Tea Polyphenols as Prospective Natural Attenuators of Brain Aging
Abstract
:1. Introduction
2. Possible Triggers and Mechanisms of Cognitive Decline in the Process of Brain Aging
2.1. Dysregulation of Neuronal Calcium Homeostasis
2.2. Mitochondrial Oxidative Stress in Brain Cells
2.3. Telomere Shortening and Telomerase Dysfunction
3. TP Suppress Brain Aging-Related Signaling Pathways and Hallmarks
3.1. TP Increase the Expression of Neurotrophic Factors and Their Receptors
3.2. TP Activate Signal Pathways for Neurotrophic Function
4. Crosstalk between Brain and Gut Microbiome Have Certain Impact on Aging
5. Effects of TP on Brain Aging by Modulating Gut Microbiota
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- World Population Ageing 2020 Highlights. 2020. Available online: https://www.un.org/development/desa/pd/ (accessed on 15 June 2022).
- Naoi, M.; Inaba-Hasegawa, K.; Shamoto-Nagai, M.; Maruyama, W. Neurotrophic function of phytochemicals for neuroprotection in aging and neurodegenerative disorders: Modulation of intracellular signaling and gene expression. J. Neural Transm. 2017, 124, 1515–1527. [Google Scholar] [CrossRef] [PubMed]
- Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- d’Avila, J.C.; Siqueira, L.D.; Mazeraud, A.; Azevedo, E.P.; Foguel, D.; Castro-Faria-Neto, H.C.; Sharshar, T.; Chrétien, F.; Bozza, F.A. Age-related cognitive impairment is associated with long-term neuroinflammation and oxidative stress in a mouse model of episodic systemic inflammation. J. Neuroinflamm. 2018, 15, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, X.; Li, B.; Lou, P.; Dai, T.; Chen, Y.; Zhuge, A.; Yuan, Y.; Li, L. The relationship between the gut microbiome and neurodegenerative diseases. Neurosci. Bull. 2021, 37, 1510–1522. [Google Scholar] [CrossRef]
- Konjevod, M.; Nikolac Perkovic, M.; Sáiz, J.; Svob Strac, D.; Barbas, C.; Rojo, D. Metabolomics analysis of microbiota-gut-brain axis in neurodegenerative and psychiatric diseases. J. Pharm. Biomed. Anal. 2021, 194, 113681. [Google Scholar] [CrossRef]
- He, D.; Wu, H.; Xiang, J.; Ruan, X.; Peng, P.; Ruan, Y.; Chen, Y.G.; Wang, Y.; Yu, Q.; Zhang, H.; et al. Gut stem cell aging is driven by mTORC1 via a p38 MAPK-p53 pathway. Nat. Commun. 2020, 11, 37. [Google Scholar] [CrossRef] [Green Version]
- Funk, M.C.; Zhou, J.; Boutros, M. Ageing, metabolism and the intestine. EMBO Rep. 2020, 21, e50047. [Google Scholar] [CrossRef]
- Mandel, S.A.; Amit, T.; Kalfon, L.; Reznichenko, L.; Weinreb, O.; Youdim, M.B. Cell signaling pathways and iron chelation in the neurorestorative activity of green tea polyphenols: Special reference to epigallocatechin gallate (EGCG). J. Alzheimers Dis. 2008, 15, 211–222. [Google Scholar] [CrossRef]
- Huang, Y.; Wei, Y.; Xu, J.; Wei, X. A comprehensive review on the prevention and regulation of Alzheimer’s disease by tea and its active ingredients. Crit. Rev. Food Sci. Nutr. 2022, 1–25. [Google Scholar] [CrossRef]
- Li, H.; Christman, L.M.; Li, R.; Gu, L. Synergic interactions between polyphenols and gut microbiota in mitigating inflammatory bowel diseases. Food Funct. 2020, 11, 4878–4891. [Google Scholar] [CrossRef]
- Pérez-Burillo, S.; Navajas-Porras, B.; López-Maldonado, A.; Hinojosa-Nogueira, D.; Pastoriza, S.; Rufián-Henares, J.Á. Green tea and its relation to human gut microbiome. Molecules 2021, 26, 3907. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.C.; Li, X.Y.; Shen, L. Modulation effect of tea consumption on gut microbiota. Appl. Microbiol. Biotechnol. 2020, 104, 981–987. [Google Scholar] [CrossRef] [PubMed]
- Larrosa, M.; Luceri, C.; Vivoli, E.; Pagliuca, C.; Lodovici, M.; Moneti, G.; Dolara, P. Polyphenol metabolites from colonic microbiota exert anti-inflammatory activity on different inflammation models. Mol. Nutr. Food Res. 2009, 53, 1044–1054. [Google Scholar] [CrossRef]
- Wu, M.; Luo, Q.; Nie, R.; Yang, X.; Tang, Z.; Chen, H. Potential implications of polyphenols on aging considering oxidative stress, inflammation, autophagy, and gut microbiota. Crit. Rev. Food Sci. Nutr. 2021, 61, 2175–2193. [Google Scholar] [CrossRef] [PubMed]
- Almeida, S.; Alves, M.G.; Sousa, M.; Oliveira, P.F.; Silva, B.M. Are polyphenols strong dietary agents against neurotoxicity and neurodegeneration? Neurotox. Res. 2016, 30, 345–366. [Google Scholar] [CrossRef] [PubMed]
- Malar, D.S.; Prasanth, M.I.; Brimson, J.M.; Sharika, R.; Sivamaruthi, B.S.; Chaiyasut, C.; Tencomnao, T. Neuroprotective properties of green tea (Camellia sinensis) in Parkinson’s disease: A review. Molecules 2020, 25, 3926. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Ning, L.; Yin, Y.; Wang, R.; Zhang, Z.; Hao, L.; Wang, B.; Zhao, X.; Yang, X.; Yin, L.; et al. Age-related shifts in gut microbiota contribute to cognitive decline in aged rats. Aging 2020, 12, 7801–7817. [Google Scholar] [CrossRef] [PubMed]
- McCay, C.M.; Maynard, L.A.; Sperling, G.; Barnes, L.L. Retarded growth, life span, ultimate body size and age changes in the albino rat after feeding diets restricted in calories: Four figures. J. Nutr. 1939, 18, 1–13. [Google Scholar] [CrossRef]
- Roe, J.M.; Vidal-Piñeiro, D.; Sørensen, Ø.; Brandmaier, A.M.; Düzel, S.; Gonzalez, H.A.; Kievit, R.A.; Knights, E.; Kühn, S.; Lindenberger, U.; et al. Asymmetric thinning of the cerebral cortex across the adult lifespan is accelerated in Alzheimer’s disease. Nat. Commun. 2022, 13, 834. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Yang, D.S.; Goulbourne, C.N.; Im, E.; Stavrides, P.; Pensalfini, A.; Chan, H.; Bouchet-Marquis, C.; Bleiwas, C.; Berg, M.J.; et al. Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat. Neurosci. 2022, 25, 688–701. [Google Scholar] [CrossRef]
- Xu, Y.; Sun, J.; Yang, L.; Zhao, S.; Liu, X.; Su, Y.; Zhang, J.; Zhao, M. Gangliosides play important roles in the nervous system by regulating ion concentrations. Neurochem. Res. 2022, 47, 1791–1798. [Google Scholar] [CrossRef] [PubMed]
- Mattson, M.P.; Arumugam, T.V. Hallmarks of brain aging: Adaptive and pathological modification by metabolic atates. Cell Metab. 2018, 27, 1176–1199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toescu, E.C.; Verkhratsky, A. Neuronal ageing in long-term cultures: Alterations of Ca2+ homeostasis. Neuroreport 2000, 11, 3725–3729. [Google Scholar] [CrossRef] [PubMed]
- Thibault, O.; Gant, J.C.; Landfield, P.W. Expansion of the calcium hypothesis of brain aging and Alzheimer’s disease: Minding the store. Aging Cell 2007, 6, 307–317. [Google Scholar] [CrossRef]
- Makhezer, N.; Ben Khemis, M.; Liu, D.; Khichane, Y.; Marzaioli, V.; Tlili, A.; Mojallali, M.; Pintard, C.; Letteron, P.; Hurtado-Nedelec, M.; et al. NOX1-derived ROS drive the expression of Lipocalin-2 in colonic epithelial cells in inflammatory conditions. Mucosal Immunol. 2019, 12, 117–131. [Google Scholar] [CrossRef] [Green Version]
- Geng, L.; Fan, L.M.; Liu, F.; Smith, C.; Li, J.M. Nox2 dependent redox-regulation of microglial response to amyloid-β stimulation and microgliosis in aging. Sci. Rep. 2020, 10, 1582. [Google Scholar] [CrossRef] [Green Version]
- Madreiter-Sokolowski, C.T.; Thomas, C.; Ristow, M. Interrelation between ROS and Ca2+ in aging and age-related diseases. Redox Biol. 2020, 36, 101678. [Google Scholar] [CrossRef]
- Zaidi, A. Plasma membrane Ca-ATPases: Targets of oxidative stress in brain aging and neurodegeneration. World J. Biol. Chem. 2010, 1, 271–280. [Google Scholar] [CrossRef]
- Bruce, J.I.E. Metabolic regulation of the PMCA: Role in cell death and survival. Cell Calcium 2018, 69, 28–36. [Google Scholar] [CrossRef]
- Garrosa, J.; Paredes, I.; Marambaud, P.; López, M.G.; Cano-Abad, M.F. Molecular and pharmacological modulation of CALHM1 promote neuroprotection against oxygen and glucose deprivation in a model of hippocampal slices. Cells 2020, 9, 664. [Google Scholar] [CrossRef] [Green Version]
- Mossad, O.; Batut, B.; Yilmaz, B.; Dokalis, N.; Mezö, C.; Nent, E.; Nabavi, L.S.; Mayer, M.; Maron, F.; Buescher, J.M.; et al. Gut microbiota drives age-related oxidative stress and mitochondrial damage in microglia via the metabolite N6-carboxymethyllysine. Nat. Neurosci. 2022, 25, 295–305. [Google Scholar] [CrossRef] [PubMed]
- Stefanatos, R.; Sanz, A. The role of mitochondrial ROS in the aging brain. FEBS Lett. 2018, 592, 743–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Swerdlow, R.H. Brain aging, Alzheimer’s disease, and mitochondria. Biochim. Biophys. Acta 2011, 1812, 1630–1639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Chen, W.; Zhao, Y.; Yang, Y. Spatiotemporal imaging of cellular energy metabolism with genetically-encoded fluorescent sensors in brain. Neurosci. Bull. 2018, 34, 875–886. [Google Scholar] [CrossRef] [PubMed]
- Bennett, J.P.; Onyango, I.G. Energy, entropy and quantum tunneling of protons and electrons in brain mitochondria: Relation to mitochondrial impairment in aging-related human brain diseases and therapeutic measures. Biomedicines 2021, 9, 225. [Google Scholar] [CrossRef]
- Kandimalla, R.; Manczak, M.; Fry, D.; Suneetha, Y.; Sesaki, H.; Reddy, P.H. Reduced dynamin-related protein 1 protects against phosphorylated Tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum. Mol. Genet. 2016, 25, 4881–4897. [Google Scholar] [CrossRef] [Green Version]
- Isaev, N.K.; Genrikhs, E.E.; Oborina, M.V.; Stelmashook, E.V. Accelerated aging and aging process in the brain. Rev. Neurosci. 2018, 29, 233–240. [Google Scholar] [CrossRef]
- Braidy, N.; Poljak, A.; Grant, R.; Jayasena, T.; Mansour, H.; Chan-Ling, T.; Guillemin, G.J.; Smythe, G.; Sachdev, P. Mapping NAD+ metabolism in the brain of ageing Wistar rats: Potential targets for influencing brain senescence. Biogerontology 2014, 15, 177–198. [Google Scholar] [CrossRef]
- Fang, E.F.; Lautrup, S.; Hou, Y.; Demarest, T.G.; Croteau, D.L.; Mattson, M.P.; Bohr, V.A. NAD+ in aging: Molecular mechanisms and translational implications. Trends Mol. Med. 2017, 23, 899–916. [Google Scholar] [CrossRef]
- Covarrubias, A.J.; Perrone, R.; Grozio, A.; Verdin, E. NAD+ metabolism and its roles in cellular processes during ageing. Nat. Rev. Mol. Cell Biol. 2021, 22, 119–141. [Google Scholar] [CrossRef]
- Kim, C.K.; Sachdev, P.S.; Braidy, N. Recent neurotherapeutic strategies to promote healthy brain aging: Are we there yet? Aging Dis. 2022, 13, 175–214. [Google Scholar] [CrossRef] [PubMed]
- Lautrup, S.; Sinclair, D.A.; Mattson, M.P.; Fang, E.F. NAD(+) in brain aging and neurodegenerative disorders. Cell Metab. 2019, 30, 630–655. [Google Scholar] [CrossRef] [PubMed]
- Luceri, C.; Bigagli, E.; Femia, A.P.; Caderni, G.; Giovannelli, L.; Lodovici, M. Aging related changes in circulating reactive oxygen species (ROS) and protein carbonyls are indicative of liver oxidative injury. Toxicol. Rep. 2017, 5, 141–145. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Halliwell, B. Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat. Rev. Neurosci. 2019, 20, 148–160. [Google Scholar] [CrossRef] [PubMed]
- Jeong, Y.J.; Son, Y.; Han, N.K.; Choi, H.D.; Pack, J.K.; Kim, N.; Lee, Y.S.; Lee, H.J. Impact of long-term RF-EMF on oxidative stress and neuroinflammation in aging brains of C57BL/6 mice. Int. J. Mol. Sci. 2018, 19, 2103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsoukalas, D.; Buga, A.M.; Docea, A.O.; Sarandi, E.; Mitrut, R.; Renieri, E.; Spandidos, D.A.; Rogoveanu, I.; Cercelaru, L.; Niculescu, M.; et al. Reversal of brain aging by targeting telomerase: A nutraceutical approach. Int. J. Mol. Med. 2021, 48, 199. [Google Scholar] [CrossRef] [PubMed]
- Rossiello, F.; Jurk, D.; Passos, J.F.; d’Adda di Fagagna, F. Telomere dysfunction in ageing and age-related diseases. Nat. Cell Biol. 2022, 24, 135–147. [Google Scholar] [CrossRef]
- Boccardi, V.; Arosio, B.; Cari, L.; Bastiani, P.; Scamosci, M.; Casati, M.; Ferri, E.; Bertagnoli, L.; Ciccone, S.; Rossi, P.D.; et al. Beta-carotene, telomerase activity and Alzheimer’s disease in old age subjects. Eur. J. Nutr. 2020, 59, 119–126. [Google Scholar] [CrossRef]
- Jolivet, P.; Serhal, K.; Graf, M.; Eberhard, S.; Xu, Z.; Luke, B.; Teixeira, M.T. A subtelomeric region affects telomerase-negative replicative senescence in Saccharomyces cerevisiae. Sci. Rep. 2019, 9, 1845. [Google Scholar] [CrossRef]
- Chakravarti, D.; LaBella, K.A.; DePinho, R.A. Telomeres: History, health, and hallmarks of aging. Cell 2021, 184, 306–322. [Google Scholar] [CrossRef]
- Petr, M.A.; Tulika, T.; Carmona-Marin, L.M.; Scheibye-Knudsen, M. Protecting the aging genome. Trends Cell Biol. 2020, 30, 117–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schumacher, B.; Pothof, J.; Vijg, J.; Hoeijmakers, J.H.J. The central role of DNA damage in the ageing process. Nature 2021, 592, 695–703. [Google Scholar] [CrossRef] [PubMed]
- Boccardi, V.; Pelini, L.; Ercolani, S.; Ruggiero, C.; Mecocci, P. From cellular senescence to Alzheimer’s disease: The role of telomere shortening. Ageing Res. Rev. 2015, 22, 1–8. [Google Scholar] [CrossRef]
- Blinkouskaya, Y.; Caçoilo, A.; Gollamudi, T.; Jalalian, S.; Weickenmeier, J. Brain aging mechanisms with mechanical manifestations. Mech. Ageing Dev. 2021, 200, 111575. [Google Scholar] [CrossRef]
- Pini, L.; Pievani, M.; Bocchetta, M.; Altomare, D.; Bosco, P.; Cavedo, E.; Galluzzi, S.; Marizzoni, M.; Frisoni, G.B. Brain atrophy in Alzheimer’s disease and aging. Ageing Res. Rev. 2016, 30, 25–48. [Google Scholar] [CrossRef] [PubMed]
- Truong, V.L.; Jeong, W.S. Antioxidant and anti-inflammatory roles of tea polyphenols in inflammatory bowel diseases. Food Sci. Hum. Wellness 2022, 11, 502–511. [Google Scholar] [CrossRef]
- Khan, N.; Mukhtar, H. Tea polyphenols in promotion of human health. Nutrients 2018, 11, 39. [Google Scholar] [CrossRef] [Green Version]
- Luo, J.; Si, H.; Jia, Z.; Liu, D. Dietary anti-aging polyphenols and potential mechanisms. Antioxidants 2021, 10, 283. [Google Scholar] [CrossRef]
- Perron, N.R.; Brumaghim, J.L. A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef]
- Weinreb, O.; Amit, T.; Mandel, S.; Youdim, M.B. Neuroprotective molecular mechanisms of (-)-epigallocatechin-3-gallate: A reflective outcome of its antioxidant, iron chelating and neuritogenic properties. Genes Nutr. 2009, 4, 283–296. [Google Scholar] [CrossRef] [Green Version]
- Ritchie, K.; Lovestone, S. The dementias. Lancet 2002, 360, 1759–1766. [Google Scholar] [CrossRef]
- Chowdhury, A.; Sarkar, J.; Chakraborti, T.; Pramanik, P.K.; Chakraborti, S. Protective role of epigallocatechin-3-gallate in health and disease: A perspective. Biomed. Pharmacother. 2016, 78, 50–59. [Google Scholar] [CrossRef] [PubMed]
- Weinreb, O.; Mandel, S.; Amit, T.; Youdim, M.B. Neurological mechanisms of green tea polyphenols in Alzheimer’s and Parkinson’s diseases. J. Nutr. Biochem. 2004, 15, 506–516. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.W.; Lee, Y.K.; Ban, J.O.; Ha, T.Y.; Yun, Y.P.; Han, S.B.; Oh, K.W.; Hong, J.T. Green tea (-)-epigallocatechin-3-gallate inhibits beta-amyloid-induced cognitive dysfunction through modification of secretase activity via inhibition of ERK and NF-kappaB pathways in mice. J. Nutr. 2009, 139, 1987–1993. [Google Scholar] [CrossRef] [PubMed]
- Ng, T.P.; Feng, L.; Niti, M.; Kua, E.H.; Yap, K.B. Tea consumption and cognitive impairment and decline in older Chinese adults. Am. J. Clin. Nutr. 2008, 88, 224–231. [Google Scholar] [CrossRef] [PubMed]
- Nurk, E.; Refsum, H.; Drevon, C.A.; Tell, G.S.; Nygaard, H.A.; Engedal, K.; Smith, A.D. Intake of flavonoid-rich wine, tea, and chocolate by elderly men and women is associated with better cognitive test performance. J. Nutr. 2009, 139, 120–127. [Google Scholar] [CrossRef]
- Mandel, S.A.; Amit, T.; Weinreb, O.; Youdim, M.B. Understanding the broad-spectrum neuroprotective action profile of green tea polyphenols in aging and neurodegenerative diseases. J. Alzheimers Dis. 2011, 25, 187–208. [Google Scholar] [CrossRef]
- Wightman, E.L.; Haskell, C.F.; Forster, J.S.; Veasey, R.C.; Kennedy, D.O. Epigallocatechin gallate, cerebral blood flow parameters, cognitive performance and mood in healthy humans: A double-blind, placebo-controlled, crossover investigation. Hum. Psychopharmacol. 2012, 27, 177–186. [Google Scholar] [CrossRef]
- Ni, L.; Zhao, M.; Hu, Z.; Yang, K.; Zhao, X.; Niu, H.; Lin, H. Neural mechanism of shentai tea polyphenols on cognitive improvements for individuals with subjective cognitive decline: A functional near-infrared spectroscopy study. J. Alzheimers Dis. 2021, 82, 1137–1145. [Google Scholar] [CrossRef]
- Zhu, X.; Wei, Y.; Yang, B.; Yin, X.; Guo, X. The mitohormetic response as part of the cytoprotection mechanism of berberine: Berberine induces mitohormesis and mechanisms. Mol. Med. 2020, 26, 10. [Google Scholar] [CrossRef]
- Tian, J.; Geiss, C.; Zarse, K.; Madreiter-Sokolowski, C.T.; Ristow, M. Green tea catechins EGCG and ECG enhance the fitness and lifespan of Caenorhabditis elegans by complex I inhibition. Aging 2021, 13, 22629–22648. [Google Scholar] [CrossRef] [PubMed]
- Xiong, L.G.; Chen, Y.J.; Tong, J.W.; Gong, Y.S.; Huang, J.A.; Liu, Z.H. Epigallocatechin-3-gallate promotes healthy lifespan through mitohormesis during early-to-mid adulthood in Caenorhabditis elegans. Redox Biol. 2018, 14, 305–315. [Google Scholar] [CrossRef] [PubMed]
- Singh, B.N.; Shankar, S.; Srivastava, R.K. Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem. Pharmacol. 2011, 82, 1807–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lambert, J.D.; Kennett, M.J.; Sang, S.; Reuhl, K.R.; Ju, J.; Yang, C.S. Hepatotoxicity of high oral dose (-)-epigallocatechin-3-gallate in mice. Food Chem. Toxicol. 2010, 48, 409–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Forrester, S.J.; Kikuchi, D.S.; Hernandes, M.S.; Xu, Q.; Griendling, K.K. Reactive oxygen species in metabolic and inflammatory signaling. Circ. Res. 2018, 122, 877–902. [Google Scholar] [CrossRef]
- Ranneh, Y.; Ali, F.; Akim, A.M.; Hamid, H.A.; Khazaai, H.; Fadel, A. Crosstalk between reactive oxygen species and pro-inflammatory markers in developing various chronic diseases: A review. Appl. Biol. Chem. 2017, 60, 327–338. [Google Scholar] [CrossRef]
- Lin, S.M.; Wang, S.W.; Ho, S.C.; Tang, Y.L. Protective effect of green tea (-)-epigallocatechin-3-gallate against the monoamine oxidase B enzyme activity increase in adult rat brains. Nutrition 2010, 26, 1195–1200. [Google Scholar] [CrossRef]
- Jeong, Y.H.; Oh, Y.C.; Pak, M.E.; Li, W.; Go, Y.; Lee, J.J. Pu’er tea water extract protects against cognitive impairment in a mouse model of lipopolysaccharide-induced neuroinflammation. Phytomedicine 2020, 79, 153338. [Google Scholar] [CrossRef]
- Gundimeda, U.; McNeill, T.H.; Fan, T.K.; Deng, R.; Rayudu, D.; Chen, Z.; Cadenas, E.; Gopalakrishna, R. Green tea catechins potentiate the neuritogenic action of brain-derived neurotrophic factor: Role of 67-kDa laminin receptor and hydrogen peroxide. Biochem. Biophys. Res. Commun. 2014, 445, 218–224. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Moosavi, F.; Hosseini, R.; Saso, L.; Firuzi, O. Modulation of neurotrophic signaling pathways by polyphenols. Drug Des. Devel. Ther. 2015, 10, 23–42. [Google Scholar] [PubMed] [Green Version]
- Castrén, E.; Monteggia, L.M. Brain-derived neurotrophic factor signaling in depression and antidepressant action. Biol. Psychiatry 2021, 90, 128–136. [Google Scholar] [CrossRef]
- Sampaio, T.B.; Savall, A.S.; Gutierrez, M.; Pinton, S. Neurotrophic factors in Alzheimer’s and Parkinson’s diseases: Implications for pathogenesis and therapy. Neural Regen. Res. 2017, 12, 549–557. [Google Scholar] [PubMed]
- Webster, N.J.; Pirrung, M.C. Small molecule activators of the Trk receptors for neuroprotection. BMC Neurosci. 2008, 9, S1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirose, M.; Kuroda, Y.; Murata, E. NGF/TrkA signaling as a therapeutic target for pain. Pain Pract. 2016, 16, 175–182. [Google Scholar] [CrossRef] [PubMed]
- Chow, H.H.; Cai, Y.; Hakim, I.A.; Crowell, J.A.; Shahi, F.; Brooks, C.A.; Dorr, R.T.; Hara, Y.; Alberts, D.S. Pharmacokinetics and safety of green tea polyphenols after multiple-dose administration of epigallocatechin gallate and polyphenon E in healthy individuals. Clin. Cancer Res. 2003, 9, 3312–3319. [Google Scholar] [PubMed]
- Assuncao, M.; Andrade, J.P. Protective action of green tea catechins in neuronal mitochondria during aging. Front. Biosci. 2015, 20, 247–262. [Google Scholar]
- Panickar, K.S.; Anderson, R.A. Effect of polyphenols on oxidative stress and mitochondrial dysfunction in neuronal death and brain edema in cerebral ischemia. Int. J. Mol. Sci. 2011, 12, 8181–8207. [Google Scholar] [CrossRef] [Green Version]
- Saleh, I.G.; Ali, Z.; Abe, N.; Wilson, F.D.; Hamada, F.M.; Abd-Ellah, M.F.; Walker, L.A.; Khan, I.A.; Ashfaq, M.K. Effect of green tea and its polyphenols on mouse liver. Fitoterapia 2013, 90, 151–159. [Google Scholar] [CrossRef]
- Isbrucker, R.A.; Edwards, J.A.; Wolz, E.; Davidovich, A.; Bausch, J. Safety studies on epigallocatechin gallate (EGCG) preparations. Part 2: Dermal, acute and short-term toxicity studies. Food Chem. Toxicol. 2006, 44, 636–650. [Google Scholar] [CrossRef]
- Gundimeda, U.; McNeill, T.H.; Schiffman, J.E.; Hinton, D.R.; Gopalakrishna, R. Green tea polyphenols potentiate the action of nerve growth factor to induce neuritogenesis: Possible role of reactive oxygen species. J. Neurosci. Res. 2010, 88, 3644–3655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Uddin, M.S.; Mamun, A.A.; Rahman, M.M.; Jeandet, P.; Alexiou, A.; Behl, T.; Sarwar, M.S.; Sobarzo-Sánchez, E.; Ashraf, G.M.; Sayed, A.A.; et al. Natural products for neurodegeneration: Regulating neurotrophic signals. Oxidative Med. Cell. Longev. 2021, 2021, 8820406. [Google Scholar] [CrossRef] [PubMed]
- More, S.V.; Koppula, S.; Kim, I.S.; Kumar, H.; Kim, B.W.; Choi, D.K. The role of bioactive compounds on the promotion of neurite outgrowth. Molecules 2012, 17, 6728–6753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salminen, A.; Kauppinen, A.; Kaarniranta, K. Phytochemicals suppress nuclear factor-κB signaling: Impact on health span and the aging process. Curr. Opin. Clin. Nutr. Metab. Care 2012, 15, 23–28. [Google Scholar] [CrossRef] [PubMed]
- Pires, B.R.B.; Silva, R.C.M.C.; Ferreira, G.M.; Abdelhay, E. NF-kappaB: Two sides of the same coin. Genes 2018, 9, 24. [Google Scholar] [CrossRef] [Green Version]
- Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
- Chung, H.Y.; Kim, D.H.; Lee, E.K.; Chung, K.W.; Chung, S.; Lee, B.; Seo, A.Y.; Chung, J.H.; Jung, Y.S.; Im, E.; et al. Redefining chronic inflammation in aging and age-related diseases: Proposal of the senoinflammation concept. Aging Dis. 2019, 10, 367–382. [Google Scholar] [CrossRef] [Green Version]
- Kauppinen, A.; Suuronen, T.; Ojala, J.; Kaarniranta, K.; Salminen, A. Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell. Signal. 2013, 25, 1939–1948. [Google Scholar] [CrossRef]
- Zhang, H.S.; Wu, T.C.; Sang, W.W.; Ruan, Z. EGCG inhibits Tat-induced LTR transactivation: Role of Nrf2, AKT, AMPK signaling pathway. Life Sci. 2012, 90, 747–754. [Google Scholar] [CrossRef]
- Matsushita, T.; Sasaki, H.; Takayama, K.; Ishida, K.; Matsumoto, T.; Kubo, S.; Matsuzaki, T.; Nishida, K.; Kurosaka, M.; Kuroda, R. The overexpression of SIRT1 inhibited osteoarthritic gene expression changes induced by interleukin-1β in human chondrocytes. J. Orthop. Res. 2013, 31, 531–537. [Google Scholar] [CrossRef]
- de Gregorio, E.; Colell, A.; Morales, A.; Marí, M. Relevance of SIRT1-NF-κB axis as therapeutic target to ameliorate inflammation in liver disease. Int. J. Mol. Sci. 2020, 21, 3858. [Google Scholar] [CrossRef] [PubMed]
- Olszowy, M. What is responsible for antioxidant properties of polyphenolic compounds from plants? Plant Physiol. Biochem. 2019, 144, 135–143. [Google Scholar] [CrossRef] [PubMed]
- Alharbi, K.S.; Afzal, O.; Almalki, W.H.; Kazmi, I.; Javed Shaikh, M.A.; Thangavelu, L.; Gulati, M.; Singh, S.K.; Jha, N.K.; Gupta, P.K.; et al. Nuclear factor-kappa B (NF-κB) inhibition as a therapeutic target for plant nutraceuticals in mitigating inflammatory lung diseases. Chem. Biol. Interact. 2022, 354, 109842. [Google Scholar] [CrossRef] [PubMed]
- Joo, S.Y.; Song, Y.A.; Park, Y.L.; Myung, E.; Chung, C.Y.; Park, K.J.; Cho, S.B.; Lee, W.S.; Kim, H.S.; Rew, J.S.; et al. Epigallocatechin-3-gallate inhibits LPS-induced NF-κB and MAPK signaling pathways in bone marrow-derived macrophages. Gut Liver 2012, 6, 188–196. [Google Scholar] [CrossRef] [Green Version]
- Bellezza, I.; Giambanco, I.; Minelli, A.; Donato, R. Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim. Biophys. Acta Mol. Cell Res. 2018, 1865, 721–733. [Google Scholar] [CrossRef] [PubMed]
- Song, M.Y.; Lee, D.Y.; Chun, K.S.; Kim, E.H. The role of NRF2/KEAP1 signaling pathway in cancer metabolism. Int. J. Mol. Sci. 2021, 22, 4376. [Google Scholar] [CrossRef]
- Dempke, W.C.M.; Reck, M. KEAP1/NRF2 (NFE2L2) mutations in NSCLC-Fuel for a superresistant phenotype? Lung Cancer 2021, 159, 10–17. [Google Scholar] [CrossRef]
- Rojo, A.I.; Innamorato, N.G.; Martín-Moreno, A.M.; De Ceballos, M.L.; Yamamoto, M.; Cuadrado, A. Nrf2 regulates microglial dynamics and neuroinflammation in experimental Parkinson’s disease. Glia 2010, 58, 588–598. [Google Scholar] [CrossRef]
- Cordaro, M.; D’Amico, R.; Morabito, R.; Fusco, R.; Siracusa, R.; Peritore, A.F.; Impellizzeri, D.; Genovese, T.; Crupi, R.; Gugliandolo, E.; et al. Physiological and biochemical changes in NRF2 pathway in aged animals subjected to brain injury. Cell. Physiol. Biochem. 2021, 55, 160–179. [Google Scholar]
- Cheng, C.Y.; Barro, L.; Tsai, S.T.; Feng, T.W.; Wu, X.Y.; Chao, C.W.; Yu, R.S.; Chin, T.Y.; Hsieh, M.F. Epigallocatechin-3-gallate-loaded liposomes favor anti-inflammation of microglia cells and promote neuroprotection. Int. J. Mol. Sci. 2021, 22, 3037. [Google Scholar] [CrossRef]
- Scapagnini, G.; Vasto, S.; Abraham, N.G.; Caruso, C.; Zella, D.; Fabio, G. Modulation of Nrf2/ARE pathway by food polyphenols: A nutritional neuroprotective strategy for cognitive and neurodegenerative disorders. Mol. Neurobiol. 2011, 44, 192–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Song, D.; Ge, J.; Wang, Y.; Yan, Q.; Wu, C.; Yu, H.; Yang, M.; Yang, H.; Zou, J. Tea polyphenol attenuates oxidative stress-induced degeneration of intervertebral discs by regulating the Keap1/Nrf2/ARE pathway. Oxidative Med. Cell. Longev. 2021, 2021, 6684147. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Yang, T.; Leak, R.K.; Chen, J.; Zhang, F. Preventive and protective roles of dietary Nrf2 activators against central nervous system diseases. CNS Neurol. Disord. Drug Targets 2017, 16, 326–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, X.; Shi, X.; Liu, Q.; Li, X. Tea polyphenols alleviates acetochlor-induced apoptosis and necroptosis via ROS/MAPK/NF-κB signaling in Ctenopharyngodon idellus kidney cells. Aquat. Toxicol. 2022, 246, 106153. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Gong, X.; Wang, J.; Wang, Y.; Zhang, Y.; Li, T.; Yan, J.; Zhou, M.; Zhang, B. Investigation of nickel sulfate-induced cytotoxicity and underlying toxicological mechanisms in human umbilical vein endothelial cells through oxidative stress, inflammation, apoptosis, and MAPK signaling pathways. Environ. Toxicol. 2022, 37, 2058–2071. [Google Scholar] [CrossRef]
- Zhao, M.; Howard, E.W.; Parris, A.B.; Guo, Z.; Zhao, Q.; Yang, X. Alcohol promotes migration and invasion of triple-negative breast cancer cells through activation of p38 MAPK and JNK. Mol. Carcinog. 2017, 56, 849–862. [Google Scholar] [CrossRef]
- Cicenas, J.; Zalyte, E.; Rimkus, A.; Dapkus, D.; Noreika, R.; Urbonavicius, S. JNK, p38, ERK, and SGK1 inhibitors in cancer. Cancers 2017, 10, 1. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Mei, X.; Tu, Y.Y. Effects of tea polyphenols and their polymers on MAPK signaling pathways in cancer research. Mini Rev. Med. Chem. 2012, 12, 120–126. [Google Scholar] [CrossRef]
- He, Y.; Yang, Z.; Pi, J.; Cai, T.; Xia, Y.; Cao, X.; Liu, J. EGCG attenuates the neurotoxicity of methylglyoxal via regulating MAPK and the downstream signaling pathways and inhibiting advanced glycation end products formation. Food Chem. 2022, 384, 132358. [Google Scholar] [CrossRef]
- Du, Y.; Gao, Y.; Zeng, B.; Fan, X.; Yang, D.; Yang, M. Effects of anti-aging interventions on intestinal microbiota. Gut Microbes 2021, 13, 1994835. [Google Scholar] [CrossRef]
- Maynard, C.; Weinkove, D. The gut microbiota and ageing. Subcell. Biochem. 2018, 90, 351–371. [Google Scholar] [PubMed]
- Cserép, C.; Pósfai, B.; Dénes, Á. Shaping neuronal fate: Functional heterogeneity of direct microglia-neuron interactions. Neuron 2021, 109, 222–240. [Google Scholar] [CrossRef] [PubMed]
- Zhou, R.; Qian, S.; Cho, W.C.S.; Zhou, J.; Jin, C.; Zhong, Y.; Wang, J.; Zhang, X.; Xu, Z.; Tian, M.; et al. Microbiota-microglia connections in age-related cognition decline. Aging Cell 2022, 21, e13599. [Google Scholar] [CrossRef]
- Schächtle, M.A.; Rosshart, S.P. The microbiota-gut-brain axis in health and disease and its implications for translational research. Front. Cell. Neurosci. 2021, 15, 698172. [Google Scholar] [CrossRef]
- Moor, K.; Diard, M.; Sellin, M.E.; Felmy, B.; Wotzka, S.Y.; Toska, A.; Bakkeren, E.; Arnoldini, M.; Bansept, F.; Co, A.D.; et al. High-avidity IgA protects the intestine by enchaining growing bacteria. Nature 2017, 544, 498–502. [Google Scholar] [CrossRef] [PubMed]
- Hepworth, M.R.; Greenhalgh, A.D.; Cook, P.C. B cells on the brain: Meningeal IgA and a novel gut-brain firewall. Immunol. Cell Biol. 2021, 99, 17–20. [Google Scholar] [CrossRef] [PubMed]
- Fitzpatrick, Z.; Frazer, G.; Ferro, A.; Clare, S.; Bouladoux, N.; Ferdinand, J.; Tuong, Z.K.; Negro-Demontel, M.L.; Kumar, N.; Suchanek, O.; et al. Gut-educated IgA plasma cells defend the meningeal venous sinuses. Nature 2020, 587, 472–476. [Google Scholar] [CrossRef] [PubMed]
- Gabanyi, I.; Lepousez, G.; Wheeler, R.; Vieites-Prado, A.; Nissant, A.; Wagner, S.; Moigneu, C.; Dulauroy, S.; Hicham, S.; Polomack, B.; et al. Bacterial sensing via neuronal Nod2 regulates appetite and body temperature. Science 2022, 376, eabj3986. [Google Scholar] [CrossRef] [PubMed]
- Cryan, J.F.; Mazmanian, S.K. Microbiota-brain axis: Context and causality. Science 2022, 376, 938–939. [Google Scholar] [CrossRef] [PubMed]
- Parker, A.; Romano, S.; Ansorge, R.; Aboelnour, A.; Le Gall, G.; Savva, G.M.; Pontifex, M.G.; Telatin, A.; Baker, D.; Jones, E.; et al. Fecal microbiota transfer between young and aged mice reverses hallmarks of the aging gut, eye, and brain. Microbiome 2022, 10, 68. [Google Scholar] [CrossRef]
- Lee, Y.; Kim, Y.K. Understanding the connection between the gut-brain axis and stress/anxiety disorders. Curr. Psychiatry Rep. 2021, 23, 22. [Google Scholar] [CrossRef]
- Hong, M.; Cheng, L.; Liu, Y.; Wu, Z.; Zhang, P.; Zhang, X. Mechanisms underlying the interaction between chronic neurological disorders and microbial metabolites via tea polyphenols therapeutics. Front. Microbiol. 2022, 13, 823902. [Google Scholar] [CrossRef]
- Boehme, M.; Guzzetta, K.E.; Bastiaanssen, T.F.S.; van de Wouw, M.; Moloney, G.M.; Gual-Grau, A.; Spichak, S.; Olavarría-Ramírez, L.; Fitzgerald, P.; Morillas, E.; et al. Microbiota from young mice counteracts selective age-associated behavioral deficits. Nat. Aging 2021, 1, 666–676. [Google Scholar] [CrossRef]
- Zhou, N.; Gu, X.; Zhuang, T.; Xu, Y.; Yang, L.; Zhou, M. Gut microbiota: A pivotal hub for polyphenols as antidepressants. J. Agric. Food Chem. 2020, 68, 6007–6020. [Google Scholar] [CrossRef]
- Pervin, M.; Unno, K.; Takagaki, A.; Isemura, M.; Nakamura, Y. Function of green tea catechins in the brain: Epigallocatechin gallate and its metabolites. Int. J. Mol. Sci. 2019, 20, 3630. [Google Scholar] [CrossRef] [Green Version]
- Dey, P. Gut microbiota in phytopharmacology: A comprehensive overview of concepts, reciprocal interactions, biotransformations and mode of actions. Pharmacol. Res. 2019, 147, 104367. [Google Scholar] [CrossRef]
- Davinelli, S.; Scapagnini, G. Interactions between dietary polyphenols and aging gut microbiota: A review. BioFactors 2022, 48, 274–284. [Google Scholar] [CrossRef]
- Santangelo, R.; Silvestrini, A.; Mancuso, C. Ginsenosides, catechins, quercetin and gut microbiota: Current evidence of challenging interactions. Food Chem. Toxicol. 2019, 123, 42–49. [Google Scholar] [CrossRef]
- Carregosa, D.; Carecho, R.; Figueira, I.; Santos, C.N. Low-molecular weight metabolites from polyphenols as effectors for attenuating neuroinflammation. J. Agric. Food Chem. 2020, 68, 1790–1807. [Google Scholar] [CrossRef] [Green Version]
- Mithul Aravind, S.; Wichienchot, S.; Tsao, R.; Ramakrishnan, S.; Chakkaravarthi, S. Role of dietary polyphenols on gut microbiota, their metabolites and health benefits. Food Res. Int. 2021, 142, 110189. [Google Scholar] [CrossRef]
- Zhao, Y.; Jia, M.; Chen, W.; Liu, Z. The neuroprotective effects of intermittent fasting on brain aging and neurodegenerative diseases via regulating mitochondrial function. Free Radic. Biol. Med. 2022, 182, 206–218. [Google Scholar] [CrossRef]
- DeJong, E.N.; Surette, M.G.; Bowdish, D.M.E. The gut microbiota and unhealthy aging: Disentangling cause from consequence. Cell Host Microbe 2020, 28, 180–189. [Google Scholar] [CrossRef]
- Bazinet, R.P.; Layé, S. Polyunsaturated fatty acids and their metabolites in brain function and disease. Nat. Rev. Neurosci. 2014, 15, 771–785. [Google Scholar] [CrossRef]
- Russo, G.L.; Spagnuolo, C.; Russo, M.; Tedesco, I.; Moccia, S.; Cervellera, C. Mechanisms of aging and potential role of selected polyphenols in extending healthspan. Biochem. Pharmacol. 2020, 173, 113719. [Google Scholar] [CrossRef]
- Song, C.; Zhang, Y.; Cheng, L.; Shi, M.; Li, X.; Zhang, L.; Zhao, H. Tea polyphenols ameliorates memory decline in aging model rats by inhibiting brain TLR4/NF-κB inflammatory signaling pathway caused by intestinal flora dysbiosis. Exp. Gerontol. 2021, 153, 111476. [Google Scholar] [CrossRef]
- Lagha, A.B.; Grenier, D. Tea polyphenols protect gingival keratinocytes against TNF-α-induced tight junction barrier dysfunction and attenuate the inflammatory response of monocytes/macrophages. Cytokine 2019, 115, 64–75. [Google Scholar] [CrossRef]
- Shal, B.; Ding, W.; Ali, H.; Kim, Y.S.; Khan, S. Anti-neuroinflammatory potential of natural products in attenuation of Alzheimer’s disease. Front. Pharmacol. 2018, 9, 548. [Google Scholar] [CrossRef]
- Branca, J.J.V.; Gulisano, M.; Nicoletti, C. Intestinal epithelial barrier functions in ageing. Ageing Res. Rev. 2019, 54, 100938. [Google Scholar] [CrossRef]
- Mao, L.; Hochstetter, D.; Yao, L.; Zhao, Y.; Zhou, J.; Wang, Y.; Xu, P. Green tea polyphenol (-)-epigallocatechin gallate (EGCG) attenuates neuroinflammation in palmitic acid-stimulated BV-2 microglia and high-fat diet-induced obese mice. Int. J. Mol. Sci. 2019, 20, 5081. [Google Scholar] [CrossRef] [Green Version]
- Zhou, C.; Wu, Z.; Du, W.; Que, H.; Wang, Y.; Ouyang, Q.; Jian, F.; Yuan, W.; Zhao, Y.; Tian, R.; et al. Recycling of autophagosomal components from autolysosomes by the recycler complex. Nat. Cell Biol. 2022, 24, 497–512. [Google Scholar] [CrossRef]
- Zhou, H.; Chen, Y.; Huang, S.W.; Hu, P.F.; Tang, L.J. Regulation of autophagy by tea polyphenols in diabetic cardiomyopathy. J. Zhejiang Univ. Sci. B. 2018, 19, 333–341. [Google Scholar] [CrossRef] [PubMed]
- Xie, X.; Yi, W.; Zhang, P.; Wu, N.; Yan, Q.; Yang, H.; Tian, C.; Xiang, S.; Du, M.; Getachew Assefa, E.; et al. Green tea polyphenols, mimicking the effects of dietary restriction, ameliorate high-fat diet-induced kidney injury via regulating autophagy flux. Nutrients 2017, 9, 497. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yuan, H.; Li, Y.; Ling, F.; Guan, Y.; Zhang, D.; Zhu, Q.; Liu, J.; Wu, Y.; Niu, Y. The phytochemical epigallocatechin gallate prolongs the lifespan by improving lipid metabolism, reducing inflammation and oxidative stress in high-fat diet-fed obese rats. Aging Cell 2020, 19, e13199. [Google Scholar] [CrossRef] [PubMed]
- Mi, Y.; Qi, G.; Fan, R.; Qiao, Q.; Sun, Y.; Gao, Y.; Liu, X. EGCG ameliorates high-fat- and high-fructose-induced cognitive defects by regulating the IRS/AKT and ERK/CREB/BDNF signaling pathways in the CNS. FASEB J. 2017, 31, 4998–5011. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Shi, M.; Song, C.; Cheng, L.; Li, X.; Yang, Q.; Zhang, Y.; Dong, R.; Kou, J.; Lv, C.; et al. Tea polyphenols improve the memory in aging ovariectomized rats by regulating brain glucose metabolism in vivo and in vitro. J. Funct. Foods 2021, 87, 104856. [Google Scholar] [CrossRef]
- Rathor, L.; Pant, A.; Awasthi, H.; Mani, D.; Pandey, R. An antidiabetic polyherbal phytomedicine confers stress resistance and extends lifespan in Caenorhabditis elegans. Biogerontology 2017, 18, 131–147. [Google Scholar] [CrossRef]
- Kian, K.; Khalatbary, A.R.; Ahmadvand, H.; Karimpour Malekshah, A.; Shams, Z. Neuroprotective effects of (-)-epigallocatechin-3-gallate (EGCG) against peripheral nerve transection-induced apoptosis. Nutr. Neurosci. 2019, 22, 578–586. [Google Scholar] [CrossRef]
- Yildirim, A.E.; Dalgic, A.; Divanlioglu, D.; Akdag, R.; Cetinalp, N.E.; Alagoz, F.; Helvacioglu, F.; Take, G.; Guvenc, Y.; Koksal, I.; et al. Biochemical and histopathological effects of catechin on experimental peripheral nerve injuries. Turk. Neurosurg. 2015, 25, 453–460. [Google Scholar] [CrossRef]
- Wang, S.; Qiu, Y.; Gan, R.Y.; Zhu, F. Chemical constituents and biological properties of Pu-erh tea. Food Res. Int. 2022, 154, 110899. [Google Scholar] [CrossRef]
Research Environment | Model Used | Neuroprotective Effects | Reference |
---|---|---|---|
In vivo | Sprague–Dawley male rats | Inhibition of PI3K/AKT/mTOR pathway and upregulate autophagy process | [15] |
C57BL/6J | Enhance memory and extend lifespan | [59] | |
APPsw transgenic mice | Aβ and plaques levels decreased | [16] | |
Male Long–Evans rats | Inhibits MAO-B enzyme activity in rat brain | [78] | |
ICR mice | Enhance spatial memory, inhibit neuronal damage in the brain | [79] | |
Human | Improve brain compensatory response and cognitive reserve ability | [70] | |
In vitro | U118MG cells | Increase BDNF gene expression significantly | [2] |
Human neuroblastoma SH-SY5Y cells | Inhibit neuronal cell death caused by the neurotoxins 6-hydroxydopamine (6-OHDA) and 1-methyl-4 phenylpyridinium (MPP+) | [9] | |
PC12 cells | Inhibit the aggregation of α-synuclein | [17] | |
PC12 cells | Reduce cell death and promote neurite outgrowth | [80] | |
BV2 microglial cells | Inhibit LPS-induced inflammation, reduce TNF-α secretion, iNOS and COX-2 protein expression | [79,81] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Hong, M.; Yu, J.; Wang, X.; Liu, Y.; Zhan, S.; Wu, Z.; Zhang, X. Tea Polyphenols as Prospective Natural Attenuators of Brain Aging. Nutrients 2022, 14, 3012. https://doi.org/10.3390/nu14153012
Hong M, Yu J, Wang X, Liu Y, Zhan S, Wu Z, Zhang X. Tea Polyphenols as Prospective Natural Attenuators of Brain Aging. Nutrients. 2022; 14(15):3012. https://doi.org/10.3390/nu14153012
Chicago/Turabian StyleHong, Mengyu, Jing Yu, Xuanpeng Wang, Yanan Liu, Shengnan Zhan, Zufang Wu, and Xin Zhang. 2022. "Tea Polyphenols as Prospective Natural Attenuators of Brain Aging" Nutrients 14, no. 15: 3012. https://doi.org/10.3390/nu14153012
APA StyleHong, M., Yu, J., Wang, X., Liu, Y., Zhan, S., Wu, Z., & Zhang, X. (2022). Tea Polyphenols as Prospective Natural Attenuators of Brain Aging. Nutrients, 14(15), 3012. https://doi.org/10.3390/nu14153012