Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview
Abstract
:1. Introduction
2. Polyphenols: Anti-Senescence Nutraceuticals
3. Action of Polyphenols on Some Hallmarks of Aging
3.1. Polyphenols and Mitochondria
3.2. Polyphenols and Telomeres
3.3. Polyphenols and Sirt-1
3.4. Polyphenols and Male Fertility
3.5. Polyphenols and Inflammation, Apoptosis, and Autophagy
3.6. Polyphenols, Nrf2, and Proteostasis
4. Examples of Polyphenolic-Food: Anti-Aging Super Food
5. Blue Zone, Polyphenolic-Food, and Longevity
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Jin, K.; Rose, M.R. Modern Biological Theories of Aging. Aging Dis. 1988, 1, 220–221. [Google Scholar]
- Queen, B.L.; Tollefsbol, T.O. Polyphenols and aging. Curr. Aging Sci. 2010, 3, 34–42. [Google Scholar] [CrossRef]
- Aunan, J.R.; Watson, M.M.; Hagland, H.R.; Søreide, K. Molecular and biological hallmarks of aging. Br. J. Surg. 2016, 103, e29–e46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bocheva, G.; Slominski, R.M.; Slominski, A.T. Neuroendocrine aspects of skin aging. Int. J. Mol. Sci. 2019, 20, 2798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhanjal, D.S.; Bhardwaj, S.; Sharma, R.; Bhardwaj, K.; Kumar, D.; Chopra, C.; Nepovimova, E.; Singh, R.; Kuca, K. Plant Fortification of the Diet for Anti-Aging Effects: A Review. Nutrients 2020, 12, 3008. [Google Scholar] [CrossRef] [PubMed]
- Da Costa, J.P.; Vitorino, R.; Silva, G.M.; Vogel, C.; Duarte, A.C.; Rocha-Santos, T. Asynopsis on aging—Theories, mechanisms and future prospects. Aging Res. Rev. 2016, 29, 90–112. [Google Scholar] [CrossRef] [PubMed]
- Gladyshev, V.N. The free radical theory of aging is dead. Long live the damage theory! Antioxid. Redox Signal. 2014, 20, 727–731. [Google Scholar] [CrossRef] [PubMed]
- Liochev, S.I. Reactive oxygen species and the free radical theory of aging. Free Radic. Biol. Med. 2013, 60, 1–4. [Google Scholar] [CrossRef] [PubMed]
- Di Meo, S.; Venditti, P. Evolution of the Knowledge of Free Radicals and Other Oxidants. Oxid. Med. Cell. Longev. 2020, 2020, 9829176. [Google Scholar] [CrossRef] [PubMed]
- Viña, J. The free radical theory of frailty: Mechanisms and opportunities for interventions to promote successful aging. Free Radic. Biol. Med. 2019, 134, 690–694. [Google Scholar] [CrossRef]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sies, H. Oxidative stress: A concept in redox biology and medicine. Redox Biol. 2015, 4, 180–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McHugh, D.; Gil, J. Senescence and aging: Causes, consequences, and therapeutic avenues. J. Cell Biol. 2018, 217, 65–77. [Google Scholar] [CrossRef]
- Gutteridge, J.M.; Halliwell, B. Free radicals and antioxidants in the year 2000. A historical look to the future. Ann. N. Y. Acad. Sci. 2000, 899, 136–147. [Google Scholar] [CrossRef] [PubMed]
- Reczek, C.R.; Chandel, N.S. ROS-dependent signal transduction. Curr. Opin. Cell Biol. 2015, 33, 8–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ingrosso, D.; D’Angelo, S.; Perna, A.F.; Iolascon, A.; Miraglia del Giudice, E.; Perrotta, S.; Zappia, V.; Galletti, P. Increased membrane-protein methylation in hereditary spherocytosis. A marker of cytoskeletal disarray. Eur. J. Biochem. 1995, 228, 894–898. [Google Scholar] [CrossRef]
- Ingrosso, D.; D’Angelo, S.; Perrotta, S.; d’Urzo, G.; Iolascon, A.; Perna, A.F.; Galletti, P.; Zappia, V.; Miraglia del Giudice, E. Cytoskeletal behaviour in Spectrin and Band 3 deficient spherocytic red cells: Evidence for a differentiated splenic conditioning role. Br. J. Haematol. 1996, 93, 38–41. [Google Scholar] [CrossRef]
- D’Angelo, S.; Lembo, S.; Flora, F.; De Bonis, M.L.; Balato, A.; Ayala, F.; Balato, N.; Galletti, P.; Zappia, V. Abnormal isoaspartyl residues in erythrocyte membranes from psoriatic patients. Arch. Dermatol. Res. 2012, 304, 475–479. [Google Scholar] [CrossRef]
- D’Angelo, S.; Trojsi, F.; Salvatore, A.; Daniele, L.; Raimo, M.; Galletti, P.; Monsurrò, M.R. Accumulation of altered aspartyl residues in erythrocyte membrane proteins from patients with sporadic amyotrophic lateral sclerosis. Neurochem. Int. 2013, 63, 626–634. [Google Scholar] [CrossRef]
- D’Angelo, S.; Rosa, R. Oxidative stress and sport performance. Sport Sci. 2020, 13 (Suppl. S1), 18–22. [Google Scholar]
- D’Angelo, S. Polyphenols and athletic performance: A review on human data. In Plant Physiological Aspects of Phenolic Compounds; Soto-Hernández, M., García-Mateos, R., Tenango, M.P., Eds.; IntechOpen: London, UK, 2019; pp. 1–24. ISBN 978-1-78984-033-9. [Google Scholar] [CrossRef]
- Jarrett, S.G.; Boulton, M.E. Consequences of oxidative stress in age-related macular degeneration. Mol. Asp. Med. 2012, 33, 399–417. [Google Scholar] [CrossRef] [Green Version]
- Kirkwood, T.B.L. Why and how are we living longer? Exp. Physiol. 2017, 102, 1067–1074. [Google Scholar] [CrossRef] [Green Version]
- López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Z.; Ren, Z.; Zhang, J.; Chuang, C.C.; Kandaswamy, E.; Zhou, T.; Zuo, L. Role of ROS and nutritional antioxidants in human diseases. Front. Physiol. 2018, 9, 477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2015, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
- Minuz, P.; Velo, G.; Violi, F.; Ferro, A. Are nutraceuticals the modern panacea? From myth to science. Br. J. Clin. Pharmacol. 2017, 83, 5–7. [Google Scholar] [CrossRef] [Green Version]
- Sachdeva, V.; Roy, A.; Bharadvaja, N. Current Prospects of Nutraceuticals: A Review. Curr. Pharm. Biotechnol. 2020, 21, 884–896. [Google Scholar] [CrossRef]
- Da Costa, J.P. A current look at nutraceuticals–Key concepts and future prospects. Trends Food Sci. Technol. 2017, 62, 68–78. [Google Scholar] [CrossRef]
- D’Angelo, S.; Tafuri, D. Nutraceutical: Their role in improving sports performance. Sport Sci. 2020, 13 (Suppl. S1), 7–12. [Google Scholar]
- Pérez-Jiménez, J.; Neveu, V.; Vos, F.; Scalbert, A. Identification of the 100 richest dietary sources of polyphenols: An application of the Phenol-Explorer database. Eur. J. Clin. Nutr. 2010, 64 (Suppl. S3), S112–S120. [Google Scholar] [CrossRef]
- 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, 87. [Google Scholar] [CrossRef] [Green Version]
- Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fraga, C.G.; Croft, K.D.; Kennedy, 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] [Green Version]
- Ahuja, I.; Kissen, R.; Bones, A.M. Phytoalexins in defense against pathogens. Trends Plant Sci. 2012, 17, 73–90. [Google Scholar] [CrossRef] [PubMed]
- Gutiérrez-Grijalva, E.P.; Ambriz-Pére, D.L.; Leyva-López, N.; Castillo-López, R.I.; Heredia, J.B. Review: Dietary phenolic compounds, health benefits and bioaccessibility. Arch. Latinoam. Nutr. 2016, 66, 87–100. [Google Scholar] [PubMed]
- Bravo, L. Polyphenols: Chemistry, dietary sources, metabolism, and nutritional significance. Nutr. Rev. 1998, 56, 317–333. [Google Scholar] [CrossRef]
- Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
- Quero, J.; Mármol, I.; Cerrada, E.; Rodríguez-Yoldi, M.J. Insight into the potential application of polyphenol-rich dietary intervention in degenerative disease management. Food Funct. 2020, 11, 2805–2825. [Google Scholar] [CrossRef]
- Devi, S.A.; Chamoli, A. Polyphenols as an Effective Therapeutic Intervention against Cognitive Decline during Normal and Pathological Brain Aging. Adv. Exp. Med. Biol. 2020, 1260, 159–174. [Google Scholar] [CrossRef]
- 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]
- Menaa, F.; Menaa, A.; Tréton, J. Polyphenols against Skin Aging, Polyphenols in Human Health and Disease; Elsevier: Amsterdam, The Netherlands, 2014; pp. 819–830. [Google Scholar]
- Ignat, I.; Volf, I.; Popa, V.I. A critical review of methods for characterization of polyphenolic compounds in fruits and vegetables. Food Chem. 2011, 126, 1821–1835. [Google Scholar] [CrossRef] [PubMed]
- Tešic, Ž.; Gašic, U.; Milojkovic-Opsenica, D. Polyphenolic Profile of the Fruits Grown in Serbia. In Advances in Plant. Phenolics: From Chemistry to Human Health, 1st ed.; ACS Symposium Series; Jayaprakasha, G.K., Patil, B.S., Gattuso, G., Eds.; American Chemical Society: Washington, DC, USA, 2018; Chapter 3; Volume 1286, pp. 47–66. [Google Scholar]
- Haminiuk, C.W.I.; Maciel, G.M.; Plata-Oviedo, M.S.V.; Peralta, R.M. Phenolic compounds in fruits—An overview. Int. J. Food Sci. Technol. 2012, 47, 2023–2044. [Google Scholar] [CrossRef]
- Gašić, U.; Ćirić, I.; Pejčić, T.; Radenković, D.; Djordjević, V.; Radulović, S.; Tešić, Ž. Polyphenols as Possible Agents for Pancreatic Diseases. Antioxidants 2020, 9, 547. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Nichols, J.A.; Katiyar, S.K. Skin photoprotection by natural polyphenols: Anti-inflammatory, antioxidant and DNA repair mechanisms. Arch. Dermatol. Res. 2010, 302, 71–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dunaway, S.; Odin, R.; Zhou, L.; Ji, L.; Zhang, Y.; Kadekaro, A.L. Natural antioxidants: Multiple mechanisms to protect skin from solar radiation. Front. Pharmacol. 2018, 9, 392. [Google Scholar] [CrossRef] [Green Version]
- Pangestuti, R.; Siahaan, E.A.; Kim, S.K. Photoprotective Substances Derived from Marine Algae. Mar. Drugs 2018, 16, 399. [Google Scholar] [CrossRef] [Green Version]
- Barbosa, M.; Valentão, P.; Andrade, P.B. Polyphenols from Brown Seaweeds (Ochrophyta, Phaeophyceae): Phlorotannins in the Pursuit of Natural Alternatives to Tackle Neurodegeneration. Mar. Drugs. 2020, 18, 654. [Google Scholar] [CrossRef]
- Thring, T.; Hili, P.; Naughton, D. Anti-collagenase, anti-elastase and anti-oxidant activities of extracts from 21 plants. BMC Complement. Altern. Med. 2009, 9, 27. [Google Scholar] [CrossRef] [Green Version]
- Zillich, O.V.; Schweiggert-Weisz, U.; Eisner, P.; Kerscher, M. Polyphenols as active ingredients for cosmetic products. Int. J. Cosmet. Sci. 2015, 37, 455–464. [Google Scholar] [CrossRef]
- Zappia, V.; Galletti, P.; Manna, C.; D’Angelo, S.; Napoli, D.; De Bonis, M.L.; Capasso, G. Effects of Hydroxytyrosol on Cyclosporine Nephrotoxicity. In Olives and Olive Oil in Health and Disease Prevention; Preedy, V.R., Watson, R.R., Eds.; Academic Press: Oxford, UK, 2010; pp. 1245–1252. [Google Scholar]
- D’Angelo, S.; Sammartino, D. Protective Effect of Annurca Apple Extract against Oxidative Damage in Human Erythrocytes. Curr. Nutr. Food Sci. 2015, 11, 248–256. [Google Scholar] [CrossRef]
- D’Angelo, S.; Martino, E.; Ilisso, C.P.; Bagarolo, M.L.; Porcelli, M.; Cacciapuoti, G. Pro-oxidant and pro-apoptotic activity of polyphenol extract from Annurca apple and its underlying mechanisms in human breast cancer cells. Int. J. Oncol. 2017, 51, 939–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Angelo, S.; Martino, E.; Cacciapuoti, G. Effects of Annurca Apple (Malus pumila cv Annurca) Polyphenols on Breast Cancer Cells. Curr. Nutr. Food Sci. 2019, 15, 745–751. [Google Scholar] [CrossRef]
- Vuoso, D.C.; D’Angelo, S.; Ferraro, R.; Caserta, S.; Guido, S.; Cammarota, M.; Porcelli, M.; Cacciapuoti, G. Annurca apple polyphenol extract promotes mesenchymal-to-epithelial transition and inhibits migration in triple-negative breast cancer cells through ROS/JNK signaling. Sci. Rep. 2020, 10, 15921. [Google Scholar] [CrossRef]
- Boccellino, M.; Quagliuolo, L.; D’Angelo, S. Annurca Apple Biophenols’ Effects in Combination with Cisplatin on A549 Cells. Curr. Nutr. Food Sci. 2021, 17, 111–120. [Google Scholar] [CrossRef]
- Martino, E.; Vuoso, D.C.; D’Angelo, S.; Mele, L.; D’Onofrio, N.; Porcelli, M.; Cacciapuoti, G. Annurca apple polyphenol extract selectively kills MDA-MB-231 cells through ROS generation, sustained JNK activation and cell growth and survival inhibition. Sci. Rep. 2019, 10, 13045. [Google Scholar] [CrossRef]
- D’Angelo, S.; Morana, A.; Salvatore, A.; Zappia, V.; Galletti, P. Protective effect of polyphenols from Glycyrrhiza glabra against oxidative stress in Caco-2 cells. J. Med. Food 2009, 12, 1326–1333. [Google Scholar] [CrossRef]
- D’Angelo, S.; Cimmino, A.; Raimo, M.; Salvatore, A.; Zappia, V.; Galletti, P. Effect of reddening-ripening on the antioxidant activity of polyphenol extracts from cv. ‘Annurca’ apple fruits. J. Agric. Food Chem. 2007, 55, 9977–9985. [Google Scholar] [CrossRef]
- Silva, R.F.M.; Pogacnik, L. Polyphenols fromfood and natural products: Neuroprotection and safety. Antioxidants 2020, 9, 61. [Google Scholar] [CrossRef] [Green Version]
- Williamson, G. The role of polyphenols in modern nutrition. Nutr. Bull. 2017, 42, 226–235. [Google Scholar] [CrossRef]
- D’Angelo, S. Polyphenols: Potential beneficial effects of these phytochemicals in athletes. Curr. Sports Med. Rep. 2020, 19, 260–265. [Google Scholar] [CrossRef] [PubMed]
- D’Angelo, S.; Rosa, R. The impact of supplementation with Pomegranate fruit (Punica Granatum L.) on sport performance. Sport Sci. 2020, 13 (Suppl. S1), 29–37. [Google Scholar]
- D’Angelo, S.; Ascione, A. Guaranà and physical performance: A myth or reality? J. Hum. Sport Exerc. 2020, 15, S539–S551. [Google Scholar] [CrossRef]
- D’Angelo, S. Current Evidence on the Effect of Dietary Polyphenols Intake on Brain Health. Curr. Nutr. Food Sci. 2020, 16, 1170–1182. [Google Scholar] [CrossRef]
- Gulcin, I. Antioxidant activity of food constituents: An overview. Arch. Toxicol. 2012, 86, 345–391. [Google Scholar] [CrossRef]
- Bhat, M.I.; Kapila, R. Dietary metabolites derived from gut microbiota: Critical modulators of epigenetic changes in mammals. Nutr. Rev. 2017, 75, 374–389. [Google Scholar] [CrossRef] [PubMed]
- Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S.; Giustina, A.; Gazzaruso, C. Microbiota and metabolic diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef]
- Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, 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]
- D’Angelo, S.; Donini, L. The relationship between microbiota and exercise. Sport Sci. 2020, 14 (Suppl. S1), 24–29. [Google Scholar]
- Bohn, T. Dietary factors affecting polyphenol bioavailability. Nutr. Rev. 2014, 72, 429–452. [Google Scholar] [CrossRef]
- Singh, R.K.; Chang, H.W.; Yan, D.; Lee, K.M.; Ucmak, D.; Wong, K.; Abrouk, M.; Farahnik, B.; Nakamura, M.; Zhu, T.H.; et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 2017, 8, 73. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Etxeberria, U.; Arias, N.; Boqué, N.; Macarulla, M.T.; Portillo, M.P.; Martínez, J.A.; Milagro, F.I. Reshaping faecal gut microbiota composition by the intake of trans-resveratrol and quercetin in high-fat sucrose diet-fed rats. J. Nutr. Biochem 2015, 26, 651–660. [Google Scholar] [CrossRef]
- Sung, M.M.; Kim, T.T.; Denou, E.; Soltys, C.M.; Hamza, S.M.; Byrne, N.J.; Masson, G.; Park, H.; Wishart, D.S.; Madsen, K.L.; et al. Improved glucose homeostasis in obese mice treated with resveratrol is associated with alterations in the gut microbiome. Diabetes 2017, 66, 418–425. [Google Scholar] [CrossRef] [Green Version]
- Espley, R.V.; Butts, C.A.; Laing, W.A.; Martell, S.; Smith, H.; McGhie, T.K.; Zhang, J.; Paturi, G.; Hedderley, D.; Bovy, A.; et al. Dietary flavonoids from modified apple reduce inflammation markers and modulate gut microbiota in mice. J. Nutr. 2014, 144, 146–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, M.A.; Shukitt-Hale, B.; Kalt, W.; Ingram, D.K.; Joseph, J.A.; Wolkow, C.A. Blueberry polyphenols increase lifespan and thermotolerance in Caenorhabditis elegans. Aging Cell 2006, 5, 59–68. [Google Scholar] [CrossRef] [Green Version]
- Peng, C.; Chan, H.Y.; Li, Y.M.; Huang, Y.; Chen, Z.Y. Black tea theaflavins extend the lifespan of fruit flies. Exp. Gerontol. 2009, 44, 773–783. [Google Scholar] [CrossRef] [PubMed]
- Sunagawa, T.; Shimizu, T.; Kanda, T.; Tagashira, M.; Sami, M.; Shirasawa, T. Procyanidins from apples (Malus pumila Mill.) extend the lifespan of Caenorhabditis elegans. Planta Med. 2011, 77, 122–127. [Google Scholar] [CrossRef] [Green Version]
- Bass, T.M.; Weinkove, D.; Houthoofd, K.; Gems, D.; Partridge, L. Effects of resveratrol on lifespan in Drosophila melanogaster and Caenorhabditis elegans. Mech. Aging Dev. 2007, 128, 546–552. [Google Scholar] [CrossRef]
- Liao, V.H.; Yu, C.W.; Chu, Y.J.; Li, W.H.; Hsieh, Y.C.; Wang, T.T. Curcumin-mediated lifespan extension in Caenorhabditis elegans. Mech. Aging Dev. 2011, 132, 480–487. [Google Scholar] [CrossRef]
- Abbas, S.; Wink, M. Epigallocatechin gallate inhibits beta amyloid oligomerization in Caenorhabditis elegans and affects the daf-2/insulin-like signaling pathway. Phytomedicine 2010, 17, 902–909. [Google Scholar] [CrossRef]
- Gomes, E.C.; Silva, A.N.; Oliveira, M.R.D. Oxidants, antioxidants, and the beneficial roles of exercise-induced production of reactive species. Oxidative Med. Cell. Longev. 2012, 2012, 756132. [Google Scholar] [CrossRef] [PubMed]
- Salehi, A.; Emami, S.; Keighobadi, M.; Mirzaei, H. An overview of the effects of polyphenols on cardiac mitochondrial function. J. Maz. Univ. Med. Sci. 2019, 28, 211–224. [Google Scholar]
- Maleki, M.; Khelghati, N.; Alemi, F.; Bazdar, M.; Asemi, Z.; Majidinia, M.; Sadeghpoor, A.; Mahmoodpoor, A.; Jadidi-Niaragh, F.; Targhazeh, N.; et al. Stabilization of telomere by the antioxidant property of polyphenols: Anti-aging potential. Life Sci. 2020, 259, 118341. [Google Scholar] [CrossRef] [PubMed]
- Barbosa, M.C.; Grosso, R.A.; Fader, C.M. Hallmarks of aging: An autophagic perspective. Front. Endocrinol. 2019, 9, 790. [Google Scholar] [CrossRef] [PubMed]
- Höhn, A.; Weber, D.; Jung, T.; Ott, C.; Hugo, M.; Kochlik, B.; Kehm, R.; König, J.; Grune, T.; Castro, J.P. Happily (n) ever after: Aging in the context of oxidative stress, proteostasis loss and cellular senescence. Redox Biol. 2017, 11, 482–501. [Google Scholar] [CrossRef]
- Rolt, A.; Cox, L.S. Structural basis of the anti-aging effects of polyphenolics: Mitigation of oxidative stress. BMC Chem. 2020, 14, 50. [Google Scholar] [CrossRef]
- Hernandez-Segura, A.; Nehme, J.; Demaria, M. Hallmarks of cellular senescence. Trends Cell Biol. 2018, 28, 436–453. [Google Scholar] [CrossRef] [PubMed]
- Horvath, S. DNA methylation age of human tissues and cell types. Genome Biol. 2013, 14, R115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeppesen, D.K.; Bohr, V.A.; Stevnsner, T. DNA repair deficiency in neurodegeneration. Prog. Neurobiol. 2011, 94, 166–200. [Google Scholar] [CrossRef] [Green Version]
- Kennedy, S.R.; Salk, J.J.; Schmitt, M.W.; Loeb, L.A. Ultra-sensitive sequencing reveals an age-related increase in somatic mitochondrial mutations that are inconsistent with oxidative damage. PLoS Genet. 2013, 9, e1003794. [Google Scholar] [CrossRef] [Green Version]
- Kudryavtseva, A.V.; Krasnov, G.S.; Dmitriev, A.A.; Alekseev, B.Y.; Kardymon, O.L.; Sadritdinova, A.F.; Fedorova, M.S.; Pokrovsky, A.V.; Melnikova, N.V.; Kaprin, A.D.; et al. Mitochondrial dysfunction and oxidative stress in aging and cancer. Oncotarget 2016, 7, 44879–44905. [Google Scholar] [CrossRef] [Green Version]
- Santo, A.; Zhu, H.; Li, Y.R. Free radicals: From health to disease. React. Oxyg. Species 2016, 2, 245–263. [Google Scholar] [CrossRef]
- Nissanka, N.; Moraes, C.T. Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 2018, 592, 728–742. [Google Scholar] [CrossRef]
- Quijano, C.; Cao, L.; Fergusson, M.M.; Romero, H.; Liu, J.; Gutkind, S.; Rovira, I.I.; Mohney, R.P.; Karoly, E.D.; Finkel, T. Oncogene-induced senescence results in marked metabolic and bioenergetic alterations. Cell Cycle 2012, 11, 1383–1392. [Google Scholar] [CrossRef] [Green Version]
- Bakula, D.; Scheibye-Knudsen, M. MitophAging: Mitophagy in Aging and Disease. Front. Cell Dev. Biol. 2020, 8, 239. [Google Scholar] [CrossRef] [Green Version]
- Evans, C.S.; Holzbaur, E.L.F. Autophagy and mitophagy in ALS. Neurobiol. Dis. 2019, 122, 35–40. [Google Scholar] [CrossRef] [PubMed]
- Zole, E.; Ranka, R. Mitochondria, its DNA and telomeres in aging and human population. Biogerontology 2018, 19, 189–208. [Google Scholar] [CrossRef] [PubMed]
- Peng, K.; Tao, Y.; Zhang, J.; Wang, J.; Ye, F.; Dan, G.; Zhao, Y.; Cai, Y.; Zhao, J.; Wu, Q.; et al. Resveratrol Regulates Mitochondrial Biogenesis and Fission/Fusion to Attenuate Rotenone-Induced Neurotoxicity. Oxid. Med. Cell. Longev. 2016, 2016, 6705621. [Google Scholar] [CrossRef] [Green Version]
- Ferrara, L.; Joksimovic, M.; D’Angelo, S. Modulation of mitochondrial biogenesis: Action of physical activity and phytochemicals. J. Phys. Educ. Sport 2021, 21, 425–433. [Google Scholar] [CrossRef]
- Fivenson, E.M.; Lautrup, S.; Sun, N.; Scheibye-Knudsen, M.; Stevnsner, T.; Nilsen, H.; Bohr, V.A.; Fang, E.F. Mitophagy in neurodegeneration and aging. Neurochem. Int. 2017, 109, 202–209. [Google Scholar] [CrossRef]
- Ames, B.N. Endogenous oxidative DNA damage, aging, and cancer. Free Radic. Res. Commun. 1989, 7, 121–128. [Google Scholar] [CrossRef]
- Pourahmad, J.; Salimi, A.; Seydi, E. Role of Oxygen Free Radicals in Cancer Development and Treatment. In Free Radicals and Diseases; InTech: London, UK, 2016. [Google Scholar] [CrossRef] [Green Version]
- Hornsby, P.J. Telomerase and the aging process. Exp. Gerontol. 2007, 42, 575–581. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, J.C.; Cech, T.R. Human telomerase: Biogenesis, trafficking, recruitment, and activation. Genes Dev. 2015, 29, 1095–1105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shay, J.W.; Wright, W.E. Hallmarks of telomeres in aging research. J. Pathol. 2007, 211, 114–123. [Google Scholar] [CrossRef]
- Boccardi, V.; Paolisso, G.; Mecocci, P. Nutrition and lifestyle in healthy aging: The telomerase challenge. Aging 2016, 8, 12–15. [Google Scholar] [CrossRef] [Green Version]
- Gomez-Delgado, F.; Delgado-Lista, J.; Lopez-Moreno, J.; Rangel-Zuñiga, O.A.; Alcala-Diaz, J.F.; Leon-Acuña, A.; Corina, A.; Yubero-Serrano, E.; Torres-Peña, J.D.; Camargo, A.; et al. Telomerase RNA Component Genetic Variants Interact With the Mediterranean Diet Modifying the Inflammatory Status and its Relationship With Aging: CORDIOPREV Study. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 327–332. [Google Scholar] [CrossRef] [Green Version]
- Shi, J.; Yu, J.; Pohorly, J.E.; Kakuda, Y. Polyphenolics in grape seeds-biochemistry and functionality. J. Med. Food 2003, 6, 291–299. [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]
- Gardner, E.; Ruxton, C.; Leeds, A. Black tea–helpful or harmful? A review of the evidence. Eur. J. Clin. Nutr. 2007, 61, 3–18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, S.C.; Huang, S.M.; Chen, A.; Sun, C.Y.; Lin, S.H.; Chen, J.S.; Liu, S.T.; Hsu, Y.J. Resveratrol increases anti-aging Klotho gene expression via the activating transcription factor 3/c-Jun complex-mediated signaling pathway. Int. J. Biochem. Cell Biol. 2014, 53, 361–371. [Google Scholar] [CrossRef] [PubMed]
- Ong, A.L.C.; Ramasamy, T.S. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Aging Res. Rev. 2018, 43, 64–80. [Google Scholar] [CrossRef]
- Imai, S.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef] [PubMed]
- Gurău, F.; Baldoni, S.; Prattichizzo, F.; Espinosa, E.; Amenta, F.; Procopio, A.D.; Albertini, M.C.; Bonafè, M.; Olivieri, F. Anti-senescence compounds: A potential nutraceutical approach to healthy aging. Aging Res. Rev. 2018, 46, 14–31. [Google Scholar] [CrossRef]
- Davalli, P.; Mitic, T.; Caporali, A.; Lauriola, A.; D’Arca, D. ROS, Cell Senescence, and Novel Molecular Mechanisms in Aging and Age-Related Diseases. Oxid. Med. Cell. Longev. 2016, 2016, 3565127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sarubbo, F.; Esteban, S.; Miralles, A.; Moranta, D. Effects of Resveratrol and other Polyphenols on Sirt1: Relevance to Brain Function during Aging. Curr. Neuropharmacol. 2018, 16, 126–136. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.H.; Lee, J.H.; Lee, H.Y.; Min, K.J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Angelo, S.; Mele, E.; Di Filippo, F.; Viggiano, A.; Meccariello, R. Sirt1 activity in the brain: Simultaneous effects on energy homeostasis and reproduction. Int. J. Environ. Res. Public Health 2021, 18, 1243. [Google Scholar] [CrossRef] [PubMed]
- Zou, P.; Liu, X.; Li, G.; Wang, Y. Resveratrol pretreatment attenuates traumatic brain injury in rats by suppressing NLRP3 inflammasome activation via SIRT1. Mol. Med. Rep. 2018, 17, 3212–3217. [Google Scholar] [CrossRef]
- Sarubbo, F.; Ramis, M.R.; Kienzer, C.; Aparicio, S.; Esteban, S.; Miralles, A.; Moranta, D. Chronic Silymarin, Quercetin and Naringenin Treatments Increase Monoamines Synthesis and Hippocampal Sirt1 Levels Improving Cognition in Aged Rats. J. Neuroimmune Pharmacol. 2018, 13, 24–38. [Google Scholar] [CrossRef]
- Yao, H.; Rahman, I. Perspectives on translational and therapeutic aspects of SIRT1 in inflammaging and senescence. Biochem. Pharmacol. 2012, 84, 1332–1339. [Google Scholar] [CrossRef] [Green Version]
- Pierantoni, R.; Cobellis, G.; Meccariello, R.; Fasano, S. Evolutionary aspects of cellular communication in the vertebrate hypothalamo-hypophysio-gonadal axis. Int. Rev. Cytol. 2002, 218, 69–141. [Google Scholar] [CrossRef] [PubMed]
- Chianese, R.; Cobellis, G.; Chioccarelli, T.; Ciaramella, V.; Migliaccio, M.; Fasano, S.; Pierantoni, R.; Meccariello, R. Kisspeptins, Estrogens and Male Fertility. Curr. Med. Chem. 2016, 23, 4070–4091. [Google Scholar] [CrossRef] [PubMed]
- Almeida, S.; Rato, L.; Sousa, M.; Alves, M.-G.; Oliveira, P.F. Fertility and Sperm Quality in the Aging Male. Curr. Pharm. Des. 2017, 23, 442–4437. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, T.G.; Aston, K.I.; Meyer, T.; Carrell, D.T. The Sperm Epigenome, Male Aging, and Potential Effects on the Embryo. Adv. Exp. Med. Biol. 2015, 868, 81–93. [Google Scholar] [CrossRef]
- Paoli, D.; Pecora, G.; Pallotti, F.; Faja, F.; Pelloni, M.; Lenzi, A.; Lombardo, F. Cytological and molecular aspects of the aging sperm. Hum. Reprod. 2019, 34, 218–227. [Google Scholar] [CrossRef]
- Chianese, R.; Troisi, J.; Richards, S.; Scafuro, M.; Fasano, S.; Guida, M.; Pierantoni, R.; Meccariello, R. Bisphenol A in Reproduction: Epigenetic Effects. Curr. Med. Chem. 2018, 25, 748–770. [Google Scholar] [CrossRef]
- Skoracka, K.; Eder, P.; Łykowska-Szuber, L.; Dobrowolska, A.; Krela-Kaźmierczak, I. Diet and Nutritional Factors in Male (In)fertility-Underestimated Factors. J. Clin. Med. 2020, 9, 1400. [Google Scholar] [CrossRef]
- Ahmadi, S.; Bashiri, R.; Ghadiri-Anari, A.; Nadjarzadeh, A. Antioxidant supplements and semen parameters: An evidence based review. Int. J. Reprod. Biomed. 2016, 14, 729–736. [Google Scholar] [CrossRef]
- Truong, T.; Gardner, D.K. Antioxidants improve IVF outcome and subsequent embryo development in the mouse. Hum. Reprod. 2017, 32, 2404–2413. [Google Scholar] [CrossRef]
- Wu, Z.H.; Ke, X.W.; Feng, S.Y.; Zhang, L.; Wu, J.F.; Cheng, W.; Cheng, J.J.; Zhang, J.D.; Zhang, Y.G. Tea polyphenols reduces the apoptosis of spermatogenic cells in rats with experimental varicocele. Zhonghua Nan Ke Xue 2015, 21, 702–707. [Google Scholar]
- Opuwari, C.; Monsees, T. Green tea consumption increases sperm concentration and viability in male rats and is safe for reproductive, liver and kidney health. Sci. Rep. 2020, 10, 15269. [Google Scholar] [CrossRef]
- Opuwari, C.S.; Monsees, T.K. In vivo effects of black tea on the male rat reproductive system and functions of the kidney and liver. Andrologia 2020, 52, e13552. [Google Scholar] [CrossRef]
- Zhu, Y.; Yin, Q.; Yang, Y. Comprehensive Investigation of Moringa oleifera from Different Regions by Simultaneous Determination of 11 Polyphenols Using UPLC-ESI-MS/MS. Molecules 2020, 25, 676. [Google Scholar] [CrossRef] [Green Version]
- Moichela, F.T.; Adefolaju, G.A.; Henkel, R.R.; Opuwari, C.S. Aqueous leaf extract of Moringa oleifera reduced intracellular ROS production, DNA fragmentation and acrosome reaction in Human spermatozoa in vitro. Andrologia 2021, 53, e13903. [Google Scholar] [CrossRef] [PubMed]
- Azadi, L.; Tavalaee, M.; Deemeh, M.R.; Arbabian, M.; Nasr-Esfahani, M.H. Effects of Tempol and Quercetin on Human Sperm Function after Cryopreservation. Cryo Lett. 2017, 38, 29–36. [Google Scholar]
- Kawasaki, Y.; Sakurai, D.; Yoshihara, T.; Tsuchida, M.; Harakawa, S.; Suzuki, H. Effect of quercetin on the motility of cryopreserved canine spermatozoa. Cryobiology 2020, 96, 50–54. [Google Scholar] [CrossRef]
- Ahmed, H.; Jahan, S.; Salman, M.M.; Ullah, F. Stimulating effects of Quercetin (QUE) in tris citric acid extender on post thaw quality and in vivo fertility of buffalo (Bubalus bubalis) bull spermatozoa. Theriogenology 2019, 134, 18–23. [Google Scholar] [CrossRef] [PubMed]
- Mao, T.; Han, C.; Wei, B.; Zhao, L.; Zhang, Q.; Deng, R.; Liu, J.; Luo, Y.; Zhang, Y. Protective Effects of Quercetin against Cadmium Chloride-Induced Oxidative Injury in Goat Sperm and Zygotes. Biol. Trace Elem. Res. 2018, 185, 344–355. [Google Scholar] [CrossRef]
- Cui, X.; Jing, X.; Wu, X.; Yan, M. Protective effect of resveratrol on spermatozoa function in male infertility induced by excess weight and obesity. Mol. Med. Rep. 2016, 14, 4659–4665. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alamo, A.; Condorelli, R.A.; Mongioì, L.M.; Cannarella, R.; Giacone, F.; Calabrese, V.; La Vignera, S.; Calogero, A.E. Environment and Male Fertility: Effects of Benzo-α-Pyrene and Resveratrol on Human Sperm Function In Vitro. J. Clin. Med. 2019, 8, 561. [Google Scholar] [CrossRef] [Green Version]
- Gadani, B.; Bucci, D.; Spinaci, M.; Tamanini, C.; Galeati, G. Resveratrol and Epigallocatechin-3-gallate addition to thawed boar sperm improves in vitro fertilization. Theriogenology 2017, 90, 88–93. [Google Scholar] [CrossRef]
- Shabani Nashtaei, M.; Nekoonam, S.; Naji, M.; Bakhshalizadeh, S.; Amidi, F. Cryoprotective effect of resveratrol on DNA damage and crucial human sperm messenger RNAs, possibly through 5′ AMP-activated protein kinase activation. Cell Tissue Bank. 2018, 19, 87–95. [Google Scholar] [CrossRef] [PubMed]
- Bucci, D.; Spinaci, M.; Yeste, M.; Mislei, B.; Gadani, B.; Prieto Martinez, N.; Love, C.; Mari, G.; Tamanini, C.; Galeati, G. Combined effects of resveratrol and epigallocatechin-3-gallate on post thaw boar sperm and IVF parameters. Theriogenology 2018, 117, 16–25. [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] [PubMed] [Green Version]
- Wang, X.B.; Zhu, L.; Huang, J.; Yin, Y.G.; Kong, X.Q.; Rong, Q.F.; Shi, A.W.; Cao, K.J. Resveratrol-induced augmentation of telomerase activity delays senescence of endothelial progenitor cells. Chin. Med. J. 2011, 124, 4310–4315. [Google Scholar]
- Tili, E.; Michaille, J.J.; Adair, B.; Alder, H.; Limagne, E.; Taccioli, C.; Ferracin, M.; Delmas, D.; Latruffe, N.; Croce, C.M. Resveratrol decreases the levels of miR-155 by upregulating miR-663, a microRNA targeting JunB and JunD. Carcinogenesis 2010, 31, 1561–1566. [Google Scholar] [CrossRef]
- Boccellino, M.; D’Angelo, S. Anti-Obesity Effects of Polyphenol Intake: Current Status and Future Possibilities. Int. J. Mol. Sci. 2020, 21, 5642. [Google Scholar] [CrossRef]
- Benvenuto, M.; Albonici, L.; Focaccetti, C.; Ciuffa, S.; Fazi, S.; Cifaldi, L.; Miele, M.T.; De Maio, F.; Tresoldi, I.; Manzari, V.; et al. Polyphenol-Mediated Autophagy in Cancer: Evidence of In Vitro and In Vivo Studies. Int. J. Mol. Sci. 2020, 21, 6635. [Google Scholar] [CrossRef]
- Kiruthiga, C.; Devi, K.P.; Nabavi, S.M.; Bishayee, A. Autophagy: A Potential Therapeutic Target of Polyphenols in Hepatocellular Carcinoma. Cancers 2020, 12, 562. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Ma, K.; Wu, H.Y.; Wu, Y.P.; Li, B.X. Isoflavones Induce BEX2-Dependent Autophagy to Prevent ATR-Induced Neurotoxicity in SH-SY5Y Cells. Cell. Physiol. Biochem. 2017, 43, 1866–1879. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Zhu, S.; Li, J.; Assa, A.; Jundoria, A.; Xu, J.; Fan, S.; Eissa, N.T.; Tracey, K.J.; Sama, A.E.; et al. EGCG stimulates autophagy and reduces cytoplasmic HMGB1 levels in endotoxin-stimulated macrophages. Biochem. Pharmacol. 2011, 81, 1152–1163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trivedi, P.C.; Bartlett, J.J.; Perez, L.J.; Brunt, K.R.; Legare, J.F.; Hassan, A.; Kienesberger, P.C.; Pulinilkunnil, T. Glucolipotoxicity diminishes cardiomyocyte TFEB and inhibits lysosomal autophagy during obesity and diabetes. Biochim. Biophys. Acta 2016, 1861, 1893–1910. [Google Scholar] [CrossRef]
- Holczer, M.; Besze, B.; Zámbó, V.; Csala, M.; Bánhegyi, G.; Kapuy, O. Epigallocatechin-3-Gallate (EGCG) Promotes Autophagy-Dependent Survival via Influencing the Balance of mTOR-AMPK Pathways upon Endoplasmic Reticulum Stress. Oxid. Med. Cell. Longev. 2018, 2018, 6721530. [Google Scholar] [CrossRef] [Green Version]
- Sykiotis, G.P.; Habeos, I.G.; Samuelson, A.V.; Bohmann, D. The role of the antioxidant and longevity-promoting Nrf2 pathway in metabolic regulation. Curr. Opin. Clin. Nutr. Metab. Care 2011, 14, 41–48. [Google Scholar] [CrossRef] [Green Version]
- Itoh, K.; Wakabayashi, N.; Katoh, Y.; Ishii, T.; Igarashi, K.; Engel, J.D.; Yamamoto, M. Keap1 represses nuclear activation of antioxidant responsive elements by Nrf2 through binding to the amino-terminal Neh2 domain. Genes Dev. 1999, 13, 76–86. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, T.; Nioi, P.; Pickett, C.B. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J. Biol. Chem. 2009, 284, 13291–13295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eggler, A.L.; Savinov, S.N. Chemical and biological mechanisms of phytochemical activation of Nrf2 and importance in disease prevention. Recent Adv. Phytochem. 2013, 43, 121–155. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Y.; Jiang, Z.; Lu, H.; Xu, Z.; Tong, R.; Shi, J.; Jia, G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2 Signaling Pathway. Chem. Biodivers. 2019, 16, e1900400. [Google Scholar] [CrossRef]
- Kim, E.N.; Lim, J.H.; Kim, M.Y.; Ban, T.H.; Jang, I.A.; Yoon, H.E.; Park, C.W.; Chang, Y.S.; Choi, B.S. Resveratrol, an Nrf2 activator, ameliorates aging-related progressive renal injury. Aging 2018, 10, 83–99. [Google Scholar] [CrossRef] [Green Version]
- Kwak, M.-K.; Wakabayashi, N.; Greenlaw, J.L.; Yamamoto, M.; Kensler, T.W. Antioxidants enhance mammalian proteasome expression through the Keap1-Nrf2 signaling pathway. Mol. Cell Biol. 2003, 23, 8786–8794. [Google Scholar] [CrossRef] [Green Version]
- Klaips, C.L.; Jayaraj, G.G.; Hartl, F.U. Pathways of cellular proteostasis in aging and disease. J. Cell Biol. 2018, 217, 51–63. [Google Scholar] [CrossRef]
- Korovila, I.; Hugo, M.; Castro, J.P.; Weber, D.; Höhn, A.; Grune, T.; Jung, T. Proteostasis, oxidative stress and aging. Redox Biol. 2017, 13, 550–567. [Google Scholar] [CrossRef]
- Dikic, I. Proteasomal and Autophagic Degradation Systems. Annu. Rev. Biochem. 2017, 86, 193–224. [Google Scholar] [CrossRef] [PubMed]
- Hipp, M.S.; Kasturi, P.; Hartl, F.U. The proteostasis network and its decline in aging. Nat. Rev. Mol. Cell Biol. 2019, 20, 421–435. [Google Scholar] [CrossRef] [PubMed]
- Aguilaniu, H.; Gustafsson, L.; Rigoulet, M.; Nyström, T. Asymmetric inheritance of oxidatively damaged proteins during cytokinesis. Science 2003, 299, 1751–1753. [Google Scholar] [CrossRef] [PubMed]
- Macedo, D.; Jardim, C.; Figueira, I.; Almeida, A.F.; McDougall, G.J.; Stewart, D.; Yuste, J.E.; Tomás-Barberán, F.A.; Tenreiro, S.; Outeiro, T.F.; et al. (Poly)phenol-digested metabolites modulate alpha-synuclein toxicity by regulating proteostasis. Sci. Rep. 2018, 8, 6965. [Google Scholar] [CrossRef]
- Hajieva, P. The Effect of Polyphenols on Protein Degradation Pathways: Implications for Neuroprotection. Molecules 2017, 22, 159. [Google Scholar] [CrossRef] [Green Version]
- Chiti, F.; Dobson, C.M. Protein Misfolding, Amyloid Formation, and Human Disease: A Summary of Progress over the Last Decade. Annu. Rev. Biochem. 2017, 86, 27–68. [Google Scholar] [CrossRef]
- Mennen, L.I.; Walker, R.; Bennetau-Pelissero, C.; Scalbert, A. Risks and safety of polyphenol consumption. Am. J. Clin. Nutr. 2005, 81 (Suppl. S1), 326S–329S. [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]
- Ou, J.; Wang, M.; Zheng, J.; Ou, S. Positive and negative effects of polyphenol incorporation in baked foods. Food Chem. 2019, 284, 90–99. [Google Scholar] [CrossRef]
- Neveu, V.; Perez-Jiménez, J.; Vos, F.; Crespy, V.; du Chaffaut, L.; Mennen, L.; Knox, C.; Eisner, R.; Cruz, J.; Wishart, D.; et al. Phenol-Explorer: An online comprehensive database on polyphenol contents in foods. Database 2010, 2020, bap024. [Google Scholar] [CrossRef]
- Habtemariam, S. Chapter 7—Bilberries and blueberries as potential modulators of type 2 diabetes and associated diseases. In Medicinal Foods as Potential Therapies for Type-2 Diabetes and Associated Diseases; Habtemariam, S., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 135–175. [Google Scholar]
- Bell, L.; Williams, C.M. Blueberry benefits to cognitive function across the lifespan. Int. J. Food Sci. Nutr. 2020, 30, 1–3. [Google Scholar] [CrossRef]
- Tran, P.H.L.; Tran, T.T.D. Blueberry Supplementation in Neuronal Health and Protective Technologies for Efficient Delivery of Blueberry Anthocyanins. Biomolecules 2021, 11, 102. [Google Scholar] [CrossRef]
- Kalt, W.; Cassidy, A.; Howard, L.R.; Krikorian, R.; Stull, A.J.; Tremblay, F.; Zamora-Ros, R. Recent Research on the Health Benefits of Blueberries and Their Anthocyanins. Adv. Nutr. 2020, 11, 224–236. [Google Scholar] [CrossRef]
- Galli, R.L.; Bielinski, D.F.; Szprengiel, A.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplemented diet reverses age-related decline in hippocampal HSP70 neuroprotection. Neurobiol. Aging 2006, 27, 344–350. [Google Scholar] [CrossRef] [PubMed]
- Goyarzu, P.; Malin, D.H.; Lau, F.C.; Taglialatela, G.; Moon, W.D.; Jennings, R.; Moy, E.; Moy, D.; Lippold, S.; Shukitt-Hale, B.; et al. Blueberry supplemented diet: Effects on object recognition memory and nuclear factor-kappa B levels in aged rats. Nutr. Neurosci. 2004, 7, 75–83. [Google Scholar] [CrossRef] [PubMed]
- Peng, C.; Zuo, Y.; Kwan, K.M.; Liang, Y.; Ma, K.Y.; Chan, H.Y.E.; Huang, Y.; Yu, H.; Chen, Z.Y. Blueberry extract prolongs lifespan of Drosophila melanogaster. Exp. Gerontol. 2012, 47, 170–178. [Google Scholar] [CrossRef]
- Devore, E.E.; Kang, J.H.; Breteler, M.M.; Grodstein, F. Dietary intakes of berries and flavonoids in relation to cognitive decline. Ann. Neurol. 2012, 72, 135–143. [Google Scholar] [CrossRef]
- Bowtell, J.L.; Aboo-Bakkar, Z.; Conway, M.E.; Adlam, A.L.R.; Fulford, J. Enhanced task-related brain activation and resting perfusion in healthy older adults after chronic blueberry supplementation. Appl. Physiol. Nutr. Metab. 2017, 42, 773–779. [Google Scholar] [CrossRef]
- Miller, M.G.; Hamiltonm, D.A.; Joseph, J.A.; Shukitt-Hale, B. Dietary blueberry improves cognition among older adults in a randomized, double-blind, placebo-controlled trial. Eur. J. Nutr. 2018, 57, 1169–1180. [Google Scholar] [CrossRef]
- Krikorian, R.; Shidler, M.D.; Nash, T.A.; Kalt, W.; Vinqvist-Tymchuk, M.R.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplementation improves memory in older adults. J. Agric. Food Chem. 2010, 58, 3996–4000. [Google Scholar] [CrossRef] [Green Version]
- Boespflug, E.L.; Eliassen, J.C.; Dudley, J.A.; Shidler, M.D.; Kalt, W.; Summer, S.S.; Stein, A.L.; Stover, A.N.; Krikorian, R. Enhanced neural activation with blueberry supplementation in mild cognitive impairment. Nutr. Neurosci. 2018, 21, 297–305. [Google Scholar] [CrossRef]
- Rutledge, G.A.; Fisher, D.R.; Miller, M.G.; Kelly, M.E.; Bielinski, D.F.; Shukitt-Hale, B. The effects of blueberry and strawberry serum metabolites on age-related oxidative and inflammatory signaling in vitro. Food Funct. 2019, 10, 7707–7713. [Google Scholar] [CrossRef]
- Agarwal, P.; Holland, T.M.; Wang, Y.; Bennett, D.A.; Morris, M.C. Association of Strawberries and Anthocyanidin Intake with Alzheimer’s Dementia Risk. Nutrients 2019, 11, 3060. [Google Scholar] [CrossRef] [Green Version]
- Vuoso, D.C.; Porcelli, M.; Cacciapuoti, G.; D’Angelo, S. Biological Activity of MelAnnurca Flesh Apple Biophenols. Curr. Nutr. Food Sci. 2020, 16, 1149–1162. [Google Scholar] [CrossRef]
- Nasso, R.; Pagliara, V.; D’Angelo, S.; Rullo, R.; Masullo, M.; Arcone, R. Annurca Apple Polyphenol Extract Affects Acetyl- Cholinesterase and Mono-Amine Oxidase In Vitro Enzyme Activity. Pharmaceuticals 2021, 14, 62. [Google Scholar] [CrossRef]
- Boyer, J.; Liu, R.H. Apple phytochemicals and their health benefits. Nutr. J. 2004, 3, 5. [Google Scholar] [CrossRef] [Green Version]
- Sommella, E.; Badolati, N.; Riccio, G.; Salviati, E.; Bottone, S.; Dentice, M.; Campiglia, P.; Tenore, G.C.; Stornaiuolo, M.; Novellino, E. A Boost in Mitochondrial Activity Underpins the Cholesterol-Lowering Effect of Annurca Apple Polyphenols on Hepatic Cells. Nutrients 2019, 11, 163. [Google Scholar] [CrossRef] [Green Version]
- Masuzaki, H.; Kozuka, C.; Okamoto, S.; Yonamine, M.; Tanaka, H.; Shimabukuro, M. Brown rice-specific γ-oryzanol as a promising prophylactic avenue to protect against diabetes mellitus and obesity in humans. J. Diabetes Investig. 2019, 10, 18–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kozuka, C.; Kaname, T.; Shimizu-Okabe, C.; Takayama, C.; Tsutsui, M.; Matsushita, M.; Abe, K.; Masuzaki, H. Impact of brown rice-specific γ-oryzanol on epigenetic modulation of dopamine D2 receptors in brain striatum in high-fat-diet-induced obesity in mice. Diabetologia 2017, 60, 1502–1511. [Google Scholar] [CrossRef] [PubMed]
- Rungratanawanich, W.; Cenini, G.; Mastinu, A.; Sylvester, M.; Wilkening, A.; Abate, G.; Bonini, S.A.; Aria, F.; Marziano, M.; Maccarinelli, G.; et al. γ-Oryzanol Improves Cognitive Function and Modulates Hippocampal Proteome in Mice. Nutrients 2019, 11, 753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, C.; Chan, H.Y.E.; Huang, Y.; Yu, H.; Chen, Z.Y. Apple polyphenols extend the mean lifespan of Drosophila melanogaster. J. Agric. Food Chem. 2011, 59, 2097–2106. [Google Scholar] [CrossRef]
- Dias, A.L.S.; Pachikian, B.; Larondelle, Y.; Quetin-Leclercq, J. Recent advances on bioactivities of black rice. Curr. Opin. Clin. Nutr. Metab. Care 2017, 20, 470–476. [Google Scholar] [CrossRef]
- Zuo, Y.; Peng, C.; Liang, Y.; Ma, K.Y.; Yu, H.; Edwin Chan, H.Y.; Chen, Z.Y. Black rice extract extends the lifespan of fruit flies. Food Funct. 2012, 3, 1271–1279. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.J.; Zhao, S.M.; Jin, L.; Huang, L.J.; He, X.; Wei, Q. Anti-aging effect of black rice in subacute aging model mice. Chin. J. Clin. Rehabil. 2006, 10, 82–84. [Google Scholar]
- Tomata, Y.; Sugiyama, K.; Kaiho, Y.; Honkura, K.; Watanabe, T.; Zhang, S.; Sugawara, Y.; Tsuji, I. Green Tea Consumption and the Risk of Incident Dementia in Elderly Japanese: The Ohsaki Cohort 2006 Study. Am. J. Geriatr. Psychiatry 2016, 24, 881–889. [Google Scholar] [CrossRef] [Green Version]
- Mancini, E.; Beglinger, C.; Drewe, J.; Zanchi, D.; Lang, U.E.; Borgwardt, S. Green tea effects on cognition, mood and human brain function: A systematic review. Phytomedicine 2017, 34, 26–37. [Google Scholar] [CrossRef] [Green Version]
- Kakutani, S.; Watanabe, H.; Murayama, N. Green Tea Intake and Risks for Dementia, Alzheimer’s Disease, Mild Cognitive Impairment, and Cognitive Impairment: A Systematic Review. Nutrients 2019, 11, 1165. [Google Scholar] [CrossRef] [Green Version]
- Roh, E.; Kim, J.E.; Kwon, J.Y.; Park, J.S.; Bode, A.M.; Dong, Z.; Lee, K.W. Molecular mechanisms of green tea polyphenols with protective effects against skin photoaging. Crit. Rev. Food Sci. Nutr. 2017, 57, 1631–1637. [Google Scholar] [CrossRef]
- Prasanth, M.I.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. A Review of the Role of Green Tea (Camellia sinensis) in Antiphotoaging, Stress Resistance, Neuroprotection, and Autophagy. Nutrients 2019, 11, 474. [Google Scholar] [CrossRef] [Green Version]
- Elmets, C.A.; Singh, D.; Tubesing, K.; Matsui, M.; Katiyar, S.; Mukhtar, H. Cutaneous photoprotection from ultraviolet injury by green tea polyphenols. J. Am. Acad. Dermatol. 2001, 44, 425–432. [Google Scholar] [CrossRef]
- Li, Y.M.; Chan, H.Y.E.; Huang, Y.; Chen, Z.Y. Green tea catechins upregulate Superoxide dismutase and catalase in fruit flies. Mol. Nutr. Food Res. 2007, 51, 546–554. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Miranda, J.; Perez-Jimenez, F.; Ros, E.; De Caterina, R.; Badimon, L.; Covas, M.I.; Escrich, E.; Ordovas, J.M.; Soriguer, F.; Abia, R.; et al. Olive oil and health: Summary of the II international conference on olive oil and health consensus report, Jaén and Córdoba (Spain) 2008. Nutr. Metab. Cardiovasc. Dis. 2010, 20, 284–294. [Google Scholar] [CrossRef]
- Piroddi, M.; Albini, A.; Fabiani, R.; Giovannelli, L.; Luceri, C.; Natella, F.; Rosignoli, P.; Rossi, T.; Taticchi, A.; Servili, M.; et al. Nutrigenomics of extra-virgin olive oil: A review. Biofactors 2017, 43, 17–41. [Google Scholar] [CrossRef]
- Fernández del Río, L.; Gutiérrez-Casado, E.; Varela-López, A.; Villalba, J.M. Olive Oil and the Hallmarks of Aging. Molecules 2016, 21, 163. [Google Scholar] [CrossRef] [Green Version]
- D’Angelo, S.; Ingrosso, D.; Migliardi, V.; Sorrentino, A.; Donnarumma, G.; Baroni, A.; Masella, L.; Tufano, M.A.; Zappia, M.; Galletti, P. Hydroxytyrosol, a natural antioxidant from olive oil, prevents protein damage induced by long-wave ultraviolet radiation in melanoma cells. Free Radic. Biol. Med. 2005, 38, 908–919. [Google Scholar] [CrossRef]
- Buettner, D. The Blue Zones: Lessons for Living Longer from the People Who’ve Lived the Longest; National Geographic: Washington, DC, USA, 2010. [Google Scholar]
- Buettner, D.; Skemp, S. Blue Zones: Lessons from the World’s Longest Lived. Am. J. Lifestyle Med. 2016, 10, 318–321. [Google Scholar] [CrossRef]
- Nieddu, A.; Vindas, L.; Errigo, A.; Vindas, J.; Pes, G.M.; Dore, M.P. Dietary Habits, Anthropometric Features and Daily Performance in Two Independent Long-Lived Populations from Nicoya peninsula (Costa Rica) and Ogliastra (Sardinia). Nutrients 2020, 12, 1621. [Google Scholar] [CrossRef] [PubMed]
- Poulain, M.; Pes, G.M.; Grasland, C.; Carru, C.; Ferrucci, L.; Baggio, G.; Franceschi, C.; Deiana, L. Identification of a geographic area characterized by extreme longevity in the Sardinia island: The AKEA study. Exp. Gerontol. 2004, 39, 1423–1429. [Google Scholar] [CrossRef]
- Le, L.T.; Sabaté, J. Beyond meatless, the health effects of vegan diets: Findings from the Adventist cohorts. Nutrients 2014, 6, 2131–2147. [Google Scholar] [CrossRef] [Green Version]
- Rosero-Bixby, L.; Dow, W.H.; Rehkopf, D.H. The Nicoya region of Costa Rica: A high longevity island for elderly males. Vienna Yearb. Popul. Res. 2013, 11, 109–136. [Google Scholar] [CrossRef] [Green Version]
- Appel, L.J. Dietary patterns and longevity: Expanding the blue zones. Circulation 2008, 118, 214–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Poulain, M.; Herm, A.; Pes, G. The Blue Zones: Areas of exceptional longevity around the world. Vienna Yearb. Popul. Res. 2013, 11, 87–108. [Google Scholar] [CrossRef]
- Panagiotakos, D.B.; Chrysohoou, C.; Siasos, G.; Zisimos, K.; Skoumas, J.; Pitsavos, C.; Stefanadis, C. Sociodemographic and lifestyle statistics of oldest old people (>80 years) living in ikaria island: The ikaria study. Cardiol. Res. Pract. 2011, 2011, 679187. [Google Scholar] [CrossRef] [Green Version]
- Willcox, D.C.; Willcox, B.J.; Todoriki, H.; Suzuki, M. The Okinawan diet: Health implications of a low-calorie, nutrient-dense, antioxidant-rich dietary pattern low in the glycemic load. J. Am. Coll. Nutr. 2009, 28, 500S–516S. [Google Scholar] [CrossRef] [PubMed]
- Willcox, D.C.; Scapagnini, G.; Willcox, B.J. Healthy aging diets other than the Mediterranean: A focus on the Okinawan diet. Mech. Aging Dev. 2014, 136–137, 148–162. [Google Scholar] [CrossRef] [Green Version]
- Vasto, S.; Buscemi, S.; Barera, A.; Di Carlo, M.; Accardi, G.; Caruso, C. Mediterranean diet and healthy aging: A Sicilian perspective. Gerontology 2014, 60, 508–518. [Google Scholar] [CrossRef]
- Sohal, R.S.; Weindruch, R. Oxidative stress, caloric restriction, and aging. Science 1996, 273, 59–63. [Google Scholar] [CrossRef] [Green Version]
- Cantó, C.; Auwerx, J. Caloric restriction, SIRT1 and longevity. Trends Endocrinol. Metab. 2009, 20, 325–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, C.; Wang, X.; Chen, J.; Jiao, R.; Wang, L.; Li, Y.M.; Zuo, Y.; Liu, Y.; Lei, L.; Ma, K.Y.; et al. Biology of aging and role of dietary antioxidants. BioMed Res. Int. 2014, 2014, 831841. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pallauf, K.; Giller, K.; Huebbe, P.; Rimbach, G. Nutrition and healthy aging: Calorie restriction or polyphenol-rich “MediterrAsian” diet? Oxid. Med. Cell. Longev. 2013, 2013, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hidalgo-Mora, J.J.; García-Vigara, A.; Sanchez-Sanchez, M.L.; García-Pérez, M.-Á.; Tarín, J.J.; Cano, A. The Mediterranean diet: A historical perspective on food for health. Maturitas 2020, 132, 65–69. [Google Scholar] [CrossRef] [PubMed]
- Guasch-Ferré, M.; Merino, J.; Sun, Q.; Fitó, M.; Salas-Salvadó, J. Dietary Polyphenols, Mediterranean Diet, Prediabetes, and Type 2 Diabetes: A Narrative Review of the Evidence. Oxid. Med. Cell. Longev. 2017, 2017, 6723931. [Google Scholar] [CrossRef]
- Ditano-Vázquez, P.; Torres-Peña, J.D.; Galeano-Valle, F.; Pérez-Caballero, A.I.; Demelo-Rodríguez, P.; Lopez-Miranda, J.; Katsiki, N.; Delgado-Lista, J.; Alvarez-Sala-Walther, L.A. The Fluid Aspect of the Mediterranean Diet in the Prevention and Management of Cardiovascular Disease and Diabetes: The Role of Polyphenol Content in Moderate Consumption of Wine and Olive Oil. Nutrients 2019, 11, 2833. [Google Scholar] [CrossRef] [Green Version]
- D’Angelo, S.; Cusano, P. Adherence to the Mediterranean diet in athletes. Sport Sci. 2020, 13 (Suppl. S1), 58–63. [Google Scholar]
- Tosti, V.; Bertozzi, B.; Fontana, L. Health Benefits of the Mediterranean Diet: Metabolic and Molecular Mechanisms. J. Gerontol. A Biol. Sci. Med. Sci. 2018, 73, 318–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chatzianagnostou, K.; Del Turco, S.; Pingitore, A.; Sabatino, L.; Vassalle, C. The Mediterranean Lifestyle as a Non-Pharmacological and Natural Antioxidant for Healthy Aging. Antioxidants 2015, 4, 719–736. [Google Scholar] [CrossRef]
- Godos, J.; Marventano, S.; Mistretta, A.; Galvano, F.; Grosso, G. Dietary sources of polyphenols in the Mediterranean healthy Eating, Aging and Lifestyle (MEAL) study cohort. Int. J. Food Sci. Nutr. 2017, 68, 750–756. [Google Scholar] [CrossRef]
- Knoops, K.T.; Groot de, L.C.; Fidanza, F.; Alberti-Fidanza, A.; Kromhout, D.; van Staveren, W.A. Comparison of three different dietary scores in relation to 10-year mortality in elderly European subjects: The HALE project. Eur. J. Clin. Nutr. 2006, 60, 746–755. [Google Scholar] [CrossRef] [Green Version]
- Tong, T.Y.; Wareham, N.J.; Khaw, K.T.; Imamura, F.; Forouhi, N.G. Prospective association of the Mediterranean diet with cardiovascular disease incidence and mortality and its population impact in a non-Mediterranean population: The EPICNorfolk study. BMC Med. 2016, 14, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Crous-Bou, M.; Minguillón, C.; Gramunt-Fombuena, N.; Molinuevo, J.L. Alzheimer’s disease prevention: From risk factors to early intervention. Alzheimer’s Res. Ther. 2017, 9, 71. [Google Scholar] [CrossRef] [PubMed]
- Singh, B.; Parsaik, A.K.; Mielke, M.M.; Erwin, P.J.; Knopman, D.S.; Petersen, R.C.; Roberts, R.O. Association of editerranean diet with mild cognitive impairment and Alzheimer’s disease: A systematic review and meta-analysis. J. Alzheimer’s Dis. 2014, 39, 271–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wengreen, H.; Munger, R.G.; Cutler, A.; Quach, A.; Bowles, A.; Corcoran, C.; Tschanz, J.T.; Norton, M.C.; Welsh-Bohmer, K.A. Prospective study of Dietary Approaches to Stop Hypertension- and Mediterranean-style dietary patterns and age-related cognitive change: The Cache County Study on Memory, Health and Aging. Am. J. Clin. Nutr. 2013, 98, 1263–1271. [Google Scholar] [CrossRef]
- Feart, C.; Samieri, C.; Rondeau, V.; Amieva, H.; Portet, F.; Dartigues, J.F.; Scarmeas, N.; Barberger-Gateau, P. Adherence to a Mediterranean diet, cognitive decline, and risk of dementia. JAMA 2009, 302, 638–648. [Google Scholar] [CrossRef] [Green Version]
- Scarmeas, N.; Stern, Y.; Tang, M.-X.; Mayeux, R.; Luchsinger, J.A. Mediterranean diet and risk for Alzheimer’s disease. Ann. Neurol. 2006, 59, 912–921. [Google Scholar] [CrossRef] [Green Version]
- Mazza, E.; Fava, A.; Ferro, Y.; Rotundo, S.; Romeo, S.; Bosco, D.; Pujia, A.; Montalcini, T. Effect of the E replacement of dietary vegetable oils with a low dose of extravirgin olive oil in the Mediterranean Diet on cognitive functions in the elderly. J. Transl. Med. 2018, 16, 10. [Google Scholar] [CrossRef] [Green Version]
- Varghese, N.; Werner, S.; Grimm, A.; Eckert, A. Dietary Mitophagy Enhancer: A Strategy for Healthy Brain Aging? Antioxidants 2020, 9, 932. [Google Scholar] [CrossRef] [PubMed]
- Cao, L.; Tan, L.; Wang, H.-F.; Jiang, T.; Zhu, X.-C.; Lu, H.; Tan, M.-S.; Yu, J.-T. Dietary Patterns and Risk of Dementia: A Systematic Review and Meta-Analysis of Cohort Studies. Mol. Neurobiol. 2015, 53, 6144–6154. [Google Scholar] [CrossRef]
- Bajerska, J.; Woźniewicz, M.; Suwalska, A.; Jeszka, J. Eating patterns are associated with cognitive function in the elderly at risk of metabolic syndrome from rural areas. Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 3234–3245. [Google Scholar]
- Ashby-Mitchell, K.; Peeters, A.; Anstey, K.J. Role of dietary pattern analysis in determining cognitive status in elderly Australian adults. Nutrients 2015, 7, 1052–1067. [Google Scholar] [CrossRef]
- D’Angelo, S.; Motti, M.L.; Meccariello, R. ω-3 and ω-6 Polyunsaturated Fatty Acids, Obesity and Cancer. Nutrients 2020, 12, 2751. [Google Scholar] [CrossRef]
- D’Angelo, S.; Madonna, G.; Di Palma, D. Effects of fish oil supplementation in the sport performance. J. Phys. Educ. Sport 2020, 20 (Suppl. S4), 2322–2329. [Google Scholar] [CrossRef]
- Sloan, R.P.; Wall, M.; Yeung, L.K.; Feng, T.; Feng, X.; Provenzano, F.; Schroeter, H.; Lauriola, V.; Brickman, A.M.; Small, S.A. Insights into the role of diet and dietary flavanols in cognitive aging: Results of a randomized controlled trial. Sci. Rep. 2021, 11, 3837. [Google Scholar] [CrossRef] [PubMed]
- D’Angelo, S.; Cusano, P. Who practices sports can be vegetarian? J. Hum. Sport Exerc. 2020, 15, S552–S561. [Google Scholar] [CrossRef]
- Le Couteur, D.; Solon-Biet, S.M.; Wahl, D.; Cogger, V.C.; Willcox, B.J.; Willcox, D.C.; Raubenheimer, D.; Simpson, S.J. New Horizons: Dietary protein, aging and the Okinawan ratio. Age Aging 2016, 45, 443–447. [Google Scholar] [CrossRef] [Green Version]
- Sasaki, S. The value of the National Health and Nutrition Survey in Japan. Lancet 2011, 378, 1205–1206. [Google Scholar] [CrossRef]
- Wirth, M.; Schwarz, C.; Benson, G.; Horn, N.; Buchert, R.; Lange, C.; Kobe, T.; Hetzer, S.; Maglione, M.; Michael, E.; et al. Effects of spermidine supplementation on cognition and biomarkers in older adults with subjective cognitive decline (SmartAge)-study protocol for a randomized controlled trial. Alzheimer’s Res. Ther. 2019, 11, 36. [Google Scholar] [CrossRef] [PubMed]
- Hausman, D.B.; Fisher, J.G.; Johnson, M.A. Nutrition in centenarians. Maturitas 2011, 68, 203–209. [Google Scholar] [CrossRef] [PubMed]
- Guilleminault, L.; Williams, E.A.; Scott, H.; Berthon, B.S.; Jensen, M.E.; Wood, L.G. Diet and Asthma: Is It Time to Adapt Our Message? Nutrients 2017, 9, 1227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Meccariello, R.; D’Angelo, S. Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview. Antioxidants 2021, 10, 507. https://doi.org/10.3390/antiox10040507
Meccariello R, D’Angelo S. Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview. Antioxidants. 2021; 10(4):507. https://doi.org/10.3390/antiox10040507
Chicago/Turabian StyleMeccariello, Rosaria, and Stefania D’Angelo. 2021. "Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview" Antioxidants 10, no. 4: 507. https://doi.org/10.3390/antiox10040507
APA StyleMeccariello, R., & D’Angelo, S. (2021). Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview. Antioxidants, 10(4), 507. https://doi.org/10.3390/antiox10040507