The Molecular Mechanism of Polyphenols in the Regulation of Ageing Hallmarks
Abstract
:1. Introduction
2. Age-Related Effects of Polyphenols on Epigenetic Changes
3. Effects of Polyphenols on Genomic Instability
4. Effects of Polyphenols on Telomere Attrition
5. Effects of Polyphenols on Proteostasis Loss
6. Effects of Polyphenols on Deregulated Nutrient-Sensing Pathways
7. Effects of Polyphenols on Mitochondrial Dysfunction
8. Effects of Polyphenols on Cell Senescence
9. Effects of Polyphenols on Stem Cell Exhaustion
10. Effects of Polyphenols on Altered Intercellular Communication
11. Polyphenols’ Regulation of Ageing—A Clinical Perspective
12. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Gems, D.; De Magalhães, J.P. The hoverfly and the wasp: A critique of the hallmarks of aging as a paradigm. Ageing Res. Rev. 2021, 70, 101407. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Gems, D.; Partridge, L. Genetics of longevity in model organisms: Debates and paradigm shifts. Annu. Rev. Physiol. 2013, 75, 621–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kenyon, C.J. The genetics of ageing. Nature 2010, 464, 504–512. [Google Scholar] [CrossRef] [PubMed]
- Meccariello, R.; D’Angelo, S. Impact of Polyphenolic-Food on Longevity: An Elixir of Life. An Overview. Antioxidants 2021, 10, 507. [Google Scholar] [CrossRef]
- 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.E.; 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]
- 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. Ageing Dev. 2007, 128, 546–552. [Google Scholar] [CrossRef]
- Liao, V.H.-C.; Yu, C.-W.; Chu, Y.-J.; Li, W.-H.; Hsieh, Y.-C.; Wang, T.-T. Curcumin-mediated lifespan extension in Caenorhabditis elegans. Mech. Ageing 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] [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]
- Gomes, E.C.; Silva, A.N.; De Oliveira, M.R. 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]
- Barbosa, M.C.; Grosso, R.A.; Fader, C.M. Hallmarks of aging: An autophagic perspective. Front. Endocrinol. 2019, 9, 790. [Google Scholar] [CrossRef]
- 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-ageing effects of polyphenolics: Mitigation of oxidative stress. BMC Chem. 2020, 14, 50. [Google Scholar] [CrossRef]
- 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]
- 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, 3156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fraga, M.F.; Ballestar, E.; Paz, M.F.; Ropero, S.; Setien, F.; Ballestar, M.L.; Heine-Suñer, D.; Cigudosa, J.C.; Urioste, M.; Benitez, J.; et al. Epigenetic differences arise during the lifetime of monozygotic twins. Proc. Natl. Acad. Sci. USA 2005, 102, 10604–10609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guillaumet-Adkins, A.; Yañez, Y.; Peris-Diaz, M.D.; Calabria, I.; Palanca-Ballester, C.; Sandoval, J. Epigenetics and Oxidative Stress in Aging. Oxidative Med. Cell. Longev. 2017, 2017, 9175806. [Google Scholar] [CrossRef] [Green Version]
- Pal, S.; Tyler, J.K. Epigenetics and aging. Sci. Adv. 2016, 2, e1600584. [Google Scholar] [CrossRef] [Green Version]
- Denham, J.; Marques, F.Z.; O’Brien, B.J.; Charchar, F.J. Exercise: Putting action into our epigenome. Sport. Med. 2014, 44, 189–209. [Google Scholar] [CrossRef]
- Arora, I.; Sharma, M.; Sun, L.Y.; Tollefsbol, T.O. The Epigenetic Link between Polyphenols, Aging and Age-Related Diseases. Genes 2020, 11, 1094. [Google Scholar] [CrossRef]
- Donohoe, D.R.; Bultman, S.J. Metaboloepigenetics: Interrelationships between energy metabolism and epigenetic control of gene expression. J. Cell. Physiol. 2012, 227, 3169–3177. [Google Scholar] [CrossRef] [Green Version]
- Ribarič, S. Diet and aging. Oxidative Med. Cell. Longev. 2012, 2012, 741468. [Google Scholar] [CrossRef] [Green Version]
- Druesne, N.; Pagniez, A.; Mayeur, C.; Thomas, M.; Cherbuy, C.; Duée, P.-H.; Martel, P.; Chaumontet, C. Diallyl disulfide (DADS) increases histone acetylation and p21 waf1/cip1 expression in human colon tumor cell lines. Carcinogenesis 2004, 25, 1227–1236. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Wang, X.; Han, L.; Zhou, Y.; Sun, S. Green tea polyphenol EGCG reverse cisplatin resistance of A549/DDP cell line through candidate genes demethylation. Biomed. Pharmacother. 2015, 69, 285–290. [Google Scholar] [CrossRef]
- Berletch, J.B.; Liu, C.; Love, W.K.; Andrews, L.G.; Katiyar, S.K.; Tollefsbol, T.O. Epigenetic and genetic mechanisms contribute to telomerase inhibition by EGCG. J. Cell. Biochem. 2008, 103, 509–519. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nandakumar, V.; Vaid, M.; Katiyar, S.K. (−)-Epigallocatechin-3-gallate reactivates silenced tumor suppressor genes, Cip1/p21 and p 16 INK4a, by reducing DNA methylation and increasing histones acetylation in human skin cancer cells. Carcinogenesis 2011, 32, 537–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fang, M.Z.; Wang, Y.; Ai, N.; Hou, Z.; Sun, Y.; Lu, H.; Welsh, W.; Yang, C.S. Tea polyphenol (−)-epigallocatechin-3-gallate inhibits DNA methyltransferase and reactivates methylation-silenced genes in cancer cell lines. Cancer Res. 2003, 63, 7563–7570. [Google Scholar] [PubMed]
- Li, Y.; Chen, F.; Wei, A.; Bi, F.; Zhu, X.; Yin, S.; Lin, W.; Cao, W. Klotho recovery by genistein via promoter histone acetylation and DNA demethylation mitigates renal fibrosis in mice. J. Mol. Med. 2019, 97, 541–552. [Google Scholar] [CrossRef] [PubMed]
- Henning, S.M.; Wang, P.; Said, J.; Magyar, C.; Castor, B.; Doan, N.; Tosity, C.; Moro, A.; Gao, K.; Li, L.; et al. Polyphenols in brewed green tea inhibit prostate tumor xenograft growth by localizing to the tumor and decreasing oxidative stress and angiogenesis. J. Nutr. Biochem. 2012, 23, 1537–1542. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.-H.; Lee, D.-H.; Jeong, J.-H.; Guo, Z.S.; Lee, Y.J. Quercetin augments TRAIL-induced apoptotic death: Involvement of the ERK signal transduction pathway. Biochem. Pharmacol. 2008, 75, 1946–1958. [Google Scholar] [CrossRef] [Green Version]
- Du, L.; Xie, Z.; Wu, L.-c.; Chiu, M.; Lin, J.; Chan, K.K.; Liu, S.; Liu, Z. Reactivation of RASSF1A in breast cancer cells by curcumin. Nutr. Cancer 2012, 64, 1228–1235. [Google Scholar] [CrossRef] [Green Version]
- Jiang, A.; Wang, X.; Shan, X.; Li, Y.; Wang, P.; Jiang, P.; Feng, Q. Curcumin reactivates silenced tumor suppressor gene RARβ by reducing DNA methylation. Phytother. Res. 2015, 29, 1237–1245. [Google Scholar] [CrossRef]
- Bandyopadhyay, S.; Lion, J.-M.; Mentaverri, R.; Ricupero, D.A.; Kamel, S.; Romero, J.R.; Chattopadhyay, N. Attenuation of osteoclastogenesis and osteoclast function by apigenin. Biochem. Pharmacol. 2006, 72, 184–197. [Google Scholar] [CrossRef]
- Samet, I.; Villareal, M.O.; Han, J.; Sayadi, S.; Isoda, H. Effect of olive leaf components on the proliferation and viability of hematopoietic stem cells. Asian J. Biomed. Pharm. Sci. 2014, 4, 1. [Google Scholar] [CrossRef]
- Eggler, A.L.; Savinov, S.N. Chemical and biological mechanisms of phytochemical activation of Nrf2 and importance in disease prevention. In 50 Years of Phytochemistry Research; Springer: Cham, Switzerland, 2013; pp. 121–155. [Google Scholar]
- Zahedipour, F.; Hosseini, S.A.; Henney, N.C.; Barreto, G.E.; Sahebkar, A. Phytochemicals as inhibitors of tumor necrosis factor alpha and neuroinflammatory responses in neurodegenerative diseases. Neural Regen. Res. 2022, 17, 1675–1684. [Google Scholar] [PubMed]
- Zhong, Y.; Chiou, Y.-S.; Pan, M.-H.; Shahidi, F. Anti-inflammatory activity of lipophilic epigallocatechin gallate (EGCG) derivatives in LPS-stimulated murine macrophages. Food Chem. 2012, 134, 742–748. [Google Scholar] [CrossRef]
- Khoi, P.N.; Park, J.S.; Kim, J.H.; Xia, Y.; Kim, N.H.; Kim, K.K.; Jung, Y.D. (-)-Epigallocatechin-3-gallate blocks nicotine-induced matrix metalloproteinase-9 expression and invasiveness via suppression of NF-κB and AP-1 in endothelial cells. Int. J. Oncol. 2013, 43, 868–876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rahimifard, M.; Baeeri, M.; Bahadar, H.; Moini-Nodeh, S.; Khalid, M.; Haghi-Aminjan, H.; Mohammadian, H.; Abdollahi, M. Therapeutic effects of gallic acid in regulating senescence and diabetes; an in vitro study. Molecules 2020, 25, 5875. [Google Scholar] [CrossRef] [PubMed]
- Menicacci, B.; Cipriani, C.; Margheri, F.; Mocali, A.; Giovannelli, L. Modulation of the senescence-associated inflammatory phenotype in human fibroblasts by olive phenols. Int. J. Mol. Sci. 2017, 18, 2275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parzonko, A.; Czerwińska, M.E.; Kiss, A.K.; Naruszewicz, M. Oleuropein and oleacein may restore biological functions of endothelial progenitor cells impaired by angiotensin II via activation of Nrf2/heme oxygenase-1 pathway. Phytomedicine 2013, 20, 1088–1094. [Google Scholar] [CrossRef]
- Jung, Y.H.; Lee, S.-J.; Oh, S.Y.; Lee, H.J.; Ryu, J.M.; Han, H.J. Oleic acid enhances the motility of umbilical cord blood derived mesenchymal stem cells through EphB2-dependent F-actin formation. Biochim. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2015, 1853, 1905–1917. [Google Scholar] [CrossRef] [Green Version]
- Katsiki, M.; Chondrogianni, N.; Chinou, I.; Rivett, A.J.; Gonos, E.S. The olive constituent oleuropein exhibits proteasome stimulatory properties in vitro and confers life span extension of human embryonic fibroblasts. Rejuvenation Res. 2007, 10, 157–172. [Google Scholar] [CrossRef]
- Santiago-Mora, R.; Casado-Díaz, A.; De Castro, M.; Quesada-Gómez, J. Oleuropein enhances osteoblastogenesis and inhibits adipogenesis: The effect on differentiation in stem cells derived from bone marrow. Osteoporos. Int. 2011, 22, 675–684. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Hickson, L.J.; Prata, L.G.L.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bureau, G.; Longpré, F.; Martinoli, M.G. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J. Neurosci. Res. 2008, 86, 403–410. [Google Scholar] [CrossRef]
- Ghosh, B. Quercetin inhibits LPS-induced nitric oxide and tumor necrosis factor-α production in murine macrophages. Int. J. Immunopharmacol. 1999, 21, 435–443. [Google Scholar]
- Geraets, L.; Moonen, H.J.; Brauers, K.; Wouters, E.F.; Bast, A.; Hageman, G.J. Dietary flavones and flavonoles are inhibitors of poly (ADP-ribose) polymerase-1 in pulmonary epithelial cells. J. Nutr. 2007, 137, 2190–2195. [Google Scholar] [CrossRef] [Green Version]
- Kandere-Grzybowska, K.; Kempuraj, D.; Cao, J.; Cetrulo, C.L.; Theoharides, T.C. Regulation of IL-1-induced selective IL-6 release from human mast cells and inhibition by quercetin. Br. J. Pharmacol. 2006, 148, 208. [Google Scholar] [CrossRef] [Green Version]
- Matsuno, Y.; Atsumi, Y.; Alauddin, M.; Rana, M.M.; Fujimori, H.; Hyodo, M.; Shimizu, A.; Ikuta, T.; Tani, H.; Torigoe, H.; et al. Resveratrol and its Related Polyphenols Contribute to the Maintenance of Genome Stability. Sci. Rep. 2020, 10, 5388. [Google Scholar] [CrossRef] [Green Version]
- Thilakarathna, W.W.; Rupasinghe, H.V. Microbial metabolites of proanthocyanidins reduce chemical carcinogen-induced DNA damage in human lung epithelial and fetal hepatic cells in vitro. Food Chem. Toxicol. 2019, 125, 479–493. [Google Scholar] [CrossRef]
- Li, J.; Zhang, C.-X.; Liu, Y.-M.; Chen, K.-L.; Chen, G. A comparative study of anti-aging properties and mechanism: Resveratrol and caloric restriction. Oncotarget 2017, 8, 65717–65729. [Google Scholar] [CrossRef] [Green Version]
- He, S.; Zhou, M.; Zheng, H.; Wang, Y.; Wu, S.; Gao, Y.; Chen, J. Resveratrol inhibits the progression of premature senescence partially by regulating v-rel avian reticuloendotheliosis viral oncogene homolog A (RELA) and sirtuin 1 (SIRT1). Ren. Fail. 2022, 44, 171–183. [Google Scholar] [CrossRef]
- Zheng, M.; Bai, Y.; Sun, X.; Fu, R.; Liu, L.; Liu, M.; Li, Z.; Huang, X. Resveratrol Reestablishes Mitochondrial Quality Control in Myocardial Ischemia/Reperfusion Injury through Sirt1/Sirt3-Mfn2-Parkin-PGC-1α Pathway. Molecules 2022, 27, 5545. [Google Scholar] [CrossRef] [PubMed]
- Finkel, T.; Deng, C.-X.; Mostoslavsky, R. Recent progress in the biology and physiology of sirtuins. Nature 2009, 460, 587–591. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, X.; Liu, Q.; Wang, M.; Liang, M.; Yang, X.; Xu, X.; Zou, H.; Qiu, J. Activation of Sirt1 by resveratrol inhibits TNF-α induced inflammation in fibroblasts. PLoS ONE 2011, 6, e27081. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Moskalev, A.A.; Shaposhnikov, M.V.; Plyusnina, E.N.; Zhavoronkov, A.; Budovsky, A.; Yanai, H.; Fraifeld, V.E. The role of DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Res. Rev. 2013, 12, 661–684. [Google Scholar] [CrossRef] [PubMed]
- Niedernhofer, L.J.; Gurkar, A.U.; Wang, Y.; Vijg, J.; Hoeijmakers, J.H.; Robbins, P.D. Nuclear genomic instability and aging. Annu. Rev. Biochem. 2018, 87, 295–322. [Google Scholar] [CrossRef] [PubMed]
- Kubben, N.; Misteli, T. Shared molecular and cellular mechanisms of premature ageing and ageing-associated diseases. Nat. Rev. Mol. Cell Biol. 2017, 18, 595–609. [Google Scholar] [CrossRef]
- Tiwari, V.; Wilson, D.M., III. DNA damage and associated DNA repair defects in disease and premature aging. Am. J. Hum. Genet. 2019, 105, 237–257. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Vijg, J. Somatic mutagenesis in mammals and its implications for human disease and aging. Annu. Rev. Genet. 2018, 52, 397. [Google Scholar] [CrossRef]
- García-Nieto, P.E.; Morrison, A.J.; Fraser, H.B. The somatic mutation landscape of the human body. Genome Biol. 2019, 20, 298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, L.; Dong, X.; Lee, M.; Maslov, A.Y.; Wang, T.; Vijg, J. Single-cell whole-genome sequencing reveals the functional landscape of somatic mutations in B lymphocytes across the human lifespan. Proc. Natl. Acad. Sci. USA 2019, 116, 9014–9019. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Forsberg, L.A.; Gisselsson, D.; Dumanski, J.P. Mosaicism in health and disease—Clones picking up speed. Nat. Rev. Genet. 2017, 18, 128–142. [Google Scholar] [CrossRef]
- Risques, R.A.; Kennedy, S.R. Aging and the rise of somatic cancer-associated mutations in normal tissues. PLoS Genet. 2018, 14, e1007108. [Google Scholar] [CrossRef] [Green Version]
- De, S. Somatic mosaicism in healthy human tissues. Trends Genet. 2011, 27, 217–223. [Google Scholar] [CrossRef] [PubMed]
- Krimmel, J.D.; Schmitt, M.W.; Harrell, M.I.; Agnew, K.J.; Kennedy, S.R.; Emond, M.J.; Loeb, L.A.; Swisher, E.M.; Risques, R.A. Ultra-deep sequencing detects ovarian cancer cells in peritoneal fluid and reveals somatic TP53 mutations in noncancerous tissues. Proc. Natl. Acad. Sci. USA 2016, 113, 6005–6010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Young, A.L.; Challen, G.A.; Birmann, B.M.; Druley, T.E. Clonal haematopoiesis harbouring AML-associated mutations is ubiquitous in healthy adults. Nat. Commun. 2016, 7, 12484. [Google Scholar] [CrossRef]
- Qiu, G.-H.; Zheng, X.; Fu, M.; Huang, C.; Yang, X. The protective function of non-coding DNA in DNA damage accumulation with age and its roles in age-related diseases. Biogerontology 2019, 20, 741–761. [Google Scholar] [CrossRef]
- Qiu, G.-H.; Huang, C.; Zheng, X.; Yang, X. The protective function of noncoding DNA in genome defense of eukaryotic male germ cells. Epigenomics 2018, 10, 499–517. [Google Scholar] [CrossRef]
- Janssen, A.; Colmenares, S.U.; Karpen, G.H. Heterochromatin: Guardian of the genome. Annu. Rev. Cell Dev. Biol. 2018, 34, 265–288. [Google Scholar] [CrossRef] [Green Version]
- Ferrucci, L.; Gonzalez-Freire, M.; Fabbri, E.; Simonsick, E.; Tanaka, T.; Moore, Z.; Salimi, S.; Sierra, F.; de Cabo, R. Measuring biological aging in humans: A quest. Aging Cell 2020, 19, e13080. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Proshkina, E.; Shaposhnikov, M.; Moskalev, A. Genome-Protecting Compounds as Potential Geroprotectors. Int. J. Mol. Sci. 2020, 21, 4484. [Google Scholar] [CrossRef] [PubMed]
- Olinski, R.; Siomek, A.; Rozalski, R.; Gackowski, D.; Foksinski, M.; Guz, J.; Dziaman, T.; Szpila, A.; Tudek, B. Oxidative damage to DNA and antioxidant status in aging and age-related diseases. Acta Biochim. Pol. 2007, 54, 11–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Humphreys, V.; Martin, R.M.; Ratcliffe, B.; Duthie, S.; Wood, S.; Gunnell, D.; Collins, A.R. Age-related increases in DNA repair and antioxidant protection: A comparison of the Boyd Orr Cohort of elderly subjects with a younger population sample. Age Ageing 2007, 36, 521–526. [Google Scholar] [CrossRef] [Green Version]
- Reddy, K.; Reddy, T.; Somasekharaiah, B.; Kumarl, K.S. Changes in antioxidant enzyme levels and DNA damage during aging. Indian J. Clin. Biochem. 1998, 13, 20–26. [Google Scholar] [CrossRef] [Green Version]
- De Caterina, R.; Martinez, A.J.; Kohlmeier, M. Principles of Nutrigenetics and Nutrigenomics: Fundamentals of Individualized Nutrition; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar]
- Fenech, M. Vitamins associated with brain aging, mild cognitive impairment, and alzheimer disease: Biomarkers, epidemiological and experimental evidence, plausible mechanisms, and knowledge gaps. Adv. Nutr. 2017, 8, 958–970. [Google Scholar] [CrossRef] [Green Version]
- Vaid, M.; Sharma, S.D.; Katiyar, S.K. Proanthocyanidins Inhibit Photocarcinogenesis through Enhancement of DNA Repair and Xeroderma Pigmentosum Group A–Dependent MechanismProanthocyanidins Stimulate DNA Repair. Cancer Prev. Res. 2010, 3, 1621–1629. [Google Scholar] [CrossRef] [Green Version]
- Vaid, M.; Prasad, R.; Singh, T.; Katiyar, S.K. Dietary grape seed proanthocyanidins inactivate regulatory T cells by promoting NER-dependent DNA repair in dendritic cells in UVB-exposed skin. Oncotarget 2017, 8, 49625–49636. [Google Scholar] [CrossRef] [Green Version]
- Rossiello, F.; Jurk, D. Telomere dysfunction in ageing and age-related diseases. Nature 2022, 24, 135–147. [Google Scholar] [CrossRef]
- Whittemore, K.; Vera, E.; Martínez-Nevado, E.; Sanpera, C.; Blasco, M.A. Telomere shortening rate predicts species life span. Proc. Natl. Acad. Sci. USA 2019, 116, 15122–15127. [Google Scholar] [CrossRef] [Green Version]
- Zole, E.; Ranka, R. Mitochondria, its DNA and telomeres in ageing and human population. Biogerontology 2018, 19, 189–208. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Wang, Z.; Wu, X. Abnormal function of telomere protein TRF2 induces cell mutation and the effects of environmental tumor-promoting factors (Review). Oncol. Rep. 2021, 46, 184. [Google Scholar] [CrossRef]
- Sahin, E.; Colla, S.; Liesa, M.; Moslehi, J.; Müller, F.L.; Guo, M.; Cooper, M.; Kotton, D.; Fabian, A.J.; Walkey, C.; et al. Telomere dysfunction induces metabolic and mitochondrial compromise. Nature 2011, 470, 359–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amano, H.; Chaudhury, A.; Rodriguez-Aguayo, C.; Lu, L.; Akhanov, V.; Catic, A.; Popov, Y.V.; Verdin, E.; Johnson, H.; Stossi, F.; et al. Telomere dysfunction induces sirtuin repression that drives telomere-dependent disease. Cell Metab. 2019, 29, 1274–1290.e9. [Google Scholar] [CrossRef] [PubMed]
- Chakravarti, D.; LaBella, K.A.; DePinho, R.A. Telomeres: History, health, and hallmarks of aging. Cell 2021, 184, 306–322. [Google Scholar] [CrossRef]
- Kondo, H.; Kim, H.W.; Wang, L.; Okada, M.; Paul, C.; Millard, R.W.; Wang, Y. Blockade of senescence-associated microRNA-195 in aged skeletal muscle cells facilitates reprogramming to produce induced pluripotent stem cells. Aging Cell 2016, 15, 56–66. [Google Scholar] [CrossRef]
- Sun, C.; Wang, K.; Stock, A.J.; Gong, Y.; Demarest, T.G.; Yang, B.; Giri, N.; Harrington, L.; Alter, B.P.; Savage, S.A.; et al. Re-equilibration of imbalanced NAD metabolism ameliorates the impact of telomere dysfunction. EMBO J. 2020, 39, e103420. [Google Scholar] [CrossRef]
- Lin, J.; Epel, E. Stress and telomere shortening: Insights from cellular mechanisms. Ageing Res. Rev. 2022, 73, 101507. [Google Scholar] [CrossRef]
- Boccardi, V.; Paolisso, G.; Mecocci, P. Nutrition and lifestyle in healthy aging: The telomerase challenge. Aging 2016, 8, 12. [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. Ser. A 2018, 73, 327–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheng, R.; Gu, Z.-L.; Xie, M.-L. Epigallocatechin gallate, the major component of polyphenols in green tea, inhibits telomere attrition mediated cardiomyocyte apoptosis in cardiac hypertrophy. Int. J. Cardiol. 2013, 162, 199–209. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Omidifar, N.; Moghadami, M. Trends in Natural Nutrients for Oxidative Stress and Cell Senescence. Oxidative Med. Cell. Longev. 2021, 2021, 7501424. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.R.; Li, S.; Lin, C.C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors 2018, 44, 69–82. [Google Scholar] [CrossRef] [PubMed]
- Morselli, E.; Maiuri, M.; Markaki, M.; Megalou, E.; Pasparaki, A.; Palikaras, K.; Criollo, A.; Galluzzi, L.; Malik, S.; Vitale, I.; et al. Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy. Cell Death Dis. 2010, 1, e10. [Google Scholar] [CrossRef] [Green Version]
- Porquet, D.; Casadesús, G.; Bayod, S.; Vicente, A.; Canudas, A.M.; Vilaplana, J.; Pelegrí, C.; Sanfeliu, C.; Camins, A.; Pallàs, M.; et al. Dietary resveratrol prevents Alzheimer’s markers and increases life span in SAMP8. Age 2013, 35, 1851–1865. [Google Scholar] [CrossRef] [Green Version]
- He, F.; Ru, X.; Wen, T. NRF2, a Transcription Factor for Stress Response and Beyond. Int. J. Mol. Sci. 2020, 21, 4777. [Google Scholar] [CrossRef]
- Dodson, M.; Anandhan, A.; Zhang, D.D.; Madhavan, L. An NRF2 Perspective on Stem Cells and Ageing. Front. Aging 2021, 2, 690686. [Google Scholar] [CrossRef]
- Khacho, M.; Clark, A.; Svoboda, D.S.; Azzi, J.; MacLaurin, J.G.; Meghaizel, C.; Sesaki, H.; Lagace, D.C.; Germain, M.; Harper, M.-E.; et al. Mitochondrial dynamics impacts stem cell identity and fate decisions by regulating a nuclear transcriptional program. Cell Stem Cell 2016, 19, 232–247. [Google Scholar] [CrossRef] [Green Version]
- Huang, Y.; Li, W.; Su, Z.Y.; Kong, A.N. The complexity of the Nrf2 pathway: Beyond the antioxidant response. J. Nutr. Biochem. 2015, 26, 1401–1413. [Google Scholar] [CrossRef]
- Lewis, K.N.; Wason, E.; Edrey, Y.H.; Kristan, D.M.; Nevo, E.; Buffenstein, R. Regulation of Nrf2 signaling and longevity in naturally long-lived rodents. Proc. Natl. Acad. Sci. USA 2015, 112, 3722–3727. [Google Scholar] [CrossRef] [Green Version]
- Lewis, K.N.; Mele, J.; Hayes, J.D.; Buffenstein, R. Nrf2, a guardian of healthspan and gatekeeper of species longevity. Integr. Comp. Biol. 2010, 50, 829–843. [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]
- Yu, C.; Xiao, J.H. The Keap1-Nrf2 System: A Mediator between Oxidative Stress and Aging. Oxidative Med. Cell. Longev. 2021, 2021, 6635460. [Google Scholar] [CrossRef]
- Yan, X.; Fu, X. Nrf2/Keap1/ARE Signaling Mediated an Antioxidative Protection of Human Placental Mesenchymal Stem Cells of Fetal Origin in Alveolar Epithelial Cells. Oxidative Med. Cell. Longev. 2019, 2019, 2654910. [Google Scholar] [CrossRef]
- Ulasov, A.V.; Rosenkranz, A.A.; Georgiev, G.P.; Sobolev, A.S. Nrf2/Keap1/ARE signaling: Towards specific regulation. Life Sci 2022, 291, 120111. [Google Scholar] [CrossRef]
- 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] [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]
- 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] [PubMed]
- 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] [PubMed]
- Fedorova, M.; Bollineni, R.C.; Hoffmann, R. Protein carbonylation as a major hallmark of oxidative damage: Update of analytical strategies. Mass Spectrom. Rev. 2014, 33, 79–97. [Google Scholar] [CrossRef] [PubMed]
- Semchyshyn, H.M. Reactive carbonyl species in vivo: Generation and dual biological effects. Sci. World J. 2014, 2014, 417842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dikic, I. Proteasomal and autophagic degradation systems. Annu. Rev. Biochem. 2017, 86, 193–224. [Google Scholar] [CrossRef]
- Hipp, M.S.; Kasturi, P.; Hartl, F.U. The proteostasis network and its decline in ageing. Nat. Rev. Mol. Cell Biol. 2019, 20, 421–435. [Google Scholar] [CrossRef]
- Aguilaniu, H.; Gustafsson, L.; Rigoulet, M.; Nystrom, T. Asymmetric inheritance of oxidatively damaged proteins during cytokinesis. Science 2003, 299, 1751–1753. [Google Scholar] [CrossRef]
- 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] [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]
- Hajieva, P. The Effect of Polyphenols on Protein Degradation Pathways: Implications for Neuroprotection. Molecules 2017, 22, 159. [Google Scholar] [CrossRef] [Green Version]
- Pignatti, C.; D’Adamo, S.; Stefanelli, C.; Flamigni, F. Nutrients and Pathways that Regulate Health Span and Life Span. Geriatrics 2020, 5, 95. [Google Scholar] [CrossRef]
- 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]
- Cui, Z.; Zhao, X.; Amevor, F.K.; Du, X.; Wang, Y.; Li, D.; Shu, G.; Tian, Y.; Zhao, X. Therapeutic application of quercetin in aging-related diseases: SIRT1 as a potential mechanism. Front. Immunol. 2022, 13, 943321. [Google Scholar] [CrossRef]
- Yu, A.; Dang, W. Regulation of stem cell aging by SIRT1-Linking metabolic signaling to epigenetic modifications. Mol. Cell. Endocrinol. 2017, 455, 75–82. [Google Scholar] [CrossRef]
- Xu, C.; Wang, L.; Fozouni, P.; Evjen, G.; Chandra, V.; Jiang, J.; Lu, C.; Nicastri, M.; Bretz, C.; Winkler, J.D.; et al. SIRT1 is downregulated by autophagy in senescence and ageing. Nature 2020, 22, 1170–1179. [Google Scholar] [CrossRef]
- Ong, A.L.; Ramasamy, T.S. Role of Sirtuin1-p53 regulatory axis in aging, cancer and cellular reprogramming. Ageing Res. Rev. 2018, 43, 64–80. [Google Scholar] [CrossRef]
- Imai, S.-i.; Guarente, L. NAD+ and sirtuins in aging and disease. Trends Cell Biol. 2014, 24, 464–471. [Google Scholar] [CrossRef]
- 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. Ageing 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. Oxidative Med. Cell. Longev. 2016, 2016, 3565127. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.-H.; Lee, J.-H.; Lee, H.-Y.; Min, K.-J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24. [Google Scholar] [CrossRef] [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] [PubMed] [Green Version]
- Sarubbo, F.; Ramis, M.; 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] [PubMed]
- 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]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Nissanka, N.; Moraes, C.T. Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 2018, 592, 728–742. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Evans, C.S.; Holzbaur, E.L. Autophagy and mitophagy in ALS. Neurobiol. Dis. 2019, 122, 35–40. [Google Scholar] [CrossRef]
- Pourahmad, J.; Salimi, A.; Seydi, E. Free Radicals and Diseases; Intech Open: Rijeka, Croatia, 2016. [Google Scholar]
- Chodari, L.; Dilsiz Aytemir, M. Targeting Mitochondrial Biogenesis with Polyphenol Compounds. Oxidative Med. Cell. Longev. 2021, 2021, 4946711. [Google Scholar] [CrossRef] [PubMed]
- Ashkar, F.; Bhullar, K.S.; Wu, J. The Effect of Polyphenols on Kidney Disease: Targeting Mitochondria. Nutrients 2022, 14, 3115. [Google Scholar] [CrossRef]
- Yessenkyzy, A.; Saliev, T. Polyphenols as Caloric-Restriction Mimetics and Autophagy Inducers in Aging Research. Nutrients 2020, 12, 1344. [Google Scholar] [CrossRef] [PubMed]
- Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef] [Green Version]
- Dilberger, B.; Passon, M. Polyphenols and Metabolites Enhance Survival in Rodents and Nematodes-Impact of Mitochondria. Nutrients 2019, 11, 1886. [Google Scholar] [CrossRef] [Green Version]
- 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. Oxidative 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]
- Gherardi, G.; Corbioli, G.; Ruzza, F.; Rizzuto, R. CoQ(10) and Resveratrol Effects to Ameliorate Aged-Related Mitochondrial Dysfunctions. Nutrients 2022, 14, 4326. [Google Scholar] [CrossRef]
- Ungvari, Z.; Sonntag, W.E.; de Cabo, R.; Baur, J.A.; Csiszar, A. Mitochondrial protection by resveratrol. Exerc. Sport Sci. Rev. 2011, 39, 128–132. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed]
- Goñi, I.; Hernández-Galiot, A. Intake of Nutrient and Non-Nutrient Dietary Antioxidants. Contribution of Macromolecular Antioxidant Polyphenols in an Elderly Mediterranean Population. Nutrients 2019, 11, 2165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lourenço, S.C.; Moldão-Martins, M. Antioxidants of Natural Plant Origins: From Sources to Food Industry Applications. Molecules 2019, 24, 4132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, J.; Si, H.; Jia, Z.; Liu, D. Dietary Anti-Aging Polyphenols and Potential Mechanisms. Antioxidants 2021, 10, 283. [Google Scholar] [CrossRef] [PubMed]
- Truong, V.L.; Jeong, W.S. Cellular Defensive Mechanisms of Tea Polyphenols: Structure-Activity Relationship. Int. J. Mol. Sci. 2021, 22, 9109. [Google Scholar] [CrossRef]
- Yu, Z.; Zhang, F. Association between Circulating Antioxidants and Longevity: Insight from Mendelian Randomization Study. BioMed Res. Int. 2022, 2022, 4012603. [Google Scholar] [CrossRef]
- Dodig, S.; Čepelak, I.; Pavić, I. Hallmarks of senescence and aging. Biochem. Med. 2019, 29, 483–497. [Google Scholar] [CrossRef]
- Herranz, N.; Gil, J. Mechanisms and functions of cellular senescence. J. Clin. Investig. 2018, 128, 1238–1246. [Google Scholar] [CrossRef] [Green Version]
- Vicencio, J.M.; Galluzzi, L.; Tajeddine, N.; Ortiz, C.; Criollo, A.; Tasdemir, E.; Morselli, E.; Younes, A.B.; Maiuri, M.C.; Lavandero, S.; et al. Senescence, apoptosis or autophagy? Gerontology 2008, 54, 92–99. [Google Scholar] [CrossRef]
- Yanagi, S.; Tsubouchi, H.; Miura, A.; Matsuo, A.; Matsumoto, N.; Nakazato, M. The impacts of cellular senescence in elderly pneumonia and in age-related lung diseases that increase the risk of respiratory infections. Int. J. Mol. Sci. 2017, 18, 503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawless, C.; Wang, C.; Jurk, D.; Merz, A.; von Zglinicki, T.; Passos, J.F. Quantitative assessment of markers for cell senescence. Exp. Gerontol. 2010, 45, 772–778. [Google Scholar] [CrossRef] [PubMed]
- Sone, H.; Kagawa, Y. Pancreatic beta cell senescence contributes to the pathogenesis of type 2 diabetes in high-fat diet-induced diabetic mice. Diabetologia 2005, 48, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Fyhrquist, F.; Saijonmaa, O.; Strandberg, T. The roles of senescence and telomere shortening in cardiovascular disease. Nat. Rev. Cardiol. 2013, 10, 274–283. [Google Scholar] [CrossRef] [PubMed]
- Minamino, T.; Orimo, M.; Shimizu, I.; Kunieda, T.; Yokoyama, M.; Ito, T.; Nojima, A.; Nabetani, A.; Oike, Y.; Matsubara, H.; et al. A crucial role for adipose tissue p53 in the regulation of insulin resistance. Nat. Med. 2009, 15, 1082–1087. [Google Scholar] [CrossRef] [PubMed]
- Unterluggauer, H.; Hampel, B.; Zwerschke, W.; Jansen-Dürr, P. Senescence-associated cell death of human endothelial cells: The role of oxidative stress. Exp. Gerontol. 2003, 38, 1149–1160. [Google Scholar] [CrossRef]
- Joseph, J.; Cutler, R. The role of oxidative stress in signal transduction changes and cell loss in senescence. Ann. N. Y. Acad. Sci. 1994, 738, 37–43. [Google Scholar] [CrossRef]
- Roger, L.; Tomas, F.; Gire, V. Mechanisms and Regulation of Cellular Senescence. Int. J. Mol. Sci. 2021, 22, 13173. [Google Scholar] [CrossRef]
- Bayram, B.; Ozcelik, B.; Grimm, S.; Roeder, T.; Schrader, C.; Ernst, I.M.; Wagner, A.E.; Grune, T.; Frank, J.; Rimbach, G. A diet rich in olive oil phenolics reduces oxidative stress in the heart of SAMP8 mice by induction of Nrf2-dependent gene expression. Rejuvenation Res. 2012, 15, 71–81. [Google Scholar] [CrossRef] [Green Version]
- Maharajan, N.; Vijayakumar, K.; Jang, C.H.; Cho, G.-W. Caloric restriction maintains stem cells through niche and regulates stem cell aging. J. Mol. Med. 2020, 98, 25–37. [Google Scholar] [CrossRef]
- Giovannelli, L. Beneficial effects of olive oil phenols on the aging process: Experimental evidence and possible mechanisms of action. Nutr. Aging 2012, 1, 207–223. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- Puca, F.; Fedele, M. Role of Diet in Stem and Cancer Stem Cells. Int. J. Mol. Sci. 2022, 23, 8108. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Huang, H.; Li, B.; Wu, D.; Wang, F.; Zheng, X.H.; Chen, Q.; Wu, B.; Fan, X. Olive oil in the prevention and treatment of osteoporosis after artificial menopause. Clin. Interv. Aging 2014, 9, 2087. [Google Scholar] [CrossRef] [Green Version]
- Cardoso, C.R.; Favoreto, S., Jr.; Oliveira, L.L.D.; Vancim, J.O.; Barban, G.B.; Ferraz, D.B.; Silva, J.S.D. Oleic acid modulation of the immune response in wound healing: A new approach for skin repair. Immunobiology 2011, 216, 409–415. [Google Scholar] [CrossRef] [PubMed]
- Shannon, O.M.; Ashor, A.W.; Scialo, F.; Saretzki, G.; Martin-Ruiz, C.; Lara, J.; Matu, J.; Griffiths, A.; Robinson, N.; Lillà, L.; et al. Mediterranean diet and the hallmarks of ageing. Eur. J. Clin. Nutr. 2021, 75, 1176–1192. [Google Scholar] [CrossRef]
- Cesari, F.; Sofi, F.; Lova, R.M.; Vannetti, F.; Pasquini, G.; Cecchi, F.; Marcucci, R.; Gori, A.; Macchi, C.; Boni, R.; et al. Aging process, adherence to Mediterranean diet and nutritional status in a large cohort of nonagenarians: Effects on endothelial progenitor cells. Nutr. Metab. Cardiovasc. Dis. 2018, 28, 84–90. [Google Scholar] [CrossRef] [PubMed]
- Marin, C.; Ramirez, R.; Delgado-Lista, J.; Yubero-Serrano, E.M.; Perez-Martinez, P.; Carracedo, J.; Garcia-Rios, A.; Rodriguez, F.; Gutierrez-Mariscal, F.M.; Gomez, P.; et al. Mediterranean diet reduces endothelial damage and improves the regenerative capacity of endothelium. Am. J. Clin. Nutr. 2011, 93, 267–274. [Google Scholar] [CrossRef] [Green Version]
- Fernández, J.M.; Rosado-Álvarez, D.; Da Silva Grigoletto, M.E.; Rangel-Zúñiga, O.A.; Landaeta-Díaz, L.L.; Caballero-Villarraso, J.; López-Miranda, J.; Pérez-Jiménez, F.; Fuentes-Jiménez, F. Moderate-to-high-intensity training and a hypocaloric Mediterranean diet enhance endothelial progenitor cells and fitness in subjects with the metabolic syndrome. Clin. Sci. 2012, 123, 361–373. [Google Scholar] [CrossRef]
- Kim, H.J.; Kim, W.J.; Shin, H.R.; Yoon, H.I.; Moon, J.I.; Lee, E.; Lim, J.M.; Cho, Y.D.; Lee, M.H.; Kim, H.G.; et al. ROS-induced PADI2 downregulation accelerates cellular senescence via the stimulation of SASP production and NFκB activation. Cell. Mol. Life Sci. 2022, 79, 155. [Google Scholar] [CrossRef]
- Chien, Y.; Scuoppo, C.; Wang, X.; Fang, X.; Balgley, B.; Bolden, J.E.; Premsrirut, P.; Luo, W.; Chicas, A.; Lee, C.S.; et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev. 2011, 25, 2125–2136. [Google Scholar] [CrossRef] [Green Version]
- Rea, I.M.; Gibson, D.S.; McGilligan, V.; McNerlan, S.E.; Alexander, H.D.; Ross, O.A. Age and Age-Related Diseases: Role of Inflammation Triggers and Cytokines. Front. Immunol. 2018, 9, 586. [Google Scholar] [CrossRef] [PubMed]
- Koppula, S.; Akther, M.; Haque, M.E. Potential Nutrients from Natural and Synthetic Sources Targeting Inflammaging—A Review of Literature, Clinical Data and Patents. Nutrients 2021, 13, 4058. [Google Scholar] [CrossRef]
- Calder, P.C.; Bosco, N.; Bourdet-Sicard, R.; Capuron, L.; Delzenne, N.; Doré, J.; Franceschi, C.; Lehtinen, M.J.; Recker, T.; Salvioli, S.; et al. Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res. Rev. 2017, 40, 95–119. [Google Scholar] [CrossRef]
- Miquel, J. An update of the oxidation-inflammation theory of aging: The involvement of the immune system in oxi-inflamm-aging. Curr. Pharm. Des. 2009, 15, 3003–3026. [Google Scholar]
- Salminen, A.; Huuskonen, J.; Ojala, J.; Kauppinen, A.; Kaarniranta, K.; Suuronen, T. Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Res. Rev. 2008, 7, 83–105. [Google Scholar] [CrossRef]
- Shakoor, H.; Feehan, J. Immunomodulatory Effects of Dietary Polyphenols. Nutrients 2021, 13, 728. [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] [Green Version]
- Said, R.S.; El-Demerdash, E.; Nada, A.S.; Kamal, M.M. Resveratrol inhibits inflammatory signaling implicated in ionizing radiation-induced premature ovarian failure through antagonistic crosstalk between silencing information regulator 1 (SIRT1) and poly (ADP-ribose) polymerase 1 (PARP-1). Biochem. Pharmacol. 2016, 103, 140–150. [Google Scholar] [CrossRef]
- Tung, B.T.; Rodríguez-Bies, E.; Talero, E.; Gamero-Estevez, E.; Motilva, V.; Navas, P.; López-Lluch, G. Anti-inflammatory effect of resveratrol in old mice liver. Exp. Gerontol. 2015, 64, 1–7. [Google Scholar] [CrossRef]
- Jeong, S.I.; Shin, J.A.; Cho, S.; Kim, H.W.; Lee, J.Y.; Kang, J.L.; Park, E.-M. Resveratrol attenuates peripheral and brain inflammation and reduces ischemic brain injury in aged female mice. Neurobiol. Aging 2016, 44, 74–84. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Wiczkowski, W.; Romaszko, J.; Bucinski, A.; Szawara-Nowak, D.; Honke, J.; Zielinski, H.; Piskula, M.K. Quercetin from shallots (Allium cepa L. var. aggregatum) is more bioavailable than its glucosides. J. Nutr. 2008, 138, 885–888. [Google Scholar]
- Aguirre, L.; Arias, N.; Teresa Macarulla, M.; Gracia, A.; P Portillo, M. Beneficial effects of quercetin on obesity and diabetes. Open Nutraceuticals J. 2011, 4, 189–198. [Google Scholar]
- Liu, S.; Liu, J.; He, L.; Liu, L.; Cheng, B.; Zhou, F.; Cao, D.; He, Y. A Comprehensive Review on the Benefits and Problems of Curcumin with Respect to Human Health. Molecules 2022, 27, 4400. [Google Scholar] [CrossRef]
- Xu, X.Y.; Meng, X.; Li, S.; Gan, R.Y. Bioactivity, Health Benefits, and Related Molecular Mechanisms of Curcumin: Current Progress, Challenges and Perspectives. Nutrients 2018, 10, 1553. [Google Scholar] [CrossRef] [Green Version]
- Kong, W.Y.; Ngai, S.C.; Goh, B.H. Is Curcumin the Answer to Future Chemotherapy Cocktail? Molecules 2021, 26, 4329. [Google Scholar] [CrossRef]
- Peng, Y.; Ao, M.; Dong, B.; Jiang, Y.; Yu, L.; Chen, Z.; Hu, C.; Xu, R. Anti-Inflammatory Effects of Curcumin in the Inflammatory Diseases: Status, Limitations and Countermeasures. Drug Des. Dev. Ther. 2021, 15, 4503–4525. [Google Scholar] [CrossRef]
- Pulido-Moran, M.; Moreno-Fernandez, J.; Ramirez-Tortosa, C.; Ramirez-Tortosa, M. Curcumin and Health. Molecules 2016, 21, 264. [Google Scholar] [CrossRef]
- Kunnumakkara, A.B.; Bordoloi, D.; Padmavathi, G.; Monisha, J.; Roy, N.K.; Prasad, S.; Aggarwal, B.B. Curcumin, the golden nutraceutical: Multitargeting for multiple chronic diseases. Br. J. Pharmacol. 2017, 174, 1325–1348. [Google Scholar] [CrossRef] [Green Version]
- Lakey-Beitia, J.; González, Y.; Doens, D.; Stephens, D.E.; Santamaría, R.; Murillo, E.; Gutiérrez, M.; Fernández, P.L.; Rao, K.S.; Larionov, O.V.; et al. Assessment of Novel Curcumin Derivatives as Potent Inhibitors of Inflammation and Amyloid-β Aggregation in Alzheimer’s Disease. J. Alzheimer’s Dis. 2017, 60 (Suppl. S1), S59–S68. [Google Scholar] [CrossRef] [Green Version]
- Hussain, Y.; Khan, H.; Alotaibi, G.; Khan, F. How Curcumin Targets Inflammatory Mediators in Diabetes: Therapeutic Insights and Possible Solutions. Molecules 2022, 27, 4058. [Google Scholar] [CrossRef]
- Basnet, P.; Skalko-Basnet, N. Curcumin: An anti-inflammatory molecule from a curry spice on the path to cancer treatment. Molecules 2011, 16, 4567–4598. [Google Scholar] [CrossRef] [Green Version]
- Zhou, H.; Beevers, C.S.; Huang, S. The targets of curcumin. Curr. Drug Targets 2011, 12, 332–347. [Google Scholar] [CrossRef]
- de Alcântara, G.F.; Simões-Neto, E.; da Cruz, G.M.; Nobre, M.E.; Neves, K.R.; de Andrade, G.M.; Brito, G.A.; Viana, G.S. Curcumin reverses neurochemical, histological and immuno-histochemical alterations in the model of global brain ischemia. J. Tradit. Complement. Med. 2017, 7, 14–23. [Google Scholar] [CrossRef] [Green Version]
- Ojha, R.P.; Rastogi, M.; Devi, B.P.; Agrawal, A.; Dubey, G.P. Neuroprotective effect of curcuminoids against inflammation-mediated dopaminergic neurodegeneration in the MPTP model of Parkinson’s disease. J. Neuroimmune Pharmacol. 2012, 7, 609–618. [Google Scholar] [CrossRef]
- Chi, X.; Ma, X.; Li, Z.; Zhang, Y.; Wang, Y.; Yuan, L.; Wu, Y.; Xu, W. Protective Effect of Epigallocatechin-3-Gallate in Hydrogen Peroxide-Induced Oxidative Damage in Chicken Lymphocytes. Oxidative Med. Cell. Longev. 2020, 2020, 7386239. [Google Scholar] [CrossRef]
- Mut-Salud, N.; Álvarez, P.J.; Garrido, J.M.; Carrasco, E.; Aránega, A.; Rodríguez-Serrano, F. Antioxidant Intake and Antitumor Therapy: Toward Nutritional Recommendations for Optimal Results. Oxidative Med. Cell. Longev. 2016, 2016, 6719534. [Google Scholar] [CrossRef] [Green Version]
- Farhan, M. Green Tea Catechins: Nature’s Way of Preventing and Treating Cancer. Int J Mol Sci 2022, 23, 10713. [Google Scholar] [CrossRef]
- Fechtner, S.; Singh, A.; Chourasia, M.; Ahmed, S. Molecular insights into the differences in anti-inflammatory activities of green tea catechins on IL-1β signaling in rheumatoid arthritis synovial fibroblasts. Toxicol. Appl. Pharmacol. 2017, 329, 112–120. [Google Scholar] [CrossRef]
- Wu, Y.R.; Choi, H.J.; Kang, Y.G.; Kim, J.K.; Shin, J.W. In vitro study on anti-inflammatory effects of epigallocatechin-3-gallate-loaded nano-and microscale particles. Int. J. Nanomed. 2017, 12, 7007–7013. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Lai, Y.J.; Chan, Y.L.; Li, T.L.; Wu, C.J. Epigallocatechin-3-gallate effectively attenuates skeletal muscle atrophy caused by cancer cachexia. Cancer Lett. 2011, 305, 40–49. [Google Scholar] [CrossRef]
- Rahman, M.M.; Rahaman, M.S. Role of Phenolic Compounds in Human Disease: Current Knowledge and Future Prospects. Molecules 2021, 27, 233. [Google Scholar] [CrossRef]
- Ide, K.; Yamada, H.; Takuma, N.; Park, M.; Wakamiya, N.; Nakase, J.; Ukawa, Y.; Sagesaka, Y.M. Green tea consumption affects cognitive dysfunction in the elderly: A pilot study. Nutrients 2014, 6, 4032–4042. [Google Scholar] [CrossRef]
- Baker, L.D.; Manson, J.E.; Rapp, S.R.; Sesso, H.D.; Gaussoin, S.A.; Shumaker, S.A.; Espeland, M.A. Effects of cocoa extract and a multivitamin on cognitive function: A randomized clinical trial. Alzheimer’s Dement. 2022. [Google Scholar] [CrossRef]
- Rezende, C.; Oliveira, G.V.; Volino-Souza, M.; Castro, P.; Murias, J.M. Turmeric root extract supplementation improves pre-frontal cortex oxygenation and blood volume in older males and females: A randomised cross-over, placebo-controlled study. Int. J. Food Sci. Nutr. 2022, 73, 274–283. [Google Scholar] [CrossRef]
- Anton, S.D.; Embry, C.; Marsiske, M.; Lu, X.; Doss, H.; Leeuwenburgh, C.; Manini, T.M. Safety and metabolic outcomes of resveratrol supplementation in older adults: Results of a twelve-week, placebo-controlled pilot study. Exp. Gerontol. 2014, 57, 181–187. [Google Scholar] [CrossRef] [Green Version]
- Harper, S.A.; Bassler, J.R.; Peramsetty, S.; Yang, Y.; Roberts, L.M.; Drummer, D.; Mankowski, R.T.; Leeuwenburgh, C.; Ricart, K.; Patel, R.P.; et al. Resveratrol and exercise combined to treat functional limitations in late life: A pilot randomized controlled trial. Exp. Gerontol. 2021, 143, 111111. [Google Scholar] [CrossRef]
Polyphenols | Models | Main Effects | Ref |
---|---|---|---|
Apigenin | MSCs Hematopoietic stem cell | ↓Adipogenesis/↑osteoblastogenesis ↑Lifespan ↑Asymmetric divisions | [40] [41] |
Catechins | Hepa1c1c7 cells | ↑ARE/Nrf2 | [42] |
Curcumin | MCF-7 cells A549 cells Microglial cells Astrocytes cells | ↓DNMT1; ↑RASSF1A ↓DNMT3b; ↑RARβ ↓NF-κB ↓TNF-α ↓IL-6 ↓COX-2 | [36] [38] [39] [43] |
DADS | Caco-2 cells HT-29 cells | ↑HDAC activity; ↑H3 and H4 acetylation; ↑p21waf1/cip1 | [30] |
EGCG | A549 cells HL60 cells KYSE-150 cells PC3 cells MCF-7cells Macrophages ECV304 cells | ↓Cell proliferation; ↑Cell cycle arrest in G1 phase; ↑Apoptotic activity; ↓hTERTmethylation; ↓H3K9 acetylation; ↓DNMT1, DNMT3a and DNMT3b; ↓HDAC activity. ↓NF-κB ↓TNF-α ↓iNOS ↓COX-2 ↓AP-1 | [31] [32] [33] [34] [44] [45] |
Gallic acid | Rat embryonic fibroblast cells | ↓β-galactosidase activity ↓ROS | [46] |
Hydroxytyrosol | MRC5 cells NHDF cells | ↓NFκB ↓β-galactosidadse activity ↓Accumulation of senescent cells ↓p16 ↓SASP (IL-6, COX2, TNFα) | [47] |
Oleacein | SPCs | ↑ Angiotensin II ↑Nrf2/heme oxygenase-1 | [48] |
Oleic acid | MSCs | ↑EphB2 | [49] |
Oleuropein | MRC5 cells NHDF cells IMR90 cells WI38 cells MSCs Hematopoietic stem cell SPCs | ↓NFκB ↓β-galactosidadse activity ↓Accumulation of senescent cells ↓p16 ↓SASP(IL-6, COX2, TNFα) ↓ROS ↑Proteasome activity ↓Adipogenesis/↑osteoblastogenesis ↑Lifespan ↑AsyMmetricdivisions ↑Angiotensin II ↑Nrf2/heme oxygenase-1 | [47] [50] [51] [41] [48] |
Procyanidins | BEAS-2B WRL-68 | ↓DNA damage ↑ATM and ATR activity | [52] |
Quercetin | Primary human fibroblasts HUVEC cultures Murine glial cells Macrophages A549 Mast cells | ↓SASP ↓β-galactosidadse activity ↓Accumulation of senescent cells ↓p16 ↓p21 ↓TNF-α ↓IL-8 | [53] [54] [55] [56] [57] [58] |
Resveratrol | Mouse embryonic fibroblasts IMR-90 cells BMMSCs Rat cardiomyocyte cultures 3T3 | ↓Mutations in the ARF/p53 pathway ↓DNA double-strand breaks ↑Telomerase activity ↑SIRT1 ↑Sirt1/Sirt3-FoxO pathway ↑Sirt1/Sirt3-Mfn2-Parkin-PGC-1α Pathway ↓NF-κB ↓TNF-α, IL-1β and IL-6 expression ↓mTOR phosphorylation) ↓S6RP | [59] [60] [61] [62] [63] [64] [65] |
Sulphoraphane | Embryonic fibroblast cells | ↑Nrf2/ARE | [66] |
Treatment | Trial ID | Phase | No Pts | Condition | Results | Ref |
---|---|---|---|---|---|---|
Green tea powder 2 g/day for 3 months | NCT01594086 | NA | 15 | Cognitive dysfunction in elderly | Improved cognitive function and reduced progression of vascular dementia | [227] |
Cocoa extract (500 mg/d flavanols, including 80 mg. (-)-epicatechins) 3-year trial | NCT03035201 | NA | 2262 | Cognitive dysfunction in elderly | Cocoa extract did not benefit cognition | [228] |
Cocoa extract (500 mg/d flavanols, including 80 mg. (-)-epicatechins) 2-year trial | NCT04582617 | NA | 4000 | Cognitive changes and brain structure | No results posted | |
10 g of curcumin supplementation | NCT04119752 | NA | 28 | Elderly with cardiometabolic risks | Improved cerebral oxygenation and blood volume | [229] |
Curcumin (1000 mg/day) 3-month trial | NCT03085680 | Phase 2 | 17 | Ageing adults at increased risk for disability | Improvement in attention, memory, and physical function grip strength | |
300 or 1000 mg/d of resveratrol, 3-month trial | NCT01126229 | Phase 1 | 32 | Overweight, older adults | Supports the safety of resveratrol supplementation in this at-risk population | [230] |
500 or 1000 mg of resveratrol per day, 3-month trial | NCT02523274 | Phase 2 | 60 | Ageing | Improved skeletal muscle mitochondrial function and mobility-related indices of physical function | [231] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Pereira, Q.C.; dos Santos, T.W.; Fortunato, I.M.; Ribeiro, M.L. The Molecular Mechanism of Polyphenols in the Regulation of Ageing Hallmarks. Int. J. Mol. Sci. 2023, 24, 5508. https://doi.org/10.3390/ijms24065508
Pereira QC, dos Santos TW, Fortunato IM, Ribeiro ML. The Molecular Mechanism of Polyphenols in the Regulation of Ageing Hallmarks. International Journal of Molecular Sciences. 2023; 24(6):5508. https://doi.org/10.3390/ijms24065508
Chicago/Turabian StylePereira, Quélita Cristina, Tanila Wood dos Santos, Isabela Monique Fortunato, and Marcelo Lima Ribeiro. 2023. "The Molecular Mechanism of Polyphenols in the Regulation of Ageing Hallmarks" International Journal of Molecular Sciences 24, no. 6: 5508. https://doi.org/10.3390/ijms24065508
APA StylePereira, Q. C., dos Santos, T. W., Fortunato, I. M., & Ribeiro, M. L. (2023). The Molecular Mechanism of Polyphenols in the Regulation of Ageing Hallmarks. International Journal of Molecular Sciences, 24(6), 5508. https://doi.org/10.3390/ijms24065508