Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1
Abstract
:1. Introduction
2. Berries as Source of Quercetin
3. Metabolism of Quercetin
3.1. Effects of Quercetin on Cognitive Impairments
3.2. Effects of Quercetin on Oxidative Stress
3.3. Effects of Quercetin on P-53-Mediated Apoptosis
3.4. Effects of Quercetin on Neuroinflammation
3.5. Effects of Quercetin on Amyloid-β and Tau
3.6. Effects of Quercetin on Apolipoprotein E ε4
3.7. Effects of Quercetin on α-Synuclein
4. Clinical Trial of Quercetin
5. Limitations and Challenges of Quercetin for Neuroprotection
6. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Pedersen, J.T.; Chen, S.W.; Borg, C.B.; Ness, S.; Bahl, J.M.; Heegaard, N.H.; Teilum, K. Amyloid-β and α-synuclein decrease the level of metal-catalyzed reactive oxygen species by radical scavenging and redox silencing. J. Am. Chem. Soc. 2016, 138, 3966–3969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, D.C. Mitochondria: Dynamic Organelles in Disease, Aging, and Development. Cell 2006, 125, 1241–1252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.-W.; Thompson, R.; Zhang, H.; Xu, H. APP processing in Alzheimer’s disease. Mol. Brain 2011, 4, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luo, J.-F.; Shen, X.Y.; Lio, C.K.; Dai, Y.; Cheng, C.S.; Liu, J.X.; Zhou, H. Activation of Nrf2/HO-1 Pathway by Nardochinoid C Inhibits Inflammation and Oxidative Stress in Lipopolysaccharide-Stimulated Macrophages. Front. Pharmacol. 2018, 9, 911. [Google Scholar] [CrossRef] [Green Version]
- Arnold, S. Cytochrome c Oxidase and Its Role in Neurodegeneration and Neuroprotection. In Mitochondrial Oxidative Phosphorylation: Nuclear-Encoded Genes, Enzyme Regulation, and Pathophysiology; Kadenbach, B., Ed.; Springer: New York, NY, USA, 2012; pp. 305–339. [Google Scholar]
- Forman, H.J.; Zhang, H. Targeting oxidative stress in disease: Promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 2021, 20, 689–709. [Google Scholar] [CrossRef] [PubMed]
- Moosecker, S.; Gomes, P.; Dioli, C.; Yu, S.; Sotiropoulos, I.; Almeida, O.F. Activated PPARγ abrogates misprocessing of amyloid precursor protein, Tau missorting and synaptotoxicity. Front. Cell. Neurosci. 2019, 13, 239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koh, E.-J.; Kim, K.-J.; Choi, J.; Kang, D.-H.; Lee, B.-Y. Spirulina maxima extract prevents cell death through BDNF activation against amyloid beta 1-42 (Aβ 1-42) induced neurotoxicity in PC12 cells. Neurosci. Lett. 2018, 673, 33–38. [Google Scholar] [CrossRef]
- Bayazid, A.B.; Jang, Y.A.; Kim, Y.M.; Kim, J.G.; Lim, B.O. Neuroprotective Effects of Sodium Butyrate through Suppressing Neuroinflammation and Modulating Antioxidant Enzymes. Neurochem. Res. 2021, 46, 2348–2358. [Google Scholar] [CrossRef]
- Tonelli, C.; Chio, I.I.C.; Tuveson, D.A. Transcriptional Regulation by Nrf2. Antioxid. Redox Signal. 2018, 29, 1727–1745. [Google Scholar] [CrossRef] [Green Version]
- Jabri, M.-A.; Rtibi, K.; Sebai, H. Chamomile decoction mitigates high fat diet-induced anxiety-like behavior, neuroinflammation and cerebral ROS overload. Nutr. Neurosci. 2020, 25, 1350–1361. [Google Scholar] [CrossRef]
- Brandes, M.S.; Gray, N.E. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro 2020, 12, 1759091419899782. [Google Scholar] [CrossRef] [PubMed]
- Watson, R.R. Complementary and Alternative Therapies and the Aging Population: An Evidence-based Approach; Academic Press: Cambridge, MA, USA, 2011. [Google Scholar]
- Bayazid, A.B.; Jang, Y.A. The Role of Andrographolide on Skin Inflammations and Modulation of Skin Barrier Functions in Human Keratinocyte. Biotechnol. Bioprocess Eng. 2021, 26, 804–813. [Google Scholar] [CrossRef]
- Bayazid, A.B.; Jang, Y.A.; Jeong, S.A.; Lim, B.O. Cypress tree (Chamaecyparis obtusa) Bark extract inhibits melanogenesis through repressing CREB and MITF signalling pathways in α-MSH-stimulated B16F10 cells. Food Agric. Immunol. 2022, 33, 498–510. [Google Scholar] [CrossRef]
- Valle, I.F.D.; Roweth, H.G.; Malloy, M.W.; Moco, S.; Barron, D.; Battinelli, E.; Loscalzo, J.; Barabási, A.-L. Network medicine framework shows that proximity of polyphenol targets and disease proteins predicts therapeutic effects of polyphenols. Nat. Food 2021, 2, 143–155. [Google Scholar] [CrossRef]
- Redford, K.E.; Abbott, G.W. The ubiquitous flavonoid quercetin is an atypical KCNQ potassium channel activator. Commun. Biol. 2020, 3, 356. [Google Scholar] [CrossRef]
- Dabeek, W.M.; Marra, M.V. Dietary Quercetin and Kaempferol: Bioavailability and Potential Cardiovascular-Related Bioactivity in Humans. Nutrients 2019, 11, 2288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- García-Viñuales, S.; Ahmed, R.; Sciacca, M.F.M.; Lanza, V.; Giuffrida, M.L.; Zimbone, S.; Romanucci, V.; Zarrelli, A.; Bongiorno, C.; Spinella, N.; et al. Trehalose Conjugates of Silybin as Prodrugs for Targeting Toxic Aβ Aggregates. ACS Chem. Neurosci. 2020, 11, 2566–2576. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Hu, J.; Zhong, L.; Wang, N.; Yang, L.; Liu, C.-C.; Li, H.; Wang, X.; Zhou, Y.; Zhang, Y.; et al. Quercetin stabilizes apolipoprotein E and reduces brain Aβ levels in amyloid model mice. Neuropharmacology 2016, 108, 179–192. [Google Scholar] [CrossRef] [PubMed]
- Rauf, A.; Imran, M.; Khan, I.A.; Ur-Rehman, M.; Gilani, S.A.; Mehmood, Z.; Mubarak, M.S. Anticancer potential of quercetin: A comprehensive review. Phytotherapy Res. 2018, 32, 2109–2130. [Google Scholar] [CrossRef]
- Lesjak, M.; Beara, I.; Simin, N.; Pintać, D.; Majkić, T.; Bekvalac, K.; Orčić, D.; Mimica-Dukić, N. Antioxidant and anti-inflammatory activities of quercetin and its derivatives. J. Funct. Foods 2018, 40, 68–75. [Google Scholar] [CrossRef]
- Singh, P.; Arif, Y.; Bajguz, A.; Hayat, S. The role of quercetin in plants. Plant Physiol. Biochem. 2021, 166, 10–19. [Google Scholar] [CrossRef] [PubMed]
- Sellappan, S.; Akoh, C.C.; Krewer, G. Phenolic Compounds and Antioxidant Capacity of Georgia-Grown Blueberries and Blackberries. J. Agric. Food Chem. 2002, 50, 2432–2438. [Google Scholar] [CrossRef] [PubMed]
- Häkkinen, S.H.; Kärenlampi, S.O.; Heinonen, I.M.; Mykkänen, H.M.; Törrönen, A.R. Content of the Flavonols Quercetin, Myricetin, and Kaempferol in 25 Edible Berries. J. Agric. Food Chem. 1999, 47, 2274–2279. [Google Scholar] [CrossRef] [PubMed]
- Russo, G.L.; Russo, M.; Spagnuolo, C. The pleiotropic flavonoid quercetin: From its metabolism to the inhibition of protein kinases in chronic lymphocytic leukemia. Food Funct. 2014, 5, 2393–2401. [Google Scholar] [CrossRef] [PubMed]
- Docampo, M.; Olubu, A.; Wang, X.; Pasinetti, G.; Dixon, R.A. Glucuronidated Flavonoids in Neurological Protection: Structural Analysis and Approaches for Chemical and Biological Synthesis. J. Agric. Food Chem. 2017, 65, 7607–7623. [Google Scholar] [CrossRef]
- Lines, T.C. Quercetin-Containing Compositions. US Patents US8440704B2, 14 May 2013. [Google Scholar]
- Ulusoy, H.G.; Sanlier, N. A minireview of quercetin: From its metabolism to possible mechanisms of its biological activities. Crit. Rev. Food Sci. Nutr. 2019, 60, 3290–3303. [Google Scholar] [CrossRef]
- Lakhanpal, P.; Rai, D.K. Quercetin: A versatile flavonoid. Internet J. Med. Update 2007, 2, 22–37. [Google Scholar] [CrossRef] [Green Version]
- Terao, J.; Murota, K.; Kawai, Y. Conjugated quercetin glucuronides as bioactive metabolites and precursors of aglycone in vivo. Food Funct. 2011, 2, 11–17. [Google Scholar] [CrossRef]
- Xiao, J.; Kai, G. A Review of Dietary Polyphenol-Plasma Protein Interactions: Characterization, Influence on the Bioactivity, and Structure-Affinity Relationship. Crit. Rev. Food Sci. Nutr. 2012, 52, 85–101. [Google Scholar] [CrossRef]
- Bakoyiannis, I.; Daskalopoulou, A.; Pergialiotis, V.; Perrea, D. Phytochemicals and cognitive health: Are flavonoids doing the trick? Biomed. Pharmacother. 2019, 109, 1488–1497. [Google Scholar] [CrossRef]
- Robertson, D.A.; Savva, G.M.; Kenny, R.A. Frailty and cognitive impairment—A review of the evidence and causal mechanisms. Ageing Res. Rev. 2013, 12, 840–851. [Google Scholar] [CrossRef] [PubMed]
- Broman-Fulks, J.J.; Canu, W.H.; Trout, K.L.; Nieman, D.C. The effects of quercetin supplementation on cognitive functioning in a community sample: A randomized, placebo-controlled trial. Ther. Adv. Psychopharmacol. 2012, 2, 131–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Subash, S.; Essa, M.M.; Al-Adawi, S.; Memon, M.A.; Manivasagam, T.; Akbar, M. Neuroprotective effects of berry fruits on neurodegenerative diseases. Neural Regener. Res. 2014, 9, 1557–1566. [Google Scholar] [CrossRef]
- Wang, D.-M.; Li, S.-Q.; Wu, W.-L.; Zhu, X.-Y.; Wang, Y.; Yuan, H.-Y. Effects of Long-Term Treatment with Quercetin on Cognition and Mitochondrial Function in a Mouse Model of Alzheimer’s Disease. Neurochem. Res. 2014, 39, 1533–1543. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Wang, G.; Ma, Z.F.; Qin, L.-Q.; Zhai, Y.-J.; Yu, Z.-L.; Xue, M.; Zhang, Y.-H.; Wan, Z. DietaryAdvancedGlycationEnd Products-InducedCognitive Impairment in Aged ICR Mice: Protective Role of Quercetin. Mol. Nutr. Food Res. 2019, 64, e1901019. [Google Scholar] [CrossRef]
- Zingales, V.; Sirerol-Piquer, M.S.; Fernández-Franzón, M.; Ruiz, M.-J. Role of quercetin on sterigmatocystin-induced oxidative stress-mediated toxicity. Food Chem. Toxicol. 2021, 156, 112498. [Google Scholar] [CrossRef]
- Halder, S.; Kar, R.; Galav, V.; Mehta, A.K.; Bhattacharya, S.K.; Mediratta, P.K.; Banerjee, B.D. Cadmium exposure during lactation causes learning and memory-impairment in F1 generation mice: Amelioration by quercetin. Drug Chem. Toxicol. 2015, 39, 272–278. [Google Scholar] [CrossRef]
- Priprem, A.; Watanatorn, J.; Sutthiparinyanont, S.; Phachonpai, W.; Muchimapura, S. Anxiety and cognitive effects of quercetin liposomes in rats. Nanomed. Nanotechnol. Biol. Med. 2008, 4, 70–78. [Google Scholar] [CrossRef]
- Choi, G.N.; Kim, J.H.; Kwak, J.H.; Jeong, C.-H.; Jeong, H.R.; Lee, U.; Heo, H.J. Effect of quercetin on learning and memory performance in ICR mice under neurotoxic trimethyltin exposure. Food Chem. 2011, 132, 1019–1024. [Google Scholar] [CrossRef]
- Pu, F.; Mishima, K.; Irie, K.; Motohashi, K.; Tanaka, Y.; Orito, K.; Egawa, T.; Kitamura, Y.; Egashira, N.; Iwasaki, K.; et al. Neuroprotective Effects of Quercetin and Rutin on Spatial Memory Impairment in an 8-Arm Radial Maze Task and Neuronal Death Induced by Repeated Cerebral Ischemia in Rats. J. Pharmacol. Sci. 2007, 104, 329–334. [Google Scholar] [CrossRef]
- Mert, D.G.; Turgut, N.H.; Arslanbas, E.; Gungor, H.; Kara, H. The influence of quercetin on recognition memory and brain oxidative damage in a ketamine model of schizophrenia. Psychiatry Clin. Psychopharmacol. 2018, 29, 1–7. [Google Scholar] [CrossRef] [Green Version]
- Cavaliere, G.; Trinchese, G.; Penna, E.; Cimmino, F.; Pirozzi, C.; Lama, A.; Annunziata, C.; Catapano, A.; Mattace Raso, G.; Meli, R.; et al. High-Fat Diet Induces Neuroinflammation and Mitochondrial Impairment in Mice Cerebral Cortex and Synaptic Fraction. Front. Cell. Neurosci. 2019, 13, 509. [Google Scholar] [CrossRef] [PubMed]
- Emerit, J.; Edeas, M.; Bricaire, F. Neurodegenerative diseases and oxidative stress. Biomed. Pharmacother. 2003, 58, 39–46. [Google Scholar] [CrossRef]
- Khoubnasabjafari, M.; Ansarin, K.; Jouyban, A. Reliability of malondialdehyde as a biomarker of oxidative stress in psychological disorders. BioImpacts 2015, 5, 123–127. [Google Scholar] [PubMed]
- Gumeni, S.; Papanagnou, E.-D.; Manola, M.S.; Trougakos, I.P. Nrf2 activation induces mitophagy and reverses Parkin/Pink1 knock down-mediated neuronal and muscle degeneration phenotypes. Cell Death Dis. 2021, 12, 671. [Google Scholar] [CrossRef] [PubMed]
- Butterfield, D.A.; Drake, J.; Pocernich, C.; Castegna, A. Evidence of oxidative damage in Alzheimer’s disease brain: Central role for amyloid β-peptide. Trends Mol. Med. 2001, 7, 548–554. [Google Scholar] [CrossRef]
- Yousof Ali, M.; Zaib, S.; Jannat, S.; Khan, I. Discovery of potent and selective dual cholinesterases and β-secretase inhibitors in pomegranate as a treatment for Alzheimer’s disease. Bioorganic Chem. 2022, 129, 106137. [Google Scholar] [CrossRef]
- Le, K.; Song, Z.; Deng, J.; Peng, X.; Zhang, J.; Wang, L.; Zhou, L.; Bi, H.; Liao, Z.; Feng, Z. Quercetin alleviates neonatal hypoxic-ischemic brain injury by inhibiting microglia-derived oxidative stress and TLR4-mediated inflammation. Agents Actions 2020, 69, 1201–1213. [Google Scholar] [CrossRef]
- Benameur, T.; Soleti, R.; Porro, C. The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases. Nutrients 2021, 13, 1318. [Google Scholar] [CrossRef]
- Yu, X.; Li, Y.; Mu, X. Effect of quercetin on PC12 Alzheimer’s disease cell model induced by Aβ25-35 and its mechanism based on sirtuin1/Nrf2/HO-1 pathway. BioMed Res. Int. 2020, 2020, 8210578. [Google Scholar] [CrossRef]
- Song, J.; Du, G.; Wu, H.; Gao, X.; Yang, Z.; Liu, B.; Cui, S. Protective effects of quercetin on traumatic brain injury induced inflammation and oxidative stress in cortex through activating Nrf2/HO-1 pathway. Restor. Neurol. Neurosci. 2021, 39, 73–84. [Google Scholar] [CrossRef] [PubMed]
- Saw, C.L.L.; Guo, Y.; Yang, A.Y.; Paredes-Gonzalez, X.; Ramirez, C.; Pung, D.; Kong, A.-N.T. The berry constituents quercetin, kaempferol, and pterostilbene synergistically attenuate reactive oxygen species: Involvement of the Nrf2-ARE signaling pathway. Food Chem. Toxicol. 2014, 72, 303–311. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, A.; Khan, M.M.; Hoda, N.; Raza, S.S.; Javed, H.; Ishrat, T.; Ashafaq, M.; Ahmad, E.; Safhi, M.M.; Islam, F. Quercetin Protects Against Oxidative Stress Associated Damages in a Rat Model of Transient Focal Cerebral Ischemia and Reperfusion. Neurochem. Res. 2011, 36, 1360–1371. [Google Scholar] [CrossRef]
- Jembrek, M.J.; Vlainić, J.; Čadež, V.; Šegota, S. Atomic force microscopy reveals new biophysical markers for monitoring subcellular changes in oxidative injury: Neuroprotective effects of quercetin at the nanoscale. PLoS ONE 2018, 13, e0200119. [Google Scholar] [CrossRef]
- Roshanzamir, F.; Yazdanparast, R. Quercetin attenuates cell apoptosis of oxidant-stressed SK-N-MC cells while suppressing up-regulation of the defensive element, HIF-1α. Neuroscience 2014, 277, 780–793. [Google Scholar] [CrossRef]
- Tanigawa, S.; Fujii, M.; Hou, D.-X. Stabilization of p53 Is Involved in Quercetin-Induced Cell Cycle Arrest and Apoptosis in HepG2 Cells. Biosci. Biotechnol. Biochem. 2008, 72, 797–804. [Google Scholar] [CrossRef]
- Agarwal, M.L.; Taylor, W.R.; Chernov, M.V.; Chernova, O.B.; Stark, G.R. The p53 Network. J. Biol. Chem. 1998, 273, 1–4. [Google Scholar] [CrossRef] [Green Version]
- Xue, W.; Zender, L.; Miething, C.; Dickins, R.A.; Hernando, E.; Krizhanovsky, V.; Cordon-Cardo, C.; Lowe, S.W. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 2007, 445, 656–660. [Google Scholar] [CrossRef] [Green Version]
- Sablina, A.A.; Budanov, A.V.; Ilyinskaya, G.V.; Agapova, L.S.; Kravchenko, J.E.; Chumakov, P. The antioxidant function of the p53 tumor suppressor. Nat. Med. 2005, 11, 1306–1313. [Google Scholar] [CrossRef] [Green Version]
- Hafner, A.; Bulyk, M.L.; Jambhekar, A.; Lahav, G. The multiple mechanisms that regulate p53 activity and cell fate. Nat. Rev. Mol. Cell Biol. 2019, 20, 199–210. [Google Scholar] [CrossRef]
- Sharma, D.; Wani, W.; Sunkaria, A.; Kandimalla, R.; Sharma, R.; Verma, D.; Bal, A.; Gill, K. Quercetin attenuates neuronal death against aluminum-induced neurodegeneration in the rat hippocampus. Neuroscience 2016, 324, 163–176. [Google Scholar] [CrossRef] [PubMed]
- Fridman, J.S.; Lowe, S.W. Control of apoptosis by p53. Oncogene 2003, 22, 9030–9040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Gu, S.; Ling, Y.; Shen, C.; Cao, X.; Xie, R. p53 inhibition provides a pivotal protective effect against ischemia-reperfusion injury in vitro via mTOR signaling. Brain Res. 2015, 1605, 31–38. [Google Scholar] [CrossRef]
- Wolff, S.; Erster, S.; Palacios, G.; Moll, U.M. p53’s mitochondrial translocation and MOMP action is independent of Puma and Bax and severely disrupts mitochondrial membrane integrity. Cell Res. 2008, 18, 733–744. [Google Scholar] [CrossRef] [Green Version]
- Sun, J.; Wang, F.; Li, H.; Zhang, H.; Jin, J.; Chen, W.; Pang, M.; Yu, J.; He, Y.; Liu, J.; et al. Neuroprotective Effect of Sodium Butyrate against Cerebral Ischemia/Reperfusion Injury in Mice. BioMed Res. Int. 2015, 2015, 395895. [Google Scholar] [CrossRef] [Green Version]
- Maor-Nof, M.; Shipony, Z.; Lopez-Gonzalez, R.; Nakayama, L.; Zhang, Y.-J.; Couthouis, J.; Blum, J.A.; Castruita, P.A.; Linares, G.R.; Ruan, K.; et al. p53 is a central regulator driving neurodegeneration caused by C9orf72 poly(PR). Cell 2021, 184, 689–708.e20. [Google Scholar] [CrossRef] [PubMed]
- Bayazid, A.B.; Kim, J.G.; Azam, S.; Jeong, S.A.; Kim, D.H.; Park, C.W.; Lim, B.O. Sodium butyrate ameliorates neurotoxicity and exerts anti-inflammatory effects in high fat diet-fed mice. Food Chem. Toxicol. 2021, 159, 112743. [Google Scholar] [CrossRef]
- Wu, H.; Wang, Y.; Zhang, Y.; Xu, F.; Chen, J.; Duan, L.; Zhang, T.; Wang, J.; Zhang, F. Breaking the vicious loop between inflammation, oxidative stress and coagulation, a novel anti-thrombus insight of nattokinase by inhibiting LPS-induced inflammation and oxidative stress. Redox Biol. 2020, 32, 101500. [Google Scholar] [CrossRef]
- Xu, J.; Gao, H.; Zhang, L.; Rong, S.; Yang, W.; Ma, C.; Chen, M.; Huang, Q.; Deng, Q.; Huang, F. Melatonin alleviates cognition impairment by antagonizing brain insulin resistance in aged rats fed a high-fat diet. J. Pineal Res. 2019, 67, e12584. [Google Scholar] [CrossRef]
- Bayazid, A.B.; Jeong, S.A.; Park, C.W.; Kim, D.H.; Lim, B.O. The Anti-Inflammatory Activities of Fermented Curcuma That Contains Butyrate Mitigate DSS-Induced Colitis in Mice. Molecules 2022, 27, 4745. [Google Scholar] [CrossRef]
- Lee, B.; Yeom, M.; Shim, I.; Lee, H.; Hahm, D.-H. Protective Effects of Quercetin on Anxiety-Like Symptoms and Neuroinflammation Induced by Lipopolysaccharide in Rats. Evidence-Based Complement. Altern. Med. 2020, 2020, 4892415. [Google Scholar] [CrossRef] [PubMed]
- Wu, M.; Liu, F.; Guo, Q. Quercetin attenuates hypoxia-ischemia induced brain injury in neonatal rats by inhibiting TLR4/NF-κB signaling pathway. Int. Immunopharmacol. 2019, 74, 105704. [Google Scholar] [CrossRef]
- Ashrafizadeh, M.; Ahmadi, Z.; Farkhondeh, T.; Samarghandian, S. Autophagy as a molecular target of quercetin underlying its protective effects in human diseases. Arch. Physiol. Biochem. 2019, 128, 200–208. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Xu, T.; Fang, Q.; Zhang, H.; Yue, L.; Hu, G.; Sun, L. Quercetin hinders microglial activation to alleviate neurotoxicity via the interplay between NLRP3 inflammasome and mitophagy. Redox Biol. 2021, 44, 102010. [Google Scholar] [CrossRef]
- Crews, L.; Masliah, E. Molecular mechanisms of neurodegeneration in Alzheimer’s disease. Hum. Mol. Genet. 2010, 19, R12–R20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- By, S. World Alzheimer Report 2013; Alzheimer’s Disease International (ADI): London, UK, 2013. [Google Scholar]
- Wei, W.; Nguyen, L.N.; Kessels, H.; Hagiwara, H.; Sisodia, S.; Malinow, R. Amyloid beta from axons and dendrites reduces local spine number and plasticity. Nat. Neurosci. 2009, 13, 190–196. [Google Scholar] [CrossRef]
- Stoothoff, W.H.; Johnson, G.V. Tau phosphorylation: Physiological and pathological consequences. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2005, 1739, 280–297. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Deng, X.; Liu, N.; Li, M.; Liu, B.; Fu, Q.; Qu, R.; Ma, S. Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of ER stress in a manner dependent on AMPK pathway. J. Funct. Foods 2016, 22, 463–476. [Google Scholar] [CrossRef]
- Suganthy, N.; Devi, K.P.; Nabavi, S.F.; Braidy, N. Bioactive effects of quercetin in the central nervous system: Focusing on the mechanisms of actions. Biomed. Pharmacother. 2016, 84, 892–908. [Google Scholar] [CrossRef]
- Ku, T.; Li, B.; Gao, R.; Zhang, Y.; Yan, W.; Ji, X.; Sang, N. NF-κB-regulated microRNA-574-5p underlies synaptic and cognitive impairment in response to atmospheric PM2.5 aspiration. Part. Fibre Toxicol. 2017, 14, 34. [Google Scholar] [CrossRef]
- Pang, K.; Jiang, R.; Zhang, W.; Yang, Z.; Li, L.L.; Shimozawa, M.; Lu, B. An App knock-in rat model for Alzheimer’s disease exhibiting Aβ and tau pathologies, neuronal death and cognitive impairments. Cell Res. 2022, 32, 157–175. [Google Scholar] [CrossRef] [PubMed]
- Zheng, H.; Koo, E.H. The amyloid precursor protein: Beyond amyloid. Mol. Neurodegener. 2006, 1, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, M.; Huang, H.; Mo, X.; Zhu, Y.; Chen, X.; Li, X.; Liu, L. Quercetin-3-O-Glucuronide Alleviates Cognitive Deficit and Toxicity in Aβ1-42-Induced AD-Like Mice and SH-SY5Y Cells. Mol. Nutr. Food Res. 2021, 65, 2000660. [Google Scholar] [CrossRef] [PubMed]
- Regitz, C.; Dußling, L.M.; Wenzel, U. Amyloid-beta (Aβ1–42)-induced paralysis in Caenorhabditis elegans is inhibited by the polyphenol quercetin through activation of protein degradation pathways. Mol. Nutr. Food Res. 2014, 58, 1931–1940. [Google Scholar] [CrossRef]
- Ansari, M.A.; Abdul, H.M.; Joshi, G.; Opii, W.O.; Butterfield, D.A. Protective effect of quercetin in primary neurons against Aβ(1–42): Relevance to Alzheimer’s disease. J. Nutr. Biochem. 2009, 20, 269–275. [Google Scholar] [CrossRef] [Green Version]
- Eremenko, E.; Mittal, K.; Berner, O.; Kamenetsky, N.; Nemirovsky, A.; Elyahu, Y.; Monsonego, A. BDNF-producing, amyloid β-specific CD4 T cells as targeted drug-delivery vehicles in Alzheimer’s disease. EBioMedicine 2019, 43, 424–434. [Google Scholar] [CrossRef] [PubMed]
- Yao, R.-Q.; Qi, D.-S.; Yu, H.-L.; Liu, J.; Yang, L.-H.; Wu, X.-X. Quercetin Attenuates Cell Apoptosis in Focal Cerebral Ischemia Rat Brain Via Activation of BDNF–TrkB–PI3K/Akt Signaling Pathway. Neurochem. Res. 2012, 37, 2777–2786. [Google Scholar] [CrossRef]
- Arias, N.; Macarulla, M.T.; Aguirre, L.; Martínez-Castaño, M.G.; Portillo, M.P. Quercetin can reduce insulin resistance without decreasing adipose tissue and skeletal muscle fat accumulation. Genes Nutr. 2013, 9, 361. [Google Scholar] [CrossRef] [Green Version]
- Osama, A.; Zhang, J.; Yao, J.; Yao, X.; Fang, J. Nrf2: A dark horse in Alzheimer’s disease treatment. Ageing Res. Rev. 2020, 64, 101206. [Google Scholar] [CrossRef]
- Bettens, K.; Sleegers, K.; van Broeckhoven, C. Genetic insights in Alzheimer’s disease. Lancet Neurol. 2013, 12, 92–104. [Google Scholar] [CrossRef]
- Association, A.S. 2019 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2019, 15, 321–387. [Google Scholar] [CrossRef]
- Liu, L.; MacKenzie, K.R.; Putluri, N.; Maletić-Savatić, M.; Bellen, H.J. The Glia-Neuron Lactate Shuttle and Elevated ROS Promote Lipid Synthesis in Neurons and Lipid Droplet Accumulation in Glia via APOE/D. Cell Metab. 2017, 26, 719–737.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boesch-Saadatmandi, C.; Wolffram, S.; Minihane, A.M.; Rimbach, G. Effect of apoE genotype and dietary quercetin on blood lipids and TNF-α levels in apoE3 and apoE4 targeted gene replacement mice. Br. J. Nutr. 2008, 101, 1440–1443. [Google Scholar] [CrossRef] [PubMed]
- Plump, A.S.; Breslow, J.L. Apolipoprotein E and the apolipoprotein E-deficient mouse. Annu. Rev. Nutr. 1995, 15, 495–518. [Google Scholar] [CrossRef]
- Liu, C.-C.; Kanekiyo, T.; Xu, H.; Bu, G. Apolipoprotein E and Alzheimer disease: Risk, mechanisms and therapy. Nat. Rev. Neurol. 2013, 9, 106–118, Correction in Nat. Rev. Neurol. 2013, 9, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rhinn, H.; Fujita, R.; Qiang, L.; Cheng, R.; Lee, J.H.; Abeliovich, A. Integrative genomics identifies APOE ε4 effectors in Alzheimer’s disease. Nature 2013, 500, 45–50. [Google Scholar]
- Bellenguez, C.; Küçükali, F.; Jansen, I.E.; Kleineidam, L.; Moreno-Grau, S.; Amin, N.; Naj, A.C.; Campos-Martin, R.; Grenier-Boley, B.; Andrade, V.; et al. New insights into the genetic etiology of Alzheimer’s disease and related dementias. Nat. Genet. 2022, 54, 412–436. [Google Scholar] [CrossRef]
- Tachibana, M.; Holm, M.-L.; Liu, C.-C.; Shinohara, M.; Aikawa, T.; Oue, H.; Yamazaki, Y.; Martens, Y.A.; Murray, M.E.; Sullivan, P.M.; et al. APOE4-mediated amyloid-β pathology depends on its neuronal receptor LRP1. J. Clin. Investig. 2019, 129, 1272–1277. [Google Scholar] [CrossRef] [Green Version]
- Carmona, S.; Zahs, K.; Wu, E.; Dakin, K.; Bras, J.; Guerreiro, R. The role of TREM2 in Alzheimer’s disease and other neurodegenerative disorders. Lancet Neurol. 2018, 17, 721–730. [Google Scholar]
- Lee, J.-H.; Yang, D.S.; Goulbourne, C.N.; Im, E.; Stavrides, P.; Pensalfini, A.; Nixon, R.A. 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]
- Wadhwani, A.R.; Affaneh, A.; Van Gulden, S.; Kessler, J.A. Neuronal apolipoprotein E4 increases cell death and phosphorylated tau release in alzheimer disease. Ann. Neurol. 2019, 85, 726–739. [Google Scholar] [CrossRef] [PubMed]
- Zheng, X.J.; Chen, W.L.; Yi, J.; Li, W.; Liu, J.Y.; Fu, W.Q.; Wang, J.H. Apolipoprotein C1 promotes glioblastoma tumorigenesis by reducing KEAP1/NRF2 and CBS-regulated ferroptosis. Acta Pharmacol. Sin. 2022, 43, 2977–2992. [Google Scholar] [CrossRef]
- He, K.; Nie, L.; Zhou, Q.; Rahman, S.U.; Liu, J.; Yang, X.; Li, S. Proteomic Profiles of the Early Mitochondrial Changes in APP/PS1 and ApoE4 Transgenic Mice Models of Alzheimer’s Disease. J. Proteome Res. 2019, 18, 2632–2642. [Google Scholar] [CrossRef] [PubMed]
- Zhu, M.; Han, S.; Fink, A.L. Oxidized quercetin inhibits α-synuclein fibrillization. Biochim. Biophys. Acta (BBA)-Gen. Subj. 2013, 1830, 2872–2881. [Google Scholar] [CrossRef] [PubMed]
- Stefanis, L. α-Synuclein in Parkinson’s disease. Cold Spring Harb. Perspect. Med. 2012, 2, a009399. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.-W.; Han, R.; He, H.J.; Li, J.; Chen, S.Y.; Gu, Y.; Xie, C. Administration of quercetin improves mitochondria quality control and protects the neurons in 6-OHDA-lesioned Parkinson’s disease models. Aging 2021, 13, 11738. [Google Scholar] [CrossRef]
- Das, N.; Sharma, S. Peroxisome Proliferator Activated Receptor Gamma Coactivator 1 Alpha: An Emerging Target for Neuroprotection in Parkinson’s Disease. CNS Neurol. Disord-Drug Targets 2015, 14, 1024–1030. [Google Scholar] [CrossRef]
- Xiao, N.; Mei, F.; Sun, Y.; Pan, G.; Liu, B.; Liu, K. Quercetin, Luteolin, and Epigallocatechin Gallate Promote Glucose Disposal in Adipocytes with Regulation of AMP-Activated Kinase and/or Sirtuin 1 Activity. Planta Medica 2014, 80, 993–1000. [Google Scholar] [CrossRef] [Green Version]
- Gonzales, M.M.; Garbarino, V.R.; Marques Zilli, E.; Petersen, R.C.; Kirkland, J.L.; Tchkonia, T.; Orr, M.E. Senolytic Therapy to Modulate the Progression of Alzheimer’s Disease (SToMP-AD): A Pilot Clinical Trial. J. Prev. Alzheimer’s Dis. 2022, 9, 22–29. [Google Scholar] [CrossRef]
- Nishihira, J.; Nishimura, M.; Kurimoto, M.; Kagami-Katsuyama, H.; Hattori, H.; Nakagawa, T.; Muro, T.; Kobori, M. The effect of 24-week continuous intake of quercetin-rich onion on age-related cognitive decline in healthy elderly people: A randomized, double-blind, placebo-controlled, parallel-group comparative clinical trial. J. Clin. Biochem. Nutr. 2021, 69, 203–215. [Google Scholar] [CrossRef]
- Rinwa, P.; Kumar, A. Quercetin along with piperine prevents cognitive dysfunction, oxidative stress and neuro-inflammation associated with mouse model of chronic unpredictable stress. Arch. Pharmacal Res. 2013, 40, 1166–1175. [Google Scholar] [CrossRef] [PubMed]
- Amanzadeh, E.; Esmaeili, A.; Rahgozar, S.; Nourbakhshnia, M. Application of quercetin in neurological disorders: From nutrition to nanomedicine. Rev. Neurosci. 2019, 30, 555–572. [Google Scholar] [CrossRef] [PubMed]
- Collaboration, B.P.L.T.T. Effects of different blood-pressure-lowering regimens on major cardiovascular events: Results of prospectively-designed overviews of randomised trials. Lancet 2003, 362, 1527–1535. [Google Scholar]
- Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.F.; Flamm, G.W.; Williams, G.M.; Lines, T.C. A critical review of the data related to the safety of quercetin and lack of evidence of in vivo toxicity, including lack of genotoxic/carcinogenic properties. Food Chem. Toxicol. 2007, 45, 2179–2205. [Google Scholar] [CrossRef] [PubMed]
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
Bayazid, A.B.; Lim, B.O. Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1. Nutrients 2022, 14, 5132. https://doi.org/10.3390/nu14235132
Bayazid AB, Lim BO. Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1. Nutrients. 2022; 14(23):5132. https://doi.org/10.3390/nu14235132
Chicago/Turabian StyleBayazid, Al Borhan, and Beong Ou Lim. 2022. "Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1" Nutrients 14, no. 23: 5132. https://doi.org/10.3390/nu14235132
APA StyleBayazid, A. B., & Lim, B. O. (2022). Quercetin Is An Active Agent in Berries against Neurodegenerative Diseases Progression through Modulation of Nrf2/HO1. Nutrients, 14(23), 5132. https://doi.org/10.3390/nu14235132