Deciphering the Role of Polyphenols in Sports Performance: From Nutritional Genomics to the Gut Microbiota toward Phytonutritional Epigenomics
Abstract
:1. Introduction
2. Nutritional Genomics and Sports Performance
Diet, Microbiota, and Sports Performance
3. Polyphenols in Sports Performance: A Holistic View
3.1. Polyphenols and Sports Performance
3.1.1. Curcumin
3.1.2. Resveratrol
3.1.3. Cocoa Flavanols
3.1.4. Quercetin
3.1.5. Other Polyphenols
3.2. Polyphenol Epigenetic Mechanisms and Sports Performance: A Roadmap for Future Practical Applications
4. Conclusions and Future Perspectives
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Williams, J.M.E. Applied Sport Psychology: Personal Growth to Peak Performance; Mayfield Publishing Co.: California City, CA, USA, 1993. [Google Scholar]
- Abbott, A.; Button, C.; Pepping, G.-J.; Collins, D. Unnatural selection: Talent identification and development in sport. Nonlinear Dyn. Psychol. Life Sci. 2005, 9, 61–88. [Google Scholar]
- Guest, N.S.; Horne, J.; Vanderhout, S.; El-Sohemy, A. Sport nutrigenomics: Personalized nutrition for athletic performance. Front. Nutr. 2019, 6, 8. [Google Scholar] [CrossRef] [PubMed]
- Sorrenti, V.; Caudullo, G.; Lucignano, F.; Fortinguerra, S.; Zusso, M.; Giusti, P.; Buriani, A. Personalized sports nutrition: Role of nutrients in athletic performance. In Sports, Exercise, and Nutritional Genomics; Debmalya Barh, I.I.A., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 411–431. [Google Scholar] [CrossRef]
- Lippi, G.; Longo, U.G.; Maffulli, N. Genetics and sports. Br. Med. Bull. 2010, 93, 27–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
- Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front. Nutr. 2018, 5. [Google Scholar] [CrossRef] [Green Version]
- Fraga, C.G.; Galleano, M.; Verstraeten, S.V.; Oteiza, P.I. Basic biochemical mechanisms behind the health benefits of polyphenols. Mol. Asp. Med. 2010, 31, 435–445. [Google Scholar] [CrossRef]
- Leri, M.; Scuto, M.; Ontario, M.L.; Calabrese, V.; Calabrese, E.J.; Bucciantini, M.; Stefani, M. Healthy Effects of Plant Polyphenols: Molecular Mechanisms. Int. J. Mol. Sci. 2020, 21, 1250. [Google Scholar] [CrossRef] [Green Version]
- Kawabata, K.; Yoshioka, Y.; Terao, J. Role of Intestinal Microbiota in the Bioavailability and Physiological Functions of Dietary Polyphenols. Molecules 2019, 24, 370. [Google Scholar] [CrossRef] [Green Version]
- Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
- WHO. Nutrient Requirements and Dietary Guidelines. Available online: https://www.who.int/nutrition/publications/nutrient/en/ (accessed on 8 March 2020).
- Mariman, E.C. Nutrigenomics and nutrigenetics: The ‘omics’ revolution in nutritional science. Biotechnol. Appl. Biochem. 2006, 44, 119–128. [Google Scholar] [CrossRef] [PubMed]
- Fenech, M.; El-Sohemy, A.; Cahill, L.; Ferguson, L.R.; French, T.A.; Tai, E.S.; Milner, J.; Koh, W.P.; Xie, L.; Zucker, M.; et al. Nutrigenetics and nutrigenomics: Viewpoints on the current status and applications in nutrition research and practice. J. Nutrigenet. Nutr. 2011, 4, 69–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Puthucheary, Z.; Skipworth, J.R.; Rawal, J.; Loosemore, M.; Van Someren, K.; Montgomery, H.E. Genetic influences in sport and physical performance. Sports Med. 2011, 41, 845–859. [Google Scholar] [CrossRef] [PubMed]
- Joyner, M.J. Genetic Approaches for Sports Performance: How Far Away Are We? Sports Med. 2019, 49, 199–204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carbone, J.W.; Pasiakos, S.M. Dietary Protein and Muscle Mass: Translating Science to Application and Health Benefit. Nutrients 2019, 11, 1136. [Google Scholar] [CrossRef] [Green Version]
- Merritt, D.C.; Jamnik, J.; El-Sohemy, A. FTO genotype, dietary protein intake, and body weight in a multiethnic population of young adults: A cross-sectional study. Genes Nutr. 2018, 13, 4. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Proud, C.G. The mTOR pathway in the control of protein synthesis. Physiology (Bethesda) 2006, 21, 362–369. [Google Scholar] [CrossRef] [Green Version]
- Kuhne, A.; Kaiser, R.; Schirmer, M.; Heider, U.; Muhlke, S.; Niere, W.; Overbeck, T.; Hohloch, K.; Trumper, L.; Sezer, O.; et al. Genetic polymorphisms in the amino acid transporters LAT1 and LAT2 in relation to the pharmacokinetics and side effects of melphalan. Pharm. Genom. 2007, 17, 505–517. [Google Scholar] [CrossRef]
- Zining, J.; Lu, X.; Caiyun, H.; Yuan, Y. Genetic polymorphisms of mTOR and cancer risk: A systematic review and updated meta-analysis. Oncotarget 2016, 7, 57464–57480. [Google Scholar] [CrossRef] [Green Version]
- Ambrosone, C.B.; Freudenheim, J.L.; Thompson, P.A.; Bowman, E.; Vena, J.E.; Marshall, J.R.; Graham, S.; Laughlin, R.; Nemoto, T.; Shields, P.G. Manganese superoxide dismutase (MnSOD) genetic polymorphisms, dietary antioxidants, and risk of breast cancer. Cancer Res. 1999, 59, 602–606. [Google Scholar]
- Pourvali, K.; Abbasi, M.; Mottaghi, A. Role of Superoxide Dismutase 2 Gene Ala16Val Polymorphism and Total Antioxidant Capacity in Diabetes and its Complications. Avicenna J. Med. Biotechnol. 2016, 8, 48–56. [Google Scholar] [PubMed]
- Pereira, D.S.; Mateo, E.C.; De Queiroz, B.Z.; Assumpcao, A.M.; Miranda, A.S.; Felicio, D.C.; Rocha, N.P.; Da Cruz dos Anjos, D.M.; Pereira, D.A.; Teixeira, A.L.; et al. TNF-alpha, IL6, and IL10 polymorphisms and the effect of physical exercise on inflammatory parameters and physical performance in elderly women. Age (Dordr) 2013, 35, 2455–2463. [Google Scholar] [CrossRef] [Green Version]
- Fedotovskaya, O.N.; Mustafina, L.J.; Popov, D.V.; Vinogradova, O.L.; Ahmetov, I.I. A common polymorphism of the MCT1 gene and athletic performance. Int. J. Sports Physiol. Perform. 2014, 9, 173–180. [Google Scholar] [CrossRef]
- Eckburg, P.B.; Bik, E.M.; Bernstein, C.N.; Purdom, E.; Dethlefsen, L.; Sargent, M.; Gill, S.R.; Nelson, K.E.; Relman, D.A. Diversity of the human intestinal microbial flora. Science 2005, 308, 1635–1638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mariat, D.; Firmesse, O.; Levenez, F.; Guimaraes, V.; Sokol, H.; Dore, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123. [Google Scholar] [CrossRef] [PubMed]
- Arboleya, S.; Watkins, C.; Stanton, C.; Ross, R.P. Gut Bifidobacteria Populations in Human Health and Aging. Front. Microbiol. 2016, 7, 1204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sekirov, I.; Russell, S.L.; Antunes, L.C.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [Green Version]
- Rothschild, D.; Weissbrod, O.; Barkan, E.; Kurilshikov, A.; Korem, T.; Zeevi, D.; Costea, P.I.; Godneva, A.; Kalka, I.N.; Bar, N.; et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018, 555, 210–215. [Google Scholar] [CrossRef]
- Costea, P.I.; Hildebrand, F.; Arumugam, M.; Backhed, F.; Blaser, M.J.; Bushman, F.D.; De Vos, W.M.; Ehrlich, S.D.; Fraser, C.M.; Hattori, M.; et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol. 2018, 3, 8–16. [Google Scholar] [CrossRef] [Green Version]
- Daien, C.I.; Pinget, G.V.; Tan, J.K.; Macia, L. Detrimental Impact of Microbiota-Accessible Carbohydrate-Deprived Diet on Gut and Immune Homeostasis: An Overview. Front. Immunol. 2017, 8, 548. [Google Scholar] [CrossRef] [Green Version]
- Sonnenburg, E.D.; Sonnenburg, J.L. Starving our microbial self: The deleterious consequences of a diet deficient in microbiota-accessible carbohydrates. Cell Metab. 2014, 20, 779–786. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sonnenburg, E.D.; Smits, S.A.; Tikhonov, M.; Higginbottom, S.K.; Wingreen, N.S.; Sonnenburg, J.L. Diet-induced extinctions in the gut microbiota compound over generations. Nature 2016, 529, 212–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fernandes, J.; Su, W.; Rahat-Rozenbloom, S.; Wolever, T.M.; Comelli, E.M. Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans. Nutr. Diabetes 2014, 4, e121. [Google Scholar] [CrossRef] [PubMed]
- Allen, J.M.; Berg Miller, M.E.; Pence, B.D.; Whitlock, K.; Nehra, V.; Gaskins, H.R.; White, B.A.; Fryer, J.D.; Woods, J.A. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. J. Appl. Physiol. (1985) 2015, 118, 1059–1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campbell, S.C.; Wisniewski, P.J., 2nd. Exercise is a Novel Promoter of Intestinal Health and Microbial Diversity. Exerc. Sport Sci. Rev. 2017, 45, 41–47. [Google Scholar] [CrossRef] [PubMed]
- Scheiman, J.; Luber, J.M.; Chavkin, T.A.; MacDonald, T.; Tung, A.; Pham, L.D.; Wibowo, M.C.; Wurth, R.C.; Punthambaker, S.; Tierney, B.T.; et al. Meta-omics analysis of elite athletes identifies a performance-enhancing microbe that functions via lactate metabolism. Nat. Med. 2019, 25, 1104–1109. [Google Scholar] [CrossRef] [PubMed]
- Cardona, F.; Andres-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuno, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Del Rio, D.; Costa, L.G.; Lean, M.E.; Crozier, A. Polyphenols and health: What compounds are involved? Nutr. Metab. Cardiovasc. Dis. 2010, 20, 1–6. [Google Scholar] [CrossRef]
- Koch, W. Dietary Polyphenols-Important Non-Nutrients in the Prevention of Chronic Noncommunicable Diseases. A Systematic Review. Nutrients 2019, 11, 1039. [Google Scholar] [CrossRef] [Green Version]
- Halliwell, B. Dietary polyphenols: Good, bad, or indifferent for your health? Cardiovasc. Res. 2007, 73, 341–347. [Google Scholar] [CrossRef] [PubMed]
- Bors, W.; Michel, C. Chemistry of the antioxidant effect of polyphenols. Ann. N. Y. Acad. Sci. 2002, 957, 57–69. [Google Scholar] [CrossRef] [PubMed]
- Denaro, M.; Smeriglio, A.; Barreca, D.; De Francesco, C.; Occhiuto, C.; Milano, G.; Trombetta, D. Antiviral activity of plants and their isolated bioactive compounds: An update. Phytother. Res. 2019. [Google Scholar] [CrossRef]
- Harms, L.M.; Scalbert, A.; Zamora-Ros, R.; Rinaldi, S.; Jenab, M.; Murphy, N.; Achaintre, D.; Tjonneland, A.; Olsen, A.; Overvad, K.; et al. Plasma polyphenols associated with lower high-sensitivity C-reactive protein concentrations: A cross-sectional study within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. Br. J. Nutr. 2020, 123, 198–208. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Visioli, F. Polyphenols in Sport: Facts or Fads? In Antioxidants in Sport Nutrition; Lamprecht, M., Ed.; CRC Press: Boca Raton, FL, USA, 2015. [Google Scholar]
- Malaguti, M.; Angeloni, C.; Hrelia, S. Polyphenols in exercise performance and prevention of exercise-induced muscle damage. Oxid. Med. Cell Longev. 2013, 2013, 825928. [Google Scholar] [CrossRef] [Green Version]
- Costa Pereira, C.; Duraes, C.; Coelho, R.; Gracio, D.; Silva, M.; Peixoto, A.; Lago, P.; Pereira, M.; Catarino, T.; Pinho, S.; et al. Association between Polymorphisms in Antioxidant Genes and Inflammatory Bowel Disease. PLoS ONE 2017, 12, e0169102. [Google Scholar] [CrossRef] [PubMed]
- Yeh, H.L.; Kuo, L.T.; Sung, F.C.; Yeh, C.C. Association between Polymorphisms of Antioxidant Gene (MnSOD, CAT, and GPx1) and Risk of Coronary Artery Disease. BioMed Res. Int. 2018, 2018, 5086869. [Google Scholar] [CrossRef]
- Vecchio, M.; Curro, M.; Trimarchi, F.; Naccari, S.; Caccamo, D.; Ientile, R.; Barreca, D.; Di Mauro, D. The Oxidative Stress Response in Elite Water Polo Players: Effects of Genetic Background. BioMed Res. Int. 2017, 2017, 7019694. [Google Scholar] [CrossRef] [PubMed]
- Miranda-Vilela, A.L.; Lordelo, G.S.; Akimoto, A.K.; Alves, P.C.; Pereira, L.C.; Klautau-Guimaraes Mde, N.; Grisolia, C.K. Genetic polymorphisms influence runners’ responses to the dietary ingestion of antioxidant supplementation based on pequi oil (Caryocar brasiliense Camb.): A before-after study. Genes Nutr. 2011, 6, 369–395. [Google Scholar] [CrossRef] [Green Version]
- Shunmoogam, N.; Naidoo, P.; Chilton, R. Paraoxonase (PON)-1: A brief overview on genetics, structure, polymorphisms and clinical relevance. Vasc. Health Risk Manag. 2018, 14, 137–143. [Google Scholar] [CrossRef] [Green Version]
- Silberberg, M.; Morand, C.; Mathevon, T.; Besson, C.; Manach, C.; Scalbert, A.; Remesy, C. The bioavailability of polyphenols is highly governed by the capacity of the intestine and of the liver to secrete conjugated metabolites. Eur. J. Nutr. 2006, 45, 88–96. [Google Scholar] [CrossRef]
- Sissung, T.M.; Gardner, E.R.; Gao, R.; Figg, W.D. Pharmacogenetics of membrane transporters: A review of current approaches. Methods Mol. Biol. 2008, 448, 41–62. [Google Scholar] [CrossRef] [PubMed]
- Alvarez, A.I.; Real, R.; Perez, M.; Mendoza, G.; Prieto, J.G.; Merino, G. Modulation of the activity of ABC transporters (P-glycoprotein, MRP2, BCRP) by flavonoids and drug response. J. Pharm. Sci. 2010, 99, 598–617. [Google Scholar] [CrossRef] [PubMed]
- Wu, A.H. Drug metabolizing enzyme activities versus genetic variances for drug of clinical pharmacogenomic relevance. Clin. Proteom. 2011, 8, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barnes, S. Nutritional genomics, polyphenols, diets, and their impact on dietetics. J. Am. Diet. Assoc. 2008, 108, 1888–1895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Drobnic, F.; Riera, J.; Appendino, G.; Togni, S.; Franceschi, F.; Valle, X.; Pons, A.; Tur, J. Reduction of delayed onset muscle soreness by a novel curcumin delivery system (Meriva(R)): A randomised, placebo-controlled trial. J. Int. Soc. Sports Nutr. 2014, 11, 31. [Google Scholar] [CrossRef] [Green Version]
- Ozdal, T.; Sela, D.A.; Xiao, J.; Boyacioglu, D.; Chen, F.; Capanoglu, E. The Reciprocal Interactions between Polyphenols and Gut Microbiota and Effects on Bioaccessibility. Nutrients 2016, 8, 78. [Google Scholar] [CrossRef]
- Cassidy, A.; Minihane, A.M. The role of metabolism (and the microbiome) in defining the clinical efficacy of dietary flavonoids. Am. J. Clin. Nutr. 2017, 105, 10–22. [Google Scholar] [CrossRef] [Green Version]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
- Ma, G.; Chen, Y. Polyphenol supplementation benefits human health via gut microbiota: A systematic review via meta-analysis. J. Funct. Foods 2020, 66, 103829. [Google Scholar] [CrossRef]
- Hassan, F.U.; Rehman, M.S.; Khan, M.S.; Ali, M.A.; Javed, A.; Nawaz, A.; Yang, C. Curcumin as an Alternative Epigenetic Modulator: Mechanism of Action and Potential Effects. Front. Genet. 2019, 10, 514. [Google Scholar] [CrossRef] [Green Version]
- Fernandez-Lazaro, D.; Mielgo-Ayuso, J.; Seco Calvo, J.; Cordova Martinez, A.; Caballero Garcia, A.; Fernandez-Lazaro, C.I. Modulation of Exercise-Induced Muscle Damage, Inflammation, and Oxidative Markers by Curcumin Supplementation in a Physically Active Population: A Systematic Review. Nutrients 2020, 12, 501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Asher, G.N.; Spelman, K. Clinical utility of curcumin extract. Altern. Ther. Health Med. 2013, 19, 20–22. [Google Scholar] [PubMed]
- Takahashi, M.; Suzuki, K.; Kim, H.K.; Otsuka, Y.; Imaizumi, A.; Miyashita, M.; Sakamoto, S. Effects of curcumin supplementation on exercise-induced oxidative stress in humans. Int. J. Sports Med. 2014, 35, 469–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jager, R.; Purpura, M.; Kerksick, C.M. Eight Weeks of a High Dose of Curcumin Supplementation May Attenuate Performance Decrements Following Muscle-Damaging Exercise. Nutrients 2019, 11, 1692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amalraj, A.; Pius, A.; Gopi, S.; Gopi, S. Biological activities of curcuminoids, other biomolecules from turmeric and their derivatives—A review. J. Tradit. Complement. Med. 2017, 7, 205–233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scapagnini, G.S.V. Curcumina; EDRA: Perignano, Italy, 2019; p. 32. [Google Scholar]
- Kunnumakkara, A.B.; Harsha, C.; Banik, K.; Vikkurthi, R.; Sailo, B.L.; Bordoloi, D.; Gupta, S.C.; Aggarwal, B.B. Is curcumin bioavailability a problem in humans: Lessons from clinical trials. Expert Opin. Drug Metab. Toxicol. 2019, 15, 705–733. [Google Scholar] [CrossRef]
- Lopresti, A.L. The Problem of Curcumin and Its Bioavailability: Could Its Gastrointestinal Influence Contribute to Its Overall Health-Enhancing Effects? Adv. Nutr. 2018, 9, 41–50. [Google Scholar] [CrossRef] [Green Version]
- Ren, L.; Zhan, P.; Wang, Q.; Wang, C.; Liu, Y.; Yu, Z.; Zhang, S. Curcumin upregulates the Nrf2 system by repressing inflammatory signaling-mediated Keap1 expression in insulin-resistant conditions. Biochem. Biophys. Res. Commun. 2019, 514, 691–698. [Google Scholar] [CrossRef]
- Alamdari, N.; O’Neal, P.; Hasselgren, P.O. Curcumin and muscle wasting: A new role for an old drug? Nutrition 2009, 25, 125–129. [Google Scholar] [CrossRef] [Green Version]
- Ray Hamidie, R.D.; Yamada, T.; Ishizawa, R.; Saito, Y.; Masuda, K. Curcumin treatment enhances the effect of exercise on mitochondrial biogenesis in skeletal muscle by increasing cAMP levels. Metabolism 2015, 64, 1334–1347. [Google Scholar] [CrossRef]
- Delecroix, B.; Abaidia, A.E.; Leduc, C.; Dawson, B.; Dupont, G. Curcumin and Piperine Supplementation and Recovery Following Exercise Induced Muscle Damage: A Randomized Controlled Trial. J. Sports Sci. Med. 2017, 16, 147–153. [Google Scholar] [PubMed]
- Kawanishi, N.; Kato, K.; Takahashi, M.; Mizokami, T.; Otsuka, Y.; Imaizumi, A.; Shiva, D.; Yano, H.; Suzuki, K. Curcumin attenuates oxidative stress following downhill running-induced muscle damage. Biochem. Biophys. Res. Commun. 2013, 441, 573–578. [Google Scholar] [CrossRef] [PubMed]
- Gault, M.L.; Willems, M.E. Aging, functional capacity and eccentric exercise training. Aging Dis. 2013, 4, 351–363. [Google Scholar] [CrossRef] [PubMed]
- McFarlin, B.K.; Venable, A.S.; Henning, A.L.; Sampson, J.N.; Pennel, K.; Vingren, J.L.; Hill, D.W. Reduced inflammatory and muscle damage biomarkers following oral supplementation with bioavailable curcumin. BBA Clin. 2016, 5, 72–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nicol, L.M.; Rowlands, D.S.; Fazakerly, R.; Kellett, J. Curcumin supplementation likely attenuates delayed onset muscle soreness (DOMS). Eur. J. Appl. Physiol. 2015, 115, 1769–1777. [Google Scholar] [CrossRef] [PubMed]
- Drobnic, F. Curcumin reduces pain in DOMS. Pain 2016, 157, 2390–2391. [Google Scholar] [CrossRef]
- Huang, W.C.; Chiu, W.C.; Chuang, H.L.; Tang, D.W.; Lee, Z.M.; Wei, L.; Chen, F.A.; Huang, C.C. Effect of curcumin supplementation on physiological fatigue and physical performance in mice. Nutrients 2015, 7, 905–921. [Google Scholar] [CrossRef] [Green Version]
- Prasad, S.; Tyagi, A.K.; Aggarwal, B.B. Recent developments in delivery, bioavailability, absorption and metabolism of curcumin: The golden pigment from golden spice. Cancer Res. Treat. 2014, 46, 2–18. [Google Scholar] [CrossRef] [Green Version]
- Gan, Z.; Wei, W.; Li, Y.; Wu, J.; Zhao, Y.; Zhang, L.; Wang, T.; Zhong, X. Curcumin and Resveratrol Regulate Intestinal Bacteria and Alleviate Intestinal Inflammation in Weaned Piglets. Molecules 2019, 24, 1220. [Google Scholar] [CrossRef] [Green Version]
- Jin, T.; Song, Z.; Weng, J.; Fantus, I.G. Curcumin and other dietary polyphenols: Potential mechanisms of metabolic actions and therapy for diabetes and obesity. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E201–E205. [Google Scholar] [CrossRef]
- Zhang, Z.; Chen, Y.; Xiang, L.; Wang, Z.; Xiao, G.G.; Hu, J. Effect of Curcumin on the Diversity of Gut Microbiota in Ovariectomized Rats. Nutrients 2017, 9, 1146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qiao, Y.; Sun, J.; Xia, S.; Tang, X.; Shi, Y.; Le, G. Effects of resveratrol on gut microbiota and fat storage in a mouse model with high-fat-induced obesity. Food Funct. 2014, 5, 1241–1249. [Google Scholar] [CrossRef] [PubMed]
- Di Meo, F.; Margarucci, S.; Galderisi, U.; Crispi, S.; Peluso, G. Curcumin, Gut Microbiota, and Neuroprotection. Nutrients 2019, 11, 2426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dey, P. Gut microbiota in phytopharmacology: A comprehensive overview of concepts, reciprocal interactions, biotransformations and mode of actions. Pharmacol. Res. 2019, 147, 104367. [Google Scholar] [CrossRef] [PubMed]
- Peterson, C.T.; Vaughn, A.R.; Sharma, V.; Chopra, D.; Mills, P.J.; Peterson, S.N.; Sivamani, R.K. Effects of Turmeric and Curcumin Dietary Supplementation on Human Gut Microbiota: A Double-Blind, Randomized, Placebo-Controlled Pilot Study. J. Evid. Based Integr. Med. 2018, 23, 2515690X18790725. [Google Scholar] [CrossRef]
- Wang, J.; Ghosh, S.S.; Ghosh, S. Curcumin improves intestinal barrier function: Modulation of intracellular signaling, and organization of tight junctions. Am. J. Physiol. Cell Physiol. 2017, 312, 438–445. [Google Scholar] [CrossRef]
- Lamming, D.W.; Wood, J.G.; Sinclair, D.A. Small molecules that regulate lifespan: Evidence for xenohormesis. Mol. Microbiol. 2004, 53, 1003–1009. [Google Scholar] [CrossRef]
- Howitz, K.T.; Sinclair, D.A. Xenohormesis: Sensing the chemical cues of other species. Cell 2008, 133, 387–391. [Google Scholar] [CrossRef] [Green Version]
- Arola-Arnal, A.; Cruz-Carrion, A.; Torres-Fuentes, C.; Avila-Roman, J.; Aragones, G.; Mulero, M.; Bravo, F.I.; Muguerza, B.; Arola, L.; Suarez, M. Chrononutrition and Polyphenols: Roles and Diseases. Nutrients 2019, 11, 2602. [Google Scholar] [CrossRef] [Green Version]
- Gambini, J.; Ingles, M.; Olaso, G.; Lopez-Grueso, R.; Bonet-Costa, V.; Gimeno-Mallench, L.; Mas-Bargues, C.; Abdelaziz, K.M.; Gomez-Cabrera, M.C.; Vina, J.; et al. 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] [Green Version]
- Anisimova, N.Y.; Kiselevsky, M.V.; Sosnov, A.V.; Sadovnikov, S.V.; Stankov, I.N.; Gakh, A.A. Trans-, cis-, and dihydro-resveratrol: A comparative study. Chem. Cent. J. 2011, 5, 88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Howitz, K.T.; Bitterman, K.J.; Cohen, H.Y.; Lamming, D.W.; Lavu, S.; Wood, J.G.; Zipkin, R.E.; Chung, P.; Kisielewski, A.; Zhang, L.L.; et al. Small molecule activators of sirtuins extend Saccharomyces cerevisiae lifespan. Nature 2003, 425, 191–196. [Google Scholar] [CrossRef] [PubMed]
- Csiszar, A.; Labinskyy, N.; Pinto, J.T.; Ballabh, P.; Zhang, H.; Losonczy, G.; Pearson, K.; De Cabo, R.; Pacher, P.; Zhang, C.; et al. Resveratrol induces mitochondrial biogenesis in endothelial cells. Am. J. Physiol. Heart Circ. Physiol. 2009, 297, H13–H20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, K.; Zhou, R.; Wang, B.; Mi, M.T. Effect of resveratrol on glucose control and insulin sensitivity: A meta-analysis of 11 randomized controlled trials. Am. J. Clin. Nutr. 2014, 99, 1510–1519. [Google Scholar] [CrossRef] [Green Version]
- Berman, A.Y.; Motechin, R.A.; Wiesenfeld, M.Y.; Holz, M.K. The therapeutic potential of resveratrol: A review of clinical trials. NPJ Precis. Oncol. 2017, 1. [Google Scholar] [CrossRef] [Green Version]
- Farzaei, M.H.; Rahimi, R.; Nikfar, S.; Abdollahi, M. Effect of resveratrol on cognitive and memory performance and mood: A meta-analysis of 225 patients. Pharmacol. Res. 2018, 128, 338–344. [Google Scholar] [CrossRef]
- Kulashekar, M.; Stom, S.M.; Peuler, J.D. Resveratrol’s Potential in the Adjunctive Management of Cardiovascular Disease, Obesity, Diabetes, Alzheimer Disease, and Cancer. J. Am. Osteopath. Assoc. 2018, 118, 596–605. [Google Scholar] [CrossRef] [Green Version]
- Walle, T. Bioavailability of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 9–15. [Google Scholar] [CrossRef]
- Wang, P.; Sang, S. Metabolism and pharmacokinetics of resveratrol and pterostilbene. Biofactors 2018, 44, 16–25. [Google Scholar] [CrossRef]
- Walle, T.; Hsieh, F.; DeLegge, M.H.; Oatis, J.E., Jr.; Walle, U.K. High absorption but very low bioavailability of oral resveratrol in humans. Drug Metab. Dispos. 2004, 32, 1377–1382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Scapagnini, G.S.V. Il Resveratrolo; EDRA: Perignano, Italy, 2019; p. 28. [Google Scholar]
- Du, Q.H.; Peng, C.; Zhang, H. Polydatin: A review of pharmacology and pharmacokinetics. Pharm. Biol. 2013, 51, 1347–1354. [Google Scholar] [CrossRef] [PubMed]
- Ventura-Clapier, R. Potentiating exercise training with resveratrol. J. Physiol. 2012, 590, 3215–3216. [Google Scholar] [CrossRef] [PubMed]
- Dolinsky, V.W.; Jones, K.E.; Sidhu, R.S.; Haykowsky, M.; Czubryt, M.P.; Gordon, T.; Dyck, J.R. Improvements in skeletal muscle strength and cardiac function induced by resveratrol during exercise training contribute to enhanced exercise performance in rats. J. Physiol. 2012, 590, 2783–2799. [Google Scholar] [CrossRef] [PubMed]
- Bennett, B.T.; Mohamed, J.S.; Alway, S.E. Effects of resveratrol on the recovery of muscle mass following disuse in the plantaris muscle of aged rats. PLoS ONE 2013, 8, e83518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rodriguez-Bies, E.; Tung, B.T.; Navas, P.; Lopez-Lluch, G. Resveratrol primes the effects of physical activity in old mice. Br. J. Nutr. 2016, 116, 979–988. [Google Scholar] [CrossRef] [Green Version]
- Polley, K.R.; Jenkins, N.; O’Connor, P.; McCully, K. Influence of exercise training with resveratrol supplementation on skeletal muscle mitochondrial capacity. Appl. Physiol. Nutr. Metab. 2016, 41, 26–32. [Google Scholar] [CrossRef] [Green Version]
- Kan, N.W.; Lee, M.C.; Tung, Y.T.; Chiu, C.C.; Huang, C.C.; Huang, W.C. The Synergistic Effects of Resveratrol combined with Resistant Training on Exercise Performance and Physiological Adaption. Nutrients 2018, 10, 1360. [Google Scholar] [CrossRef] [Green Version]
- Carpene, C.; Les, F.; Casedas, G.; Peiro, C.; Fontaine, J.; Chaplin, A.; Mercader, J.; Lopez, V. Resveratrol Anti-Obesity Effects: Rapid Inhibition of Adipocyte Glucose Utilization. Antioxidants (Basel) 2019, 8, 74. [Google Scholar] [CrossRef] [Green Version]
- Wong, R.H.X.; Howe, P.R.C. Resveratrol Counteracts Insulin Resistance-Potential Role of the Circulation. Nutrients 2018, 10, 1160. [Google Scholar] [CrossRef] [Green Version]
- Ramirez-Garza, S.L.; Laveriano-Santos, E.P.; Marhuenda-Munoz, M.; Storniolo, C.E.; Tresserra-Rimbau, A.; Vallverdu-Queralt, A.; Lamuela-Raventos, R.M. Health Effects of Resveratrol: Results from Human Intervention Trials. Nutrients 2018, 10, 1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chaplin, A.; Carpene, C.; Mercader, J. Resveratrol, Metabolic Syndrome, and Gut Microbiota. Nutrients 2018, 10, 1651. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, J.; Zhang, C.; Kim, M.; Su, Y.; Qin, L.; Dong, J.; Zhou, Y.; Ding, S. Early potential effects of resveratrol supplementation on skeletal muscle adaptation involved in exercise-induced weight loss in obese mice. BMB Rep. 2018, 51, 200–205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Zhou, L.; Xiao, X.; Zhang, Q.; Zheng, J.; Deng, M. Deciphering the Anti-obesity Benefits of Resveratrol: The “Gut Microbiota-Adipose Tissue” Axis. Front. Endocrinol. (Lausanne) 2019, 10, 413. [Google Scholar] [CrossRef] [Green Version]
- Naito, Y.; Uchiyama, K.; Takagi, T. A next-generation beneficial microbe: Akkermansia muciniphila. J. Clin. Biochem. Nutr. 2018, 63, 33–35. [Google Scholar] [CrossRef] [Green Version]
- Wood, G.A.R.; Lass, R. Cocoa; John Wiley & Sons: Hoboken, NJ, USA, 2008. [Google Scholar]
- Young, A.M. The chocolate Tree: A Natural History Of Cacao; Smithsonian Institution Press: Washington, DC, USA, 1994. [Google Scholar]
- Fraga, C.G.; Litterio, M.C.; Prince, P.D.; Calabro, V.; Piotrkowski, B.; Galleano, M. Cocoa flavanols: Effects on vascular nitric oxide and blood pressure. J. Clin. Biochem. Nutr. 2011, 48, 63–67. [Google Scholar] [CrossRef] [Green Version]
- Socci, V.; Tempesta, D.; Desideri, G.; De Gennaro, L.; Ferrara, M. Enhancing Human Cognition with Cocoa Flavonoids. Front. Nutr. 2017, 4, 19. [Google Scholar] [CrossRef] [Green Version]
- Blumberg, J.B.; Ding, E.L.; Dixon, R.; Pasinetti, G.M.; Villarreal, F. The science of cocoa flavanols: Bioavailability, emerging evidence, and proposed mechanisms. Adv. Nutr. 2014, 5, 547–549. [Google Scholar] [CrossRef] [Green Version]
- Oracz, J.; Nebesny, E.; Zyzelewicz, D.; Budryn, G.; Luzak, B. Bioavailability and metabolism of selected cocoa bioactive compounds: A comprehensive review. Crit. Rev. Food Sci. Nutr. 2019, 24, 1–39. [Google Scholar] [CrossRef]
- Jaramillo Flores, M.E. Cocoa Flavanols: Natural Agents with Attenuating Effects on Metabolic Syndrome Risk Factors. Nutrients 2019, 11, 751. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mena, P.; Bresciani, L.; Brindani, N.; Ludwig, I.A.; Pereira-Caro, G.; Angelino, D.; Llorach, R.; Calani, L.; Brighenti, F.; Clifford, M.N.; et al. Phenyl-gamma-valerolactones and phenylvaleric acids, the main colonic metabolites of flavan-3-ols: Synthesis, analysis, bioavailability, and bioactivity. Nat. Prod. Rep. 2019, 36, 714–752. [Google Scholar] [CrossRef] [PubMed]
- Decroix, L.; Soares, D.D.; Meeusen, R.; Heyman, E.; Tonoli, C. Cocoa Flavanol Supplementation and Exercise: A Systematic Review. Sports Med. 2018, 48, 867–892. [Google Scholar] [CrossRef] [PubMed]
- De Carvalho, F.G.; Fisher, M.G.; Thornley, T.T.; Roemer, K.; Pritchett, R.; Freitas, E.C.; Pritchett, K. Cocoa flavanol effects on markers of oxidative stress and recovery after muscle damage protocol in elite rugby players. Nutrition 2019, 62, 47–51. [Google Scholar] [CrossRef]
- Decroix, L.; Tonoli, C.; Lespagnol, E.; Balestra, C.; Descat, A.; Drittij-Reijnders, M.J.; Blackwell, J.R.; Stahl, W.; Jones, A.M.; Weseler, A.R.; et al. One-week cocoa flavanol intake increases prefrontal cortex oxygenation at rest and during moderate-intensity exercise in normoxia and hypoxia. J. Appl. Physiol. (1985) 2018, 125, 8–18. [Google Scholar] [CrossRef]
- Li, L.; Somerset, S. Associations between Flavonoid Intakes and Gut Microbiota in a Group of Adults with Cystic Fibrosis. Nutrients 2018, 10, 1264. [Google Scholar] [CrossRef] [Green Version]
- Jang, S.; Sun, J.; Chen, P.; Lakshman, S.; Molokin, A.; Harnly, J.M.; Vinyard, B.T.; Urban, J.F.; Davis, C.D.; Solano-Aguilar, G. Flavanol-Enriched Cocoa Powder Alters the Intestinal Microbiota, Tissue and Fluid Metabolite Profiles, and Intestinal Gene Expression in Pigs1234. J. Nutr. 2016, 146, 673–680. [Google Scholar] [CrossRef] [Green Version]
- Strat, K.M.; Rowley, T.J.t.; Smithson, A.T.; Tessem, J.S.; Hulver, M.W.; Liu, D.; Davy, B.M.; Davy, K.P.; Neilson, A.P. Mechanisms by which cocoa flavanols improve metabolic syndrome and related disorders. J. Nutr. Biochem. 2016, 35, 1–21. [Google Scholar] [CrossRef]
- Tzounis, X.; Rodriguez-Mateos, A.; Vulevic, J.; Gibson, G.R.; Kwik-Uribe, C.; Spencer, J.P. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 2011, 93, 62–72. [Google Scholar] [CrossRef] [Green Version]
- Andres-Lacueva, C.; Monagas, M.; Khan, N.; Izquierdo-Pulido, M.; Urpi-Sarda, M.; Permanyer, J.; Lamuela-Raventos, R.M. Flavanol and flavonol contents of cocoa powder products: Influence of the manufacturing process. J. Agric. Food Chem. 2008, 56, 3111–3117. [Google Scholar] [CrossRef]
- Boots, A.W.; Haenen, G.R.; Bast, A. Health effects of quercetin: From antioxidant to nutraceutical. Eur. J. Pharmacol. 2008, 585, 325–337. [Google Scholar] [CrossRef] [PubMed]
- Cooray, H.C.; Janvilisri, T.; Van Veen, H.W.; Hladky, S.B.; Barrand, M.A. Interaction of the breast cancer resistance protein with plant polyphenols. Biochem. Biophys. Res. Commun. 2004, 317, 269–275. [Google Scholar] [CrossRef] [PubMed]
- Carrasco-Pozo, C.; Cires, M.J.; Gotteland, M. Quercetin and Epigallocatechin Gallate in the Prevention and Treatment of Obesity: From Molecular to Clinical Studies. J. Med. Food 2019, 22, 753–770. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, Inflammation and Immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef]
- Davis, J.M.; Murphy, E.A.; Carmichael, M.D.; Davis, B. Quercetin increases brain and muscle mitochondrial biogenesis and exercise tolerance. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2009, 296, R1071–R1077. [Google Scholar] [CrossRef] [Green Version]
- Davis, J.M.; Murphy, E.A.; Carmichael, M.D. Effects of the dietary flavonoid quercetin upon performance and health. Curr. Sports Med. Rep. 2009, 8, 206–213. [Google Scholar] [CrossRef]
- Somerville, V.; Bringans, C.; Braakhuis, A. Polyphenols and Performance: A Systematic Review and Meta-Analysis. Sports Med. 2017, 47, 1589–1599. [Google Scholar] [CrossRef]
- Askari, G.; Ghiasvand, R.; Paknahad, Z.; Karimian, J.; Rabiee, K.; Sharifirad, G.; Feizi, A. The effects of quercetin supplementation on body composition, exercise performance and muscle damage indices in athletes. Int. J. Prev. Med. 2013, 4, 21–26. [Google Scholar]
- Scholten, S.D.; Sergeev, I.N. Long-term quercetin supplementation reduces lipid peroxidation but does not improve performance in endurance runners. Open Access J. Sports Med. 2013, 4, 53–61. [Google Scholar] [CrossRef] [Green Version]
- Patrizio, F.; Ditroilo, M.; Felici, F.; Duranti, G.; De Vito, G.; Sabatini, S.; Sacchetti, M.; Bazzucchi, I. The acute effect of Quercetin on muscle performance following a single resistance training session. Eur. J. Appl. Physiol. 2018, 118, 1021–1031. [Google Scholar] [CrossRef]
- Bazzucchi, I.; Patrizio, F.; Ceci, R.; Duranti, G.; Sgro, P.; Sabatini, S.; Di Luigi, L.; Sacchetti, M.; Felici, F. The Effects of Quercetin Supplementation on Eccentric Exercise-Induced Muscle Damage. Nutrients 2019, 11, 205. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riva, A.; Vitale, J.A.; Belcaro, G.; Hu, S.; Feragalli, B.; Vinciguerra, G.; Cacchio, M.; Bonanni, E.; Giacomelli, L.; Eggenhoffner, R.; et al. Quercetin phytosome(R) in triathlon athletes: A pilot registry study. Minerva. Med. 2018, 109, 285–289. [Google Scholar] [CrossRef] [PubMed]
- Andres, S.; Pevny, S.; Ziegenhagen, R.; Bakhiya, N.; Schafer, B.; Hirsch-Ernst, K.I.; Lampen, A. Safety Aspects of the Use of Quercetin as a Dietary Supplement. Mol. Nutr. Food Res. 2018, 62, 1700447. [Google Scholar] [CrossRef] [PubMed]
- Egert, S.; Wolffram, S.; Bosy-Westphal, A.; Boesch-Saadatmandi, C.; Wagner, A.E.; Frank, J.; Rimbach, G.; Mueller, M.J. Daily quercetin supplementation dose-dependently increases plasma quercetin concentrations in healthy humans. J. Nutr. 2008, 138, 1615–1621. [Google Scholar] [CrossRef] [Green Version]
- Kressler, J.; Millard-Stafford, M.; Warren, G.L. Quercetin and endurance exercise capacity: A systematic review and meta-analysis. Med. Sci. Sports Exerc. 2011, 43, 2396–2404. [Google Scholar] [CrossRef]
- Jowko, E.; Dlugolecka, B.; Makaruk, B.; Cieslinski, I. The effect of green tea extract supplementation on exercise-induced oxidative stress parameters in male sprinters. Eur J Nutr. 2015, 54, 783–791. [Google Scholar] [CrossRef] [Green Version]
- Da Silva, W.; Machado, A.S.; Souza, M.A.; Mello-Carpes, P.B.; Carpes, F.P. Effect of green tea extract supplementation on exercise-induced delayed onset muscle soreness and muscular damage. Physiol. Behav. 2018, 194, 77–82. [Google Scholar] [CrossRef]
- Sadowska-Krepa, E.; Domaszewski, P.; Pokora, I.; Zebrowska, A.; Gdanska, A.; Podgorski, T. Effects of medium-term green tea extract supplementation combined with CrossFit workout on blood antioxidant status and serum brain-derived neurotrophic factor in young men: A pilot study. J. Int. Soc. Sports Nutr. 2019, 16, 13. [Google Scholar] [CrossRef] [Green Version]
- Machado, A.S.; Da Silva, W.; Souza, M.A.; Carpes, F.P. Green Tea Extract Preserves Neuromuscular Activation and Muscle Damage Markers in Athletes Under Cumulative Fatigue. Front. Physiol. 2018, 9, 1137. [Google Scholar] [CrossRef] [Green Version]
- Jowko, E.; Sacharuk, J.; Balasinska, B.; Ostaszewski, P.; Charmas, M.; Charmas, R. Green tea extract supplementation gives protection against exercise-induced oxidative damage in healthy men. Nutr. Res. 2011, 31, 813–821. [Google Scholar] [CrossRef]
- Park, C.H.; Kwak, Y.S.; Seo, H.K.; Kim, H.Y. Assessing the Values of Blueberries Intake on Exercise Performance, TAS, and Inflammatory Factors. Iran. J. Public Health 2018, 47, 27–32. [Google Scholar] [PubMed]
- Curtis, P.J.; Van der Velpen, V.; Berends, L.; Jennings, A.; Feelisch, M.; Umpleby, A.M.; Evans, M.; Fernandez, B.O.; Meiss, M.S.; Minnion, M.; et al. 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] [Green Version]
- McLeay, Y.; Barnes, M.J.; Mundel, T.; Hurst, S.M.; Hurst, R.D.; Stannard, S.R. Effect of New Zealand blueberry consumption on recovery from eccentric exercise-induced muscle damage. J. Int. Soc. Sports Nutr. 2012, 9, 19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.; Keirsey, K.I.; Kirkland, R.; Grunewald, Z.I.; Fischer, J.G.; De La Serre, C.B. Blueberry Supplementation Influences the Gut Microbiota, Inflammation, and Insulin Resistance in High-Fat-Diet-Fed Rats. J. Nutr. 2018, 148, 209–219. [Google Scholar] [CrossRef] [Green Version]
- Chandra, P.; Rathore, A.S.; Kay, K.L.; Everhart, J.L.; Curtis, P.; Burton-Freeman, B.; Cassidy, A.; Kay, C.D. Contribution of Berry Polyphenols to the Human Metabolome. Molecules 2019, 24, 4220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pajuelo, D.; Quesada, H.; Diaz, S.; Fernandez-Iglesias, A.; Arola-Arnal, A.; Blade, C.; Salvado, J.; Arola, L. Chronic dietary supplementation of proanthocyanidins corrects the mitochondrial dysfunction of brown adipose tissue caused by diet-induced obesity in Wistar rats. Br. J. Nutr. 2012, 107, 170–178. [Google Scholar] [CrossRef]
- Zhang, L.; Carmody, R.N.; Kalariya, H.M.; Duran, R.M.; Moskal, K.; Poulev, A.; Kuhn, P.; Tveter, K.M.; Turnbaugh, P.J.; Raskin, I.; et al. Grape proanthocyanidin-induced intestinal bloom of Akkermansia muciniphila is dependent on its baseline abundance and precedes activation of host genes related to metabolic health. J. Nutr. Biochem. 2018, 56, 142–151. [Google Scholar] [CrossRef]
- Casanova-Marti, A.; Serrano, J.; Portune, K.J.; Sanz, Y.; Blay, M.T.; Terra, X.; Ardevol, A.; Pinent, M. Grape seed proanthocyanidins influence gut microbiota and enteroendocrine secretions in female rats. Food Funct. 2018, 9, 1672–1682. [Google Scholar] [CrossRef] [Green Version]
- Masumoto, S.; Terao, A.; Yamamoto, Y.; Mukai, T.; Miura, T.; Shoji, T. Non-absorbable apple procyanidins prevent obesity associated with gut microbial and metabolomic changes. Sci. Rep. 2016, 6, 31208. [Google Scholar] [CrossRef]
- Bialonska, D.; Kasimsetty, S.G.; Schrader, K.K.; Ferreira, D. The effect of pomegranate (Punica granatum L.) byproducts and ellagitannins on the growth of human gut bacteria. J. Agric. Food Chem. 2009, 57, 8344–8349. [Google Scholar] [CrossRef]
- Tabasco, R.; Sanchez-Patan, F.; Monagas, M.; Bartolome, B.; Victoria Moreno-Arribas, M.; Pelaez, C.; Requena, T. Effect of grape polyphenols on lactic acid bacteria and bifidobacteria growth: Resistance and metabolism. Food Microbiol. 2011, 28, 1345–1352. [Google Scholar] [CrossRef] [PubMed]
- Aldret Rl EdD, A.C.; Bellar D PhD, F.C.R. A Double-Blind, Cross-Over Study to Examine the Effects of Maritime Pine Extract on Exercise Performance and Postexercise Inflammation, Oxidative Stress, Muscle Soreness, and Damage. J. Diet. Suppl. 2019, 19, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Vinciguerra, G.; Belcaro, G.; Bonanni, E.; Cesarone, M.R.; Rotondi, V.; Ledda, A.; Hosoi, M.; Dugall, M.; Cacchio, M.; Cornelli, U. Evaluation of the effects of supplementation with Pycnogenol(R) on fitness in normal subjects with the Army Physical Fitness Test and in performances of athletes in the 100-minute triathlon. J. Sports Med. Phys. Fitness 2013, 53, 644–654. [Google Scholar] [PubMed]
- Piccolella, S.; Fiorentino, A.; Pacifico, S.; D’Abrosca, B.; Uzzo, P.; Monaco, P. Antioxidant properties of sour cherries (Prunus cerasus L.): Role of colorless phytochemicals from the methanolic extract of ripe fruits. J. Agric. Food Chem. 2008, 56, 1928–1935. [Google Scholar] [CrossRef] [PubMed]
- Bell, P.G.; Walshe, I.H.; Davison, G.W.; Stevenson, E.; Howatson, G. Montmorency cherries reduce the oxidative stress and inflammatory responses to repeated days high-intensity stochastic cycling. Nutrients 2014, 6, 829–843. [Google Scholar] [CrossRef] [Green Version]
- McCormick, R.; Peeling, P.; Binnie, M.; Dawson, B.; Sim, M. Effect of tart cherry juice on recovery and next day performance in well-trained Water Polo players. J. Int. Soc. Sports Nutr. 2016, 13, 41. [Google Scholar] [CrossRef] [Green Version]
- Bell, P.G.; McHugh, M.P.; Stevenson, E.; Howatson, G. The role of cherries in exercise and health. Scand. J. Med. Sci. Sports 2014, 24, 477–490. [Google Scholar] [CrossRef]
- Howatson, G.; McHugh, M.P.; Hill, J.A.; Brouner, J.; Jewell, A.P.; Van Someren, K.A.; Shave, R.E.; Howatson, S.A. Influence of tart cherry juice on indices of recovery following marathon running. Scand. J. Med. Sci. Sports 2010, 20, 843–852. [Google Scholar] [CrossRef]
- Keane, K.M.; Bailey, S.J.; Vanhatalo, A.; Jones, A.M.; Howatson, G. Effects of montmorency tart cherry (L. Prunus Cerasus) consumption on nitric oxide biomarkers and exercise performance. Scand. J. Med. Sci. Sports 2018, 28, 1746–1756. [Google Scholar] [CrossRef]
- Oh, J.K.; Shin, Y.O.; Yoon, J.H.; Kim, S.H.; Shin, H.C.; Hwang, H.J. Effect of supplementation with Ecklonia cava polyphenol on endurance performance of college students. Int. J. Sport Nutr. Exerc. Metab. 2010, 20, 72–79. [Google Scholar] [CrossRef]
- Weinhold, B. Epigenetics: The science of change. Environ. Health Perspect. 2006, 114, A160–A167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.; Kutateladze, T.G. Diet and the epigenome. Nat. Commun. 2018, 9, 3375. [Google Scholar] [CrossRef] [PubMed]
- Barron-Cabrera, E.; Ramos-Lopez, O.; Gonzalez-Becerra, K.; Riezu-Boj, J.I.; Milagro, F.I.; Martinez-Lopez, E.; Martinez, J.A. Epigenetic Modifications as Outcomes of Exercise Interventions Related to Specific Metabolic Alterations: A Systematic Review. Lifestyle Genom. 2019, 12, 25–44. [Google Scholar] [CrossRef]
- Ehlert, T.; Simon, P.; Moser, D.A. Epigenetics in sports. Sports Med. 2013, 43, 93–110. [Google Scholar] [CrossRef] [PubMed]
- Voisin, S.; Harvey, N.R.; Haupt, L.M.; Griffiths, L.R.; Ashton, K.J.; Coffey, V.G.; Doering, T.M.; Thompson, J.M.; Benedict, C.; Cedernaes, J.; et al. An epigenetic clock for human skeletal muscle. J. Cachexia Sarcopenia Muscle 2020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ntanasis-Stathopoulos, J.; Tzanninis, J.G.; Philippou, A.; Koutsilieris, M. Epigenetic regulation on gene expression induced by physical exercise. J. Musculoskelet. Neuronal. Interact. 2013, 13, 133–146. [Google Scholar]
- Angelini, F.; Pagano, F.; Bordin, A.; Milan, M.; Chimenti, I.; Peruzzi, M.; Valenti, V.; Marullo, A.; Schirone, L.; Palmerio, S. The impact of environmental factors in influencing epigenetics related to oxidative states in the cardiovascular system. Oxid. Med. Cell. Longev. 2017, 2017, 2712751. [Google Scholar] [CrossRef]
- Abdul, Q.A.; Yu, B.P.; Chung, H.Y.; Jung, H.A.; Choi, J.S. Epigenetic modifications of gene expression by lifestyle and environment. Arch. Pharmacal. Res. 2017, 40, 1219–1237. [Google Scholar] [CrossRef]
- Liu, C.D.; Yang, L.; Pu, H.Z.; Yang, Q.; Huang, W.Y.; Zhao, X.; Zhu, L.; Zhang, S.H. Epigenetics regulates gene expression patterns of skeletal muscle induced by physical exercise. Yi Chuan 2017, 39, 888–896. [Google Scholar] [CrossRef]
- Jacques, M.; Hiam, D.; Craig, J.; Barres, R.; Eynon, N.; Voisin, S. Epigenetic changes in healthy human skeletal muscle following exercise- a systematic review. Epigenetics 2019, 14, 633–648. [Google Scholar] [CrossRef]
- Ayissi, V.B.; Ebrahimi, A.; Schluesenner, H. Epigenetic effects of natural polyphenols: A focus on SIRT1-mediated mechanisms. Mol. Nutr. Food Res. 2014, 58, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Pan, M.H.; Lai, C.S.; Wu, J.C.; Ho, C.T. Epigenetic and disease targets by polyphenols. Curr. Pharm. Des. 2013, 19, 6156–6185. [Google Scholar] [CrossRef] [PubMed]
- Malireddy, S.; Kotha, S.R.; Secor, J.D.; Gurney, T.O.; Abbott, J.L.; Maulik, G.; Maddipati, K.R.; Parinandi, N.L. Phytochemical antioxidants modulate mammalian cellular epigenome: Implications in health and disease. Antioxid. Redox Signal. 2012, 17, 327–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Russell, G.; Lightman, S. The human stress response. Nat. Rev. Endocrinol. 2019, 15, 525–534. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, J.K. Abiotic Stress Signaling and Responses in Plants. Cell 2016, 167, 313–324. [Google Scholar] [CrossRef] [Green Version]
- Voelter-Mahlknecht, S. Epigenetic associations in relation to cardiovascular prevention and therapeutics. Clin. Epigenetics 2016, 8, 4. [Google Scholar] [CrossRef] [Green Version]
- Crescenti, A.; Sola, R.; Valls, R.M.; Caimari, A.; Del Bas, J.M.; Anguera, A.; Angles, N.; Arola, L. Cocoa consumption alters the global DNA methylation of peripheral leukocytes in humans with cardiovascular disease risk factors: A randomized controlled trial. PLoS ONE 2013, 8, e65744. [Google Scholar] [CrossRef]
- Szarc vel Szic, K.; Declerck, K.; Vidakovic, M.; Vanden Berghe, W. From inflammaging to healthy aging by dietary lifestyle choices: Is epigenetics the key to personalized nutrition? Clin. Epigenet. 2015, 7, 33. [Google Scholar] [CrossRef] [Green Version]
- Cordero-Herrera, I.; Chen, X.; Ramos, S.; Devaraj, S. (−)-Epicatechin attenuates high-glucose-induced inflammation by epigenetic modulation in human monocytes. Eur. J. Nutr. 2017, 56, 1369–1373. [Google Scholar] [CrossRef]
- Milenkovic, D.; Declerck, K.; Guttman, Y.; Kerem, Z.; Claude, S.; Weseler, A.R.; Bast, A.; Schroeter, H.; Morand, C.; Berghe, W.V. (−)-Epicatechin metabolites promote vascular health through epigenetic reprogramming of endothelial-immune cell signaling and reversing systemic low-grade inflammation. Biochem. Pharmacol. 2020, 173, 113699. [Google Scholar] [CrossRef]
- Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Treatment of cardiovascular pathology with epigenetically active agents: Focus on natural and synthetic inhibitors of DNA methylation and histone deacetylation. Int. J. Cardiol. 2017, 227, 66–82. [Google Scholar] [CrossRef] [PubMed]
- Fang, M.; Chen, D.; Yang, C.S. Dietary polyphenols may affect DNA methylation. J. Nutr. 2007, 137, 223S–228S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ginsburg, I.; Kohen, R.; Koren, E. Saliva: A ‘solubilizer’ of lipophilic antioxidant polyphenols. Oral. Dis. 2013, 19, 321–322. [Google Scholar] [CrossRef] [PubMed]
- Metere, A.; Giacomelli, L. Absorption, metabolism and protective role of fruits and vegetables polyphenols against gastric cancer. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 5850–5858. [Google Scholar] [CrossRef]
- D’Archivio, M.; Filesi, C.; Vari, R.; Scazzocchio, B.; Masella, R. Bioavailability of the polyphenols: Status and controversies. Int. J. Mol. Sci. 2010, 11, 1321–1342. [Google Scholar] [CrossRef]
- Kay, C.D.; Mazza, G.; Holub, B.J.; Wang, J. Anthocyanin metabolites in human urine and serum. Br. J. Nutr. 2004, 91, 933–942. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Luca, S.V.; Macovei, I.; Bujor, A.; Miron, A.; Skalicka-Wozniak, K.; Aprotosoaie, A.C.; Trifan, A. Bioactivity of dietary polyphenols: The role of metabolites. Crit. Rev. Food Sci. Nutr. 2020, 60, 626–659. [Google Scholar] [CrossRef]
- Scalbert, A.; Williamson, G. Dietary intake and bioavailability of polyphenols. J. Nutr. 2000, 130, 2073S–2085S. [Google Scholar] [CrossRef]
- Yang, Q.; Liang, Q.; Balakrishnan, B.; Belobrajdic, D.P.; Feng, Q.J.; Zhang, W. Role of Dietary Nutrients in the Modulation of Gut Microbiota: A Narrative Review. Nutrients 2020, 12, 381. [Google Scholar] [CrossRef] [Green Version]
Average Daily Dose | Overall Benefits | References | |
---|---|---|---|
Curcumin | 80–200 mg |
| [59,65,68] |
Resveratrol | 100–500 mg |
| [100,109,114,119] |
Cocoa Flavanols | 200–500 mg |
| [131,132] |
Quercetin | 200–1000 mg |
| [146,148,150,153] |
Green tea extract | 250–1000 mg |
| [156,157,158] |
Blueberry | 75–150 g |
| [160,161] |
Pycnogenol® | 100–800 mg |
| [171] |
Montmorency cherry juice | 30 mL |
| [174,177] |
Ecklonia cava polyphenols | 40 mg |
| [178] |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sorrenti, V.; Fortinguerra, S.; Caudullo, G.; Buriani, A. Deciphering the Role of Polyphenols in Sports Performance: From Nutritional Genomics to the Gut Microbiota toward Phytonutritional Epigenomics. Nutrients 2020, 12, 1265. https://doi.org/10.3390/nu12051265
Sorrenti V, Fortinguerra S, Caudullo G, Buriani A. Deciphering the Role of Polyphenols in Sports Performance: From Nutritional Genomics to the Gut Microbiota toward Phytonutritional Epigenomics. Nutrients. 2020; 12(5):1265. https://doi.org/10.3390/nu12051265
Chicago/Turabian StyleSorrenti, Vincenzo, Stefano Fortinguerra, Giada Caudullo, and Alessandro Buriani. 2020. "Deciphering the Role of Polyphenols in Sports Performance: From Nutritional Genomics to the Gut Microbiota toward Phytonutritional Epigenomics" Nutrients 12, no. 5: 1265. https://doi.org/10.3390/nu12051265
APA StyleSorrenti, V., Fortinguerra, S., Caudullo, G., & Buriani, A. (2020). Deciphering the Role of Polyphenols in Sports Performance: From Nutritional Genomics to the Gut Microbiota toward Phytonutritional Epigenomics. Nutrients, 12(5), 1265. https://doi.org/10.3390/nu12051265