Neuro-Nutraceutical Polyphenols: How Far Are We?
Abstract
:1. Introduction
2. Chemistry and Sources of Polyphenols, a Sustainable Strategy to Support Central Nervous System (CNS) Functions
3. Polyphenols and Microbiota–Gut–Brain Axis
4. Polyphenols: Antioxidants for Brain Health
5. Polyphenols vs. Neurodegenerative Disease
5.1. Polyphenols Inhibit Amyloid Genesis
5.2. Polyphenols Inhibit tau Hyperphosphorylation
5.3. Polyphenols and Metal Dyshomeostasis
5.4. Polyphenols and Neuroinflammation
5.5. Polyphenol Based Formulations for Enhancing Their Neuroprotective goals
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Herculano-Houzel, S. The glia/neuron ratio: How it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia 2014, 62, 1377–1391. [Google Scholar] [CrossRef] [PubMed]
- Gitler, A.D.; Dhillon, P.; Shorter, J. Neurodegenerative Disease: Models, Mechanisms, and a New Hope; The Company of Biologists Ltd.: Cambridge, UK, 2017; pp. 499–502. [Google Scholar]
- Flanagan, E.; Müller, M.; Hornberger, M.; Vauzour, D. Impact of flavonoids on cellular and molecular mechanisms underlying age-related cognitive decline and neurodegeneration. Curr. Nutr. Rep. 2018, 7, 49–57. [Google Scholar] [CrossRef] [Green Version]
- Cao, Q.; Tan, C.-C.; Xu, W.; Hu, H.; Cao, X.-P.; Dong, Q.; Tan, L.; Yu, J.-T. The prevalence of dementia: A systematic review and meta-analysis. J. Alzheimer’s Dis. 2020, 73, 1157–1166. [Google Scholar] [CrossRef] [PubMed]
- Durães, F.; Pinto, M.; Sousa, E. Old drugs as new treatments for neurodegenerative diseases. Pharmaceuticals 2018, 11, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Piccolella, S.; Pacifico, S. Plant-derived polyphenols: A chemopreventive and chemoprotectant worth-exploring resource in toxicology. In Advances in Molecular Toxicology; Elsevier: Amsterdam, The Netherlands, 2015; pp. 161–214. [Google Scholar]
- Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and human health: The role of bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, F.H.; Li, Y. Mechanisms of cancer chemoprevention by soy isoflavone genistein. Cancer Metastasis Rev. 2002, 21, 265–280. [Google Scholar] [CrossRef]
- Bukhari, S.N.A. Dietary Polyphenols as Therapeutic Intervention for Alzheimer’s Disease: A Mechanistic Insight. Antioxidants 2022, 11, 554. [Google Scholar] [CrossRef]
- Formato, M.; Cimmino, G.; Brahmi-Chendouh, N.; Piccolella, S.; Pacifico, S. Polyphenols for Livestock Feed: Sustainable Perspectives for Animal Husbandry? Molecules 2022, 27, 7752. [Google Scholar] [CrossRef]
- Tresserra-Rimbau, A.; Lamuela-Raventos, R.M.; Moreno, J.J. Polyphenols, food and pharma. Current knowledge and directions for future research. Biochem. Pharmacol. 2018, 156, 186–195. [Google Scholar] [CrossRef]
- Sharma, A.; Shahzad, B.; Rehman, A.; Bhardwaj, R.; Landi, M.; Zheng, B. Response of phenylpropanoid pathway and the role of polyphenols in plants under abiotic stress. Molecules 2019, 24, 2452. [Google Scholar] [CrossRef] [Green Version]
- Šamec, D.; Karalija, E.; Šola, I.; Vujčić Bok, V.; Salopek-Sondi, B. The role of polyphenols in abiotic stress response: The influence of molecular structure. Plants 2021, 10, 118. [Google Scholar] [CrossRef] [PubMed]
- Piccolella, S.; Crescente, G.; Candela, L.; Pacifico, S. Nutraceutical polyphenols: New analytical challenges and opportunities. J. Pharm. Biomed. Anal. 2019, 175, 112774. [Google Scholar] [CrossRef] [PubMed]
- Williamson, G. The role of polyphenols in modern nutrition. Nutr. Bull. 2017, 42, 226–235. [Google Scholar] [CrossRef] [Green Version]
- Santos-Buelga, C. Polyphenols and Human Beings: From Epidemiology to Molecular Targets. Molecules 2021, 26, 4218. [Google Scholar] [CrossRef]
- Scalbert, A.; Johnson, I.; Saltmarsh, M. Polyphenols: Antioxidants and beyond. Am. J. Clin. Nutr. 2005, 81, 215S–217S. [Google Scholar] [CrossRef] [Green Version]
- Queen, B.L.; Tollefsbol, T.O. Polyphenols and aging. Curr. Aging Sci. 2010, 3, 34–42. [Google Scholar] [CrossRef] [PubMed]
- Finicelli, M.; Squillaro, T.; Di Cristo, F.; Di Salle, A.; Melone, M.A.B.; Galderisi, U.; Peluso, G. Metabolic syndrome, Mediterranean diet, and polyphenols: Evidence and perspectives. J. Cell. Physiol. 2019, 234, 5807–5826. [Google Scholar] [CrossRef]
- Ditano-Vázquez, P.; Torres-Peña, J.D.; Galeano-Valle, F.; Pérez-Caballero, A.I.; Demelo-Rodríguez, P.; Lopez-Miranda, J.; Katsiki, N.; Delgado-Lista, J.; Alvarez-Sala-Walther, L.A. The fluid aspect of the Mediterranean diet in the prevention and management of cardiovascular disease and diabetes: The role of polyphenol content in moderate consumption of wine and olive oil. Nutrients 2019, 11, 2833. [Google Scholar] [CrossRef] [Green Version]
- Román, G.C.; Jackson, R.E.; Gadhia, R.; Román, A.N.; Reis, J. Mediterranean diet: The role of long-chain ω-3 fatty acids in fish; polyphenols in fruits, vegetables, cereals, coffee, tea, cacao and wine; probiotics and vitamins in prevention of stroke, age-related cognitive decline, and Alzheimer disease. Rev. Neurol. 2019, 175, 724–741. [Google Scholar] [CrossRef]
- Perrone, L.; Sampaolo, S.; Melone, M.A.B. Bioactive phenolic compounds in the modulation of central and peripheral nervous system cancers: Facts and misdeeds. Cancers 2020, 12, 454. [Google Scholar] [CrossRef] [Green Version]
- Kwon, K.J.; Kim, H.-J.; Shin, C.Y.; Han, S.-H. Melatonin potentiates the neuroprotective properties of resveratrol against beta-amyloid-induced neurodegeneration by modulating AMP-activated protein kinase pathways. J. Clin. Neurol. 2010, 6, 127–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bastianetto, S.; Krantic, S.; Chabot, J.G.; Quirion, R. Possible involvement of programmed cell death pathways in the neuroprotective action of polyphenols. Curr. Alzheimer Res. 2011, 8, 445–451. [Google Scholar] [CrossRef] [PubMed]
- Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The role of polyphenols in human health and food systems: A mini-review. Front. Nutr. 2018, 5, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vauzour, D. Dietary polyphenols as modulators of brain functions: Biological actions and molecular mechanisms underpinning their beneficial effects. Oxid. Med. Cell. Longev. 2012, 2012, 914273. [Google Scholar] [CrossRef] [Green Version]
- Reddy, V.P.; Aryal, P.; Robinson, S.; Rafiu, R.; Obrenovich, M.; Perry, G. Polyphenols in Alzheimer’s Disease and in the Gut–Brain Axis. Microorganisms 2020, 8, 199. [Google Scholar] [CrossRef] [Green Version]
- Carmody, R.N.; Turnbaugh, P.J. Host-microbial interactions in the metabolism of therapeutic and diet-derived xenobiotics. J. Clin. Investig. 2014, 124, 4173–4181. [Google Scholar] [CrossRef] [Green Version]
- Cortés-Martín, A.; Selma, M.V.; Tomás-Barberán, F.A.; González-Sarrías, A.; Espín, J.C. Where to look into the puzzle of polyphenols and health? The postbiotics and gut microbiota associated with human metabotypes. Mol. Nutr. Food Res. 2020, 64, 1900952. [Google Scholar] [CrossRef]
- Wang, M.; Yu, F.; Zhang, Y.; Chang, W.; Zhou, M. The effects and mechanisms of flavonoids on cancer prevention and therapy: Focus on gut microbiota. Int. J. Biol. Sci. 2022, 18, 1451. [Google Scholar] [CrossRef]
- Wang, Y.; Liu, X.-J.; Chen, J.-B.; Cao, J.-P.; Li, X.; Sun, C.-D. Citrus flavonoids and their antioxidant evaluation. Crit. Rev. Food Sci. Nutr. 2022, 62, 3833–3854. [Google Scholar] [CrossRef]
- Kaakoush, N.O.; Morris, M.J. More flavor for flavonoid-based interventions? Trends Mol. Med. 2017, 23, 293–295. [Google Scholar] [CrossRef]
- Amaretti, A.; Raimondi, S.; Leonardi, A.; Quartieri, A.; Rossi, M. Hydrolysis of the rutinose-conjugates flavonoids rutin and hesperidin by the gut microbiota and bifidobacteria. Nutrients 2015, 7, 2788–2800. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wiese, S.; Esatbeyoglu, T.; Winterhalter, P.; Kruse, H.P.; Winkler, S.; Bub, A.; Kulling, S.E. Comparative biokinetics and metabolism of pure monomeric, dimeric, and polymeric flavan-3-ols: A randomized cross-over study in humans. Mol. Nutr. Food Res. 2015, 59, 610–621. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Keranmu, A.; Pan, L.-B.; Fu, J.; Han, P.; Yu, H.; Zhang, Z.-W.; Xu, H.; Yang, X.-Y.; Hu, J.-C.; Zhang, H.-J. Biotransformation of Liquiritigenin into Characteristic Metabolites by the Gut Microbiota. Molecules 2022, 27, 3057. [Google Scholar] [CrossRef] [PubMed]
- Ramalingam, M.; Kim, H.; Lee, Y.; Lee, Y.-I. Phytochemical and pharmacological role of liquiritigenin and isoliquiritigenin from radix glycyrrhizae in human health and disease models. Front. Aging Neurosci. 2018, 10, 348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gowd, V.; Karim, N.; Shishir, M.R.I.; Xie, L.; Chen, W. Dietary polyphenols to combat the metabolic diseases via altering gut microbiota. Trends Food Sci. Technol. 2019, 93, 81–93. [Google Scholar] [CrossRef]
- Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly) phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef]
- Shandilya, S.; Kumar, S.; Jha, N.K.; Kesari, K.K.; Ruokolainen, J. Interplay of gut microbiota and oxidative stress: Perspective on neurodegeneration and neuroprotection. J. Adv. Res. 2022, 38, 223–244. [Google Scholar] [CrossRef]
- Rastmanesh, R. High polyphenol, low probiotic diet for weight loss because of intestinal microbiota interaction. Chem.-Biol. Interact. 2011, 189, 1–8. [Google Scholar] [CrossRef]
- Dueñas, M.; Muñoz-González, I.; Cueva, C.; Jiménez-Girón, A.; Sánchez-Patán, F.; Santos-Buelga, C.; Moreno-Arribas, M.; Bartolomé, B. A survey of modulation of gut microbiota by dietary polyphenols. BioMed Res. Int. 2015, 2015, 850902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dong, R.; Liu, S.; Xie, J.; Chen, Y.; Zheng, Y.; Zhang, X.; Zhao, E.; Wang, Z.; Xu, H.; Yu, Q. The recovery, catabolism and potential bioactivity of polyphenols from carrot subjected to in vitro simulated digestion and colonic fermentation. Food Res. Int. 2021, 143, 110263. [Google Scholar] [CrossRef] [PubMed]
- Sun, H.; Chen, Y.; Cheng, M.; Zhang, X.; Zheng, X.; Zhang, Z. The modulatory effect of polyphenols from green tea, oolong tea and black tea on human intestinal microbiota in vitro. J. Food Sci. Technol. 2018, 55, 399–407. [Google Scholar] [CrossRef] [PubMed]
- Jia, M.; Li, D.; Wang, R.; Wang, A.; Strappe, P.; Wu, Q.; Shang, W.; Wang, X.; Zhuang, M.; Blanchard, C. Gut microbiota derived structural changes of phenolic compounds from colored rice and its corresponding fermentation property. Food Funct. 2022, 13, 10759–10768. [Google Scholar] [CrossRef]
- Rosario, D.; Bidkhori, G.; Lee, S.; Bedarf, J.; Hildebrand, F.; Le Chatelier, E.; Uhlen, M.; Ehrlich, S.D.; Proctor, G.; Wüllner, U. Systematic analysis of gut microbiome reveals the role of bacterial folate and homocysteine metabolism in Parkinson’s disease. Cell Rep. 2021, 34, 108807. [Google Scholar] [CrossRef]
- Cao, Y.; Ren, G.; Zhang, Y.; Qin, H.; An, X.; Long, Y.; Chen, J.; Yang, L. A new way for punicalagin to alleviate insulin resistance: Regulating gut microbiota and autophagy. Food Nutr. Res. 2021, 65. [Google Scholar] [CrossRef]
- Huang, F.; Zhao, R.; Xia, M.; Shen, G.X. Impact of cyanidin-3-glucoside on gut microbiota and relationship with metabolism and inflammation in high fat-high sucrose diet-induced insulin resistant mice. Microorganisms 2020, 8, 1238. [Google Scholar] [CrossRef]
- Sukprasansap, M.; Chanvorachote, P.; Tencomnao, T. Cyanidin-3-glucoside activates Nrf2-antioxidant response element and protects against glutamate-induced oxidative and endoplasmic reticulum stress in HT22 hippocampal neuronal cells. BMC Complement. Med. Ther. 2020, 20, 46. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Song, X.; Hu, X.; Chen, F.; Ma, C. Health benefits of proanthocyanidins linking with gastrointestinal modulation: An updated review. Food Chem. 2022, 404, 134596. [Google Scholar] [CrossRef]
- Li, C.; Wang, N.; Zheng, G.; Yang, L. Oral administration of resveratrol-selenium-peptide nanocomposites alleviates Alzheimer’s disease-like pathogenesis by inhibiting Aβ aggregation and regulating gut microbiota. ACS Appl. Mater. Interfaces 2021, 13, 46406–46420. [Google Scholar] [CrossRef]
- Filosa, S.; Di Meo, F.; Crispi, S. Polyphenols-gut microbiota interplay and brain neuromodulation. Neural Regen. Res. 2018, 13, 2055–2059. [Google Scholar] [PubMed]
- Molino, S.; Lerma-Aguilera, A.; Jiménez-Hernández, N.; Rufián Henares, J.Á.; Francino, M.P. Evaluation of the effects of a short supplementation with tannins on the gut microbiota of healthy subjects. Front. Microbiol. 2022, 13, 848611. [Google Scholar] [CrossRef] [PubMed]
- Bains, J.S.; Shaw, C.A. Neurodegenerative disorders in humans: The role of glutathione in oxidative stress-mediated neuronal death. Brain Res. Rev. 1997, 25, 335–358. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Y.; Zheng, J.; Li, Y.; Xu, D.-P.; Li, S.; Chen, Y.-M.; Li, H.-B. Natural polyphenols for prevention and treatment of cancer. Nutrients 2016, 8, 515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Subramaniam, S.R.; Chesselet, M.-F. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease. Prog. Neurobiol. 2013, 106, 17–32. [Google Scholar] [CrossRef] [Green Version]
- Grinan-Ferre, C.; Bellver-Sanchis, A.; Izquierdo, V.; Corpas, R.; Roig-Soriano, J.; Chillón, M.; Andres-Lacueva, C.; Somogyvári, M.; Sőti, C.; Sanfeliu, C. The pleiotropic neuroprotective effects of resveratrol in cognitive decline and Alzheimer’s disease pathology: From antioxidant to epigenetic therapy. Ageing Res. Rev. 2021, 67, 101271. [Google Scholar] [CrossRef]
- Kölliker-Frers, R.; Udovin, L.; Otero-Losada, M.; Kobiec, T.; Herrera, M.I.; Palacios, J.; Razzitte, G.; Capani, F. Neuroinflammation: An integrating overview of reactive-neuroimmune cell interactions in health and disease. Mediat. Inflamm. 2021, 2021, 9999146. [Google Scholar] [CrossRef]
- Wake, H.; Moorhouse, A.J.; Nabekura, J. Functions of microglia in the central nervous system—Beyond the immune response. Neuron Glia Biol. 2011, 7, 47–53. [Google Scholar] [CrossRef]
- Lull, M.E.; Block, M.L. Microglial activation and chronic neurodegeneration. Neurotherapeutics 2010, 7, 354–365. [Google Scholar] [CrossRef] [Green Version]
- Giordano, G.; Costa, L.G. Primary neurons in culture and neuronal cell lines for in vitro neurotoxicological studies. In Vitro Neurotoxicology; Springer: Berlin/Heidelberg, Germany, 2011; pp. 13–27. [Google Scholar]
- Thameem Dheen, S.; Kaur, C.; Ling, E.-A. Microglial activation and its implications in the brain diseases. Curr. Med. Chem. 2007, 14, 1189–1197. [Google Scholar] [CrossRef]
- Fernández, B.; Ferrer, I.; Gil, F.; Hilfiker, S. Biomonitorization of iron accumulation in the substantia nigra from Lewy body disease patients. Toxicol. Rep. 2017, 4, 188–193. [Google Scholar] [CrossRef] [PubMed]
- Sofic, E.; Lange, K.W.; Jellinger, K.; Riederer, P. Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson’s disease. Neurosci. Lett. 1992, 142, 128–130. [Google Scholar] [CrossRef] [PubMed]
- Cooke, M.S.; Evans, M.D.; Dizdaroglu, M.; Lunec, J. Oxidative DNA damage: Mechanisms, mutation, and disease. FASEB J. 2003, 17, 1195–1214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weng, M.; Xie, X.; Liu, C.; Lim, K.L.; Zhang, C.W.; Li, L. The sources of reactive oxygen species and its possible role in the pathogenesis of Parkinson’s disease. Parkinsons Dis. 2018, 2018, 9163040. [Google Scholar] [CrossRef] [Green Version]
- Dorszewska, J.; Kowalska, M.; Prendecki, M.; Piekut, T.; Kozłowska, J.; Kozubski, W. Oxidative stress factors in Parkinson’s disease. Neural Regen. Res. 2021, 16, 1383–1391. [Google Scholar] [CrossRef]
- 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]
- Bhakkiyalakshmi, E.; Dineshkumar, K.; Karthik, S.; Sireesh, D.; Hopper, W.; Paulmurugan, R.; Ramkumar, K.M. Pterostilbene-mediated Nrf2 activation: Mechanistic insights on Keap1: Nrf2 interface. Bioorg. Med. Chem. 2016, 24, 3378–3386. [Google Scholar] [CrossRef]
- Di Meo, F.; Valentino, A.; Petillo, O.; Peluso, G.; Filosa, S.; Crispi, S. Bioactive polyphenols and neuromodulation: Molecular mechanisms in neurodegeneration. Int. J. Mol. Sci. 2020, 21, 2564. [Google Scholar] [CrossRef] [Green Version]
- Liu, C.; Chan, C.B.; Ye, K. 7,8-dihydroxyflavone, a small molecular TrkB agonist, is useful for treating various BDNF-implicated human disorders. Transl. Neurodegener. 2016, 5, 2. [Google Scholar] [CrossRef] [Green Version]
- Błaszczyk, J.W. Parkinson’s disease and neurodegeneration: GABA-collapse hypothesis. Front. Neurosci. 2016, 10, 269. [Google Scholar] [CrossRef] [Green Version]
- Hallacli, E.; Kayatekin, C.; Nazeen, S.; Wang, X.H.; Sheinkopf, Z.; Sathyakumar, S.; Sarkar, S.; Jiang, X.; Dong, X.; Di Maio, R. The Parkinson’s disease protein alpha-synuclein is a modulator of processing bodies and mRNA stability. Cell 2022, 185, 2035–2056. [Google Scholar] [CrossRef]
- Agnello, L.; Ciaccio, M. Neurodegenerative Diseases: From Molecular Basis to Therapy. Int. J. Mol. Sci. 2022, 23, 12854. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.U.; Akram, M.; Daniyal, M.; Zainab, R. Awareness and current knowledge of Parkinson’s disease: A neurodegenerative disorder. Int. J. Neurosci. 2019, 129, 55–93. [Google Scholar] [CrossRef] [PubMed]
- Moore, D. Neurocognitive Disorders. In Psychiatry; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2015; pp. 1647–1705. [Google Scholar]
- Wood, W.G.; Li, L.; Müller, W.E.; Eckert, G.P. Cholesterol as a causative factor in Alzheimer’s disease: A debatable hypothesis. J. Neurochem. 2014, 129, 559–572. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de la Torre, J. The vascular hypothesis of Alzheimer’s disease: A key to preclinical prediction of dementia using neuroimaging. J. Alzheimer’s Dis. 2018, 63, 35–52. [Google Scholar] [CrossRef] [PubMed]
- Scheffer, S.; Hermkens, D.M.A.; Van Der Weerd, L.; De Vries, H.E.; Daemen, M.J.A.P. Vascular hypothesis of Alzheimer disease: Topical review of mouse models. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 1265–1283. [Google Scholar] [CrossRef] [PubMed]
- Leng, F.; Edison, P. Neuroinflammation and microglial activation in Alzheimer disease: Where do we go from here? Nat. Rev. Neurol. 2021, 17, 157–172. [Google Scholar] [CrossRef] [PubMed]
- Zarotsky, V.; Sramek, J.J.; Cutler, N.R. Galantamine hydrobromide: An agent for Alzheimer’s disease. Am. J. Health-Syst. Pharm. 2003, 60, 446–452. [Google Scholar] [CrossRef]
- Berkov, S.; Bastida, J.; Sidjimova, B.; Viladomat, F.; Codina, C. Alkaloid diversity in Galanthus elwesii and Galanthus nivalis. Chem. Biodivers. 2011, 8, 115–130. [Google Scholar] [CrossRef]
- Moss, D.E.; Perez, R.G. Anti-Neurodegenerative Benefits of Acetylcholinesterase Inhibitors in Alzheimer’s Disease: Nexus of Cholinergic and Nerve Growth Factor Dysfunction. Curr. Alzheimer Res. 2021, 18, 1010–1022. [Google Scholar] [CrossRef]
- Wu, M.; Liu, M.; Wang, F.; Cai, J.; Luo, Q.; Li, S.; Zhu, J.; Tang, Z.; Fang, Z.; Wang, C. The inhibition mechanism of polyphenols from Phyllanthus emblica Linn. fruit on acetylcholinesterase: A interaction, kinetic, spectroscopic, and molecular simulation study. Food Res. Int. 2022, 158, 111497. [Google Scholar] [CrossRef] [PubMed]
- Nizzari, M.; Barbieri, F.; Gentile, M.T.; Passarella, D.; Caorsi, C.; Diaspro, A.; Taglialatela, M.; Pagano, A.; Colucci-D’Amato, L.; Florio, T. Amyloid-β protein precursor regulates phosphorylation and cellular compartmentalization of microtubule associated protein tau. J. Alzheimer’s Dis. 2012, 29, 211–227. [Google Scholar] [CrossRef] [PubMed]
- Haque, A.M.; Hashimoto, M.; Katakura, M.; Tanabe, Y.; Hara, Y.; Shido, O. Long-term administration of green tea catechins improves spatial cognition learning ability in rats. J. Nutr. 2006, 136, 1043–1047. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaur, T.; Pathak, C.M.; Pandhi, P.; Khanduja, K.L. Effects of green tea extract on learning, memory, behavior and acetylcholinesterase activity in young and old male rats. Brain Cogn. 2008, 67, 25–30. [Google Scholar] [CrossRef] [PubMed]
- Figueira, I.; Garcia, G.; Pimpão, R.C.; Terrasso, A.P.; Costa, I.; Almeida, A.F.; Tavares, L.; Pais, T.F.; Pinto, P.; Ventura, M.R. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci. Rep. 2017, 7, 11456. [Google Scholar] [CrossRef] [Green Version]
- Ono, K.; Li, L.; Takamura, Y.; Yoshiike, Y.; Zhu, L.; Han, F.; Mao, X.; Ikeda, T.; Takasaki, J.-i.; Nishijo, H. Phenolic compounds prevent amyloid β-protein oligomerization and synaptic dysfunction by site-specific binding. J. Biol. Chem. 2012, 287, 14631–14643. [Google Scholar] [CrossRef] [Green Version]
- Gopal, S.; Jahan, I. Wine-related flavonols for therapeutic use in Alzheimer’s disease, an in-silico investigation. J. Proteins Proteom. 2022, 13, 133–148. [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]
- Zhang, Z.-X.; Li, Y.-B.; Zhao, R.-P. Epigallocatechin gallate attenuates β-amyloid generation and oxidative stress involvement of PPARγ in N2a/APP695 cells. Neurochem. Res. 2017, 42, 468–480. [Google Scholar] [CrossRef]
- Rong, H.; Liang, Y.; Niu, Y. Rosmarinic acid attenuates β-amyloid-induced oxidative stress via Akt/GSK-3β/Fyn-mediated Nrf2 activation in PC12 cells. Free Radic. Biol. Med. 2018, 120, 114–123. [Google Scholar] [CrossRef]
- Zhang, L.; Fang, Y.; Cheng, X.; Lian, Y.; Zeng, Z.; Wu, C.; Zhu, H.; Xu, H. The potential protective effect of curcumin on amyloid-β-42 induced cytotoxicity in HT-22 cells. BioMed Res. Int. 2018, 2018, 8134902. [Google Scholar] [CrossRef] [Green Version]
- Feng, Y.; Wang, X.-P.; Yang, S.-G.; Wang, Y.-J.; Zhang, X.; Du, X.-T.; Sun, X.-X.; Zhao, M.; Huang, L.; Liu, R.-T. Resveratrol inhibits beta-amyloid oligomeric cytotoxicity but does not prevent oligomer formation. Neurotoxicology 2009, 30, 986–995. [Google Scholar] [CrossRef] [PubMed]
- Mori, T.; Rezai-Zadeh, K.; Koyama, N.; Arendash, G.W.; Yamaguchi, H.; Kakuda, N.; Horikoshi-Sakuraba, Y.; Tan, J.; Town, T. Tannic acid is a natural β-secretase inhibitor that prevents cognitive impairment and mitigates Alzheimer-like pathology in transgenic mice. J. Biol. Chem. 2012, 287, 6912–6927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qosa, H.; Batarseh, Y.S.; Mohyeldin, M.M.; El Sayed, K.A.; Keller, J.N.; Kaddoumi, A. Oleocanthal enhances amyloid-β clearance from the brains of TgSwDI mice and in vitro across a human blood-brain barrier model. ACS Chem. Neurosci. 2015, 6, 1849–1859. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kleinrichert, K.; Alappat, B. Comparative analysis of antioxidant and anti-amyloidogenic properties of various polyphenol rich phytoceutical extracts. Antioxidants 2019, 8, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toni, M.; Massimino, M.L.; De Mario, A.; Angiulli, E.; Spisni, E. Metal dyshomeostasis and their pathological role in prion and prion-like diseases: The basis for a nutritional approach. Front. Neurosci. 2017, 11, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carmona, V.; Martín-Aragón, S.; Goldberg, J.; Schubert, D.; Bermejo-Bescós, P. Several targets involved in Alzheimer’s disease amyloidogenesis are affected by morin and isoquercitrin. Nutr. Neurosci. 2020, 23, 575–590. [Google Scholar] [CrossRef] [PubMed]
- Jiménez-Aliaga, K.; Bermejo-Bescós, P.; Benedí, J.; Martín-Aragón, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci. 2011, 89, 939–945. [Google Scholar] [CrossRef]
- Das, S.; Datta, S.; Ghosal, A.; Chaudhuri, N.R.; Sundaram, G.; Basu, S. Screening of BACE1 inhibitors with antiamyloidogenic activity: A study of flavonoids and flavonoid derivatives. Neurosci. Lett. 2023, 792, 136965. [Google Scholar] [CrossRef]
- Windsor, P.K.; Plassmeyer, S.P.; Mattock, D.S.; Bradfield, J.C.; Choi, E.Y.; Miller Iii, B.R.; Han, B.H. Biflavonoid-induced disruption of hydrogen bonds leads to amyloid-β disaggregation. Int. J. Mol. Sci. 2021, 22, 2888. [Google Scholar] [CrossRef]
- Mithu, V.S.; Sarkar, B.; Bhowmik, D.; Das, A.K.; Chandrakesan, M.; Maiti, S.; Madhu, P.K. Curcumin alters the salt bridge-containing turn region in amyloid β (1–42) aggregates. J. Biol. Chem. 2014, 289, 11122–11131. [Google Scholar] [CrossRef] [Green Version]
- Brahmkhatri, V.P.; Sharma, N.; Sunanda, P.; D’Souza, A.; Raghothama, S.; Atreya, H.S. Curcumin nanoconjugate inhibits aggregation of N-terminal region (Aβ-16) of an amyloid beta peptide. New J. Chem. 2018, 42, 19881–19892. [Google Scholar] [CrossRef]
- Manap, A.S.A.; Madhavan, P.; Vijayabalan, S.; Chia, A.; Fukui, K. Explicating anti-amyloidogenic role of curcumin and piperine via amyloid beta (Aβ) explicit pathway: Recovery and reversal paradigm effects. PeerJ 2020, 8, e10003. [Google Scholar] [CrossRef] [PubMed]
- Cascella, M.; Bimonte, S.; Muzio, M.R.; Schiavone, V.; Cuomo, A. The efficacy of Epigallocatechin-3-gallate (green tea) in the treatment of Alzheimer’s disease: An overview of pre-clinical studies and translational perspectives in clinical practice. Infect. Agents Cancer 2017, 12, 36. [Google Scholar] [CrossRef] [PubMed]
- Guo, Y.; Zhao, Y.; Nan, Y.; Wang, X.; Chen, Y.; Wang, S. (−)-Epigallocatechin-3-gallate ameliorates memory impairment and rescues the abnormal synaptic protein levels in the frontal cortex and hippocampus in a mouse model of Alzheimer’s disease. Neuroreport 2017, 28, 590–597. [Google Scholar] [CrossRef]
- Ahmed, R.; VanSchouwen, B.; Jafari, N.; Ni, X.; Ortega, J.; Melacini, G. Molecular mechanism for the (−)-epigallocatechin gallate-induced toxic to nontoxic remodeling of Aβ oligomers. J. Am. Chem. Soc. 2017, 139, 13720–13734. [Google Scholar] [CrossRef] [PubMed]
- Bao, J.; Liu, W.; Zhou, H.-Y.; Gui, Y.-R.; Yang, Y.-H.; Wu, M.-J.; Xiao, Y.-F.; Shang, J.-T.; Long, G.-F.; Shu, X.-J. Epigallocatechin-3-gallate alleviates cognitive deficits in APP/PS1 mice. Curr. Med. Sci. 2020, 40, 18–27. [Google Scholar] [CrossRef]
- Mori, T.; Koyama, N.; Tan, J.; Segawa, T.; Maeda, M.; Town, T. Combined treatment with the phenolics (−)-epigallocatechin-3-gallate and ferulic acid improves cognition and reduces Alzheimer-like pathology in mice. J. Biol. Chem. 2019, 294, 2714–5444. [Google Scholar] [CrossRef] [Green Version]
- Pacifico, S.; Bláha, P.; Faramarzi, S.; Fede, F.; Michaličková, K.; Piccolella, S.; Ricciardi, V.; Manti, L. Differential Radiomodulating Action of Olea europaea L. cv. Caiazzana Leaf Extract on Human Normal and Cancer Cells: A Joint Chemical and Radiobiological Approach. Antioxidants 2022, 11, 1603. [Google Scholar] [CrossRef]
- Rigacci, S. Olive oil phenols as promising multi-targeting agents against Alzheimer’s disease. In Natural Compounds as Therapeutic Agents for Amyloidogenic Diseases; Springer International Publishing: Cham, Switzerland, 2015; pp. 1–20. [Google Scholar]
- Hanaki, M.; Murakami, K.; Akagi, K.-I.; Irie, K. Structural insights into mechanisms for inhibiting amyloid β42 aggregation by non-catechol-type flavonoids. Bioorganic Med. Chem. 2016, 24, 304–313. [Google Scholar] [CrossRef]
- Andarzi Gargari, S.; Barzegar, A.; Tarinejad, A. The role of phenolic OH groups of flavonoid compounds with H-bond formation ability to suppress amyloid mature fibrils by destabilizing β-sheet conformation of monomeric Aβ17-42. PLoS ONE 2018, 13, e0199541. [Google Scholar] [CrossRef] [Green Version]
- Douglas Shytle, R.; Tan, J.; Bickford, P.C.; Rezai-Zadeh, K.; Hou, L.; Zeng, J.; Sanberg, P.R.; Sanberg, C.D.; Alberte, R.S.; Fink, R.C. Optimized turmeric extract reduces β-amyloid and phosphorylated tau protein burden in Alzheimer’s transgenic mice. Curr. Alzheimer Res. 2012, 9, 500–506. [Google Scholar] [CrossRef] [Green Version]
- Smith, J.V.; Luo, Y. Elevation of oxidative free radicals in Alzheimer’s disease models can be attenuated by Ginkgo biloba extract EGb 761. J. Alzheimer’s Dis. 2003, 5, 287–300. [Google Scholar] [CrossRef]
- Gupta, V.B.; Indi, S.S.; Rao, K.S.J. Garlic extract exhibits antiamyloidogenic activity on amyloid-beta fibrillogenesis: Relevance to Alzheimer’s disease. Phytother. Res. 2009, 23, 111–115. [Google Scholar] [CrossRef]
- Figueira, I.; Tavares, L.; Jardim, C.; Costa, I.; Terrasso, A.P.; Almeida, A.F.; Govers, C.; Mes, J.J.; Gardner, R.; Becker, J.D. Blood–brain barrier transport and neuroprotective potential of blackberry-digested polyphenols: An in vitro study. Eur. J. Nutr. 2019, 58, 113–130. [Google Scholar] [CrossRef] [PubMed]
- Ho, L.; Yemul, S.; Wang, J.; Pasinetti, G.M. Grape seed polyphenolic extract as a potential novel therapeutic agent in tauopathies. J. Alzheimer’s Dis. 2009, 16, 433–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pasinetti, G.M.; Ksiezak-Reding, H.; Santa-Maria, I.; Wang, J.; Ho, L. Development of a grape seed polyphenolic extract with anti-oligomeric activity as a novel treatment in progressive supranuclear palsy and other tauopathies. J. Neurochem. 2010, 114, 1557–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pacifico, S.; Piccolella, S.; Marciano, S.; Galasso, S.; Nocera, P.; Piscopo, V.; Fiorentino, A.; Monaco, P. LC-MS/MS profiling of a mastic leaf phenol enriched extract and its effects on H2O2 and Aβ (25–35) oxidative injury in SK-B-NE (C)-2 cells. J. Agric. Food Chem. 2014, 62, 11957–11966. [Google Scholar] [CrossRef] [PubMed]
- Pacifico, S.; Gallicchio, M.; Lorenz, P.; Duckstein, S.M.; Potenza, N.; Galasso, S.; Marciano, S.; Fiorentino, A.; Stintzing, F.C.; Monaco, P. Neuroprotective potential of Laurus nobilis antioxidant polyphenol-enriched leaf extracts. Chem. Res. Toxicol. 2014, 27, 611–626. [Google Scholar] [CrossRef] [PubMed]
- Pacifico, S.; Piccolella, S.; Lettieri, A.; Nocera, P.; Bollino, F.; Catauro, M. A metabolic profiling approach to an Italian sage leaf extract (SoA541) defines its antioxidant and anti-acetylcholinesterase properties. J. Funct. Foods 2017, 29, 1–9. [Google Scholar] [CrossRef]
- Xu, B.; Chen, J.; Liu, Y. Curcumin Interacts with α-Synuclein Condensates To Inhibit Amyloid Aggregation under Phase Separation. ACS Omega 2022, 7, 30281–30290. [Google Scholar] [CrossRef] [PubMed]
- Palhano, F.L.; Lee, J.; Grimster, N.P.; Kelly, J.W. Toward the molecular mechanism (s) by which EGCG treatment remodels mature amyloid fibrils. J. Am. Chem. Soc. 2013, 135, 7503–7510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mor, D.E.; Tsika, E.; Mazzulli, J.R.; Gould, N.S.; Kim, H.; Daniels, M.J.; Doshi, S.; Gupta, P.; Grossman, J.L.; Tan, V.X. Dopamine induces soluble α-synuclein oligomers and nigrostriatal degeneration. Nat. Neurosci. 2017, 20, 1560–1568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zubčić, K.; Hof, P.R.; Šimić, G.; Jazvinšćak Jembrek, M. The role of copper in tau-related pathology in Alzheimer’s disease. Front. Mol. Neurosci. 2020, 13, 572308. [Google Scholar] [CrossRef] [PubMed]
- Hu, J.; Lin, T.; Xu, J.; Ding, R.; Wang, G.; Shen, R.; Zhang, Y.-W.; Chen, H. Polyphenols isolated from leaves of Vitis thunbergii var. taiwaniana regulate APP related pathway. Bioorg. Med. Chem. Lett. 2016, 26, 505–511. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Wu, A.; Li, X.; Qin, D.; Jin, B.; Liu, J.; Tang, Y.; Wu, J.; Yu, C. The seed of Litchi chinensis fraction ameliorates hippocampal neuronal injury in an Aβ25-35-induced Alzheimer’s disease rat model via the AKT/GSK-3β pathway. Pharm. Bio. 2020, 58, 35–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Santa-Maria, I.; Ho, L.; Ksiezak-Reding, H.; Ono, K.; Teplow, D.B.; Pasinetti, G.M. Grape derived polyphenols attenuate tau neuropathology in a mouse model of Alzheimer’s disease. J. Alzheimer’s Dis. 2010, 22, 653–661. [Google Scholar] [CrossRef] [Green Version]
- Guéroux, M.; Fleau, C.; Slozeck, M.; Laguerre, M.; Pianet, I. Epigallocatechin 3-gallate as an inhibitor of tau phosphorylation and aggregation: A molecular and structural insight. J. Prev. Alzheimers Dis. 2017, 4, 218–225. [Google Scholar]
- Gao, A.X.; Xia, T.C.-X.; Mak, M.S.-H.; Kwan, K.K.-L.; Zheng, B.Z.-Y.; Xiao, J.; Dong, T.T.-X.; Tsim, K.W.-K. Luteolin stimulates the NGF-induced neurite outgrowth in cultured PC12 cells through binding with NGF and potentiating its receptor signaling. Food Funct. 2021, 12, 11515–11525. [Google Scholar]
- Suh Yewseok, K.; Robinson, A.; Zanghi, N.; Kratz, A.; Gustetic, A.; Crow Mackenzie, M.; Ritts, T.; Hankey, W.; Segarra Verónica, A. Introducing Wound Healing Assays in the Undergraduate Biology Laboratory Using Ibidi Plates. J. Microbiol. Biol. Educ. 2022, 23, e00061-22. [Google Scholar] [CrossRef]
- Johnson, G.V.W.; Stoothoff, W.H. Tau phosphorylation in neuronal cell function and dysfunction. J. Cell Sci. 2004, 117, 5721–5729. [Google Scholar] [CrossRef] [Green Version]
- Milan, G.; Napoletano, S.; Pappatà, S.; Gentile, M.T.; Colucci-D’Amato, L.; Della Rocca, G.; Maciag, A.; Rossetti, C.P.; Fucci, L.; Puca, A. GRN deletion in familial frontotemporal dementia showing association with clinical variability in 3 familial cases. Neurobiol. Aging 2017, 53, 193.e9–193.e16. [Google Scholar] [CrossRef] [PubMed]
- Pan, L.; Meng, L.; He, M.; Zhang, Z. Tau in the pathophysiology of Parkinson’s disease. J. Mol. Neurosci. 2021, 71, 2179–2191. [Google Scholar] [CrossRef]
- Zhang, X.; Gao, F.; Wang, D.; Li, C.; Fu, Y.; He, W.; Zhang, J. Tau pathology in Parkinson’s disease. Front. Neurol. 2018, 9, 809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lakey-Beitia, J.; Burillo, A.M.; La Penna, G.; Hegde, M.L.; Rao, K.S. Polyphenols as potential metal chelation compounds against Alzheimer’s disease. J. Alzheimer’s Dis. 2021, 82, S335–S357. [Google Scholar] [CrossRef] [PubMed]
- Fukumoto, L.R.; Mazza, G. Assessing antioxidant and prooxidant activities of phenolic compounds. J. Agric. Food Chem. 2000, 48, 3597–3604. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Yang, J.; Lu, Y.; Qian, L.-L.; Yang, Z.-Y.; Han, R.-M.; Zhang, J.-P.; Skibsted, L.H. Copper (II) coordination and translocation in luteolin and effect on radical scavenging. J. Phys. Chem. B 2019, 124, 380–388. [Google Scholar] [CrossRef] [PubMed]
- Abolaji, A.O.; Fasae, K.D.; Iwezor, C.E.; Aschner, M.; Farombi, E.O. Curcumin attenuates copper-induced oxidative stress and neurotoxicity in Drosophila melanogaster. Toxicol. Rep. 2020, 7, 261–268. [Google Scholar] [CrossRef]
- Lee, V.J.; Heffern, M.C. Structure-activity assessment of flavonoids as modulators of copper transport. Front. Chem. 2022, 930. [Google Scholar] [CrossRef]
- Sadžak, A.; Vlašić, I.; Kiralj, Z.; Batarelo, M.; Oršolić, N.; Jazvinšćak Jembrek, M.; Kušen, I.; Šegota, S. Neurotoxic effect of flavonol myricetin in the presence of excess copper. Molecules 2021, 26, 845. [Google Scholar] [CrossRef]
- Pan, Y.; Qin, R.; Hou, M.; Xue, J.; Zhou, M.; Xu, L.; Zhang, Y. The interactions of polyphenols with Fe and their application in Fenton/Fenton-like reactions. Sep. Purif. Technol. 2022, 300, 121831. [Google Scholar] [CrossRef]
- Nowak, M.; Tryniszewski, W.; Sarniak, A.; Wlodarczyk, A.; Nowak, P.J.; Nowak, D. Concentration Dependence of Anti-and Pro-Oxidant Activity of Polyphenols as Evaluated with a Light-Emitting Fe2+-Egta-H2O2 System. Molecules 2022, 27, 3453. [Google Scholar] [CrossRef] [PubMed]
- Masaldan, S.; Belaidi, A.A.; Ayton, S.; Bush, A.I. Cellular senescence and iron dyshomeostasis in Alzheimer’s disease. Pharmaceuticals 2019, 12, 93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tosato, M.; Di Marco, V. Metal chelation therapy and Parkinson’s disease: A critical review on the thermodynamics of complex formation between relevant metal ions and promising or established drugs. Biomolecules 2019, 9, 269. [Google Scholar] [CrossRef] [Green Version]
- Moons, R.; Konijnenberg, A.; Mensch, C.; Van Elzen, R.; Johannessen, C.; Maudsley, S.; Lambeir, A.-M.; Sobott, F. Metal ions shape α-synuclein. Sci. Rep. 2020, 10, 16293. [Google Scholar] [CrossRef]
- Lautenschläger, J.; Stephens, A.D.; Fusco, G.; Ströhl, F.; Curry, N.; Zacharopoulou, M.; Michel, C.H.; Laine, R.; Nespovitaya, N.; Fantham, M. C-terminal calcium binding of α-synuclein modulates synaptic vesicle interaction. Nat. Commun. 2018, 9, 712. [Google Scholar] [CrossRef] [Green Version]
- Mahoney-Sánchez, L.; Bouchaoui, H.; Ayton, S.; Devos, D.; Duce, J.A.; Devedjian, J.-C. Ferroptosis and its potential role in the physiopathology of Parkinson’s Disease. Prog. Neurobiol. 2021, 196, 101890. [Google Scholar] [CrossRef]
- Sun, G.; Miao, Z.; Ye, Y.; Zhao, P.; Fan, L.; Bao, Z.; Tu, Y.; Li, C.; Chao, H.; Xu, X. Curcumin alleviates neuroinflammation, enhances hippocampal neurogenesis, and improves spatial memory after traumatic brain injury. Brain Res. Bull. 2020, 162, 84–93. [Google Scholar] [CrossRef]
- Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [Green Version]
- Guzman-Martinez, L.; Maccioni, R.B.; Andrade, V.; Navarrete, L.P.; Pastor, M.G.; Ramos-Escobar, N. Neuroinflammation as a common feature of neurodegenerative disorders. Front. Pharmacol. 2019, 10, 1008. [Google Scholar] [CrossRef] [Green Version]
- Jiao, F.; Gong, Z. The beneficial roles of SIRT1 in neuroinflammation-related diseases. Oxid. Med. Cell. Longev. 2020, 2020, 6782872. [Google Scholar] [CrossRef] [PubMed]
- Pallàs, M.; Casadesús, G.; Smith, M.A.; Coto-Montes, A.; Pelegri, C.; Vilaplana, J.; Camins, A. Resveratrol and neurodegenerative diseases: Activation of SIRT1 as the potential pathway towards neuroprotection. Curr. Neurovasc. Res. 2009, 6, 70–81. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.; Zhou, L.; Chen, J.; Chen, T.; Lei, B.; Zheng, N.; Wan, X.; Xu, J.; Wang, T. Hyperoside attenuate inflammation in HT22 cells via upregulating SIRT1 to activities wnt/β-catenin and sonic hedgehog pathways. Neural Plast. 2021, 2021, 8706400. [Google Scholar] [CrossRef] [PubMed]
- Ramis, M.R.; Sarubbo, F.; Tejada, S.; Jiménez, M.; Esteban, S.; Miralles, A.; Moranta, D. Chronic Polyphenon-60 or Catechin treatments increase brain monoamines syntheses and hippocampal SIRT1 levels improving cognition in aged rats. Nutrients 2020, 12, 326. [Google Scholar] [CrossRef] [Green Version]
- Bonechi, C.; Martini, S.; Ciani, L.; Lamponi, S.; Rebmann, H.; Rossi, C.; Ristori, S. Using liposomes as carriers for polyphenolic compounds: The case of trans-resveratrol. PLoS ONE 2012, 7, e41438. [Google Scholar] [CrossRef]
- Trotta, V.; Pavan, B.; Ferraro, L.; Beggiato, S.; Traini, D.; Des Reis, L.G.; Scalia, S.; Dalpiaz, A. Brain targeting of resveratrol by nasal administration of chitosan-coated lipid microparticles. Eur. J. Pharm. Biopharm. 2018, 127, 250–259. [Google Scholar] [CrossRef]
- Chen, Z.-L.; Huang, M.; Wang, X.-R.; Fu, J.; Han, M.; Shen, Y.-Q.; Xia, Z.; Gao, J.-Q. Transferrin-modified liposome promotes α-mangostin to penetrate the blood–brain barrier. Nanomed. Nanotechnol. Biol. Med. 2016, 12, 421–430. [Google Scholar] [CrossRef]
- Ferri, P.; Angelino, D.; Gennari, L.; Benedetti, S.; Ambrogini, P.; Del Grande, P.; Ninfali, P. Enhancement of flavonoid ability to cross the blood–brain barrier of rats by co-administration with α-tocopherol. Food Funct. 2015, 6, 394–400. [Google Scholar] [CrossRef]
- Kumari, N.; Daram, N.; Alam, M.S.; Verma, A.K. Rationalizing the Use of Polyphenol Nano-formulations in the Therapy of Neurodegenerative Diseases. CNS Neurol. Disord.-Drug Targets (Former. Curr. Drug Targets-CNS Neurol. Disord.) 2022, 21, 966–976. [Google Scholar]
- Pinheiro, R.G.R.; Granja, A.; Loureiro, J.A.; Pereira, M.C.; Pinheiro, M.; Neves, A.R.; Reis, S. RVG29-functionalized lipid nanoparticles for quercetin brain delivery and Alzheimer’s disease. Pharm. Res. 2020, 37, 139. [Google Scholar] [CrossRef]
- Dei Cas, M.; Ghidoni, R. Dietary curcumin: Correlation between bioavailability and health potential. Nutrients 2019, 11, 2147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salehi, S.; Nourbakhsh, M.S.; Yousefpour, M.; Rajabzadeh, G.; Sahab-Negah, S. Chitosan-coated niosome as an efficient curcumin carrier to cross the blood–brain barrier: An animal study. J. Liposome Res. 2022, 32, 284–292. [Google Scholar] [CrossRef] [PubMed]
- Ingallina, C.; Rinaldi, F.; Bogni, A.; Ponti, J.; Passeri, D.; Reggente, M.; Rossi, M.; Kinsner-Ovaskainen, A.; Mehn, D.; Rossi, F. Niosomal approach to brain delivery: Development, characterization and in vitro toxicological studies. Int. J. Pharm. 2016, 511, 969–982. [Google Scholar] [CrossRef] [PubMed]
- Trapani, A.; Guerra, L.; Corbo, F.; Castellani, S.; Sanna, E.; Capobianco, L.; Monteduro, A.G.; Manno, D.E.; Mandracchia, D.; Di Gioia, S. Cyto/biocompatibility of dopamine combined with the antioxidant grape seed-derived polyphenol compounds in solid lipid nanoparticles. Molecules 2021, 26, 916. [Google Scholar] [CrossRef]
- Harakeh, S.; Qari, M.H.; Ramadan, W.S.; Al Jaouni, S.K.; Almuhayawi, M.S.; Al Amri, T.; Ashraf, G.M.; Bharali, D.J.; Mousa, S.A. A Novel Nanoformulation of Ellagic Acid is Promising in Restoring Oxidative Homeostasis in Rat Brains with Alzheimer’s Disease. Curr. Drug Metab. 2021, 22, 299–307. [Google Scholar]
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
Gentile, M.T.; Camerino, I.; Ciarmiello, L.; Woodrow, P.; Muscariello, L.; De Chiara, I.; Pacifico, S. Neuro-Nutraceutical Polyphenols: How Far Are We? Antioxidants 2023, 12, 539. https://doi.org/10.3390/antiox12030539
Gentile MT, Camerino I, Ciarmiello L, Woodrow P, Muscariello L, De Chiara I, Pacifico S. Neuro-Nutraceutical Polyphenols: How Far Are We? Antioxidants. 2023; 12(3):539. https://doi.org/10.3390/antiox12030539
Chicago/Turabian StyleGentile, Maria Teresa, Iolanda Camerino, Loredana Ciarmiello, Pasqualina Woodrow, Lidia Muscariello, Ida De Chiara, and Severina Pacifico. 2023. "Neuro-Nutraceutical Polyphenols: How Far Are We?" Antioxidants 12, no. 3: 539. https://doi.org/10.3390/antiox12030539