat, H.; Newman, A.R.; Wang, R.; Yuan, J.-M.; Wu, A.H.; Kurzer, M.S. Effects of green tea catechin extract on serum lipids in postmenopausal women: A randomized, placebo-controlled clinical trial. Am. J. Clin. Nutr. 2016, 104, 1671–1682. [Google Scholar] [CrossRef]
- Suzuki-Sugihara, N.; Kishimoto, Y.; Saita, E.; Taguchi, C.; Kobayashi, M.; Ichitani, M.; Ukawa, Y.; Sagesaka, Y.M.; Suzuki, E.; Kondo, K. Green tea catechins prevent low-density lipoprotein oxidation via their accumulation in low-density lipoprotein particles in humans. Nutr. Res. 2016, 36, 16–23. [Google Scholar] [CrossRef]
- McDermott, M.M.; Criqui, M.H.; Domanchuk, K.; Ferrucci, L.; Guralnik, J.M.; Kibbe, M.R.; Kosmac, K. Cocoa to Improve Walking Performance in Older People with Peripheral Artery Disease: The COCOA-PAD Pilot Randomized Clinical Trial. Circ. Res. 2020, 126, 589–599. [Google Scholar] [CrossRef] [PubMed]
- Kishimoto, Y.; Saita, E.; Taguchi, C.; Aoyama, M.; Ikegami, Y.; Ohmori, R.; Kondo, K.; Momiyama, Y. Associations between green tea consumption and coffee consumption and the prevalence of coronary artery disease. J. Nutr. Sci. Vitaminol. 2020, 66, 237–245. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.-M.; Zhou, B.; Wang, Y.-S.; Gong, Q.-Y.; Wang, Q.-M.; Yan, J.-J.; Gao, W.; Wang, L.-S. Black and green tea consumption and the risk of coronary artery disease: A meta-analysis. Am. J. Clin. Nutr. 2011, 93, 506–515. [Google Scholar] [CrossRef] [PubMed]
- Dural, I.E.; Onrat, E.; Celik, S.; Vurmaz, A.; Yalim, Z.; Gokaslan, S.; Gokaslan, C.; Korucu, C.; Aksu, U. The Relationships Between Chocolate Consumption and Endothelial Dysfunction in Patients with Heart Failure. Türk Kardiyol. Derneği Arşivi 2022, 50, 334–339. [Google Scholar] [CrossRef] [PubMed]
- Cassidy, A.; Mukamal, K.J.; Liu, L.; Franz, M.; Eliassen, A.H.; Rimm, E.B. High anthocyanin intake is associated with a reduced risk of myocardial infarction in young and middle-aged women. Circulation 2013, 127, 188–196. [Google Scholar] [CrossRef] [PubMed]
- Cassidy, A.; Bertoia, M.; Chiuve, S.; Flint, A.; Forman, J.; Rimm, E.B. Habitual intake of anthocyanins and flavanones and risk of cardiovascular disease in men. Am. J. Clin. Nutr. 2016, 104, 587–594. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Suarez, J.M.; Giampieri, F.; Tulipani, S.; Casoli, T. One month strawberry-rich anthocyanin supplementation ameliorates cardiovascular risk, oxidative stress markers and platelet activation in humans. J. Nutr. Biochem. 2014, 25, 289–294. [Google Scholar] [CrossRef]
- Aboonabi, A.; Meyer, R.R.; Gaiz, A.; Singh, I. Anthocyanins in berries exhibited anti-atherogenicity and antiplatelet activities in a metabolic syndrome population. Nutr. Res. 2020, 76, 82–93. [Google Scholar] [CrossRef]
- Santhakumar, A.; Kundur, A.; Fanning, K.; Netzel, M.; Stanley, R.; Singh, I. Consumption of anthocyanin-rich Queen Garnet plum juice reduces platelet activation related thrombogenesis in healthy volunteers. J. Func. Foods 2015, 12, 11–22. [Google Scholar] [CrossRef]
- Tian, Z.; Li, K.; Fan, D.; Zhao, Y.; Gao, X.; Ma, X.; Xu, L.; Shi, Y.; Ya, F.; Zou, J.; et al. Dose-dependent effects of anthocyanin supplementation on platelet function in subjects with dyslipidemia: A randomized clinical trial. BioMedicine 2021, 70, 103533. [Google Scholar] [CrossRef]
- Jennings, A.; Welch, A.A.; Fairweather-Tait, S.J.; Kay, C.; Minihane, A.; Chowienczyk, P. Higher anthocyanin intake is associated with lower arterial stiffness and central blood pressure in women. Am. J. Clin. Nutr. 2012, 96, 781–788. [Google Scholar] [CrossRef] [PubMed]
- McKay, D.L.; Chen, C.-Y.O.; Saltzman, E.; Blumberg, J.B. Hibiscus sabdaria L. Tea (Tisane) lowers blood pressure in prehypertensive and mildly hypertensive adults. J. Nutr. 2009, 140, 298–303. [Google Scholar] [CrossRef] [PubMed]
- Basu, A.; Du, M.; Leyva, M.J.; Sanchez, K.; Betts, N.M.; Wu, M. Blueberries decrease cardiovascular risk factors in obese men and women with metabolic syndrome. J. Nutr. 2010, 140, 1582–1587. [Google Scholar] [CrossRef] [PubMed]
- Johnson, S.A.; Figueroa, A.; Navaei, N.; Wong, A.; Kalfon, R.; Ormsbee, L.T. Daily blueberry consumption improves blood pressure and arterial stiffness in postmenopausal women with pre- and stage 1-hypertension: A randomized, double-blind, placebo-controlled clinical trial. J. Acad. Nutr. Diet. 2015, 115, 369–377. [Google Scholar] [CrossRef] [PubMed]
- Emamat, H.; Zahedmehr, A.; Asadian, S.; Nasrollahzadeh, J. The effect of purple-black barberry (Berberis integerrima) on blood pressure in subjects with cardiovascular risk factors: A randomized controlled trial. J. Ethnopharmacol. 2022, 289, 115097. [Google Scholar] [CrossRef] [PubMed]
- Dohadwala, M.M.; Holbrook, M.; Hamburg, N.M.; Shenouda, S.M.; Chung, W.B.; Titas, M.; Kluge, M.A.; Na Wang, N.; Palmisano, J.; Milbury, P.E.; et al. Effects of cranberry juice consumption on vascular function in patients with coronary artery disease. Am. J. Clin. Nutr. 2011, 93, 934–940. [Google Scholar] [CrossRef] [PubMed]
- Huang, H.; Chen, G.; Liao, D.; Zhu, Y.; Xue, X. Effects of berries consumption on cardiovascular risk factors: A meta-analysis with trial sequential analysis of randomized controlled trials. Sci. Rep. 2016, 6, 23625. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Ling, W.; Yang, Y.; Chen, Y.; Tian, Z.; Du, Z. Role of purified anthocyanins in improving cardiometabolic risk factors in Chinese men and women with prediabetes or early untreated diabetes—A randomized controlled trial. Nutrients 2017, 9, 1104. [Google Scholar] [CrossRef]
- Liu, C.; Sun, J.; Lu, Y.; Bo, Y. Effects of anthocyanin on serum lipids in dyslipidemia patients: A systematic review and meta-analysis. PLoS ONE 2016, 11, e0162089. [Google Scholar] [CrossRef]
- Gordish, K.L.; Beierwaltes, W.H. Chronic resveratrol reverses a mild angiotensin II-induced pressor effect in a rat model. Integr. Blood Press. Control 2016, 9, 23–31. [Google Scholar]
- Zhang, L.-X.; Li, C.-X.; Kakar, M.U.; Khan, M.S.; Wu, P.-F.; Amir, R.M.; Dai, D.-F.; Naveed, M.; Li, Q.-Y.; Saeed, M.; et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed. Pharmacother. 2021, 143, 112164. [Google Scholar] [CrossRef] [PubMed]
- Cheng, P.W.; Ho, W.Y.; Su, Y.T.; Lu, P.J.; Chen, B.Z.; Cheng, W.H.; Lu, W.H.; Sun, G.C.; Yeh, T.C.; Hsiao, M.; et al. Resveratrol decreases fructose-induced oxidative stress, mediated by NADPH oxidase via an AMPK-dependent mechanism. Br. J. Pharmacol. 2014, 171, 2739–2750. [Google Scholar] [CrossRef] [PubMed]
- Yu, L.; Tu, Y.; Jia, X.; Fang, K.; Liu, L.; Wan, L.; Xiang, C.; Wang, Y.; Sun, X.; Liu, T.; et al. Resveratrol protects against pulmonary arterial hypertension in rats via activation of silent information regulator 1. Cell Physiol. Biochem. 2017, 42, 55–67. [Google Scholar] [CrossRef]
- Dolinsky, V.W.; Chakrabarti, S.; Pereira, T.J.; Oka, T.; Levasseur, J.; Beker, D. Resveratrol prevents hypertension and cardiac hypertrophy in hypertensive rats and mice. Biochim. Biophys. Acta 2013, 1832, 1723–1733. [Google Scholar] [CrossRef] [PubMed]
- Grujić-Milanović, J.; Jaćević, V.; Miloradović, Z.; Milanović, S.D.; Jovović, D.; Ivanov, M.; Karanović, D.; Vajić, U.-J.; Mihailović-Stanojević, N. Resveratrol improved kidney function and structure in malignantly hypertensive rats by restoration of antioxidant capacity and nitric oxide bioavailability. Biomed. Pharmacother. 2022, 154, 113642. [Google Scholar] [CrossRef] [PubMed]
- Franco, J.G.; Lisboa, P.C.; Lima, N.S.; Amaral, T.A.; Peixoto-Silva, N.; Resende, A.C.; Oliveira, E.; Passos, M.C.; Moura, E.G. Resveratrol attenuates oxidative stress and prevents steatosis and hypertension in obese rats programmed by early weaning. J. Nutr. Biochem. 2013, 24, 960–966. [Google Scholar] [CrossRef] [PubMed]
- Mozafari, M.; Nekooeian, A.A.; Panjeshahin, M.R.; Zare, H.R. The effects of resveratrol in rats with simultaneous type 2 diabetes and renal hypertension: A study of antihypertensive mechanisms. Iran. J. Med. Sci. 2015, 40, 152–160. [Google Scholar] [PubMed]
- Tain, Y.L.; Lee, W.C.; Wu, K.L.H.; Leu, S.; Chan, J.Y.H. Resveratrol prevents the development of hypertension programmed by maternal plus post-weaning high-fructose consumption through modulation of oxidative stress, nutrient-sensing signals, and gut microbiota. Mol. Nutr. Food Res. 2018, 62, e1800066. [Google Scholar] [CrossRef]
- Prysyazhna, O.; Wolhuter, K.; Switzer, C.; Santos, C.; Yang, X.; Lynham, S.; Shah, A.M.; Eaton, P.; Burgoyne, J.R. Blood Pressure-Lowering by the Antioxidant Resveratrol Is Counterintuitively Mediated by Oxidation of cGMP-Dependent Protein Kinase. Circulation 2019, 140, 126–137. [Google Scholar] [CrossRef]
- Li, H.; Xia, N.; Hasselwander, S.; Daiber, A. Resveratrol and vascular function. Int. J. Mol. Sci. 2019, 20, 2155. [Google Scholar] [CrossRef]
- Lee, D.I.; Acosta, C.; Anderson, C.M.; Anderson, H.D. Peripheral and cerebral resistance arteries in the spontaneously hypertensive heart failure rat: Effects of stilbenoid polyphenols. Molecules 2017, 22, 380. [Google Scholar] [CrossRef]
- Li, H.; Förstermann, U. Uncoupling of endothelial NO synthase in atherosclerosis and vascular disease. Curr. Opin. Pharmacol. 2013, 13, 161–167. [Google Scholar] [CrossRef] [PubMed]
- Fabricio, V.; Oishi, J.C.; Biffe, B.G.; Ruffoni, L.D.; Silva, K.A.; Nonaka, K.O.; Rodrigues, G.J. Resveratrol treatment normalizes the endothelial function and blood pressure in ovariectomized rats. Arq. Bras. Cardiol. 2017, 108, 116–121. [Google Scholar] [CrossRef] [PubMed]
- Tsutsui, H.; Kinugawa, S.; Matsushimam, S. Oxidative stress and heart failure. Am. J. Physiol. Heart Circ. Physiol. 2011, 301, H2181–H2190. [Google Scholar] [CrossRef] [PubMed]
- Gu, X.; Wang, Z.; Ye, Z.; Lei, J.; Li, L.; Su, D.; Zheng, X. Resveratrol, an activator of SIRT1, upregulates AMPK and improves cardiac function in heart failure. Genet. Mol. Res. 2014, 13, 323–335. [Google Scholar] [CrossRef] [PubMed]
- Ahmet, I.; Tae, H.J.; Lakatta, E.G.; Talan, M. Long-term low dose dietary resveratrol supplement reduces cardiovascular structural and functional deterioration in chronic heart failure in rats. Can. J. Physiol. Pharmacol. 2017, 95, 268–274. [Google Scholar] [CrossRef] [PubMed]
- Kanamori, H.; Takemura, G.; Goto, K.; Tsujimoto, A.; Ogino, A.; Takeyama, T.; Kawaguchi, T.; Watanabe, T.; Morishita, K.; Kawasaki, M.; et al. Resveratrol reverses remodeling in hearts with large, old myocardial infarctions through enhanced autophagy-activating AMP kinase pathway. Am. J. Pathol. 2013, 182, 701–713. [Google Scholar] [CrossRef] [PubMed]
- Matsumura, N.; Takahara, S.; Maayah, Z.H.; Parajuli, N.; Byrne, N.J.; Shoieb, S.M.; Soltys, C.M.; Beker, D.L.; Masson, G.; El-Kadi, A.O.S.; et al. Resveratrol improves cardiac function and exercise performance in MI-induced heart failure through the inhibition of cardiotoxic HETE metabolites. J. Mol. Cell. Cardiol. 2018, 125, 162–173. [Google Scholar] [CrossRef] [PubMed]
- Wojciechowski, P.; Juric, D.; Louis, X.L.; Thandapilly, S.J.; Yu, L.; Taylor, C.; Netticadan, T. Resveratrol arrests and regresses the development of pressure overload- but not volume overload-induced cardiac hypertrophy in rats. J. Nutr. 2010, 140, 962–968. [Google Scholar] [CrossRef]
- Rimbaud, S.; Ruiz, M.; Piquereau, J.; Mateo, P.; Fortin, D.; Veksler, V.; Garnier, A.; Ventura-Clapier, R. Resveratrol improves survival, hemodynamics and energetics in a rat model of hypertension leading to heart failure. PLoS ONE 2011, 6, e26391. [Google Scholar] [CrossRef]
- Sung, M.M.; Das, S.K.; Levasseur, J.; Byrne, N.J.; Fung, D.; Kim, T.T.; Masson, G.; Boisvenue, J.; Soltys, C.-L. Resveratrol treatment of mice with pressure-overload-induced heart failure improves diastolic function and cardiac energy metabolism. Circ. Heart Fail. 2015, 8, 128–137. [Google Scholar] [CrossRef] [PubMed]
- Sung, M.M.; Byrne, N.J.; Robertson, I.M.; Kim, T.T.; Samokhvalov, V.; Levasseur, J.; Soltys, C.-L.; Fung, D.; Tyreman, N.; Denou, E.; et al. Resveratrol improves exercise performance and skeletal muscle oxidative capacity in heart failure. Am. J. Physiol. 2017, 312, H842–H853. [Google Scholar] [CrossRef] [PubMed]
- Feng, H.; Mou, S.-Q.; Li, W.-J.; Zhang, N.; Zhou, Z.-Y.; Ding, W.; Bian, Z.-Y.; Liao, H.-H. Resveratrol Inhibits Ischemia-Induced Myocardial Senescence Signals and NLRP3 Inflammasome Activation. Oxid. Med. Cell. Longev. 2020, 2020, 2647807. [Google Scholar] [CrossRef] [PubMed]
- Fourny, N.; Lan, C.; Eric, S.; Bernard, M.; Desrois, M. Protective effect of resveratrol against ischemia-reperfusion injury via enhanced high energy compounds and eNOS-SIRT1 expression in type 2 diabetic female rat heart. Nutrients 2019, 11, 105. [Google Scholar] [CrossRef] [PubMed]
- Mao, Z.-J.; Lin, H.; Hou, J.-W.; Zhou, Q.; Wang, Q.; Chen, Y.-H. A meta-analysis of resveratrol protects against myocardial ischemia/reperfusion injury: Evidence from small animal studies and insight into molecular mechanisms. Oxid. Med. Cell. Longev. 2019, 2019, 5793867. [Google Scholar] [CrossRef] [PubMed]
- Gurusamy, N.; Lekli, I.; Mukherjee, S.; Ray, D.; Ahsan, M.K.; Gherghiceanu, M.; Popescu, L.M.; Das, D.K. Cardioprotection by resveratrol: A novel mechanism via autophagy involving the mTORC2 pathway. Cardiovasc. Res. 2010, 86, 103–112. [Google Scholar] [CrossRef] [PubMed]
- Gupta, P.K.; DiPette, D.J.; Supowit, S.C. Protective effect of resveratrol against pressure overload-induced heart failure. Food Sci. Nutr. 2014, 2, 218–229. [Google Scholar] [CrossRef] [PubMed]
- Gao, Y.; Kang, L.; Li, C.; Liu, R.; Wang, J. Resveratrol ameliorates diabetes-induced cardiac dysfunction through AT1R-ERK/p38 MAPK signaling pathway. Cardiovasc. Toxicol. 2016, 16, 130–137. [Google Scholar] [CrossRef]
- Dolinsky, V.W.; Rogan, K.J.; Sung, M.M.; Zordoky, B.N.; Haykowsky, M.J.; Young, M.E.; Jones, L.W.; Dyck, J.R.B. Both aerobic exercise and resveratrol supplementation attenuate doxorubicin-induced cardiac injury in mice. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E243–E253. [Google Scholar] [CrossRef]
- Zeng, Y.; Cao, G.; Lin, L.; Zhang, Y.; Luo, X.; Ma, X.; Aiyisake, A.; Cheng, Q. Resveratrol Attenuates Sepsis-Induced Cardiomyopathy in Rats through Anti-Ferroptosis via the Sirt1/Nrf2 Pathway. J. Investig. Surg. 2023, 36, 2157521. [Google Scholar] [CrossRef]
- Yoshida, Y.; Shioi, T.; Izumi, T. Resveratrol ameliorates experimental autoimmune myocarditis. Circulation 2007, 71, 397–404. [Google Scholar] [CrossRef]
- Tanno, M.; Kuno, A.; Yano, T.; Miura, T.; Hisahara, S.; Ishikawa, S.; Shimamoto, K.; Horio, Y. Induction of manganese superoxide dismutase by nuclear translocation and activation of SIRT1 promotes cell survival in chronic heart failure. J. Biol. Chem. 2010, 285, 8375–8382. [Google Scholar] [CrossRef] [PubMed]
- Pan, Q.R.; Ren, Y.L.; Liu, W.X.; Hu, Y.J.; Zheng, J.S.; Xu, Y.; Wang, G. Resveratrol prevents hepatic steatosis and endoplasmic reticulum stress and regulates the expression of genes involved in lipid metabolism, insulin resistance, and inflammation in rats. Nutr. Res. 2015, 35, 576–584. [Google Scholar] [CrossRef] [PubMed]
- Chen, Q.; Wang, E.; Ma, L.; Zhai, P. Dietary resveratrol increases the expression of hepatic 7α-hydroxylase and ameliorates hypercholesterolemia in high-fat fed C57BL/6J mice. Lipids Health Dis. 2012, 11, 56. [Google Scholar] [CrossRef] [PubMed]
- Tanko, Y.; Jimoh, A.; Ahmed, A.; Mohammed, A.; Ayo, J.O. Resveratrol Protects Rabbits Against Cholesterol Diet-Induced Hyperlipidaemia. Niger. J. Physiol. Sci. 2016, 31, 71–75. [Google Scholar] [PubMed]
- Singh, A.P.; Singh, R.; Verma, S.S.; Rai, V.; Kaschula, C.H.; Maiti, P.; Gupta, S.C. Health benefits of resveratrol: Evidence from clinical studies. Med. Res. Rev. 2019, 39, 1851–1891. [Google Scholar] [CrossRef] [PubMed]
- Zhao, W.; Li, A.; Feng, X.; Hou, T.; Liu, K.; Liu, B.; Zhang, N. Metformin and resveratrol ameliorate muscle insulin resistance through preventing lipolysis and inflammation in hypoxic adipose tissue. Cell Signal. 2016, 28, 1401–1411. [Google Scholar] [CrossRef] [PubMed]
- Berrougui, H.; Grenier, G.; Loued, S.; Drouin, G.; Khalil, A. A new insight into resveratrol as an atheroprotective compound: Inhibition of lipid peroxidation and enhancement of cholesterol efflux. Atherosclerosis 2009, 207, 420–427. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, X.; Li, J. Resveratrol protects against oxidized low-density lipoprotein-induced human umbilical vein endothelial cell apoptosis via inhibition of mitochondrial-derived oxidative stress. Mol. Med. Rep. 2017, 15, 2457–2464. [Google Scholar] [CrossRef]
- Deng, Z.Y.; Hu, M.M.; Xin, Y.F.; Gang, C. Resveratrol alleviates vascular inflammatory injury by inhibiting inflammasome activation in rats with hypercholesterolemia and vitamin D2 treatment. Inflamm Res. 2015, 64, 321–332. [Google Scholar] [CrossRef]
- Brawerman, G.M.; Kereliuk, S.M.; Brar, N.; Cole, L.K.; Seshadri, N.; Pereira, T.J.; Xiang, B.; Hunt, K.L.; Fonseca, M.A.; Hatch, G.M.; et al. Maternal resveratrol administration protects against gestational diabetes-induced glucose intolerance and islet dysfunction in the rat on spring. J. Physiol. 2019, 597, 4175–4192. [Google Scholar] [CrossRef] [PubMed]
- Heebøll, S.; Kreuzfeldt, M.; Hamilton-Dutoit, S.; Kjær Poulsen, M.; Stødkilde-Jørgensen, H.; Møller, H.J.; Jessen, N.; Thorsen, K.; Kristina Hellberg, Y.; Bønløkke Pedersen, S.; et al. Placebo-controlled, randomised clinical trial: High-dose resveratrol treatment for non-alcoholic fatty liver disease. Scand. J. Gastroenterol. 2016, 51, 456–464. [Google Scholar] [CrossRef] [PubMed]
- Fujitaka, K.; Otani, H.; Jo, F.; Jo, H.; Nomura, E.; Iwasaki, M.; Nishikawa, M.; Iwasaka, T.; Das, D.K. Modified resveratrol Longevinex improves endothelial function in adults with metabolic syndrome receiving standard treatment. Nutr. Res. 2011, 31, 842–847. [Google Scholar] [CrossRef] [PubMed]
- Agarwal, B.; Campen, M.J.; Channell, M.M.; Wherry, S.J.; Varamini, B.; Davis, J.G.; Baur, J.A.; Smoliga, J.A. Resveratrol for primary prevention of atherosclerosis: Clinical trial evidence for improved gene expression in vascular endothelium. Int. J. Cardiol. 2013, 166, 246–248. [Google Scholar] [CrossRef] [PubMed]
- Militaru, C.; Donoiu, I.; Craciun, A.; Scorei, I.D.; Bulearca, A.M.; Scorei, R.I. Oral resveratrol and calcium fructoborate supplementation in subjects with stable angina pectoris: Effects on lipid profiles, inflammation markers, and quality of life. Nutrition 2013, 29, 178–183. [Google Scholar] [CrossRef] [PubMed]
- Guven, H.; Arici, A.; Simsek, O. Flavonoids in our foods: A short review. J. Basic Clin. Health Sci. 2019, 3, 96–106. [Google Scholar] [CrossRef]
- Graf, B.A.; Milbury, P.E.; Blumberg, J.B. Flavonols, flavones, flavanones, and human health: Epidemiological evidence. J. Med. Food. 2005, 8, 281–290. [Google Scholar] [CrossRef]
- Gentile, D.; Fornai, M.; Pellegrini, C.; Colucci, R.; Blandizzi, C. Dietary flavonoids as a potential intervention to improve redox balance in obesity and related co-morbidities: A review. Nutr. Res. Rev. 2018, 31, 239–247. [Google Scholar] [CrossRef]
- Ginwala, R.; Bhavsar, R.; Chigbu, D.G.I.; Jain, P.; Khan, Z.K. Potential role of flavonoids in treating chronic inflammatory diseases with a special focus on the anti-inflammatory activity of apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef]
- Biela, M.; Rimarčík, J.; Senajová, E.; Kleinová, A.; Klein, E. Antioxidant action of deprotonated flavonoids: Thermodynamics of sequential proton-loss electron-transfer. Phytochemistry 2020, 180, 112528. [Google Scholar] [CrossRef]
- Dias, M.C.; Pinto, D.C.G.A.; Freitas, H.; Santos, C.; Silva, A.M.S. The antioxidant system in Olea europaea to enhanced UV-B radiation also depends on flavonoids and secoiridoids. Phytochemistry 2020, 170, 112199. [Google Scholar] [CrossRef] [PubMed]
- D’Amelia, V.; Aversano, R.; Chiaiese, P.; Carputo, D. The antioxidant properties of plant flavonoids: Their exploitation by molecular plant breeding. Phytochem. Rev. 2018, 17, 611–625. [Google Scholar] [CrossRef]
- Hamid, A.A.; Aminuddin, A.; Yunus, M.H.M.; Murthy, J.K.; Hui, C.K. Antioxidative and anti-inflammatory activities of Polygonum minus: A review of literature. Rev. Cardiovasc. Med. 2020, 21, 275–287. [Google Scholar] [PubMed]
- Zeng, N.; Zhang, G.; Hu, X.; Pan, J.; Gong, D. Mechanism of fisetin suppressing superoxide anion and xanthine oxidase activity. J. Funct. Foods 2019, 58, 1–10. [Google Scholar] [CrossRef]
- Arts, I.C.W.; Sesink, A.L.A.; Faassen-Peters, M.; Hollman, P.C.H. The type of sugar moiety is a major determinant of the small intestinal uptake and subsequent biliary excretion of dietary quercetin glycosides. Br. J. Nutr. 2004, 91, 841–847. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Pandey, A.K. Chemistry and biological activities of flavonoids: An Overview. Sci. World J. 2013, 2013, 162750. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, N.A.; Radwan, N.M.; Aboul Ezz, H.S.; Salama, N.A. The antioxidant effect of green tea mega EGCG against electromagnetic radiation-induced oxidative stress in the hippocampus and striatum of rats. Electromagn. Biol. Med. 2017, 36, 63–73. [Google Scholar] [CrossRef]
- Zhang, H.; Xu, Z.; Zhao, H.; Wang, X.; Pang, J. Anthocyanin supplementation improves anti-oxidative and anti-inflammatory capacity in a dose-response manner in subjects with dyslipidemia. Redox Biol. 2020, 32, 101474. [Google Scholar] [CrossRef]
- Nagarajan, S.; Nagarajan, R.; Kumar, J.; Salemme, A.; Togna, A.R.; Saso, L.; Bruno, F. Antioxidant activity of synthetic polymers of phenolic compounds. Polymers 2020, 12, 1646. [Google Scholar] [CrossRef]
- Yin, Z.; Wu, Y.; Chen, Y.; Qie, X.; Zeng, M.; Wang, Z.; Qin, F.; Chen, J.; He, Z. Analysis of the interaction between cyanidin-3-O-glucoside and casein hydrolysates and its effect on the antioxidant ability of the complexes. Food Chem. 2021, 340, 127915. [Google Scholar] [CrossRef]
- Read, M. Flavonoids: Naturally occurring anti-inflammatory agents. Am. J. Pathol. 1995, 147, 235–237. [Google Scholar] [PubMed]
- Damon, M.; Flandre, O.; Michel, F.; Perdrix, L.; Labrid, C. Effect of chronic treatment with a purified flavonoid fraction on inflammatory granuloma in the rat. Study of prostaglandin E2 and F2 alpha and thromboxane B2 release and histological changes. Arzneimittel-Forschung 1987, 37, 1149–1153. [Google Scholar] [PubMed]
- Lee, H.S.; Ko, H.R.; Ryu, S.Y.; Oh, W.K.; Kim, B.Y. Inhibition of phospholipase C Gamma 1 by the prenylated flavonoids from Sophora flavescens. Planta Med. 1997, 63, 266–268. [Google Scholar] [CrossRef] [PubMed]
- Kimata, M.; Shichijo, M.; Miura, T.; Serizawa, I.; Inagaki, N.; Nagai, H. Effects of luteolin, quercetin and baicalein on immunoglobulin E-mediated mediator release from human cultured mast cells. Clin. Exp. Allergy 2000, 30, 501–508. [Google Scholar] [CrossRef] [PubMed]
- Al-Khayri, J.M.; Sahana, G.R.; Nagella, P.; Joseph, B.V.; Alessa, F.M. Flavonoids as Potential Anti-Inflammatory Molecules: A Review. Molecules 2022, 27, 2901. [Google Scholar] [CrossRef] [PubMed]
- Serafini, M.; Peluso, I.; Raguzzini, A. Flavonoids as Anti-Inflammatory Agents. Proc. Nutr. Soc. 2010, 69, 273–278. [Google Scholar] [CrossRef] [PubMed]
- Nam, T.G.; Lim, T.-G.; Lee, B.H.; Lim, S.; Kang, H. Comparison of Anti-Inflammatory Effects of Flavonoid-Rich Common and Tartary Buckwheat Sprout Extracts in Lipopolysaccharide-Stimulated RAW264.7 and Peritoneal Macrophages. Oxid. Med. Cell. Longev. 2017, 2017, 9658030. [Google Scholar] [CrossRef]
- Ullah, A.; Munir, S.; Badshah, S.L.; Khan, N.; Ghani, L. Important Flavonoids and Their Role as a Therapeutic Agent. Molecules 2020, 25, 5243. [Google Scholar] [CrossRef]
- Li, C.; Wang, T.; Zhang, C.; Xuan, J.; Su, C.; Wang, Y. Quercetin attenuates cardiomyocyte apoptosis via inhibition of JNK and p38 mitogen-activated protein kinase signaling pathways. Gene 2016, 577, 275–280. [Google Scholar] [CrossRef]
- Tang, L.; Peng, Y.; Xu, T.; Yi, X.; Liu, Y. The effects of quercetin protect cardiomyocytes from A/R injury is related to its capability to increasing expression and activity of PKCε protein. Mol. Cell. Biochem. 2013, 382, 145–152. [Google Scholar] [CrossRef]
- Shu, Z.; Yang, Y.; Yang, L.; Jiang, H.; Yu, X. Cardioprotective effects of dihydroquercetin against ischemia reperfusion injury by inhibiting oxidative stress and endoplasmic reticulum stress-induced apoptosis via the PI3K/Akt pathway. Food Funct. 2019, 10, 203–215. [Google Scholar] [CrossRef]
- Rahman, I.; Biswas, S.; Kirkham, P. Regulation of Inflammation and Redox Signaling by Dietary Polyphenols. Biochem. Pharmacol. 2006, 72, 1439–1452. [Google Scholar] [CrossRef] [PubMed]
- Rahman, I.; Marwick, J.; Kirkham, P. Redox Modulation of Chromatin Remodeling: Impact on Histone Acetylation and Deacetylation, NF-KappaB and pro-Inflammatory Gene Expression. Biochem. Pharmacol. 2004, 68, 1255–1267. [Google Scholar] [CrossRef] [PubMed]
- Min, Y.; Choi, C.; Bark, H.; Son, H.; Park, H. Quercetin Inhibits Expression of Inflammatory Cytokines through Attenuation of NFkappaB and P38 MAPK in HMC-1 Human Mast Cell Line. Inflamm. Res. 2007, 56, 210–215. [Google Scholar] [CrossRef] [PubMed]
- Chen, T.; Zhang, X.; Zhu, G.; Liu, H.; Chen, J.; Wang, Y.; He, X. Quercetin inhibits TNF-α induced HUVECs apoptosis and inflammation via downregulating NF-kB and AP-1 signaling pathway in vitro. Medicine 2020, 99, e22241. [Google Scholar] [CrossRef] [PubMed]
- Kundu, J.K.; Surh, Y.J. Epigallocatechin Gallate Inhibits Phorbol Ester-Induced Activation of NF-KB and CREB in Mouse Skin Role of P38 MAPK. Ann. N. Y. Acad. Sci. 2007, 1095, 504–512. [Google Scholar] [CrossRef] [PubMed]
- Cho, S.; Park, S.; Kwon, M.; Jeong, T.; Bok, S.; Choi, W.Y.; Jeong, W.I.; Ryu, S.Y. Quercetin Suppresses Proinflammatory Cytokines Production through MAP Kinases And NF-Kappa B Pathway in Lipopolysaccharide-Stimulated Macrophage. Mol. Cell. Biochem. 2003, 243, 153–160. [Google Scholar] [CrossRef]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
- Siti, H.N.; Jalil, J.; Asmadi, A.Y.; Kamisah, Y. Rutin Modulates MAPK Pathway Differently from Quercetin in Angiotensin II-Induced H9c2 Cardiomyocyte Hypertrophy. Int. J. Mol. Sci. 2021, 22, 5063. [Google Scholar] [CrossRef]
- Andugulapati, S.B.; Gourishetti, K.; Tirunavalli, S.K.; Shaikh, T.B.; Sistla, R. Biochanin-A ameliorates pulmonary fibrosis by suppressing the TGF-β mediated EMT, myofibroblasts differentiation and collagen deposition in in vitro and in vivo systems. Phytomedicine 2020, 78, 153298. [Google Scholar] [CrossRef]
- Wang, J.; Ge, S.; Wang, Y.; Liu, Y.; Qiu, L.; Li, J. Puerarin Alleviates UUO-Induced Inflammation and Fibrosis by Regulating the NF-κB P65/STAT3 and TGFβ1/Smads Signaling Pathways. Drug Des. Dev. Ther. 2021, 15, 3697–3708. [Google Scholar] [CrossRef] [PubMed]
- Meng, J.; Chen, Y.; Wang, J.; Qiu, J.; Chang, C.; Bi, F.; Wu, X.; Liu, W. EGCG protects vascular endothelial cells from oxidative stress-induced damage by targeting the autophagy-dependent PI3K-AKT-mTOR pathway. Ann. Transl. Med. 2020, 8, 200. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Zhang, D. (−)-Epigallocatechin-3-gallate inhibits eNOS uncoupling and alleviates high glucose-induced dysfunction and apoptosis of human umbilical vein endothelial cells by PI3K/AKT/eNOS pathway. Diabetes Metab. Syndr. Obes. 2020, 13, 2495–2504. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Liao, P.; Liang, R.; Zheng, X.; Jian, J. Epigallocatechin gallate prevents mitochondrial impairment and cell apoptosis by regulating miR-30a/p53 axis. Phytomedicine 2019, 61, 152845. [Google Scholar] [CrossRef] [PubMed]
- Mou, Q.; Jia, Z.; Luo, M.; Liu, L.; Huang, X. Epigallocatechin-3-gallate exerts cardioprotective effects related to energy metabolism in pressure overload-induced cardiac dysfunction. Arch. Biochem. Biophys. 2022, 723, 109217. [Google Scholar] [CrossRef]
- Nan, J.; Nan, C.; Ye, J.; Qian, L.; Geng, Y. EGCG protects cardiomyocytes against hypoxia-reperfusion injury through inhibition of OMA1 activation. J. Cell Sci. 2019, 132, jcs220871. [Google Scholar] [PubMed]
- Larson, A.J.; Symons, J.D.; Jalili, T. Therapeutic potential of quercetin to decrease blood pressure: Review of efficacy and mechanisms. Adv. Nutr. 2012, 3, 39–46. [Google Scholar] [CrossRef] [PubMed]
- Igarashi, K.; Honma, K.; Yoshinari, O.; Nanjo, F.; Hara, Y. Effects of dietary catechins on glucose tolerance, blood pressure and oxidative status in Goto-Kakizaki rats. J. Nutr. Sci. Vitaminol. 2007, 53, 496–500. [Google Scholar] [CrossRef]
- Huang, Y.; Zhang, A.; Lau, C.W.; Chen, Z.Y. Vasorelaxant effects of purified green tea epicatechin derivatives in rat mesenteric artery. Life Sci. 1998, 63, 275–283. [Google Scholar] [CrossRef]
- Liu, Y.; Niu, L.; Cui, L.; Hou, X.; Li, J. Hesperetin inhibits rat coronary constriction by inhibiting Ca2+ influx and enhancing voltage-gated K+ channel currents of the myocytes. Eur. J. Pharmacol. 2014, 735, 193–201. [Google Scholar] [CrossRef]
- Arumugam, S.; Thandavarayan, R.A.; Arozal, W.; Sari, F.R.; Giridharan, V.V.; Soetikno, V.; Palaniyandi, S.S.; Harima, M. Quercetin offers cardioprotection against progression of experimental autoimmune myocarditis by suppression of oxidative and endoplasmic reticulum stress via endothelin-1/MAPK signaling. Free Radic. Res. 2012, 46, 154–163. [Google Scholar] [CrossRef] [PubMed]
- Shahbaz, A.U.; Kamalov, G.; Zhao, W.; Zhao, T.; Johnson, P.L. Mitochondria-targeted Cardioprotection in Aldosteronism. J Cardiovasc. Pharmacol. 2011, 57, 37–43. [Google Scholar] [CrossRef] [PubMed]
- Kumar, M.; Kasala, E.R.; Bodduluru, L.N.; Kumar, V.; Lahkar, M. Molecular and biochemical evidence on the protective effects of quercetin in isoproterenol-induced acute myocardial injury in rats. J. Biochem. Mol. Toxicol. 2017, 31, e21832. [Google Scholar] [CrossRef] [PubMed]
- Putakala, M.; Gujjala, S.; Nukala, S.; Bongu, S.B.R.; Chintakunta, N.; Desireddy, S. Cardioprotective effect of Phyllanthus amarus against high fructose diet induced myocardial and aortic stress in rat model. Biomed. Pharmacother. 2017, 95, 1359–1368. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Yu, Z.; Huang, X.; Gao, Y.; Wang, X.; Gu, J.; Xue, X. Peroxisome proliferator-activated receptor γ (PPARγ) mediates the protective effect of quercetin against myocardial ischemia-reperfusion injury via suppressing the NF-κB pathway. Am. J. Transl. Res. 2016, 8, 5169. [Google Scholar] [PubMed]
- Han, J.J.; Hao, J.; Kim, C.H.; Hong, J.S.; Ahn, H.Y. Quercetin prevents cardiac hypertrophy induced by pressure overload in rats. J. Vet. Med. Sci. 2009, 71, 737–743. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Tan, A.; An, X.; Xia, Y.; Xie, Y. Quercetin dihydrate inhibition of cardiac fibrosis induced by angiotensin II in vivo and in vitro. Biomed. Pharmacother. 2020, 127, 110205. [Google Scholar] [CrossRef]
- Chen, K.; Rekep, M.; Wei, W.; Wu, Q.; Xue, Q.; Li, S.; Tian, J.; Yi, Q.; Zhang, G.; Zhang, G.; et al. Quercetin prevents in vivo and in vitro myocardial hypertrophy through the proteasome-GSK-3 pathway. Cardiovasc. Drugs Ther. 2018, 32, 5–21. [Google Scholar] [CrossRef]
- Chang, X.; Zhang, T.; Wang, J.; Liu, Y.; Yan, P.; Meng, Q.; Yin, Y.; Wang, S. SIRT5-related Desuccinylation modification contributes to quercetin-induced protection against heart failure and high-glucose-prompted cardiomyocytes injured through regulation of mitochondrial quality surveillance. Oxid. Med. Cell. Longev. 2021, 2021, 5876841. [Google Scholar] [CrossRef]
- Machha, A.; Achike, F.I.; Mustafa, A.M.; Mustafa, M.R. Quercetin, a flavonoid antioxidant, modulates endothelium-derived nitric oxide bioavailability in diabetic rat aortas. Nitric Oxide 2007, 16, 442–447. [Google Scholar] [CrossRef]
- Sánchez, M.; Galisteo, M.; Vera, R.; Villar, I.C.; Zarzuelo, A.; Tamargo, J.; Pérez-Vizcaíno, F.; Duarte, J. Quercetin downregulates NADPI I oxidase, increases eNOS activity and prevents endothelial dysfunction in spontaneously hypertensive rats. J. Hypertens. 2006, 24, 75–84. [Google Scholar] [CrossRef]
- Hackl, L.P.; Cuttle, G.; Dovichi, S.S.; Lima-Landman, M.T.; Nicolau, M. Inhibition of angiotensin-converting enzyme by quercetin alters the vascular response to bradykinin and angiotensin. J. Pharmacol. 2002, 65, 182–186. [Google Scholar] [CrossRef] [PubMed]
- Linz, W.; Wiemer, G.; Gohike, P.; Unger, T.; Scholkens, B.A. Contribution of kinins to the cardiovascular actions of angiotensin-converting enzyme inhibitors. Pharmacol Rev. 1995, 47, 25–49. [Google Scholar]
- Soman, S.; Rajamanickam, C.; Rauf, A.A.; Madambath, I. Molecular mechanisms of the antiglycative and cardioprotective activities of Psidium guajava leaves in the rat diabetic myocardium. Pharm. Biol. 2016, 54, 3078–3085. [Google Scholar] [CrossRef]
- Roslan, J.; Giribabu, N.; Karim, K.; Salleh, N. Quercetin ameliorates oxidative stress, inflammation, and apoptosis in the heart of streptozotocin-nicotinamide-induced adult male diabetic rats. Biomed. Pharmacother. 2017, 86, 570–582. [Google Scholar] [CrossRef] [PubMed]
- Najafi, M.; Noroozi, E.; Javadi, A.; Badalzadeh, R. Anti-arrhythmogenic and anti-inflammatory effects of troxerutin in ischemia/reperfusion injury of diabetic myocardium. Biomed. Pharmacother. 2018, 102, 385–391. [Google Scholar] [CrossRef] [PubMed]
- Ulasova, E.; Perez, J.; Hill, B.G.; Bradley, W.E.; Garber, D.W.; Landar, A.; Barnes, S.; Prasain, J.; Parks, D.A.; Dell’Italia, L.J.; et al. Quercetin prevents left ventricular hypertrophy in the Apo E knockout mouse. Redox Biol. 2013, 1, 381–386. [Google Scholar] [CrossRef]
- Zhang, J.; Liang, R.; Wang, L.; Yan, R.; Hou, R.; Gao, S.; Yang, B. Effects of an aqueous extract of Crataegus pinnatifida Bge. var. major N.E.Br. fruit on experimental atherosclerosis in rats. J. Ethnopharmacol. 2013, 148, 563–569. [Google Scholar]
- Garcia, M.L.; Pontes, R.B.; Nishi, E.E.; Ibuki, F.K.; Oliveira, V.; Sawaya, A.C.; Carvalho, P.O. The antioxidant effects of green tea reduces blood pressure and sympathoexcitation in an experimental model of hypertension. J. Hypertens. 2017, 35, 348–354. [Google Scholar] [CrossRef]
- Huang, Y.; Chan, N.W.; Lau, C.W.; Yao, X.Q.; Chan, F.L.; Chen, Z.Y. Involvement of endothelium/nitric oxide in vasorelaxation induced by purified green tea (-) epicatechin. Biochim. Biophys. Acta 1999, 1427, 322–328. [Google Scholar] [CrossRef]
- Muñoz, M.d.l.F.; Fernández, M.d.l.F.; Román-Carmena, M.; Cruz, M.d.C.I.d.l.; Amor, S.; Martorell, P.; Enrique-López, M.; García-Villalón, A.L.; Inarejos-García, A.M.; Granado, M. Supplementation with two new standardized tea extracts prevents the development of hypertension in mice with metabolic syndrome. Antioxidants 2022, 11, 1573. [Google Scholar] [CrossRef] [PubMed]
- Gao, J.; Akbari, A.; Wang, T. Green tea could improve elderly hypertension by modulating arterial stiffness, the activity of the renin/angiotensin/aldosterone axis, and the sodium-potassium pumps in old male rats. J. Food Biochem. 2022, 46, e14398. [Google Scholar] [CrossRef] [PubMed]
- Sabri, N.A.M.; Lee, S.-K.; Murugan, D.D.; Ling, W.C. Epigallocatechin gallate (EGCG) alleviates vascular dysfunction in angiotensin II-infused hypertensive mice by modulating oxidative stress and eNOS. Sci. Rep. 2022, 12, 17633. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.X.; Lin, Q.L.; Shi, Z.P. Effect of catechin and epicatechin on serum lipid level in mice. J. Hunan Agric. Univ. 2002, 28, 232–233. [Google Scholar]
- Ding, S.; Jiang, J.; Yu, P.; Zhang, G.; Zhang, G.; Liu, X. Green tea polyphenol treatment attenuates atherosclerosis in high-fat diet-fed apolipoprotein E-knockout mice via alleviating dyslipidemia and up-regulating autophagy. PLoS ONE 2017, 12, e0181666. [Google Scholar] [CrossRef] [PubMed]
- Wahyudi, S.; Sargowo, D. Green tea polyphenols inhibit oxidized LDL-induced NF-KB activation in human umbilical vein endothelial cells. Acta Med. Indones. 2007, 39, 66–70. [Google Scholar] [PubMed]
- Liao, Z.-L.; Zeng, B.-H.; Wang, W.; Li, G.-H.; Wu, F.; Wang, L.; Zhong, Q.-P.; Wei, H.; Fang, X. Impact of the consumption of tea polyphenols on early atherosclerotic lesion formation and intestinal Bifidobacteria in high-fat-fed ApoE−/− mice. Front Nutr. 2016, 3, 42. [Google Scholar] [CrossRef] [PubMed]
- Yu, J.; Li, W.; Xiao, X.; Huang, Q.; Yu, J.; Yang, Y.; Han, T.; Zhang, D.; Niu, X. (−)-Epicatechin gallate blocks the development of atherosclerosis by regulating oxidative stress in vivo and in vitro. Food Funct. 2021, 12, 8715–8727. [Google Scholar] [CrossRef]
- Ferenczyová, K.; Kindernay, L.; Vlkovicová, J.; Kalocayová, B.; Rajtík, T.; Barteková, M. Pharmacology of Catechins in Ischemia-Reperfusion Injury of the Heart. Antioxidants 2021, 10, 1390. [Google Scholar] [CrossRef]
- Muhammed, I.; Sankar, S.; Govindaraj, S. Ameliorative effect of epigallocatechin gallate on cardiac hypertrophy and fibrosis in aged rats. J. Cardiovasc. Pharmacol. 2018, 71, 65–75. [Google Scholar] [CrossRef]
- Sheng, R.; Gu, Z.L.; Xie, M.L. Epigallocatechin gallate, the major component of polyphenols in green tea, inhibits telomere attrition mediated cardiomyocyte apoptosis in cardiac hypertrophy. Int. J. Cardiol. 2013, 162, 199–209. [Google Scholar] [CrossRef] [PubMed]
- Shen, C.L.; Samathanam, C.; Tatum, O.L.; Graham, S.; Tubb, C.; Cao, J.J.; Dunn, D.M.; Wang, J.S. Green tea polyphenols avert chronic inflammation-induced myocardial fibrosis of female rats. Inflamm. Res. 2011, 60, 665–672. [Google Scholar] [CrossRef] [PubMed]
- Cai, Y.; Yu, S.S.; He, Y.; Bi, X.Y.; Gao, S.; Yan, T.D.; Zheng, G.D.; Chen, T.T.; Ye, J.T.; Liu, P.Q. EGCG inhibits pressure overload-induced cardiac hypertrophy via the PSMB5/Nmnat2/SIRT6-dependent signalling pathways. Acta Physiol. 2021, 231, e13602. [Google Scholar] [CrossRef] [PubMed]
- Ried, K.; Fakler, P.; Stocks, N.P.; Cochrane Hypertension Group. Effect of cocoa on blood pressure. Cochrane Database Syst. Rev. 2017, 2017, CD008893. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Mateos, A.; Weber, T.; Skene, S.S.; Ottaviani, J.I.; Crozier, A.; Kelm, M. Assessing the respective contributions of dietary flavanol monomers and procyanidins in mediating cardiovascular effects in humans: Randomized, controlled, double-masked intervention trial. Am. J. Clin. Nutr. 2018, 108, 1229–1237. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Zimmermann, D.; De Castro, C.A.; Actis-Goretta, L. Dose-response relationship between cocoa flavanols and human endothelial function: A systematic review and meta-analysis of randomized trials. Food Funct. 2019, 10, 6322–6330. [Google Scholar] [CrossRef] [PubMed]
- Huang, S.; Li, J.; Wu, Y.; Ranjbar, S.; Xing, A.; Zhao, H.; Wang, Y.; Shearer, G.C.; Bao, L.; Lichtenstein, A.H.; et al. Tea consumption and longitudinal change in high-density lipoprotein cholesterol concentration in Chinese adults. J. Am. Heart Assoc. 2018, 7, e008814. [Google Scholar] [CrossRef]
- Zamora-Ros, R.; Knaze, V.; Lujan-Barroso, L.; Slimani, N.; Romieu, I.; Touillaud, M. Estimation of the intake of anthocyanidins and their food sources in the European Prospective Investigation into Cancer and Nutrition (EPIC) study. Br. J. Nutr. 2011, 106, 1090–1099. [Google Scholar] [CrossRef]
- Chen, Y.; Ge, Z.; Huang, S.; Zhou, L.; Zhai, C.; Chen, Y.; Hu, Q.; Cao, W.; Weng, Y.; Li, Y. Delphinidin attenuates pathological cardiac hypertrophy via the AMPK/NOX/MAPK signaling pathway. Aging 2020, 12, 5362. [Google Scholar] [CrossRef]
- Hu, W.; Wang, W.; Ma, Q.; Liu, T.; Zhang, J.; Zhang, J. Blueberry anthocyanin-enriched extract ameliorates transverse aortic constriction-induced myocardial dysfunction via the DDAH1/ADMA/NO signaling pathway in mice. Mol. Med. Rep. 2020, 21, 454–462. [Google Scholar] [CrossRef]
- Aloud, B.M.; Raj, P.; McCallum, J.; Kirby, C.; Louis, X.L.; Jahan, F.; Yu, L.; Hiebert, B.; Duhamel, T.A.; Wigle, J.T.; et al. Cyanidin 3-O-glucoside prevents the development of maladaptive cardiac hypertrophy and diastolic heart dysfunction in 20-week-old spontaneously hypertensive rats. Food Funct. 2018, 9, 3466–3480. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-F.; Shibu, M.A.; Fan, M.-J.; Chen, M.-C.; Viswanadha, V.P.; Lin, Y.-L.; Lai, C.-H.; Lin, K.-H.; Ho, T.-J.; Kuo, W.-W.; et al. Purple rice anthocyanin extract protects cardiac function in STZ-induced diabetes rat hearts by inhibiting cardiac hypertrophy and fibrosis. J. Nutr. Biochem. 2016, 31, 98–105. [Google Scholar] [CrossRef] [PubMed]
- Yue, E.; Yu, Y.; Wang, X.; Liu, B.; Bai, Y.; Yang, B. Anthocyanin protects cardiac function and cardiac fibroblasts from high-glucose induced inflammation and myocardial fibrosis by inhibiting IL-17. Front. Pharmacol. 2021, 11, 593633. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Tan, D.; Shi, L.; Liu, X.; Zhang, Y.; Tong, C.; Song, D.; Hou, M. Blueberry Anthocyanins-Enriched Extracts Attenuate Cyclophosphamide-Induced Cardiac Injury. PLoS ONE 2015, 10, e0127813. [Google Scholar] [CrossRef] [PubMed]
- Kim, H.-Y.; Yoon, J.-J.; Lee, H.-K.; Tai, A.-L.; Lee, Y.-J.; Kim, D.-S.; Kang, D.-G.; Lee, H.-S. Blackcurrant Improves Diabetic Cardiovascular Dysfunction by Reducing Inflammatory Cytokines in Type 2 Diabetes Mellitus Mice. Nutrients 2021, 13, 4177. [Google Scholar] [CrossRef] [PubMed]
- Toufektsian, M.-C.; de Lorgeril, M.; Nagy, N.; Salen, P.; Donati, M.B.; Giordano, L.; Mock, H.-P.; Peterek, S.; Matros, A.; Petroni, K.; et al. Chronic dietary intake of plant-derived anthocyanins protects the rat heart against ischemia-reperfusion injury. J. Nutr. 2008, 138, 747–752. [Google Scholar] [CrossRef] [PubMed]
- Guler, A.; Sahin, M.A.; Yucel, O.; Yokusoglu, M.; Gamsizkan, M.; Ozal, E.; Demirkilic, U.; Arslan, M. Proanthocyanidin prevents myocardial ischemic injury in adult rats. Med. Sci. Monit. 2011, 17, BR326–BR331. [Google Scholar] [CrossRef] [PubMed]
- Petersen, C.; Bharat, D.; Cutler, B.R.; Gholami, S.; Denetso, C.; Mueller, J.E.; Cho, J.M.; Kim, J.-S.; Symons, J.D.; Babu, P.V.A. Circulating metabolites of strawberry mediate reductions in vascular inflammation and endothelial dysfunction in db/db mice. Int. J. Cardiol. 2018, 263, 111–117. [Google Scholar] [CrossRef]
- Joo, H.K.; Choi, S.; Lee, Y.R.; Lee, E.O.; Park, M.S.; Park, K.B.; Kim, C.-S.; Lim, Y.P.; Park, J.-T.; Jeon, B.H. Anthocyanin-Rich Extract from Red Chinese Cabbage Alleviates Vascular Inflammation in Endothelial Cells and Apo E−/− Mice. Int. J. Mol. Sci. 2018, 19, 816. [Google Scholar] [CrossRef]
- Nasri, S.; Roghani, M.; Baluchnejadmojarad, T.; Rabani, T.; Balvardi, M. Vascular mechanisms of cyanidin-3-glucoside response in streptozotocin-diabetic rats. Pathophysiology 2011, 18, 273–278. [Google Scholar] [CrossRef]
- Edirisinghe, I.; Banaszewski, K.; Cappozzo, J.; McCarthy, D.; Burton-Freeman, B.M. Effect of black currant anthocyanins on the activation of endothelial nitric oxide synthase (eNOS) in vitro in human endothelial cells. J. Agric. Food Chem. 2011, 59, 8616–8624. [Google Scholar] [CrossRef] [PubMed]
- Ojeda, D.; Jiménez-Ferrer, E.; Zamilpa, A.; Herrera-Arellano, A.; Tortoriello, J.; Alvarez, L. Inhibition of angiotensin converting enzyme (ACE) activity by the anthocyanins delphinidin- and cyanidin-3-o-sambubiosides from Hibiscus sabdariffa. J. Ethnopharmacol. 2010, 127, 7–10. [Google Scholar] [CrossRef] [PubMed]
- Shaughnessy, K.S.; Boswall, I.A.; Scanlan, A.P.; Gottschall-Pass, K.T.; Sweeney, M.I. Diets containing blueberry extract lower blood pressure in spontaneously hypertensive stroke-prone rats. Nutr. Res. 2009, 29, 130–138. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Zhu, J.; Gong, G.; Guo, L.; Zhang, Y.; Zhang, Z.; Ma, C. Anthocyanin attenuates high salt-induced hypertension via inhibiting the hyperactivity of the sympathetic nervous system. Clin. Exp. Hypertens. 2023, 45, 2233717. [Google Scholar] [CrossRef] [PubMed]
- Ugwu, P.; Ubom, R.; Madueke, P.; Okorie, P.; Nwachukwu, D. Anti-Hypertensive Effects of Anthocyanins from Hibiscus sabdarifa Calyx on the Renin-Angiotensin-Aldosterone System in Wistar Rats. Niger. J. Physiol. Sci. 2022, 37, 113–117. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Andrews, M.C.; Hu, Y.; Wang, D.; Qin, Y.; Zhu, Y.; Ni, H.; Ling, W. Anthocyanin extract from black rice significantly ameliorates platelet hyperactivity and hypertriglyceridemia in dyslipidemic rats induced by high fat diets. J. Agric. Food Chem. 2011, 59, 6759–6764. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Dai, M.; Nie, W.-J.; Yang, X.-R.; Zeng, X.-C. Effects of the ethanol extract of black mulberry (Morus nigra L.) fruit on experimental atherosclerosis in rats. J. Ethnopharmacol. 2017, 200, 228–235. [Google Scholar] [CrossRef] [PubMed]
- Suh, J.-H.; Romain, C.; González-Barrio, R.; Cristol, J.-P.; Teissèdre, P.-L.; Crozier, A.; Rouanet, J.-M. Raspberry juice consumption, oxidative stress and reduction of atherosclerosis risk factors in hypercholesterolemic golden Syrian hamsters. Food Funct. 2011, 2, 400. [Google Scholar] [CrossRef]
- Wang, L.; Zhu, H.; Zhao, Y.; Jiao, R.; Lei, L.; Chen, J.; Wang, X.; Zhang, Z.; Huang, Y.; Wang, T.; et al. Cranberry anthocyanin as an herbal medicine lowers plasma cholesterol by increasing excretion of fecal sterols. Phytomedicine 2018, 38, 98–106. [Google Scholar] [CrossRef]
- Vendrame, S.; Daugherty, A.; Kristo, A.S.; Klimis-Zacas, D. Wild blueberry (Vaccinium angustifolium)-enriched diet improves dyslipidaemia and modulates the expression of genes related to lipid metabolism in obese Zucker rats. Br. J. Nutr. 2014, 111, 194–200. [Google Scholar] [CrossRef]
- Gaiz, A.A.; Mosawy, S.; Colson, N.; Singh, I. Potential of Anthocyanin to Prevent Cardiovascular Disease in Diabetes. Altern. Ther. Health Med. 2018, 24, 40–47. [Google Scholar]
- Yao, Y.; Zhang, X.; Xu, Y.; Zhao, Y.; Song, F.; Tian, Z.; Zhao, M. Cyanidin-3- O-β-Glucoside Attenuates Platelet Chemokines and Their Receptors in Atherosclerotic Inflammation of ApoE−/− Mice. J. Agric. Food Chem. 2022, 70, 8254–8263. [Google Scholar] [CrossRef] [PubMed]
- Erlund, I.; Koli, R.; Alfthan, G.; Marniemi, J.; Puukka, P.; Mustonen, P. Favorable effects of berry consumption on platelet function, blood pressure, and HDL cholesterol. Am. J. Clin. Nutr. 2008, 87, 323–331. [Google Scholar] [CrossRef] [PubMed]
- Vendrame, S.; Klimis-Zacas, D. Potential Factors Influencing the Effects of Anthocyanins on Blood Pressure Regulation in Humans: A Review. Nutrients 2019, 11, 1431. [Google Scholar] [CrossRef]
- Curtis, P.J.; van der Velpen, V.; Berends, L.; Jennings, A.; Feelisch, M.; Umpleby, A.M.; Evans, M. Blueberries improve biomarkers of cardiometabolic function in participants with metabolic syndrome—Results from a 6-month, double-blind, randomized controlled trial. Am. J. Clin Nutr. 2019, 109, 1535–1545. [Google Scholar] [CrossRef] [PubMed]
- Huang, L.; Di Xiao, D.; Zhang, X.; Sandhu, A.K.; Chandra, P.; Kay, C.; Edirisinghe, I.; Burton-Freeman, B. Strawberry Consumption, Cardiometabolic Risk Factors, and Vascular Function: A Randomized Controlled Trial in Adults with Moderate Hypercholesterolemia. J. Nutr. 2021, 151, 1517–1526. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez-Mateos, A.; Istas, G.; Boschek, L.; Feliciano, R.P.; Mills, C.E.; Boby, C.; Gómez-Alonso, S.; Milenkovic, D.; Heiss, C. Circulating Anthocyanin Metabolites Mediate Vascular Benefits of Blueberries: Insights from Randomized Controlled Trials, Metabolomics, and Nutrigenomics. J. Gerontol. A Biol. Sci. Med. Sci. 2019, 74, 967–976. [Google Scholar] [CrossRef] [PubMed]
- Heiss, C.; Istas, G.; Feliciano, R.P.; Weber, T.; Wang, B.; Favari, C.; Mena, P.; Del Rio, D.; Rodriguez-Mateos, A. Daily consumption of cranberry improves endothelial function in healthy adults: A double blind randomized controlled trial. Food Funct. 2022, 13, 3812–3824. [Google Scholar] [CrossRef]
- Yang, L.; Ling, W.; Du, Z.; Chen, Y.; Li, D.; Deng, S.; Liu, Z.; Yang, L. Effects of anthocyanins on cardiometabolic health: A systematic review and meta-analysis of randomized controlled trials. Adv. Nutr. 2017, 8, 684–693. [Google Scholar] [CrossRef]
- Sesso, H.D.; Manson, J.E.; Aragaki, A.K.; Rist, P.M.; Johnson, L.G.; Friedenberg, G.; Copeland, T.; Clar, A.; Mora, S.; Moorthy, M.V.; et al. COSMOS Research Group. Effect of cocoa flavanol supplementation for the prevention of cardiovascular disease events: The COcoa Supplement and Multivitamin Outcomes Study (COSMOS) randomized clinical trial. Am. J. Clin. Nutr. 2022, 115, 1490–1500. [Google Scholar] [CrossRef]
- McCullough, M.L.; Peterson, J.J.; Patel, R.; Jacques, P.F.; Shah, R.; Dwyer, J.T. Flavonoid intake and cardiovascular disease mortality in a prospective cohort of US adults. Am. J. Clin. Nutr. 2012, 95, 454–464. [Google Scholar] [CrossRef] [PubMed]
- Grosso, G.; Micek, A.; Godos, J.; Pajak, A.; Sciacca, S.; Galvano, F.; Giovannucci, E.L. Dietary flavonoid and Lignan intake and mortality in prospective cohort studies: Systematic review and dose-response meta-analysis. Am. J. Epidemiol. 2017, 185, 1304–1316. [Google Scholar] [CrossRef] [PubMed]
- Tresserra-Rimbau, A.; Rimm, E.B.; Medina-Remón, A.; Martínez-González, M.A.; López-Sabater, M.C.; Covas, M.I. Polyphenol intake and mortality risk: A re-analysis of the PREDIMED trial. BMC Med. 2014, 12, 77. [Google Scholar] [CrossRef] [PubMed]
- Castellarin, S.D.; Bavaresco, L.; Falginella, L.; Van Zeller Gonçalves, M.I.; Di Gaspero, G. Phenolics in Grape Berry and Key Antioxidants. In The Biochemistry of the Grape Berry; Gerós, H., Chavez, M.M., Delrot, S., Eds.; Bentham e Books; Bentham Science: Sharjah, United Arab Emirates, 2012; pp. 89–110. [Google Scholar]
- Visioli, F.; Panaite, S.A.; Tomé-Carneiro, J. Wine’s Phenolic Compounds and Health: A Pythagorean View. Molecules 2020, 25, 4105. [Google Scholar] [CrossRef] [PubMed]
- Haseeb, S.; Alexander, B.; Baranchuk, A. Wine and cardiovascular health: A comprehensive review. Circulation 2017, 136, 1434–1448. [Google Scholar] [CrossRef] [PubMed]
- Di Renzo, L.; Marsella, L.T.; Carraro, A.; Valente, R.; Gualtieri, P.; Gratteri, S.; Tomasi, D.; Gaiotti, F.; De Lorenzo, A. Changes in LDL Oxidative Status and Oxidative and Inflammatory Gene Expression after Red Wine Intake in Healthy People: A Randomized Trial. Mediat. Inflamm. 2015, 2015, 317348. [Google Scholar] [CrossRef] [PubMed]
- Nova, E.; San Mauro-Martín, I.; Díaz-Prieto, L.E.; Marcos, A. Wine and beer within a moderate alcohol intake is associated with higher levels of HDL-c and adiponectin. Nutr. Res. 2019, 63, 42–50. [Google Scholar] [CrossRef] [PubMed]
- Golan, R.; Gepner, Y.; Shai, I.J.E. Wine and Health–New Evidence. Eur. J. Clin. Nutr. 2019, 72, 55–59. [Google Scholar] [CrossRef]
- Rasines-Perea, Z.; Teissedre, P.-L. Grape Polyphenols’ Effects in Human Cardiovascular Diseases and Diabetes. Molecules 2017, 22, 68. [Google Scholar] [CrossRef]
- Chiva-Blanch, G.; Urpi-Sarda, M.; Ros, E.; Valderas-Martinez, P.; Casas, R.; Arranz, S.; Guillén, M.; Lamuela-Raventós, R.M.; Llorach, R.; Andres-Lacueva, C.; et al. Effects of red wine polyphenols and alcohol on glucose metabolism and the lipid profile: A randomized clinical trial. Clin. Nutr. 2013, 32, 200–206. [Google Scholar] [CrossRef]
- Navarro-García, F.; Ponce-Ruíz, N.; Rojas-García, A.E.; Ávila-Villarreal, G.; Herrera-Moreno, J.F.; Barrón-Vivanco, B.S.; Bernal-Hernández, Y.Y.; González-Arias, C.A.; Medina-Díaz, I.M. The Role of Nutritional Habits and Moderate Red Wine Consumption in PON1 Status in Healthy Population. Appl. Sci. 2021, 11, 9503. [Google Scholar] [CrossRef]
- Toth, A.; Sandor, B.; Papp, J.; Rabai, M.; Botor, D.; Horvath, Z.; Kenyeres, P.; Juricskay, I.; Toth, K.; Czopf, L. Moderate red wine consumption improves hemorheological parameters in healthy volunteers. Clin. Hemorheol. Microcirc. 2014, 56, 13–23. [Google Scholar] [CrossRef] [PubMed]
- Naissides, M.; Mamo, J.C.; James, A.P.; Pal, S. The effect of acute red wine polyphenol consumption on postprandial lipaemia in postmenopausal women. Atherosclerosis 2004, 177, 401–408. [Google Scholar] [CrossRef] [PubMed]
- Grønbaek, M.; Becker, U.; Johansen, D.; Gottschau, A.; Schnohr, P.; Hein, H.O.; Jensen, G.; Sørensen, T.I. Type of alcohol consumed and mortality from all causes, coronary heart disease, and cancer. Ann. Intern. Med. 2000, 133, 411–419. [Google Scholar] [CrossRef]
- Stampfer, M.J.; Colditz, G.A.; Willett, W.C.; Speizer, F.E.; Hennekens, C.H. A prospective study of moderate alcohol consumption and the risk of coronary disease and stroke in women. N. Engl. J. Med. 1988, 319, 267–273. [Google Scholar] [CrossRef] [PubMed]
- Levantesi, G.; Marfisi, R.; Mozaffarian, D.; Franzosi, M.G.; Maggioni, A.; Nicolosi, G.L.; Schweiger, C.; Silletta, M.; Tavazzi, L.; Tognoni, G.; et al. Wine consumption and risk of cardiovascular events after myocardial infarction: Results from the GISSI-Prevenzione trial. Int. J. Cardiol. 2013, 163, 282–287. [Google Scholar] [CrossRef] [PubMed]
- Lucerón-Lucas-Torres, M.; Saz-Lara, A.; Díez-Fernández, A.; Martínez-García, I.; Martínez-Vizcaíno, V.; Cavero-Redondo, I.; Álvarez-Bueno, C. Association between Wine Consumption with Cardiovascular Disease and Cardiovascular Mortality: A Systematic Review and Meta-Analysis Maribel. Nutrients 2023, 15, 2785. [Google Scholar] [CrossRef]
- Torres, A.; Cachofeiro, V.; Millán, J.; Lahera, V.; Nieto, M.; Martin, R.; Bello, E.; Alvarez-Sala, L.A. Red wine intake but not other alcoholic beverages increases total antioxidant capacity and improves pro-inflammatory profile after an oral fat diet in healthy volunteers. Rev. Clín. Esp. 2015, 215, 486–494. [Google Scholar] [CrossRef]
- GBD 2020 Alcohol Collaborators. Population-level risks of alcohol consumption by amount, geography, age, sex, and year: A systematic analysis for the Global Burden of Disease Study 2020. Lancet 2022, 400, 185–235. [Google Scholar] [CrossRef]
- Anderson, P. The Impact of Alcoholic Beverages on Human Health. Nutrients 2021, 13, 4417. [Google Scholar] [CrossRef]
- WHO. Alcohol. Available online: https://www.who.int/news-room/fact-sheets/detail/alcohol (accessed on 4 September 2023).
- Rocha, K.K.R.; Souza, G.A.; Ebaid, G.X.; Seiva, F.R.F.; Cataneo, A.C.; Novelli, E.L.B. Resveratrol toxicity: Effects on risk factors for atherosclerosis and hepatic oxidative stress in standard and high-fat diets. Food Chem. Toxicol. 2009, 47, 1362–1367. [Google Scholar] [CrossRef]
- De la Lastra, C.A.; Villegas, I. Resveratrol as an antioxidant and pro-oxidant agent: Mechanisms and clinical implications. Biochem. Soc. Trans. 2007, 35 Pt 5, 1156–1160. [Google Scholar] [CrossRef] [PubMed]
- Macedo, T.S.J.; Barros, V.R.P.; Monte, A.P.O.; Gouveia, B.B.; Bezerra, M.É.S.; Cavalcante, A.Y.P.; Barberino, R.S.; Menezes, V.G.; Matos, M.H.T. Resveratrol has dose-dependent effects on DNA fragmentation and mitochondrial activity of ovine secondary follicles cultured in vitro. Zygote 2017, 25, 434–442. [Google Scholar] [CrossRef] [PubMed]
- Pignitter, M.; Schueller, K.; Burkon, A.; Knorr, V.; Esefelder, L.; Doberer, D.; Wolzt, M.; Somoza, V. Concentration-dependent effects of resveratrol and metabolites on the redox status of human erythrocytes in single-dose studies. J. Nutr. Biochem. 2016, 27, 164–170. [Google Scholar] [CrossRef]
- Berardi, V.; Ricci, F.; Castelli, M.; Galati, G.; Risuleo, G. Resveratrol exhibits a strong cytotoxic activity in cultured cells and has an antiviral action against polyomavirus: Potential clinical use. J. Exp. Clin. Cancer Res. 2009, 28, 96. [Google Scholar] [CrossRef] [PubMed]
- Martins, L.A.M.; Coelho, B.P.C.; Behr, G.; Pettenuzzo, L.F.; Souza, I.C.C.; Moreira, J.C.F.; Borojevic, R.; Gottfried, C.; Guma, F.C.R. Resveratrol induces pro-oxidant effects and time-dependent resistance to cytotoxicity in activated hepatic stellate cells. Cell Biochem. Biophys. 2014, 68, 247–257. [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]
- Crowell, J.A.; Korytko, P.J.; Morrissey, R.L.; Booth, T.D.; Levine, B.S. Resveratrol-associated renal toxicity. Toxicol. Sci. 2004, 82, 614–619. [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. Pharmacokinet. 2010, 49, 449–454. [Google Scholar] [CrossRef]
- Jiang, H.; Webster, D.; Cao, J.; Shao, A. The Safety of Green Tea and Green Tea Extract Consumption in Adults-Results of a Systematic Review. Regul. Toxicol. Pharmacol. 2018, 95, 412–433. [Google Scholar]
- Younes, M.; Aggett, P.; Aguilar, F.; Crebelli, R.; Dusemund, B.; Filipič, M.; Frutos, M.J.; Galtier, P.; Gott, D. Scientific Opinion on the Safety of Green Tea Catechins. EFSA J. 2018, 16, e05239. [Google Scholar] [PubMed]
- Cruz-Correa, M.; Shoskes, D.A.; Sanchez, P.; Zhao, R.; Hylind, L.M.; Wexner, S.D.; Giardiello, F.M. Combination treatment with curcumin and quercetin of adenomas in familial adenomatous polyposis. Clin. Gastroenterol. Hepatol. 2006, 4, 1035–1038. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Tsao, R. Dietary polyphenols, oxidative stress and antioxidant and anti-inflammatory effects. Curr. Opin. Food Sci. 2016, 8, 33–42. [Google Scholar] [CrossRef]
- Hurrell, R.; Egli, I. Iron bioavailability and dietary reference values. Am. J. Clin. Nutr. 2010, 91, 1461S–1467S. [Google Scholar] [CrossRef]
Resveratrol | ||||||
---|---|---|---|---|---|---|
Authors | Year | Area of Interest | Study | Study Design | Main Findings | Ref. |
Boa et al. | 2016 | Lipid profile | RCT |
|
| [136] |
Asgary et al. | 2019 | Lipid profile | Meta-analysis |
|
| [137] |
Magyar et al. | 2012 | Lipid profile, endothelial function | RCT |
|
| [27] |
Hoseini et al. | 2019 | Metabolic status | RCT |
|
| [138] |
Simental-Mendia et al. | 2019 | Lipid profile | RCT |
|
| [139] |
Fogacci et al. | 2018 | Blood pressure | Meta-analysis |
|
| [140] |
Liu et al. | 2015 | Blood pressure | Meta-analysis |
|
| [141] |
Bhatt et al. | 2012 | Blood pressure, diabetes mellitus | RCT |
|
| [142] |
Imamura et al. | 2017 | Blood pressure, arterial stiffness | RCT |
|
| [143] |
Timmers et al. | 2011 | Blood pressure | RCT |
|
| [144] |
Theodotou et al. | 2017 | Blood pressure | RCT |
|
| [145] |
Walker et al. | 2019 | Blood pressure, diabetes | RCT |
|
| [146] |
Wong et al. | 2011 | Endothelial function | RCT |
|
| [147] |
Marques et al. | 2018 | Endothelial function | RCT |
|
| [148] |
Gal et al. | 2020 | Heart failure, lipid profile | RCT |
|
| [66] |
Quercetin | ||||||
Authors | Year | Area of Interest | Study | Study design | Main findings | Ref. |
Edwards et al. | 2007 | Blood pressure | First RCT |
|
| [149] |
Zahedi et al. | 2013 | Blood pressure, lipid profile | RCT |
|
| [150] |
Brüll et al. | 2015 | Blood pressure | RCT |
|
| [151] |
Dower et al. | 2015 | Blood pressure | RCT |
|
| [152] |
Serban et al. | 2016 | Blood pressure | Meta-analysis |
|
| [153] |
Larson et al. | 2012 | Blood pressure | RCT |
|
| [154] |
Bondonno | 2016 | Blood pressure | RCT |
|
| [155] |
Chekalina | 2017 | Heart function | RCT |
|
| [156] |
Kondratiuk et al. | 2018 | Heart function | RCT |
|
| [157] |
Catechins | ||||||
Authors | Year | Area of interest | Study | Study design | Main findings | Ref. |
Bogdansy et al. | 2012 | Blood pressure | RCT |
|
| [158] |
Rostami et al. | 2015 | Blood pressure, lipid profile | RCT |
|
| [159] |
Dower et al. | 2015 | Blood pressure | RCT |
|
| [152] |
Chatree et al. | 2021 | Blood pressure | RCT |
|
| [160] |
Ellinger et al. | 2012 | Blood pressure | Meta-analysis |
|
| [161] |
Oyama et al. | 2010 | Vascular function | RCT |
|
| [162] |
Alañón et al. | 2020 | Vascular function | RCT |
|
| [163] |
Samavat et al. | 2016 | Lipid profile | RCT |
|
| [164] |
Suzuki-Sugihara et al. | 2016 | Redox status | RCT |
|
| [165] |
McDermott et al. | 2020 | PAD | RCT |
|
| [166] |
Kishimoto et al. | 2020 | CAD | RCT |
|
| [167] |
Wang et al. | 2011 | CAD | Meta-analysis |
|
| [168] |
Dural et al. | 2022 | Heart failure | RCT |
|
| [169] |
Anthocyanins | ||||||
Authors | Year | Area of interest | Study | Study design | Main findings | Ref. |
Cassidy et al. | 2013 | Risk of MI | OBS |
|
| [170] |
Cassidy et al. | 2016 | Risk of MI and ischemic stroke | OBS |
|
| [171] |
Alvarez-Suarez et al. | 2014 | Platelet function, lipid profile | RCT |
|
| [172] |
Aboonabi et al. | 2020 | CV risk factors, Platelet function | RCT |
|
| [173] |
Santhakumar et al. | 2015 | Platelet function | RCT |
|
| [174] |
Tian et al. | 2021 | Platelet function | RCT |
|
| [175] |
Jennings et al. | 2012 | Blood pressure | RCT |
|
| [176] |
McKay et al. | 2009 | Blood pressure | RCT |
|
| [177] |
Basu et al. | 2010 | Blood pressure | RCT |
|
| [178] |
Johnson et al. | 2015 | Blood pressure | RCT |
|
| [179] |
Emamat et al. | 2022 | Blood pressure | RCT |
|
| [180] |
Dohadwala et al. | 2010 | Vascular function | RCT |
|
| [181] |
Huang et al. | 2016 | CV risk factors | Meta-analysis |
|
| [182] |
Yang et al. | 2017 | CV risk factors | RCT |
|
| [183] |
Liu et al. | 2016 | Lipid profile | Meta-analysis |
|
| [184] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Gál, R.; Halmosi, R.; Gallyas, F., Jr.; Tschida, M.; Mutirangura, P.; Tóth, K.; Alexy, T.; Czopf, L. Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review. Biomedicines 2023, 11, 2888. https://doi.org/10.3390/biomedicines11112888
Gál R, Halmosi R, Gallyas F Jr., Tschida M, Mutirangura P, Tóth K, Alexy T, Czopf L. Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review. Biomedicines. 2023; 11(11):2888. https://doi.org/10.3390/biomedicines11112888
Chicago/Turabian StyleGál, Roland, Róbert Halmosi, Ferenc Gallyas, Jr., Michael Tschida, Pornthira Mutirangura, Kálmán Tóth, Tamás Alexy, and László Czopf. 2023. "Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review" Biomedicines 11, no. 11: 2888. https://doi.org/10.3390/biomedicines11112888
APA StyleGál, R., Halmosi, R., Gallyas, F., Jr., Tschida, M., Mutirangura, P., Tóth, K., Alexy, T., & Czopf, L. (2023). Resveratrol and beyond: The Effect of Natural Polyphenols on the Cardiovascular System: A Narrative Review. Biomedicines, 11(11), 2888. https://doi.org/10.3390/biomedicines11112888