The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases
Abstract
:1. Introduction
2. Quercetin and Its Dietary Sources
3. Broad Mechanisms of Action of Quercetin
4. Glial Cells and Quercetin-Induced Neuroprotection
5. Quercetin and microRNA
6. Strategies to Improve Quercetin Effectiveness in Neurodegeneration: Synthetic and Natural Carriers
7. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Suganthy, N.; Devi, K.P.; Nabavi, S.F.; Braidy, N.; Nabavi, S.M. Bioactive effects of quercetin in the central nervous system: Focusing on the mechanisms of actions. Biomed. Pharmacother. 2016, 84, 892–908. [Google Scholar] [CrossRef] [PubMed]
- Martel, J.; Ojcius, D.M.; Ko, Y.-F.; Young, J.D. Phytochemicals as Prebiotics and Biological Stress Inducers. Trends Biochem. Sci. 2020, 45, 462–471. [Google Scholar] [CrossRef]
- Lakhanpal, P.; Rai, D.K. Quercetin: A Versatile Flavonoid. IJMU 2007, 2, 22–37. [Google Scholar] [CrossRef] [Green Version]
- Derosa, G.; Maffioli, P.; D’Angelo, A.; Di Pierro, F. A role for quercetin in coronavirus disease 2019 (COVID-19). Phytotherapy Res. 2021, 35, 1230–1236. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Yao, J.; Han, C.; Yang, J.; Chaudhry, M.T.; Wang, S.; Liu, H.; Yin, Y. Quercetin, in-flammation and immunity. Nutrients 2016, 8, 167. [Google Scholar] [CrossRef] [PubMed]
- Shankar, G.M.; Antony, J.; Anto, R.J. Quercetin and tryptanthrin: Two broad spectrum anti-cancer agents for future chemotherapeutic interventions. Enzymes 2015, 37, 43–72. [Google Scholar]
- Rothwell, J.A.; Perez-Jimenez, J.; Neveu, V.; Medina-Remón, A.; M’hiri, N.; García-Lobato, P.; Manach, C.; Knox, C.; Eisner, R.; Wishart, D.S.; et al. Phenol-Explorer 3.0: A major up-date of the Phenol-Explorer database to incorporate data on the effects of food processing on pol-yphenol content. Database 2013, 2013, bat070. [Google Scholar] [CrossRef]
- Kawabata, K.; Mukai, R.; Ishisaka, A. Quercetin and related polyphenols: New insights and implications for their bioactivity and bioavailability. Food Funct. 2015, 6, 1399–1417. [Google Scholar] [CrossRef]
- García-Barrado, M.J.; Iglesias-Osma, M.C.; Pérez-García, E.; Carrero, S.; Blanco, E.J.; Carretero-Hernández, M.; Carretero, J. Role of Flavonoids in the Interactions among Obesity, Inflammation, and Autophagy. Pharmaceuticals 2020, 13, 342. [Google Scholar] [CrossRef]
- Manach, C.; Regerat, F.; Texier, O.; Agullo, G.; Demigne, C.; Remesy, C. Bioavailability, metabolism and physiological impact of 4-oxo-flavonoids. Nutr. Res. 1996, 16, 517–544. [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]
- Almeida, A.F.; Borge, G.I.A.; Piskula, M.; Tudose, A.; Tudoreanu, L.; Valentová, K.; Williamson, G.; Santos, C.N. Bioavailability of Quercetin in Humans with a Focus on Interindividual Variation. Compr. Rev. Food Sci. Food Saf. 2018, 17, 714–731. [Google Scholar] [CrossRef] [Green Version]
- Nabavi, S.F.; Russo, G.L.; Daglia, M. Role of quercetin as an alternative for obesity treatment: You are what you eat! Food Chem. 2015, 179, 305–310. [Google Scholar] [CrossRef]
- Andres, S.; Pevny, S.; Ziegenhagen, R.; Bakhiya, N.; Schäfer, B.; Hirsch-Ernst, K.I.; Lampen, A. Safety Aspects of the Use of Quercetin as a Dietary Supplement. Mol. Nutr. Food Res. 2018, 62. [Google Scholar] [CrossRef]
- Dajas, F.; Rivera-Megret, F.; Blasina, F.; Arredondo, F.; Abin-Carriquiry, J.; Costa, G.; Echeverry, C.; Lafon, L.; Heizen, H.; Ferreira, M.; et al. Neuroprotection by flavonoids. Braz. J. Med Biol. Res. 2003, 36, 1613–1620. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Swarts, S.G.; Yin, L.; Liu, C.; Tian, Y.; Cao, Y.; Swarts, M.; Yang, S.; Zhang, S.B.; Zhang, K.; et al. Anti-Oxidant Properties of Quercetin. In Oxygen Transport to Tissue XXXII. Advances in Experimental Medicine and Biology; LaManna, J., Puchowicz, M., Xu, K., Harrison, D., Bruley, D., Eds.; Springer: Boston, MA, USA, 2011; Volume 701. [Google Scholar]
- Wang, W.; Sun, C.; Mao, L.; Ma, P.; Liu, F.; Yang, J.; Gao, Y. The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci. Technol. 2016, 56, 21–38. [Google Scholar] [CrossRef]
- Harwood, M.; Danielewska-Nikiel, B.; Borzelleca, J.; Flamm, G.; Williams, G.; Lines, T. 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]
- Serban, M.C.; Sahebkar, A.; Zanchetti, A.; Mikhailidis, D.P.; Howard, G.; Antal, D.; Andrica, F.; Ahmed, A.; Aronow, W.S.; Muntner, P.; et al. Effects of Quercetin on Blood Pressure: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Am. Heart Assoc. 2016, 5. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Batiha, G.E.-S.; Beshbishy, A.M.; Ikram, M.; Mulla, Z.S.; El-Hack, M.E.A.; Taha, A.E.; Al-gammal, A.M.; Elewa, Y.H.A. The Pharmacological Activity, Biochemical Properties, and Phar-macokinetics of the Major Natural Polyphenolic Flavonoid: Quercetin. Foods 2020, 9, 374. [Google Scholar] [CrossRef] [Green Version]
- Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxidative Med. Cell. Longev. 2016, 2016, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Zhai, S.-W.; Liu, S.-L. Effects of Dietary Quercetin on Growth Performance, Serum Lipids Level and Body Composition of Tilapia (Oreochromis Niloticus). Ital. J. Anim. Sci. 2013, 12, e85. [Google Scholar] [CrossRef] [Green Version]
- Zhai, S.W.; Liu, S.L. Effects of dietary quercetin on the growth performance, digestive en-zymes and antioxidant potential in the hepatopancreas of tilapia (Oreochromis niloticus). Isr. J. Aquacult. Bamid. 2014, 66, 1–7. [Google Scholar]
- Kuipers, E.N.; Van Dam, A.D.; Held, N.M.; Mol, I.M.; Houtkooper, R.H.; Rensen, P.C.; Boon, M.R. Quercetin Lowers Plasma Triglycerides Accompanied by White Adipose Tissue Browning in Diet-Induced Obese Mice. Int. J. Mol. Sci. 2018, 19, 1786. [Google Scholar] [CrossRef] [Green Version]
- Wang, M.; Xiao, F.L.; Mao, Y.J.; Ying, L.L.; Zhou, B.; Li, Y. Quercetin decreases the triglycer-ide content through the PPAR signalling pathway in primary hepatocytes of broiler chickens. Biotechnol. Biotechnol. Equip. 2019, 33, 1000–1010. [Google Scholar] [CrossRef] [Green Version]
- Schadich, E.; Hlaváč, J.; Volná, T.; Varanasi, L.; Hajdúch, M.; Džubák, P. Effects of Ginger Phenylpropanoids and Quercetin on Nrf2-ARE Pathway in Human BJ Fibroblasts and HaCaT Keratinocytes. BioMed Res. Int. 2016, 2016, 1–6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, X.; Yu, Z.; Huang, X.; Gao, Y.; Wang, X.; Gu, J.; Xue, S. Peroxisome proliferator- acti-vated receptor γ (PPARγ) mediates the protective effect of quercetin against myocardial ische-mia-reperfusion injury via suppressing the NF-κB pathway. Am. J. Transl. Res. 2016, 8, 5169–5186. [Google Scholar]
- Ay, M.; Luo, J.; Langley, M.; Jin, H.; Anantharam, V.; Kanthasamy, A.; Kanthasamy, A.G. Molecular mechanisms underlying protective effects of quercetin against mitochondrial dysfunc-tion and progressive dopaminergic neurodegeneration in cell culture and MitoPark transgenic mouse models of Parkinson’s Disease. J. Neurochem. 2017, 141, 766–782. [Google Scholar] [CrossRef] [PubMed]
- Khan, H.; Ullah, H.; Aschner, M.; Cheang, W.S.; Akkol, E.K. Neuroprotective Effects of Quercetin in Alzheimer’s Disease. Biomolecules 2019, 10, 59. [Google Scholar] [CrossRef] [Green Version]
- Jeong, E.; Lee, J.Y. Intrinsic and Extrinsic Regulation of Innate Immune Receptors. Yonsei Med. J. 2011, 52, 379–392. [Google Scholar] [CrossRef] [Green Version]
- D’Andrea, G. Quercetin: A flavonol with multifaceted therapeutic applications? Fitoterapia 2015, 106, 256–271. [Google Scholar] [CrossRef]
- Sundaram, M.K.; Raina, R.; Afroze, N.; Bajbouj, K.; Hamad, M.; Haque, S.; Hussain, A. Quercetin modulates signaling pathways and induces apoptosis in cervical cancer cells. Biosci. Rep. 2019, 39, BSR20190720. [Google Scholar] [CrossRef] [Green Version]
- Zhang, F.; Feng, J.; Zhang, J.; Kang, X.; Qian, D. Quercetin modulates AMPK/SIRT1/NF‑κB signaling to inhibit inflammatory/oxidative stress responses in diabetic high fat diet‑induced atherosclerosis in the rat carotid artery. Exp. Ther. Med. 2020, 20, 1. [Google Scholar] [CrossRef]
- Hu, T.; Shi, J.J.; Fang, J.; Wang, Q.; Chen, Y.B.; Zhang, S.J. Quercetin ameliorates diabetic encephalopathy through SIRT1/ER stress pathway in db/db mice. Aging 2020, 12, 7015–7029. [Google Scholar] [CrossRef]
- Martins-Perles, J.V.C.; Bossolani, G.D.P.; Zignani, I.; de Souza, S.R.G.; Frez, F.C.V.; de Souza Melo, C.G.; Barili, E.; de Souza Neto, F.P.; Guarnier, F.A.; Armani, A.L.C.; et al. Quercetin increases bioavailability of nitric oxide in the jejunum of euglycemic and diabetic rats and induces neuronal plasticity in the myenteric plexus. Auton. Neurosci. 2020, 227, 102675. [Google Scholar] [CrossRef]
- Dong, F.; Wang, S.; Wang, Y.; Yang, X.; Jiang, J.; Wu, D.; Qu, X.; Fan, H.; Yao, R. Quercetin ameliorates learning and memory via the Nrf2-ARE signaling pathway in d-galactose-induced neurotoxicity in mice. Biochem. Biophys. Res. Commun. 2017, 491, 636–641. [Google Scholar] [CrossRef] [PubMed]
- Zaplatic, E.; Bule, M.; Shah, S.Z.A.; Uddin, M.S.; Niaz, K. Molecular mechanisms un-derlying protective role of quercetin in attenuating Alzheimer’s disease. Life Sci. 2019, 224, 109–119. [Google Scholar] [CrossRef] [PubMed]
- Kyaw, M.; Yoshizumi, M.; Tsuchiya, K.; Izawa, Y.; Kanematsu, Y.; Tamaki, T. Atheroprotective effects of antioxidants through inhibition of mitogen-activated protein kinases. Acta Pharmacol. Sin. 2004, 25, 977–985. [Google Scholar]
- Gregor, M.F.; Hotamisligil, G.S. Inflammatory Mechanisms in Obesity. Annu. Rev. Immunol. 2011, 29, 415–445. [Google Scholar] [CrossRef] [Green Version]
- Hotamisligil, G.S. Inflammation, metaflammation and immunometabolic disorders. Nature 2017, 542, 177–185. [Google Scholar] [CrossRef]
- Rogero, M.M.; Calder, P.C. Obesity, Inflammation, Toll-Like Receptor 4 and Fatty Acids. Nutrients 2018, 10, 432. [Google Scholar] [CrossRef] [Green Version]
- Schnare, M.; Barton, G.M.; Holt, A.C.; Takeda, K.; Akira, S.; Medzhitov, R. Toll-like receptors control activation of adaptive immune responses. Nat. Immunol. 2001, 2, 947–950. [Google Scholar] [CrossRef] [PubMed]
- Jagannathan, M.; Hasturk, H.; Liang, Y.; Shin, H.; Hetzel, J.T.; Kantarci, A.; Rubin, D.; McDonnell, M.E.; Van Dyke, T.E.; Ganley-Leal, L.M.; et al. TLR Cross-Talk Specifically Regulates Cytokine Production by B Cells from Chronic Inflammatory Disease Patients. J. Immunol. 2009, 183, 7461–7470. [Google Scholar] [CrossRef] [Green Version]
- Kawai, T.; Akira, S. Signaling to NF-kappaB by toll-like receptors. Trends Mol Med. 2007, 13, 460–469. [Google Scholar] [CrossRef] [PubMed]
- Panaro, M.A.; Corrado, A.; Benameur, T.; Cantatore, F.P.; Cici, D.; Porro, C. The emerging role of curcumin in the modulation of TLR-4 signaling pathway: Focus on neuroprotective and an-ti-rheumatic properties. Int. J. Mol. Sci. 2020, 21, 2299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, M.; Liu, F.; Guo, Q. Quercetin attenuates hypoxia-ischemia induced brain injury in ne-onatal rats by inhibiting TLR4/NF-κB signaling pathway. Int. Immunopharmacol. 2019, 74, 105704. [Google Scholar] [CrossRef] [PubMed]
- Cox, L.M.; Weiner, H.L. Microbiota signaling pathways that influence neurologic dis-ease. Neurotherapeutics 2018, 15, 135–145. [Google Scholar] [CrossRef] [Green Version]
- Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed. Pharmacother. 2020, 127, 110147. [Google Scholar] [CrossRef]
- Ma, Q. Role of Nrf2 in Oxidative Stress and Toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [Green Version]
- Vomhof-Dekrey, E.E.; Picklo Sr, M.J. The Nrf2-antioxidant response element pathway: A target for regulating energy metabolism. J. Nutr. Biochem. 2012, 23, 1201–1206. [Google Scholar] [CrossRef]
- Nguyen, T.; Nioi, P.; Pickett, C.B. The Nrf2-antioxidant response element signaling pathway and its activation by oxidative stress. J. Biol. Chem. 2009, 284, 13291–13295. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Gan, L.; Johnson, J.A. Oxidative damage and the Nrf2-ARE pathway in neurodegenerative diseases. Biochim. Biophys. Acta 2014, 1842, 1208–1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin Cell Dev. Biol. 2019, 94, 112–120. [Google Scholar] [CrossRef] [PubMed]
- Clarke, L.E.; Barres, B.A. Emerging roles of astrocytes in neural circuit development. Nat. Rev. Neurosci. 2013, 14, 311–321. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, L.; Miranda-Saksena, M.; Saksena, N.K. Viruses and neurodegeneration. Virol. J. 2013, 10, 172. [Google Scholar] [CrossRef] [Green Version]
- Gay, N.J.; Symmons, M.F.; Gangloff, M.; Bryant, C.E. Assembly and localization of Toll-like receptor signalling complexes. Nat. Rev. Immunol. 2014, 14, 546–558. [Google Scholar] [CrossRef]
- Chen, W.W.; Zhang, X.; Huang, W.J. Role of neuroinflammation in neurodegenerative diseases. Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef] [Green Version]
- Aïd, S.; Bosetti, F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie 2011, 93, 46–51. [Google Scholar] [CrossRef] [Green Version]
- Baufeld, C.; O’Loughlin, E.; Calcagno, N.; Madore, C.; Butovsky, O. Differential contribu-tion of microglia and monocytes in neurodegenerative diseases. J. Neural Transm. 2018, 125, 809–826. [Google Scholar] [CrossRef]
- Lawson, L.J.; Perry, V.H.; Dri, P.; Gordon, S. Heterogeneity in the distribution and morphol-ogy of microglia in the normal adult mouse brain. Neuroscience 1990, 39, 151–170. [Google Scholar] [CrossRef]
- Cherry, J.D.; Olschowka, J.A.; O’Banion, M.K. Neuroinflammation and M2 microglia: The good, the bad, and the inflamed. J. Neuroinflammation 2014, 11, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ransohoff, R.M. A polarizing question: Do M1 and M2 microglia exist? Nat. Neurosci. 2016, 19, 987–991. [Google Scholar] [CrossRef] [PubMed]
- Cianciulli, A.; Calvello, R.; Porro, C.; Trotta, T.; Panaro, M.A. Understanding the role of SOCS signaling in neurodegenerative diseases: Current and emerging concepts. Cytokine Growth Factor Rev. 2017, 37, 67–79. [Google Scholar] [CrossRef]
- Tang, Y.; Le, W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef]
- González, H.; Elgueta, D.; Montoya, A.; Pacheco, R. Neuroimmune regulation of microglial activity involved in neuroinflammation and neurodegenerative diseases. J. Neuroimmunol. 2014, 274, 1–13. [Google Scholar] [CrossRef]
- Panaro, M.A.; Lofrumento, D.D.; Saponaro, C.; De Nuccio, F.; Cianciulli, A.; Mitolo, V.; Nicolardi, G. Expression of TLR4 and CD14 in the Central Nervous System (CNS) in a MPTP Mouse Model of Parkinson’s-Like Disease. Immunopharmacol. Immunotoxicol. 2008, 30, 729–740. [Google Scholar] [CrossRef]
- Querfurth, H.W.; LaFerla, F.M. Alzheimer’s disease. N. Engl. J. Med. 2010, 362, 329–344. [Google Scholar] [CrossRef] [Green Version]
- Colombo, E.; Farina, C. Astrocytes: Key Regulators of Neuroinflammation. Trends Immunol. 2016, 37, 608–620. [Google Scholar] [CrossRef]
- Oksanen, M.; Lehtonen, S.; Jaronen, M.; Goldsteins, G.; Hamalainen, R.H.; Koistinaho, J. Astrocyte alterations in neurodegenerative pathologies and their modeling in human induced pluripotent stem cell platforms. Cell Mol. Life Sci. 2019, 76, 2739–2760. [Google Scholar] [CrossRef] [Green Version]
- Sofroniew, M.V. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009, 32, 638–647. [Google Scholar] [CrossRef] [Green Version]
- MacVicar, B.A.; Newman, E.A. Astrocyte regulation of blood flow in the brain. Cold Spring Harb. Perspect. Biol. 2015, 7, a020388. [Google Scholar] [CrossRef] [PubMed]
- Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of mi-croglia and astrocytes. Transl. Neurodegener 2020, 9, 42. [Google Scholar] [CrossRef] [PubMed]
- Hickman, S.; Izzy, S.; Sen, P.; Morsett, L.; El Khoury, J. Microglia in neurodegeneration. Nat. Neurosci. 2018, 21, 1359–1369. [Google Scholar] [CrossRef] [PubMed]
- Cianciulli, A.; Porro, C.; Calvello, R.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Microglia Mediated Neuroinflammation: Focus on PI3K Modulation. Biomolecules 2020, 10, 137. [Google Scholar] [CrossRef] [Green Version]
- Porro, C.; Cianciulli, A.; Trotta, T.; Lofrumento, D.D.; Panaro, M.A. Curcumin Regulates Anti-Inflammatory Responses by JAK/STAT/SOCS Signaling Pathway in BV-2 Microglial Cells. Biology 2019, 8, 51. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.J.; Cheang, L.C.V.; Wang, M.W.; Lee, S.M.Y. Quercetin exerts a neuroprotective effect through inhibition of the iNOS/NO system and pro-inflammation gene expression in PC12 cells and in zebrafish. Int. J. Mol. Med. 2011, 27, 195–203. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.C.; Ho, F.M.; Chao, P.D.L.; Chen, C.P.; Jeng, K.C.G.; Hsu, H.B.; Lee, S.T.; Wu, W.T.; Lin, W.W. Inhibition of iNOS gene expression by quercetin is mediated by the inhibition of I(B ki-nase, nuclear factor-kappa B and STAT1, and depends on heme oxygenase-1 induction in mouse BV-2 microglia. Eur. J. Pharmacol. 2005, 521, 9–20. [Google Scholar] [CrossRef]
- Sun, G.Y.; Chen, Z.; Jasmer, K.J.; Chuang, D.Y.; Gu, Z.; Hannink, M.; Simonyi, A. Quercetin Attenuates Inflammatory Responses in BV-2 Microglial Cells: Role of MAPKs on the Nrf2 Pathway and Induction of Heme Oxygenase-1. PLoS ONE 2015, 10, e0141509. [Google Scholar] [CrossRef] [Green Version]
- Bureau, G.; Longpré, F.; Martinoli, M.-G. Resveratrol and quercetin, two natural polyphenols, reduce apoptotic neuronal cell death induced by neuroinflammation. J. Neurosci. Res. 2008, 86, 403–410. [Google Scholar] [CrossRef]
- Fan, H.; Tang, H.B.; Shan, L.Q.; Liu, S.C.; Huang, D.G.; Chen, X.; Chen, Z.; Yang, M.; Yin, X.H.; Yang, H.; et al. Quercetin prevents necroptosis of oligodendrocytes by inhibiting mac-rophages/microglia polarization to M1 phenotype after spinal cord injury in rats. J. Neuroinflam-Mation 2019, 16, 206. [Google Scholar] [CrossRef] [Green Version]
- Yang, J.; Kim, C.S.; Tu, T.H.; Kim, M.S.; Goto, T.; Kawada, T.; Choi, M.S.; Park, T.; Sung, M.K.; Yun, J.W.; et al. Querce-tin protects obesity-induced hypothalamic inflammation by reducing microglia- mediated in-flammatory responses via HO-1 induction. Nutrients 2017, 9, 650. [Google Scholar] [CrossRef] [Green Version]
- 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 oxi-dative stress and TLR4-mediated inflammation. Inflamm. Res. 2020, 69, 1201–1213. [Google Scholar] [CrossRef]
- Sharma, V.; Mishra, M.; Ghosh, S.; Tewari, R.; Basu, A.; Seth, P.; Sen, E. Modulation of in-terleukin-1b mediated inflammatory response in human astrocytes by flavonoids: Implications in neuroprotection. Brain Res Bull 2007, 73, 55–63. [Google Scholar] [CrossRef]
- Chen, T.J.; Jeng, J.Y.; Lin, C.W.; Wu, C.Y.; Chen, Y.C. Quercetin inhibition of ROS depend-ent and-independent apoptosis in rat glioma C6 cells. Toxicology 2006, 223, 113–126. [Google Scholar] [CrossRef]
- Van Meeteren, M.E.; Hendriks, J.J.; Dijkstra, C.D.; van Tol, E.A. Dietary compounds pre-vent oxidative damage and nitric oxide production by cells involved in demyelinating disease. Biochem. Pharmacol. 2004, 67, 967–975. [Google Scholar]
- Nair, M.P.; Saiyed, Z.M.; Gandhi, N.H.; Ramchand, C. The flavonoid quercetin inhibits HIV-1 infection in normal peripheral blood mononuclear cells. Am. J. Infect. Dis. 2009, 5, 135–141. [Google Scholar] [CrossRef]
- Boesch-Saadatmandi, C.; Wagner, A.E.; Wolffram, S.; Rimbach, G. Effect of quercetin on in-flammatory gene expression in mice liver in vivo—role of redox factor 1, miRNA-122 and miR-NA-125b. Pharmacol. Res. 2012, 65, 523–530. [Google Scholar] [CrossRef] [PubMed]
- Yardim, A.; Kandemir, F.M.; Ozdemir, S.; Kucukler, S.; Comakli, S.; Gur, C.; Celik, H. Quercetin provides protection against the peripheral nerve damage caused by vincristine in rats by suppressing caspase 3, NF-κB, ATF-6 pathways and activating Nrf2, Akt pathways. Neuro-Toxicology 2020, 81, 137–146. [Google Scholar]
- Costa, L.G.; Tait, L.; de Laat, R.; Dao, K.; Giordano, G.; Pellacani, C.; Cole, T.B.; Furlong, C.E. Modulation of paraoxonase 2 (PON2) in mouse brain by the polyphenol quercetin: A mecha-nism of neuroprotection? Neurochem. Res. 2013, 38, 1809–1818. [Google Scholar] [CrossRef] [Green Version]
- Ghahremani, S.; Soodi, M.; Atashi, A. Quercetin ameliorates chlorpyrifos-induced oxidative stress in the rat brain: Possible involvment of PON2 pathway. J. Food Biochem. 2018, 42, e12530. [Google Scholar] [CrossRef]
- Selvakumar, K.; Bavithra, S.; Krishnamoorthy, G.; Arunakaran, J. Impact of quercetin on tight junctional proteins and BDNF signaling molecules in hippocampus of PCBs-exposed rats. Interdiscip. Toxicol. 2018, 11, 294–305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jin, Z.; Ke, J.; Guo, P.; Wang, Y.; Wu, H. Quercetin improves blood-brain barrier dysfunc-tion in rats with cerebral ischemia reperfusion via Wnt signaling pathway. Am. J. Transl. Res. 2019, 11, 4683–4695. [Google Scholar]
- Nunomura, A.; Perry, G. RNA and oxidative stress in Alzheimer’s disease: Focus on mi-croRNAs. Oxid. Med. Cell. Longev. 2020, 2020, 2638130. [Google Scholar] [CrossRef]
- Chang, W.S.; Wang, Y.H.; Zhu, X.T.; Wu, C.J. Genome-wide profiling of mirna and mrna expression in Alzheimer’s disease. Med. Sci. Monit. 2018, 24, 2721–2731. [Google Scholar] [CrossRef] [Green Version]
- Banzhaf-Strathmann, J.; Benito, E.; May, S.; Arzberger, T.; Tahirovic, S.; Kretzschmar, H.; Fischer, A.; Edbauer, D. MicroRNA-125b induces tau hyperphosphorylation and cognitive deficits in Alzheimer’s disease. EMBO J. 2014, 33, 1667–1680. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.; Tan, L.; Lu, Y.; Peng, J.; Zhu, Y.; Zhang, Y.; Sun, Z. MicroRNA-138 promotes tau phosphorylation by targeting retinoic acid receptor alpha. FEBS Lett. 2015, 589, 726–729. [Google Scholar] [CrossRef] [Green Version]
- Smith, P.Y.; Hernandez-Rapp, J.; Jolivette, F.; Lecours, C.; Bisht, K.; Goupil, C.; Dorval, V.; Parsi, S.; Morin, F.; Planel, E.; et al. miR-132/212 deficiency impairs tau metabolism and promotes pathological aggregation in vivo. Hum. Mo. Gen. 2015, 24, 6721–6735. [Google Scholar] [CrossRef] [Green Version]
- Santa-Maria, I.; Alaniz, M.E.; Renwick, N.; Cela, C.; Fulga, T.A.; Van Vactor, D.; Tuschl, T.L.; Clark, N.; Shelanski, M.L.; McCabe, B.D.; et al. Dysregulation of microRNA-219 pro-motes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Investig. 2015, 125, 681–686. [Google Scholar] [CrossRef]
- Hébert, S.S.; Papadopoulou, A.S.; Smith, P.; Galas, M.-C.; Planel, E.; Silahtaroglu, A.N.; Sergeant, N.; Buée, L.; De Strooper, B. Genetic ablation of Dicer in adult forebrain neurons results in abnormal tau hyperphosphorylation and neurodegeneration. Hum. Mol. Genet. 2010, 19, 3959–3969. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El Fatimy, R.; Li, S.; Chen, Z.; Mushannen, T.; Gongala, S.; Wei, Z.; Balu, D.T.; Rabinovsky, R.; Cantlon, A.; Elkhal, A.; et al. MicroRNA-132 provides neuroprotection for tauopathies via multiple signaling pathways. Acta Neuropathol. 2018, 136, 537–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Z.; Yi, P.; Yi, M.; Tong, X.; Cheng, X.; Yang, J.; Hu, Y.; Peng, W. Protective effect of quercetin against H2O2-induced oxidative damage in pc-12 cells: Comprehensive analysis of a lncRNA-associated ceRNA network. Oxid. Med. Cell. Longev. 2020, 2020, 6038919. [Google Scholar] [CrossRef]
- Konovalova, J.; Gerasymchuk, D.; Parkkinen, I.; Chmielarz, P.; Domanskyi, A. Interplay between MicroRNAs and Oxidative Stress in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 6055. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ebrahimpour, S.; Esmaeili, A.; Dehghanian, F.; Beheshti, S. Effects of quercetin- conjugated with superparamagnetic iron oxide nanoparticles on learning and memory improvement through targeting microRNAs/NF-κB pathway. Sci. Rep. 2020, 10, 15070. [Google Scholar] [CrossRef] [PubMed]
- Bilkei-Gorzo, A. Genetic mouse models of brain ageing and Alzheimer’s disease. Pharmacol. Ther. 2014, 142, 244–257. [Google Scholar] [CrossRef]
- Lv, M.; Yang, S.; Cai, L.; Qin, L.-Q.; Li, B.-Y.; Wan, Z. Effects of Quercetin Intervention on Cognition Function in APP/PS1 Mice was Affected by Vitamin D Status. Mol. Nutr. Food Res. 2018, 62, e1800621. [Google Scholar] [CrossRef]
- Yang, S.; Wang, G.; Ma, Z.F.; Qin, L.Q.; Zhai, Y.J.; Yu, Z.L.; Xue, M.; Zhang, Y.H.; Wan, Z. Dietary advanced glycation end products-induced cognitive impairment in aged ICR mice: Protective role of quercetin. Mol. Nutr. Food Res. 2020, 64, e1901019. [Google Scholar] [CrossRef]
- Zhang, X.; Abels, E.R.; Redzic, J.S.; Margulis, J.; Finkbeiner, S.; Breakefield, X.O. Potential Transfer of Polyglutamine and CAG-Repeat RNA in Extracellular Vesicles in Huntington’s Disease: Background and Evaluation in Cell Culture. Cell. Mol. Neurobiol. 2016, 36, 459–470. [Google Scholar] [CrossRef]
- Das, S.; Mandal, A.K.; Ghosh, A.; Panda, S.; Das, N.; Sarkar, S. Nanoparticulated Quercetin in Combating Age Related Cerebral Oxidative Injury. Curr. Aging Sci. 2008, 1, 169–174. [Google Scholar] [CrossRef]
- Ghosh, A.; Mandal, A.K.; Sarkar, S.; Panda, S.; Das, N. Nanoencapsulation of quercetin enhances its dietary efficacy in combating arsenic-induced oxidative damage in liver and brain of rats. Life Sci. 2009, 84, 75–80. [Google Scholar] [CrossRef]
- Ghosh, S.; Dungdung, S.R.; Chowdhury, S.T.; Mandal, A.K.; Sarkar, S.; Ghosh, D.; Das, N. Encapsulation of the flavonoid quercetin with an arsenic chelator into nanocapsules enables the simultaneous delivery of hydrophobic and hydrophilic drugs with a synergistic effect against chronic arsenic accumulation and oxidative stress. Free Radic. Biol. Med. 2011, 5, 1893–1902. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, A.; Sarkar, S.; Mandal, A.K.; Das, N. Neuroprotective role of nanoencapsulated quercetin in combating ischemia-reperfusion induced neuronal damage in young and aged rats. PLoS ONE 2013, 8, e57735. [Google Scholar] [CrossRef] [Green Version]
- Wang, G.; Wang, J.J.; Yang, G.Y.; Du, S.M.; Zeng, N.; Li, D.S.; Li, R.M.; Chen, J.Y.; Feng, J.B.; Yuan, S.H.; et al. Effects of quercetin nanoliposomes on C6 glioma cells through induction of type III programmed cell death. Int. J. Nanomed. 2012, 7, 271–280. [Google Scholar]
- Soleti, R.; Andriantsitohaina, R.; Martinez, M.C. Impact of polyphenols on extracellular vesi-cle levels and effects and their properties as tools for drug delivery for nutrition and health. Arch Biochem. Biophys. 2018, 644, 57–63. [Google Scholar] [CrossRef] [PubMed]
- Trotta, T.; Panaro, M.A.; Cianciulli, A.; Mori, G.; Di Benedetto, A.; Porro, C. Microglia-derived extracellular vesicles in Alzheimer’s Disease: A double-edged sword. Biochem. Pharmacol. 2018, 148, 184–192. [Google Scholar] [CrossRef]
- Panaro, M.A.; Benameur, T.; Porro, C. Extracellular Vesicles miRNA Cargo for Microglia Polarization in Traumatic Brain Injury. Biomol. 2020, 10, 901. [Google Scholar] [CrossRef]
- Pricci, M.; Bourget, J.-M.; Robitaille, H.; Porro, C.; Soleti, R.; Mostefai, H.A.; Auger, F.A.; Martínez, M.C.; Andriantsitohaina, R.; Germain, L. Applications of Human Tissue-Engineered Blood Vessel Models to Study the Effects of Shed Membrane Microparticles from T-Lymphocytes on Vascular Function. Tissue Eng. Part A 2009, 15, 137–145. [Google Scholar] [CrossRef] [Green Version]
- Soleti, R.; Lauret, E.; Andriantsitohaina, R.; Martínez, M.C. Internalization and induction of antioxidant messages by microvesicles contribute to the antiapoptotic effects on human endotheli-al cells. Free Radic. Biol. Med. 2012, 53, 2159–2170. [Google Scholar] [CrossRef] [Green Version]
- Trotta, T.; Panaro, M.A.; Prifti, E.; Porro, C. Modulation of Biological Activities in Glioblas-toma Mediated by Curcumin. Nutr. Cancer 2019, 71, 1241–1253. [Google Scholar] [CrossRef]
- Qi, Y.; Guo, L.; Jiang, Y.; Shi, Y.; Sui, H.; Zhao, L. Brain delivery of quercetin-loaded exo-somes improved cognitive function in AD mice by inhibiting phosphorylated tau- mediated neu-rofibrillary tangles. Drug Deliv. 2020, 27, 745–755. [Google Scholar] [CrossRef] [PubMed]
Biological Activities | Study Model | Major Findings | Signaling Pathways | References |
---|---|---|---|---|
Quercetin-induced apoptosis in cervical cancer cells and regulates tumorigenesis. | In vitro human cervical carcinoma HeLa cells | Quercetin exerts its suppressive, anti-proliferative and anti-migratory effect through MAPK, PI3K and WNT pathways | MAPK, PI3K and WNT pathways | [32] |
Neuroprotective effect against diabetes induced nerve damage, Inducer of neuronal plasticity in the myenteric plexus | STZ-induced diabetes mellitus in rats | Quercetin treatment enhanced the bioavailability of jejunal NO bioavailability in euglycemic and diabetic rats. Quercetin prevents diabetes-induced morphological changes in the myenteric plexus of diabetic rats | Neuronal NO pathway | [36] |
Anti-oxidative, anti-ER stress, neuroprotective effect against diabetic encephalopathy | db/db mouse model | Quercetin: 1. Improved learning and memory impairment 2. Alleviated impaired glucose tolerance and Insulin resistance 3. Decreased oxidative stress and protects against neuronal apoptosis in the brain of db/db mice 4. Relieved ER stress through the activation of SIRT1 | SIRT1/ER stress pathway | [34] |
Anti-inflammatory, anti-oxidative stress in the carotid arteries of diabetic rats | Diabetes-induced atherosclerosis rat model | Quercetin reduced hyperlipidemia, inflammatory cytokines and oxidative stress in the carotid arteries of diabetic rats on high-fat diet | AMPK/SIRT1/NF-κB signaling | [33] |
Anti-apoptotic effects mediated by Nrf-2 pathway against neurotoxicity. Neuroprotective effects due to up- and/or down-regulation of cytokines | Mouse mode of neurotoxicity | Quercetin: 1. Improved behavior impairment in d-galactose-induced neurotoxicity in mice. 2. Protected hippocampus neuron from damage induced by d-galactose. 3. Activated Nrf2-ARE signaling pathway in the hippocampus of d-galactose-treated mice. 4. Ameliorates Alzheimer disease via antioxidant pathway | Nrf2, Paraoxonase-2, c-Jun N-terminal kinase (JNK), PKC, MAPK signaling cascades, and PI3K/Akt pathways. | [37,38] |
Neuroprotective protective effect against the Vincristine-induced apoptosis in the sciatic nerve | Rat model of nerve injury | Quercetin reduces the ER stress caused by a vinca alkaloid antineoplastic agent (chemotherapy agent) in sciatic nerves and activates Akt, Nrf2 pathways. Quercetin may exert a protective effect against vincristine-induced peripheral neurotoxicity by suppressing NF–κB, caspase 3 and ATF-6 pathways | Akt, Nrf-2, NFκB, caspase 3, ATF-6 pathways | [90] |
Neuroprotective effects due to the activation of PON2 pathway and antagonizing the oxidative-induced neuronal toxicity. | PON2 knockout mice, Mouse striatal astrocytes. CPF-induced neurotoxicity in rats | Quercetin increased PON2 expression in striatal astrocytes; Exerts neuroprotection in vitro and in vivo, JNK/AP-1 pathways. Neuroprotective effects of quercetin has been significantly reduced on cells derived from PON2 knockout mice and CPF-induced neurotoxicity in rats. | Paraoxonase 2 (PON2) pathway, JNK and AP-1 pathway | [22,91,92] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
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. https://doi.org/10.3390/nu13041318
Benameur T, Soleti R, Porro C. The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases. Nutrients. 2021; 13(4):1318. https://doi.org/10.3390/nu13041318
Chicago/Turabian StyleBenameur, Tarek, Raffaella Soleti, and Chiara Porro. 2021. "The Potential Neuroprotective Role of Free and Encapsulated Quercetin Mediated by miRNA against Neurological Diseases" Nutrients 13, no. 4: 1318. https://doi.org/10.3390/nu13041318