Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation
<p>A general overview of the mechanisms of action of phytochemicals in neurodegenerative diseases. Intermingled as pieces of a puzzle, the multi-target effects of phytochemicals in the brain include anti-oxidant, mitochondrial protecting, anti-apoptotic, anti-aggregation, and anti-inflammatory activity.</p> "> Figure 2
<p>Oxidative stress-related pathogenic events implicated in neurodegeneration and molecular effects of phytochemicals. (<b>A</b>) summarizes the cascade of pathogenic events implicated in neurodegeneration. Oxidative stress arises from a decrease of antioxidant defense and a concomitant increase in the levels of reactive oxygen species (ROS) and lipid peroxidation products, such as malondialdehyde (MDA). This leads both to mitochondrial dysfunctions and protein oxidation/glycation fostering the accumulation of damaged mitochondria and the release of mitochondrial ROS, caspases, and mitochondrial DNA (mtDNA) fragments on the one hand and protein aggregation on the other. Altered intracellular substrates, including damaged mitochondria, and ubiquitinated proteins, and advanced glycation endproducts (AGEs) are targeted for degradation by autophagy and the proteasome. Nonetheless, the high amount of ROS along with the increasing amount of large protein aggregates and AGE-modified proteins contributes to impairing the proteasome while engulfing autophagy/lysosome vacuoles. This occludes the removal of altered substrates and impedes the release of the antioxidant factor Nrf2 from Keap1, thus fueling a vicious cycle of oxidative-related pathological events (<b>B</b>) summarizes the neuroprotective mechanisms of phytochemicals (blue stars). These consist of decreasing oxidative stress through upregulating anti-oxidant defense and decreasing ROS and MDA levels, preserving mitochondrial function, preventing protein oxidation/glycation, and aggregation through either direct molecular binding or increased degradation via autophagy induction. This includes Keap2 degradation and Nrf2 activation, which contributes to counteracting oxidative stress and mitochondrial damage along with promoting mitophagy and mitobiogenesis.</p> "> Figure 3
<p>Phytochemicals, cell-clearing machinery, and neuro-inflammation. By rescuing alterations of autophagy, phytochemicals prevent the intracellular accumulation of danger-associated molecular patterns (DAMPs, including ROS, damaged mitochondria, fragments of mtDNA, and aggregated proteins), potentially preventing their extracellular release and the subsequent neuroinflammatory events being triggered in neighboring neurons and glial cells. In fact, in neighboring cells, DAMPs bind to RAGE and TLR, triggering pro-inflammatory, pro-apoptotic cascades that contribute to recruiting the immunoproteasome while altering autophagy flux. By degrading self-proteins within antigenic sites, the immunoproteasome contributes to cytotoxic auto-immune attack, while autophagy impairment contributes to intracellular DAMPs accumulation and extracellular spreading. In this frame, phytochemicals provide beneficial effects by downregulating pro-inflammatory cascades such as AGE/RAGEs, JAK/STAT, mTOR, HMGB1/TLR-4, and NF-kB/NLRP3 while up-regulating AMPK/SIRT1/PGC-1α and Nrf2, which are bound, at least in part, to autophagy activation and blunting of immunoproteasome activity.</p> ">
Abstract
:1. Introduction
2. An Overview of Neuroprotective Phytochemical Classes
2.1. Polyphenols
2.2. Terpenes
2.3. Alkaloids
2.4. Other Nitrogen-Containing Phytochemicals
3. Phytochemicals and Oxidative Stress
3.1. Autophagy-Related Antioxidant Effects of Phytochemicals
3.1.1. An Overview of the Autophagy Pathway
3.1.2. Phytochemicals Bridging Autophagy Induction and Prevention of Oxidative Stress
4. Phytochemicals and Mitochondrial Damage
Autophagy-Related Mitochondrial Protection by Phytochemicals
5. Phytochemicals and Proteostasis
5.1. Alterations of Proteostasis in Neurodegeneration
5.2. Anti-Aggregation Mechanisms of Phytochemicals
5.3. Autophagy-Related Anti-Aggregation Effects of Phytochemicals
6. Phytochemicals and Inflammatory Pathways
Is There a Role of Autophagy and Immunoproteasome in the Anti-Inflammatory Effects of Phytochemicals?
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Sharifi-Rad, M.; Lankatillake, C.; Dias, D.A.; Docea, A.O.; Mahomoodally, M.F.; Lobine, D.; Chazot, P.L.; Kurt, B.; Tumer, T.B.; Moreira, A.C.; et al. Impact of Natural Compounds on Neurodegenerative Disorders: From Preclinical to Pharmacotherapeutics. J. Clin. Med. 2020, 9, 1061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pellegrini, C.; Fornai, M.; Antonioli, L.; Blandizzi, C.; Calderone, V. Phytochemicals as Novel Therapeutic Strategies for NLRP3 Inflammasome-Related Neurological, Metabolic, and Inflammatory Diseases. Int. J. Mol. Sci. 2019, 20, 2876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Limanaqi, F.; Biagioni, F.; Busceti, C.L.; Ryskalin, L.; Polzella, M.; Frati, A.; Fornai, F. Phytochemicals Bridging Autophagy Induction and Alpha-Synuclein Degradation in Parkinsonism. Int. J. Mol. Sci. 2019, 20, 3274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bordoni, L.; Gabbianelli, R. Mitochondrial DNA and Neurodegeneration: Any Role for Dietary Antioxidants? Antioxidants 2020, 9, 764. [Google Scholar] [CrossRef] [PubMed]
- Rajasekar, J.; Perumal, M.K.; Vallikannan, B. A critical review on anti-angiogenic property of phytochemicals. J. Nutr. Biochem. 2019, 71, 1–15. [Google Scholar] [CrossRef]
- Abdullah, A.; Mohd Murshid, N.; Makpol, S. Antioxidant Modulation of mTOR and Sirtuin Pathways in Age-Related Neurodegenerative Diseases. Mol. Neurobiol. 2020. Epub ahead of print. [Google Scholar] [CrossRef]
- Pinelli, R.; Biagioni, F.; Limanaqi, F.; Bertelli, M.; Scaffidi, E.; Polzella, M.; Busceti, C.L.; Fornai, F. A Re-Appraisal of Pathogenic Mechanisms Bridging Wet and Dry Age-Related Macular Degeneration Leads to Reconsider a Role for Phytochemicals. Int. J. Mol. Sci. 2020, 21, 5563. [Google Scholar] [CrossRef]
- Rebas, E.; Rzajew, J.; Radzik, T.; Zylinska, L. Neuroprotective Polyphenols: A Modulatory Action on Neurotransmitter Pathways. Curr. Neuropharmacol. 2020, 18, 431–445. [Google Scholar] [CrossRef]
- Momtaz, S.; Memariani, Z.; El-Senduny, F.F.; Sanadgol, N.; Golab, F.; Katebi, M.; Abdolghaffari, A.H.; Farzaei, M.H.; Abdollah, M. Targeting Ubiquitin-Proteasome Pathway by Natural Products: Novel Therapeutic Strategy for Treatment of Neurodegenerative Diseases. Front. Physiol. 2020, 11, 361. [Google Scholar] [CrossRef]
- Limanaqi, F.; Biagioni, F.; Gambardella, S.; Familiari, P.; Frati, A.; Fornai, F. Promiscuous Roles of Autophagy and Proteasome in Neurodegenerative Proteinopathies. Int. J. Mol. Sci. 2020, 21, 3028. [Google Scholar] [CrossRef]
- Limanaqi, F.; Biagioni, F.; Ryskalin, L.; Busceti, C.L.; Fornai, F. Molecular Mechanisms Linking ALS/FTD and Psychiatric Disorders, the Potential Effects of Lithium. Front. Cell. Neurosci. 2019, 13, 450. [Google Scholar] [CrossRef] [PubMed]
- Ryskalin, L.; Busceti, C.L.; Limanaqi, F.; Biagioni, F.; Gambardella, S.; Fornai, F. A Focus on the Beneficial Effects of Alpha Synuclein and a Re-Appraisal of Synucleinopathies. Curr. Protein Pept. Sci. 2018, 19, 598–611. [Google Scholar] [CrossRef]
- Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction, Oxidative Stress, and Neuroinflammation: Intertwined Roads to Neurodegeneration. Antioxidants 2020, 9, 647. [Google Scholar] [CrossRef] [PubMed]
- Giordano, S.; Darley-Usmar, V.; Zhang, J. Autophagy as an essential cellular antioxidant pathway in neurodegenerative disease. Redox Biol. 2013, 2, 82–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmad, S.; Khan, H.; Shahab, U.; Rehman, S.; Rafi, Z.; Khan, M.Y.; Ansari, A.; Siddiqui, Z.; Ashraf, J.M.; Abdullah, S.M.; et al. Protein oxidation: An overview of metabolism of sulphur containing amino acid, cysteine. Front. Biosci. (Schol. Ed.) 2017, 9, 71–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aman, Y.; Ryan, B.; Torsetnes, S.B.; Knapskog, A.B.; Watne, L.O.; McEwan, W.A.; Fang, E.F. Enhancing mitophagy as a therapeutic approach for neurodegenerative diseases. Int. Rev. Neurobiol. 2020, 155, 169–202. [Google Scholar] [CrossRef]
- Filomeni, G.; De Zio, D.; Cecconi, F. Oxidative stress and autophagy: The clash between damage and metabolic needs. Cell Death Differ. 2015, 22, 377–388. [Google Scholar] [CrossRef] [Green Version]
- Limanaqi, F.; Biagioni, F.; Gaglione, A.; Busceti, C.L.; Fornai, F. A Sentinel in the Crosstalk Between the Nervous and Immune System: The (Immuno)-Proteasome. Front. Immunol. 2019, 10, 628. [Google Scholar] [CrossRef] [Green Version]
- Lazzeri, G.; Biagioni, F.; Fulceri, F.; Busceti, C.L.; Scavuzzo, M.C.; Ippolito, C.; Salvetti, A.; Lenzi, P.; Fornai, F. mTOR Modulates Methamphetamine-Induced Toxicity through Cell Clearing Systems. Oxid. Med. Cell. Longev. 2018, 2018, 6124745. [Google Scholar] [CrossRef] [Green Version]
- Castino, R.; Lazzeri, G.; Lenzi, P.; Bellio, N.; Follo, C.; Ferrucci, M.; Fornai, F.; Isidoro, C. Suppression of autophagy precipitates neuronal cell death following low doses of methamphetamine. J. Neurochem. 2008, 106, 1426–1439. [Google Scholar] [CrossRef]
- Ryskalin, L.; Limanaqi, F.; Frati, A.; Busceti, C.L.; Fornai, F. mTOR-Related Brain Dysfunctions in Neuropsychiatric Disorders. Int. J. Mol. Sci. 2018, 19, 2226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sato, S.; Uchihara, T.; Fukuda, T.; Noda, S.; Kondo, H.; Saiki, S.; Komatsu, M.; Uchiyama, Y.; Tanaka, K.; Hattori, N. Loss of autophagy in dopaminergic neurons causes Lewy pathology and motor dysfunction in aged mice. Sci. Rep. 2018, 8, 2813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Limanaqi, F.; Biagioni, F.; Busceti, C.L.; Ryskalin, L.; Fornai, F. The effects of proteasome on baseline and methamphetamine-dependent dopamine transmission. Neurosci. Biobehav. Rev. 2019, 102, 308–317. [Google Scholar] [CrossRef] [PubMed]
- Limanaqi, F.; Biagioni, F.; Gambardella, S.; Ryskalin, L.; Fornai, F. Interdependency between Autophagy and Synaptic Vesicle Trafficking: Implications for Dopamine Release. Front. Mol. Neurosci. 2018, 11, 299. [Google Scholar] [CrossRef] [Green Version]
- Limanaqi, F.; Busceti, C.L.; Biagioni, F.; Cantini, F.; Lenzi, P.; Fornai, F. Cell-Clearing Systems Bridging Repeat Expansion Proteotoxicity and Neuromuscular Junction Alterations in ALS and SBMA. Int. J. Mol. Sci. 2020, 21, 4021. [Google Scholar] [CrossRef]
- Song, J.X.; Lu, J.H.; Liu, L.F.; Chen, L.L.; Durairajan, S.S.; Yue, Z.; Zhang, H.Q.; Li, M. HMGB1 is involved in autophagy inhibition caused by SNCA/alpha-synuclein overexpression: A process modulated by the natural autophagy inducer corynoxine B. Autophagy 2014, 10, 144–154. [Google Scholar] [CrossRef] [Green Version]
- Thibaudeau, T.A.; Anderson, R.T.; Smith, D.M. A common mechanism of proteasome impairment by neurodegenerative disease-associated oligomers. Nat. Commun. 2018, 9, 1097. [Google Scholar] [CrossRef]
- Lai, M.; Yao, H.; Shah, S.Z.A.; Wu, W.; Wang, D.; Zhao, Y.; Wang, L.; Zhou, X.; Zhao, D.; Yang, L. The NLRP3-Caspase 1 Inflammasome Negatively Regulates Autophagy via TLR4-TRIF in Prion Peptide-Infected Microglia. Front. Aging Neurosci. 2018, 10, 116. [Google Scholar] [CrossRef]
- Wang, Y.; Meng, C.; Zhang, J.; Wu, J.; Zhao, J. Inhibition of GSK-3β alleviates cerebral ischemia/reperfusion injury in rats by suppressing NLRP3 inflammasome activation through autophagy. Int. Immunopharmacol. 2019, 68, 234–241. [Google Scholar] [CrossRef]
- Lázaro, D.F.; Bellucci, A.; Brundin, P.; Outeiro, T.F. Editorial: Protein Misfolding and Spreading Pathology in Neurodegenerative Diseases. Front. Mol. Neurosci. 2020, 12, 312. [Google Scholar] [CrossRef] [Green Version]
- Maurel, C.; Dangoumau, A.; Marouillat, S.; Brulard, C.; Chami, A.; Hergesheimer, R.; Corcia, P.; Blasco, H.; Andres, C.R.; Vourc’h, P. Causative Genes in Amyotrophic Lateral Sclerosis and Protein Degradation Pathways: A Link to Neurodegeneration. Mol. Neurobiol. 2018, 55, 6480–6499. [Google Scholar] [CrossRef] [PubMed]
- Ferrucci, M.; Biagioni, F.; Ryskalin, L.; Limanaqi, F.; Gambardella, S.; Frati, A.; Fornai, F. Ambiguous Effects of Autophagy Activation Following Hypoperfusion/Ischemia. Int. J. Mol. Sci. 2018, 19, 2756. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cortes, C.J.; Miranda, H.C.; Frankowski, H.; Batlevi, Y.; Young, J.E.; Le, A.; Ivanov, N.; Sopher, B.L.; Carromeu, C.; Muotri, A.R.; et al. Polyglutamine-expanded androgen receptor interferes with TFEB to elicit autophagy defects in SBMA. Nat. Neurosci. 2014, 17, 1180–1189. [Google Scholar] [CrossRef] [Green Version]
- Rusmini, P.; Cortese, K.; Crippa, V.; Cristofani, R.; Cicardi, M.E.; Ferrari, V.; Vezzoli, G.; Tedesco, B.; Meroni, M.; Messi, E.; et al. Trehalose induces autophagy via lysosomal-mediated TFEB activation in models of motoneuron degeneration. Autophagy 2019, 15, 631–651. [Google Scholar] [CrossRef] [PubMed]
- Natale, G.; Lenzi, P.; Lazzeri, G.; Falleni, A.; Biagioni, F.; Ryskalin, L.; Fornai, F. Compartment-dependent mitochondrial alterations in experimental ALS, the effects of mitophagy and mitochondriogenesis. Front. Cell. Neurosci. 2015, 9, 434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aguzzi, A.; Rajendran, L. The transcellular spread of cytosolic amyloids, prions, and prionoids. Neuron 2009, 64, 783–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McAlary, L.; Plotkin, S.S.; Yerbury, J.J.; Cashman, N.R. Prion-Like Propagation of Protein Misfolding and Aggregation in Amyotrophic Lateral Sclerosis. Front. Mol. Neurosci. 2019, 12, 262, [published correction appears in Front. Mol. Neurosci. 2020, 12, 311]. [Google Scholar] [CrossRef] [Green Version]
- Natale, G.; Pompili, E.; Biagioni, F.; Paparelli, S.; Lenzi, P.; Fornai, F. Histochemical approaches to assess cell-to-cell transmission of misfolded proteins in neurodegenerative diseases. Eur. J. Histochem. 2013, 57, e5. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Liu, D.; Sun, L.; Lu, Y.; Zhang, Z. Advanced glycation end products and neurodegenerative diseases: Mechanisms and perspective. J. Neurol Sci. 2012, 317, 1–5. [Google Scholar] [CrossRef]
- Ott, C.; Jacobs, K.; Haucke, E.; Navarrete Santos, A.; Grune, T.; Simm, A. Role of advanced glycation end products in cellular signaling. Redox Biol. 2014, 2, 411–429. [Google Scholar] [CrossRef] [Green Version]
- Rubinsztein, D.C.; Bento, C.F.; Deretic, V. Therapeutic targeting of autophagy in neurodegenerative and infectious diseases. J. Exp. Med. 2015, 212, 979–990. [Google Scholar] [CrossRef]
- Zhang, M.; Schekman, R. Cell biology. Unconventional secretion, unconventional solutions. Science 2013, 340, 559–561. [Google Scholar] [CrossRef]
- Grimm, S.; Ernst, L.; Grotzinger, N.; Hohn, A.; Breusing, N.; Reinheckel, T.; Grune, T. Cathepsin D is one of the major enzymes involved in intracellular degradation of AGE-modified proteins. Free Radic. Res. 2010, 44, 1013–1026. [Google Scholar] [CrossRef] [PubMed]
- Grimm, S.; Ott, C.; Hörlacher, M.; Weber, D.; Höhn, A.; Grune, T. Advanced-glycation-end-product-induced formation of immunoproteasomes: Involvement of RAGE and Jak2/STAT1. Biochem. J. 2012, 448, 127–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gambardella, S.; Limanaqi, F.; Ferese, R.; Biagioni, F.; Campopiano, R.; Centonze, D.; Fornai, F. ccf-mtDNA as a Potential Link Between the Brain and Immune System in Neuro-Immunological Disorders. Front. Immunol. 2019, 10, 1064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pla, A.; Pascual, M.; Renau-Piqueras, J.; Guerri, C. TLR4 mediates the impairment of ubiquitin-proteasome and autophagy-lysosome pathways induced by ethanol treatment in brain. Cell Death Dis. 2014, 5, e1066. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Hu, L.; Jiang, J.; Li, H.; Wu, Q.; Ooi, K.; Wang, J.; Feng, Y.; Zhu, D.; Xia, C. HMGB1/RAGE axis mediates stress-induced RVLM neuroinflammation in mice via impairing mitophagy flux in microglia. J. Neuroinflamm. 2020, 17, 15. [Google Scholar] [CrossRef]
- Ray, R.; Juranek, J.K.; Rai, V. RAGE axis in neuroinflammation, neurodegeneration and its emerging role in the pathogenesis of amyotrophic lateral sclerosis. Neurosci. Biobehav. Rev. 2016, 62, 48–55. [Google Scholar] [CrossRef]
- Pietrocola, F.; Mariño, G.; Lissa, D.; Vacchelli, E.; Malik, S.A.; Niso-Santano, M.; Zamzami, N.; Galluzzi, L.; Maiuri, M.C.; Kroemer, G. Pro-autophagic polyphenols reduce the acetylation of cytoplasmic proteins. Cell Cycle 2012, 11, 3851–3860. [Google Scholar] [CrossRef] [Green Version]
- Li, P.; Ma, K.; Wu, H.Y.; Wu, Y.P.; Li, B.X. Isoflavones Induce BEX2-Dependent Autophagy to Prevent ATR-Induced Neurotoxicity in SH-SY5Y Cells. Cell Physiol. Biochem. 2017, 43, 1866–1879. [Google Scholar] [CrossRef]
- Liu, Y.; Zhou, H.; Yin, T.; Gong, Y.; Yuan, G.; Chen, L.; Liu, J. Quercetin-modified gold-palladium nanoparticles as a potential autophagy inducer for the treatment of Alzheimer’s disease. J. Colloid. Interface Sci. 2019, 552, 388–400. [Google Scholar] [CrossRef] [PubMed]
- Filomeni, G.; Graziani, I.; De Zio, D.; Dini, L.; Centonze, D.; Rotilio, G.; Ciriolo, M.R. Neuroprotection of kaempferol by autophagy in models of rotenone-mediated acute toxicity: Possible implications for Parkinson’s disease. Neurobiol. Aging 2012, 33, 767–785. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Sun, S.; Sun, Y.; Song, Q.; Zhu, J.; Song, N.; Chen, M.; Sun, T.; Xia, M.; Ding, J.; et al. Small molecule-driven NLRP3 inflammation inhibition via interplay between ubiquitination and autophagy: Implications for Parkinson disease. Autophagy 2019, 15, 1860–1881. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.Z.; Ardah, M.; Haikal, C.; Svanbergsson, A.; Diepenbroek, M.; Vaikath, N.N.; Li, W.; Wang, Z.Y.; Outeiro, T.F.; El-Agnaf, O.M.; et al. Dihydromyricetin and Salvianolic acid B inhibit alpha-synuclein aggregation and enhance chaperone-mediated autophagy. Transl. Neurodegener. 2019, 8, 18. [Google Scholar] [CrossRef]
- Kuang, L.; Cao, X.; Lu, Z. Baicalein Protects against Rotenone-Induced Neurotoxicity through Induction of Autophagy. Biol. Pharm. Bull. 2017, 40, 1537–1543. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Lin, S.; Xu, C.; Zhang, P.; Mei, X. Triggering of Autophagy by Baicalein in Response to Apoptosis after Spinal Cord Injury: Possible Involvement of the PI3K Activation. Biol. Pharm. Bull. 2018, 41, 478–486. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, W.; Yao, S.; Li, H.; Meng, Z.; Sun, X. Curcumin promotes functional recovery and inhibits neuronal apoptosis after spinal cord injury through the modulation of autophagy. J. Spinal Cord Med. 2019, 1–9, Epub ahead of print. [Google Scholar] [CrossRef]
- Lin, K.L.; Lin, K.J.; Wang, P.W.; Chuang, J.H.; Lin, H.Y.; Chen, S.D.; Chuang, Y.C.; Huang, S.T.; Tiao, M.M.; Chen, J.B.; et al. Resveratrol provides neuroprotective effects through modulation of mitochondrial dynamics and ERK1/2 regulated autophagy. Free Radic. Res. 2018, 52, 1371–1386. [Google Scholar] [CrossRef]
- Vidoni, C.; Secomandi, E.; Castiglioni, A.; Melone, M.A.B.; Isidoro, C. Resveratrol protects neuronal-like cells expressing mutant Huntingtin from dopamine toxicity by rescuing ATG4-mediated autophagosome formation. Neurochem. Int. 2018, 117, 174–187. [Google Scholar] [CrossRef]
- Moon, J.H.; Lee, J.H.; Park, J.Y.; Kim, S.W.; Lee, Y.J.; Kang, S.J.; Seol, J.W.; Ahn, D.C.; Park, S.Y. Caffeine prevents human prion protein-mediated neurotoxicity through the induction of autophagy. Int. J. Mol. Med. 2014, 34, 553–558. [Google Scholar] [CrossRef]
- Zhang, Y.; Yang, Y.; Yu, H.; Li, M.; Hang, L.; Xu, X. Apigenin Protects Mouse Retina against Oxidative Damage by Regulating the Nrf2 Pathway and Autophagy. Oxid. Med. Cell. Longev. 2020, 2020, 9420704. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wu, Q.; Zhang, L.; Wang, Q.; Yang, Z.; Liu, J.; Feng, L. Caffeic acid reduces A53T α-synuclein by activating JNK/Bcl-2-mediated autophagy in vitro and improves behaviour and protects dopaminergic neurons in a mouse model of Parkinson’s disease. Pharmacol. Res. 2019, 150, 104538. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.H.; Moon, J.H.; Kim, S.W.; Jeong, J.K.; Nazim, U.M.; Lee, Y.J.; Seol, J.W.; Park, S.Y. EGCG-mediated autophagy flux has a neuroprotection effect via a class III histone deacetylase in primary neuron cells. Oncotarget 2015, 6, 9701–9717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, J.H.; Jeong, J.K.; Park, S.Y. Sulforaphane-induced autophagy flux prevents prion protein-mediated neurotoxicity through AMPK pathway. Neuroscience 2014, 278, 31–39. [Google Scholar] [CrossRef]
- Wang, N.; He, J.; Pan, C.; Wang, J.; Ma, M.; Shi, X.; Xu, Z. Resveratrol Activates Autophagy via the AKT/mTOR Signaling Pathway to Improve Cognitive Dysfunction in Rats With Chronic Cerebral Hypoperfusion. Front. Neurosci. 2019, 13, 859. [Google Scholar] [CrossRef] [Green Version]
- Jiang, P.; Guo, Y.; Dang, R.; Yang, M.; Liao, D.; Li, H.; Sun, Z.; Feng, Q.; Xu, P. Salvianolic acid B protects against lipopolysaccharide-induced behavioral deficits and neuroinflammatory response: Involvement of autophagy and NLRP3 inflammasome. J. Neuroinflamm. 2017, 14, 239. [Google Scholar] [CrossRef]
- Wang, H.; Liu, C.; Mei, X.; Cao, Y.; Guo, Z.; Yuan, Y.; Zhao, Z.; Song, C.; Guo, Y.; Shen, Z. Berberine attenuated pro-inflammatory factors and protect against neuronal damage via triggering oligodendrocyte autophagy in spinal cord injury. Oncotarget 2017, 8, 98312–98321. [Google Scholar] [CrossRef] [Green Version]
- Sigrist, S.J.; Carmona-Gutierrez, D.; Gupta, V.K.; Bhukel, A.; Mertel, S.; Eisenberg, T.; Madeo, F. Spermidine-triggered autophagy ameliorates memory during aging. Autophagy 2014, 10, 178–179. [Google Scholar] [CrossRef] [Green Version]
- Yang, X.; Xu, S.; Qian, Y.; Xiao, Q. Resveratrol regulates microglia M1/M2 polarization via PGC-1α in conditions of neuroinflammatory injury. Brain Behav. Immun. 2017, 64, 162–172. [Google Scholar] [CrossRef]
- Zhuang, X.X.; Wang, S.F.; Tan, Y.; Song, J.X.; Zhu, Z.; Wang, Z.Y.; Wu, M.Y.; Cai, C.Z.; Huang, Z.J.; Tan, J.Q.; et al. Pharmacological enhancement of TFEB-mediated autophagy alleviated neuronal death in oxidative stress-induced Parkinson’s disease models. Cell Death Dis. 2020, 11, 128. [Google Scholar] [CrossRef] [Green Version]
- Saha, S.; Mahapatra, K.K.; Mishra, S.R.; Mallick, S.; Negi, V.D.; Sarangi, I.; Patil, S.; Patra, S.K.; Bhutia, S.K. Bacopa monnieri inhibits apoptosis and senescence through mitophagy in human astrocytes. Food Chem. Toxicol. 2020, 141, 111367. [Google Scholar] [CrossRef] [PubMed]
- Liu, R.H. Potential synergy of phytochemicals in cancer prevention: Mechanism of action. J. Nutr. 2004, 134, 3479S–3485S. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Campos-Vega, R.; Oomah, B.D. Chemistry and classification of phytochemicals. In Handbook of Plant Food Phytochemicals; Tiwari, B., Brunton, N.P., Brennan, C.S., Eds.; John Wiley & Sons: Hoboken, NJ, USA, 2013. [Google Scholar]
- Joseph, J.; Cole, G.; Head, E.; Ingram, D. Nutrition, brain aging, and neurodegeneration. J. Neurosci. 2009, 29, 12795–12801. [Google Scholar] [CrossRef] [Green Version]
- Potì, F.; Santi, D.; Spaggiari, G.; Zimetti, F.; Zanotti, I. Polyphenol Health Effects on Cardiovascular and Neurodegenerative Disorders: A Review and Meta-Analysis. Int. J. Mol. Sci. 2019, 20, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Costa, L.G.; Garrick, J.M.; Roquè, P.J.; Pellacani, C. Mechanisms of Neuroprotection by Quercetin: Counteracting Oxidative Stress and More. Oxid. Med. Cell. Longev. 2016, 2016, 2986796. [Google Scholar] [CrossRef] [Green Version]
- Bhatia, N.K.; Modi, P.; Sharma, S.; Deep, S. Quercetin and Baicalein Act as Potent Antiamyloidogenic and Fibril Destabilizing Agents for SOD1 Fibrils. ACS Chem. Neurosci. 2020, 11, 1129–1138. [Google Scholar] [CrossRef]
- Ip, P.; Sharda, P.R.; Cunningham, A.; Chakrabartty, S.; Pande, V.; Chakrabartty, A. Quercitrin and quercetin 3-β-d-glucoside as chemical chaperones for the A4V SOD1 ALS-causing mutant. Protein Eng. Des. Sel. 2017, 30, 431–440. [Google Scholar] [CrossRef]
- Huang, Y.; Chen, Y.; Shaw, A.M.; Goldfine, H.; Tian, J.; Cai, J. Enhancing TFEB-Mediated Cellular Degradation Pathways by the mTORC1 Inhibitor Quercetin. Oxid. Med. Cell. Longev. 2018, 2018, 5073420. [Google Scholar] [CrossRef] [Green Version]
- El-Horany, H.E.; El-Latif, R.N.; ElBatsh, M.M.; Emam, M.N. Ameliorative Effect of Quercetin on Neurochemical and Behavioral Deficits in Rotenone Rat Model of Parkinson’s Disease: Modulating Autophagy (Quercetin on Experimental Parkinson’s Disease). J. Biochem. Mol. Toxicol. 2016, 30, 360–369. [Google Scholar] [CrossRef]
- Pakrashi, S.; Chakraborty, J.; Bandyopadhyay, J. Neuroprotective Role of Quercetin on Rotenone-Induced Toxicity in SH-SY5Y Cell Line through Modulation of Apoptotic and Autophagic Pathways. Neurochem. Res. 2020, 45, 1962–1973. [Google Scholar] [CrossRef] [PubMed]
- Sharma, D.R.; Wani, W.Y.; Sunkaria, A.; Kandimalla, R.J.; Sharma, R.K.; Verma, D.; Bal, A.; Gill, K.D. Quercetin attenuates neuronal death against aluminum-induced neurodegeneration in the rat hippocampus. Neuroscience 2016, 324, 163–176. [Google Scholar] [CrossRef] [PubMed]
- Ueda, T.; Inden, M.; Shirai, K.; Sekine, S.I.; Masaki, Y.; Kurita, H.; Ichihara, K.; Inuzuka, T.; Hozumi, I. The effects of Brazilian green propolis that contains flavonols against mutant copper-zinc superoxide dismutase-mediated toxicity. Sci. Rep. 2017, 7, 2882. [Google Scholar] [CrossRef] [Green Version]
- Wu, B.; Luo, H.; Zhou, X.; Cheng, C.Y.; Lin, L.; Liu, B.L.; Liu, K.; Li, P.; Yang, H. Succinate-induced neuronal mitochondrial fission and hexokinase II malfunction in ischemic stroke: Therapeutical effects of kaempferol. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 2307–2318. [Google Scholar] [CrossRef]
- Jang, J.H.; Lee, S.H.; Jung, K.; Yoo, H.; Park, G. Inhibitory Effects of Myricetin on Lipopolysaccharide-Induced Neuroinflammation. Brain Sci. 2020, 10, 32. [Google Scholar] [CrossRef] [Green Version]
- Hung, K.C.; Huang, H.J.; Wang, Y.T.; Lin, A.M. Baicalein attenuates α-synuclein aggregation, inflammasome activation and autophagy in the MPP+-treated nigrostriatal dopaminergic system in vivo. J. Ethnopharmacol. 2016, 194, 522–529. [Google Scholar] [CrossRef]
- Baluchnejadmojarad, T.; Zeinali, H.; Roghani, M. Scutellarin alleviates lipopolysaccharide-induced cognitive deficits in the rat: Insights into underlying mechanisms. Int. Immunopharmacol. 2018, 54, 311–319. [Google Scholar] [CrossRef]
- Shin, J.W.; Kweon, K.J.; Kim, D.K.; Kim, P.; Jeon, T.D.; Maeng, S.; Sohn, N.W. Scutellarin Ameliorates Learning and Memory Deficit via Suppressing β-Amyloid Formation and Microglial Activation in Rats with Chronic Cerebral Hypoperfusion. Am. J. Chin. Med. 2018, 46, 1203–1223. [Google Scholar] [CrossRef]
- Che, D.N.; Cho, B.O.; Kim, J.S.; Shin, J.Y.; Kang, H.J.; Jang, S.I. Luteolin and Apigenin Attenuate LPS-Induced Astrocyte Activation and Cytokine Production by Targeting MAPK, STAT3, and NF-κB Signaling Pathways. Inflammation 2020, 43, 1716–1728. [Google Scholar] [CrossRef]
- Chesser, A.S.; Ganeshan, V.; Yang, J.; Johnson, G.V. Epigallocatechin-3-gallate enhances clearance of phosphorylated tau in primary neurons. Nutr. Neurosci. 2016, 19, 21–31. [Google Scholar] [CrossRef]
- Gu, H.F.; Nie, Y.X.; Tong, Q.Z.; Tang, Y.L.; Zeng, Y.; Jing, K.Q.; Zheng, X.L.; Liao, D.F. Epigallocatechin-3-gallate attenuates impairment of learning and memory in chronic unpredictable mild stress-treated rats by restoring hippocampal autophagic flux. PLoS ONE 2014, 9, e112683, Erratum in PLoS ONE 2015, 10, e0117649. [Google Scholar] [CrossRef] [PubMed]
- Wang, S.; Wang, J.; Wei, H.; Gu, T.; Wang, J.; Wu, Z.; Yang, Q. Genistein Attenuates Acute Cerebral Ischemic Damage by Inhibiting the NLRP3 Inflammasome in Reproductively Senescent Mice. Front. Aging Neurosci. 2020, 12, 153. [Google Scholar] [CrossRef] [PubMed]
- Qian, Y.; Cao, L.; Guan, T.; Chen, L.; Xin, H.; Li, Y.; Zheng, R.; Yu, D. Protection by genistein on cortical neurons against oxidative stress injury via inhibition of NF-kappaB, JNK and ERK signaling pathway. Pharm. Biol. 2015, 53, 1124–1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Haddadi, R.; Nayebi, A.M.; Eyvari Brooshghalan, S. Silymarin prevents apoptosis through inhibiting the Bax/caspase-3 expression and suppresses toll like receptor-4 pathway in the SNc of 6-OHDA intoxicated rats. Biomed Pharmacother. 2018, 104, 127–136. [Google Scholar] [CrossRef]
- Haddadi, R.; Eyvari-Brooshghalan, S.; Nayebi, A.M.; Sabahi, M.; Ahmadi, S.A. Neuronal degeneration and oxidative stress in the SNc of 6-OHDA intoxicated rats; improving role of silymarin long-term treatment. Naunyn Schmiedebergs Arch. Pharmacol. 2020. Online ahead of print. [Google Scholar] [CrossRef]
- Lu, X.; Dong, J.; Zheng, D.; Li, X.; Ding, D.; Xu, H. Reperfusion combined with intraarterial administration of resveratrol-loaded nanoparticles improved cerebral ischemia-reperfusion injury in rats. Nanomedicine 2020, 28, 102208. [Google Scholar] [CrossRef]
- Wu, Y.L.; Chang, J.C.; Lin, W.Y.; Li, C.C.; Hsieh, M.; Chen, H.W.; Wang, T.S.; Wu, W.T.; Liu, C.S.; Liu, K.L. Caffeic acid and resveratrol ameliorate cellular damage in cell and Drosophila models of spinocerebellar ataxia type 3 through upregulation of Nrf2 pathway. Free Radic. Biol. Med. 2018, 115, 309–317. [Google Scholar] [CrossRef]
- Wang, H.; Jiang, T.; Li, W.; Gao, N.; Zhang, T. Resveratrol attenuates oxidative damage through activating mitophagy in an in vitro model of Alzheimer’s disease. Toxicol. Lett. 2018, 282, 100–108. [Google Scholar] [CrossRef]
- Guo, D.; Xie, J.; Zhao, J.; Huang, T.; Guo, X.; Song, J. Resveratrol protects early brain injury after subarachnoid hemorrhage by activating autophagy and inhibiting apoptosis mediated by the Akt/mTOR pathway. Neuroreport 2018, 29, 368–379. [Google Scholar] [CrossRef] [Green Version]
- Le, K.; Chibaatar Daliv, E.; Wu, S.; Qian, F.; Ali, A.I.; Yu, D.; Guo, Y. SIRT1-regulated HMGB1 release is partially involved in TLR4 signal transduction: A possible anti-neuroinflammatory mechanism of resveratrol in neonatal hypoxic-ischemic brain injury. Int. Immunopharmacol. 2019, 75, 105779. [Google Scholar] [CrossRef]
- Yu, W.; Tao, M.; Zhao, Y.; Hu, X.; Wang, M. 4′-Methoxyresveratrol Alleviated AGE-Induced Inflammation via RAGE-Mediated NF-κB and NLRP3 Inflammasome Pathway. Molecules 2018, 23, 1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Diaz, A.; Muñoz-Arenas, G.; Caporal-Hernandez, K.; Vázquez-Roque, R.; Lopez-Lopez, G.; Kozina, A.; Espinosa, B.; Flores, G.; Treviño, S.; Guevara, J. Gallic acid improves recognition memory and decreases oxidative-inflammatory damage in the rat hippocampus with metabolic syndrome. Synapse 2020, e22186. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.L.; Hsu, C.C.; Huang, H.J.; Chang, C.J.; Sun, S.H.; Lin, A.M. Gallic Acid Attenuated LPS-Induced Neuroinflammation: Protein Aggregation and Necroptosis. Mol. Neurobiol. 2020, 57, 96–104. [Google Scholar] [CrossRef] [PubMed]
- Ogut, E.; Sekerci, R.; Akcay, G.; Yildirim, F.B.; Derin, N.; Aslan, M.; Sati, L. Protective effects of syringic acid on neurobehavioral deficits and hippocampal tissue damages induced by sub-chronic deltamethrin exposure. Neurotoxicol. Teratol. 2019, 76, 106839. [Google Scholar] [CrossRef] [PubMed]
- Jaroonwitchawan, T.; Chaicharoenaudomrung, N.; Namkaew, J.; Noisa, P. Curcumin attenuates paraquat-induced cell death in human neuroblastoma cells through modulating oxidative stress and autophagy. Neurosci Lett. 2017, 636, 40–47. [Google Scholar] [CrossRef] [PubMed]
- Abrahams, S.; Haylett, W.L.; Johnson, G.; Carr, J.A.; Bardien, S. Antioxidant effects of curcumin in models of neurodegeneration, aging, oxidative and nitrosative stress: A review. Neuroscience 2019, 406, 1–21. [Google Scholar] [CrossRef]
- Bohlmann, J.; Keeling, C.I. Terpenoid biomaterials. Plant J. 2008, 54, 656–669. [Google Scholar] [CrossRef]
- Aguiar, S.; Borowski, T. Neuropharmacological review of the nootropic herb Bacopa monnieri. Rejuvenation Res. 2013, 16, 313–326. [Google Scholar] [CrossRef] [Green Version]
- Srivastav, S.; Fatima, M.; Mondal, A.C. Bacopa monnieri alleviates paraquat induced toxicity in Drosophila by inhibiting jnk mediated apoptosis through improved mitochondrial function and redox stabilization. Neurochem Int. 2018, 121, 98–107. [Google Scholar] [CrossRef]
- Wadhwa, R.; Konar, A.; Kaul, S.C. Nootropic potential of Ashwagandha leaves: Beyond traditional root extracts. Neurochem. Int. 2016, 95, 109–118. [Google Scholar] [CrossRef]
- Dutta, K.; Patel, P.; Julien, J.P. Protective effects of Withania somnifera extract in SOD1G93A mouse model of amyotrophic lateral sclerosis. Exp. Neurol. 2018, 309, 193–204. [Google Scholar] [CrossRef] [PubMed]
- Manjunath, M.J.; Muralidhara. Standardized extract of Withania somnifera (Ashwagandha) markedly offsets rotenone-induced locomotor deficits, oxidative impairments and neurotoxicity in Drosophila melanogaster. J. Food Sci. Technol. 2015, 52, 1971–1981. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, K.S.; Shin, M.; Kim, S.; Lee, S.B. Recent Advances in Studies on the Therapeutic Potential of Dietary Carotenoids in Neurodegenerative Diseases. Oxid. Med. Cell. Longev. 2018, 2018, 4120458. [Google Scholar] [CrossRef] [PubMed]
- Yu, M.; Yan, W.; Beight, C. Lutein and Zeaxanthin Isomers Protect against Light-Induced Retinopathy via Decreasing Oxidative and Endoplasmic Reticulum Stress in BALB/cJ Mice. Nutrients 2018, 10, 842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hussain, G.; Rasul, A.; Anwar, H.; Aziz, N.; Razzaq, A.; Wei, W.; Ali, M.; Li, J.; Li, X. Role of Plant Derived Alkaloids and Their Mechanism in Neurodegenerative Disorders. Int. J. Biol. Sci. 2018, 14, 341–357. [Google Scholar] [CrossRef] [Green Version]
- Qin, S.; Tang, H.; Li, W.; Gong, Y.; Li, S.; Huang, J.; Fang, Y.; Yuan, W.; Liu, Y.; Wang, S.; et al. AMPK and its activator Berberine in the Treatment of Neurodegenerative Diseases. Curr. Pharm. Des. 2020. Epub ahead of print. [Google Scholar] [CrossRef]
- Chen, Y.; Chen, Y.; Liang, Y.; Chen, H.; Ji, X.; Huang, M. Berberine mitigates cognitive decline in an Alzheimer’s Disease Mouse Model by targeting both tau hyperphosphorylation and autophagic clearance. Biomed. Pharmacother. 2020, 121, 109670. [Google Scholar] [CrossRef]
- Huang, M.; Jiang, X.; Liang, Y.; Liu, Q.; Chen, S.; Guo, Y. Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of β-amyloid in APP/tau/PS1 mouse model of Alzheimer’s disease. Exp. Gerontol. 2017, 91, 25–33. [Google Scholar] [CrossRef]
- Chang, C.F.; Lee, Y.C.; Lee, K.H.; Lin, H.C.; Chen, C.L.; Shen, C.J.; Huang, C.C. Therapeutic effect of berberine on TDP-43-related pathogenesis in FTLD and ALS. J. Biomed. Sci. 2016, 23, 72. [Google Scholar] [CrossRef] [Green Version]
- Rusmini, P.; Cristofani, R.; Tedesco, B.; Ferrari, V.; Messi, E.; Piccolella, M.; Casarotto, E.; Chierichetti, M.; Cicardi, M.E.; Galbiati, M.; et al. Enhanced Clearance of Neurotoxic Misfolded Proteins by the Natural Compound Berberine and Its Derivatives. Int. J. Mol. Sci. 2020, 21, 3443. [Google Scholar] [CrossRef]
- Santín-Márquez, R.; Alarcón-Aguilar, A.; López-Diazguerrero, N.E.; Chondrogianni, N.; Königsberg, M. Sulforaphane—Role in aging and neurodegeneration. Geroscience 2019, 41, 655–670. [Google Scholar] [CrossRef]
- Zhou, Q.; Chen, B.; Wang, X.; Wu, L.; Yang, Y.; Cheng, X.; Hu, Z.; Cai, X.; Yang, J.; Sun, X.; et al. Sulforaphane protects against rotenone-induced neurotoxicity in vivo: Involvement of the mTOR, Nrf2, and autophagy pathways. Sci. Rep. 2016, 6, 32206. [Google Scholar] [CrossRef] [Green Version]
- Ren, X.; Wang, N.N.; Qi, H.; Qiu, Y.Y.; Zhang, C.H.; Brown, E.; Kong, H.; Kong, L. Up-Regulation Thioredoxin Inhibits Advanced Glycation End Products-Induced Neurodegeneration. Cell. Physiol. Biochem. 2018, 50, 1673–1686. [Google Scholar] [CrossRef]
- Yang, Y.; Chen, S.; Zhang, Y.; Lin, X.; Song, Y.; Xue, Z.; Qian, H.; Wang, S.; Wan, G.; Zheng, X.; et al. Induction of autophagy by spermidine is neuroprotective via inhibition of caspase 3-mediated Beclin 1 cleavage. Cell Death Dis. 2017, 8, e2738. [Google Scholar] [CrossRef]
- Vijayan, B.; Raj, V.; Nandakumar, S.; Kishore, A.; Thekkuveettil, A. Spermine protects alpha-synuclein expressing dopaminergic neurons from manganese-induced degeneration. Cell. Biol. Toxicol. 2019, 35, 147–159. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, I.; Sankhe, R.; Mudgal, J.; Arora, D.; Nampoothiri, M. Spermidine, an autophagy inducer, as a therapeutic strategy in neurological disorders. Neuropeptides 2020, 102083. [Google Scholar] [CrossRef] [PubMed]
- Limanaqi, F.; Gambardella, S.; Biagioni, F.; Busceti, C.L.; Fornai, F. Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. Oxid. Med. Cell. Longev. 2018, 2018, 4982453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef] [PubMed]
- Said Ahmed, M.; Hung, W.Y.; Zu, J.S.; Hockberger, P.; Siddique, T. Increased reactive oxygen species in familial amyotrophic lateral sclerosis with mutations in SOD1. J. Neurol. Sci. 2000, 176, 88–94. [Google Scholar] [CrossRef]
- Arredondo, F.; Echeverry, C.; Abin-Carriquiry, J.A.; Blasina, F.; Antúnez, K.; Jones, D.P.; Go, Y.M.; Liang, Y.L.; Dajas, F. After cellular internalization, quercetin causes Nrf2 nuclear translocation, increases glutathione levels, and prevents neuronal death against an oxidative insult. Free Radic. Biol. Med. 2010, 49, 738–747. [Google Scholar] [CrossRef]
- Wang, J.; Mao, J.; Wang, R.; Li, S.; Wu, B.; Yuan, Y. Kaempferol Protects Against Cerebral Ischemia Reperfusion Injury Through Intervening Oxidative and Inflammatory Stress Induced Apoptosis. Front. Pharmacol. 2020, 11, 424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hussein, R.M.; Mohamed, W.R.; Omar, H.A. A neuroprotective role of kaempferol against chlorpyrifos-induced oxidative stress and memory deficits in rats via GSK3β-Nrf2 signaling pathway. Pestic. Biochem. Physiol. 2018, 152, 29–37. [Google Scholar] [CrossRef] [PubMed]
- Dhanraj, V.; Karuppaiah, J.; Balakrishnan, R.; Elangovan, N. Myricetin attenuates neurodegeneration and cognitive impairment in Parkinsonism. Front. Biosci (Elite Ed.) 2018, 10, 481–494. [Google Scholar]
- Wu, S.; Yue, Y.; Peng, A.; Zhang, L.; Xiang, J.; Cao, X.; Ding, H.; Yin, S. Myricetin ameliorates brain injury and neurological deficits via Nrf2 activation after experimental stroke in middle-aged rats. Food Funct. 2016, 7, 2624–2634. [Google Scholar] [CrossRef]
- Choi, S.M.; Kim, B.C.; Cho, Y.H.; Choi, K.H.; Chang, J.; Park, M.S.; Kim, M.K.; Cho, K.H.; Kim, J.K. Effects of Flavonoid Compounds on β-amyloid-peptide-induced Neuronal Death in Cultured Mouse Cortical Neurons. Chonnam. Med. J. 2014, 50, 45–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hong, H.; Liu, G.Q. Protection against hydrogen peroxide-induced cytotoxicity in PC12 cells by scutellarin. Life Sci. 2004, 74, 2959–2973. [Google Scholar] [CrossRef]
- Chao, X.J.; Chen, Z.W.; Liu, A.M.; He, X.X.; Wang, S.G.; Wang, Y.T.; Liu, P.Q.; Ramassamy, C.; Mak, S.H.; Cui, W.; et al. Effect of tacrine-3-caffeic acid, a novel multifunctional anti-Alzheimer’s dimer, against oxidative-stress-induced cell death in HT22 hippocampal neurons: Involvement of Nrf2/HO-1 pathway. CNS Neurosci. Ther. 2014, 20, 840–850. [Google Scholar] [CrossRef] [PubMed]
- Morroni, F.; Sita, G.; Graziosi, A.; Turrini, E.; Fimognari, C.; Tarozzi, A.; Hrelia, P. Neuroprotective Effect of Caffeic Acid Phenethyl Ester in A Mouse Model of Alzheimer’s Disease Involves Nrf2/HO-1 Pathway. Aging Dis. 2018, 9, 605–622. [Google Scholar] [CrossRef] [Green Version]
- Lee, T.K.; Kang, I.J.; Kim, B.; Sim, H.J.; Kim, D.W.; Ahn, J.H.; Lee, J.C.; Ryoo, S.; Shin, M.C.; Cho, J.H.; et al. Experimental Pretreatment with Chlorogenic Acid Prevents Transient Ischemia-Induced Cognitive Decline and Neuronal Damage in the Hippocampus through Anti-Oxidative and Anti-Inflammatory Effects. Molecules 2020, 25, 3578. [Google Scholar] [CrossRef]
- Zhang, X.; Wu, Q.; Lu, Y.; Wan, J.; Dai, H.; Zhou, X.; Lv, S.; Chen, X.; Zhang, X.; Hang, C.; et al. Cerebroprotection by salvianolic acid B after experimental subarachnoid hemorrhage occurs via Nrf2- and SIRT1-dependent pathways. Free Radic. Biol. Med. 2018, 124, 504–516. [Google Scholar] [CrossRef] [PubMed]
- Xia, X.J.; Lian, Y.G.; Zhao, H.Y.; Xu, Q.L. Curcumin protects from oxidative stress and inhibits α-synuclein aggregation in MPTP induced parkinsonian mice. Int. J. Clin. Exp. Med. 2016, 9, 2654–2665. [Google Scholar]
- Cui, Q.; Li, X.; Zhu, H. Curcumin ameliorates dopaminergic neuronal oxidative damage via activation of the Akt/Nrf2 pathway. Mol. Med. Rep. 2016, 13, 1381–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palle, S.; Neerati, P. Improved neuroprotective effect of resveratrol nanoparticles as evinced by abrogation of rotenone-induced behavioral deficits and oxidative and mitochondrial dysfunctions in rat model of Parkinson’s disease. Naunyn. Schmiedebergs Arch. Pharmacol. 2018, 391, 445–453. [Google Scholar] [CrossRef] [PubMed]
- Bhardwaj, P.; Jain, C.K.; Mathur, A. Comparative evaluation of four triterpenoid glycoside saponins of bacoside A in alleviating sub-cellular oxidative stress of N2a neuroblastoma cells. J. Pharm. Pharmacol. 2018, 70, 1531–1540. [Google Scholar] [CrossRef] [PubMed]
- Parihar, P.; Shetty, R.; Ghafourifar, P.; Parihar, M.S. Increase in oxidative stress and mitochondrial impairment in hypothalamus of streptozotocin treated diabetic rat: Antioxidative effect of Withania somnifera. Cell. Mol. Biol. (Noisy-le-grand). 2016, 62, 73–83. [Google Scholar]
- Ademowo, O.S.; Dias, I.H.K.; Diaz-Sanchez, L.; Sanchez-Aranguren, L.; Stahl, W.; Griffiths, H.R. Partial Mitigation of Oxidized Phospholipid-Mediated Mitochondrial Dysfunction in Neuronal Cells by Oxocarotenoids. J. Alzheimers Dis. 2020, 74, 113–126. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wen, C.; Huang, C.; Yang, M.; Fan, C.; Li, Q.; Zhao, J.; Gan, D.; Li, A.; Zhu, L.; Lu, D. The Secretion from Bone Marrow Mesenchymal Stem Cells Pretreated with Berberine Rescues Neurons with Oxidative Damage Through Activation of the Keap1-Nrf2-HO-1 Signaling Pathway. Neurotox. Res. 2020, 38, 59–73. [Google Scholar] [CrossRef]
- Zhang, C.; Li, C.; Chen, S.; Li, Z.; Jia, X.; Wang, K.; Bao, J.; Liang, Y.; Wang, X.; Chen, M.; et al. Berberine protects against 6-OHDA-induced neurotoxicity in PC12 cells and zebrafish through hormetic mechanisms involving PI3K/AKT/Bcl-2 and Nrf2/HO-1 pathways. Redox Biol. 2017, 11, 1–11. [Google Scholar] [CrossRef] [Green Version]
- Sharma, S.; Kumar, P.; Deshmukh, R. Neuroprotective potential of spermidine against rotenone induced Parkinson’s disease in rats. Neurochem. Int. 2018, 116, 104–111. [Google Scholar] [CrossRef]
- Costa, L.G.; de Laat, R.; Dao, K.; Pellacani, C.; Cole, T.B.; Furlong, C.E. Paraoxonase-2 (PON2) in brain and its potential role in neuroprotection. Neurotoxicology 2014, 43, 3–9. [Google Scholar] [CrossRef]
- Cohen-Kaplan, V.; Ciechanover, A.; Livneh, I. p62 at the crossroad of the ubiquitin-proteasome system and autophagy. Oncotarget 2016, 7, 83833–83834. [Google Scholar] [CrossRef] [PubMed]
- Minakaki, G.; Menges, S.; Kittel, A.; Emmanouilidou, E.; Schaeffner, I.; Barkovits, K.; Bergmann, A.; Rockenstein, E.; Adame, A.; Marxreiter, F.; et al. Autophagy inhibition promotes SNCA/alpha-synuclein release and transfer via extracellular vesicles with a hybrid autophagosome-exosome-like phenotype. Autophagy 2018, 14, 98–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Menon, M.B.; Dhamija, S. Beclin 1 Phosphorylation - at the Center of Autophagy Regulation. Front. Cell. Dev. Biol. 2018, 6, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fornai, F.; Longone, P.; Ferrucci, M.; Lenzi, P.; Isidoro, C.; Ruggieri, S.; Paparelli, A. Autophagy and amyotrophic lateral sclerosis: The multiple roles of lithium. Autophagy 2008, 4, 527–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, J.; Tan, S.H.; Nicolas, V.; Bauvy, C.; Yang, N.D.; Zhang, J.; Xue, Y.; Codogno, P.; Shen, H.M. Activation of lysosomal function in the course of autophagy via mTORC1 suppression and autophagosome-lysosome fusion. Cell Res. 2013, 23, 508–523. [Google Scholar] [CrossRef] [Green Version]
- Lenzi, P.; Lazzeri, G.; Biagioni, F.; Busceti, C.L.; Gambardella, S.; Salvetti, A.; Fornai, F. The Autophagoproteasome a Novel Cell Clearing Organelle in Baseline and Stimulated Conditions. Front. Neuroanat. 2016, 10, 78. [Google Scholar] [CrossRef] [Green Version]
- Wan, F.Y.; Wang, Y.N.; Zhang, G.J. The influence of oxidation of membrane thiol groups on lysosomal proton permeability. Biochem. J. 2001, 360, 355–362. [Google Scholar] [CrossRef]
- Dodson, M.; Wani, W.Y.; Redmann, M.; Benavides, G.A.; Johnson, M.S.; Ouyang, X.; Cofield, S.S.; Mitra, K.; Darley-Usmar, V.; Zhang, J. Regulation of autophagy, mitochondrial dynamics, and cellular bioenergetics by 4-hydroxynonenal in primary neurons. Autophagy 2017, 13, 1828–1840. [Google Scholar] [CrossRef] [Green Version]
- Janda, E.; Lascala, A.; Carresi, C.; Parafati, M.; Aprigliano, S.; Russo, V.; Savoia, C.; Ziviani, E.; Musolino, V.; Morani, F.; et al. Parkinsonian toxin-induced oxidative stress inhibits basal autophagy in astrocytes via NQO2/quinone oxidoreductase 2: Implications for neuroprotection. Autophagy 2015, 11, 1063–1080. [Google Scholar] [CrossRef]
- He, X.; Yuan, W.; Li, Z.; Hou, Y.; Liu, F.; Feng, J. 6-Hydroxydopamine induces autophagic flux dysfunction by impairing transcription factor EB activation and lysosomal function in dopaminergic neurons and SH-SY5Y cells. Toxicol. Lett. 2018, 283, 58–68. [Google Scholar] [CrossRef]
- Mitter, S.K.; Song, C.; Qi, X.; Mao, H.; Rao, H.; Akin, D.; Lewin, A.; Grant, M.; Dunn, W., Jr.; Ding, J.; et al. Dysregulated autophagy in the RPE is associated with increased susceptibility to oxidative stress and AMD. Autophagy 2014, 10, 1989–2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dhingra, A.; Bell, B.A.; Peachey, N.S.; Daniele, L.L.; Reyes-Reveles, J.; Sharp, R.C.; Jun, B.; Bazan, N.G.; Sparrow, J.R.; Kim, H.J.; et al. Microtubule-Associated Protein 1 Light Chain 3B, (LC3B) Is Necessary to Maintain Lipid-Mediated Homeostasis in the Retinal Pigment Epithelium. Front. Cell. Neurosci. 2018, 12, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, Y.D.; Zhao, J.J. TFEB Participates in the Aβ-Induced Pathogenesis of Alzheimer’s Disease by Regulating the Autophagy-Lysosome Pathway. DNA Cell Biol. 2015, 34, 661–668. [Google Scholar] [CrossRef] [PubMed]
- Daniele, S.; Mangano, G.; Durando, L.; Ragni, L.; Martini, C. The Nootropic Drug A-Glyceryl-Phosphoryl-Ethanolamine Exerts Neuroprotective Effects in Human Hippocampal Cells. Int. J. Mol. Sci. 2020, 21, 941. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Karsli-Uzunbas, G.; Poillet-Perez, L.; Sawant, A.; Hu, Z.S.; Zhao, Y.; Moore, D.; Hu, W.; White, E. Autophagy promotes mammalian survival by suppressing oxidative stress and p53. Genes Dev. 2020, 34, 688–700. [Google Scholar] [CrossRef]
- Ichimura, Y.; Waguri, S.; Sou, Y.S.; Kageyama, S.; Hasegawa, J.; Ishimura, R.; Saito, T.; Yang, Y.; Kouno, T.; Fukutomi, T.; et al. Phosphorylation of p62 activates the Keap1-Nrf2 pathway during selective autophagy. Mol. Cell. 2013, 51, 618–631. [Google Scholar] [CrossRef] [Green Version]
- Taguchi, K.; Fujikawa, N.; Komatsu, M.; Ishii, T.; Unno, M.; Akaike, T.; Motohashi, H.; Yamamoto, M. Keap1 degradation by autophagy for the maintenance of redox homeostasis. Proc. Natl. Acad. Sci. USA 2012, 109, 13561–13566. [Google Scholar] [CrossRef] [Green Version]
- Di Martino, R.M.C.; Pruccoli, L.; Bisi, A.; Gobbi, S.; Rampa, A.; Martinez, A.; Pérez, C.; Martinez-Gonzalez, L.; Paglione, M.; Di Schiavi, E.; et al. Novel Curcumin-Diethyl Fumarate Hybrid as a Dualistic GSK-3β Inhibitor/Nrf2 Inducer for the Treatment of Parkinson’s Disease. ACS Chem. Neurosci. 2020, 11, 2728–2740. [Google Scholar] [CrossRef]
- Gao, J.; Wang, L.; Liu, J.; Xie, F.; Su, B.; Wang, X. Abnormalities of Mitochondrial Dynamics in Neurodegenerative Diseases. Antioxidants 2017, 6, 25. [Google Scholar] [CrossRef]
- Panov, A.V.; Dikalov, S.I. Cardiolipin, Perhydroxyl Radicals, and Lipid Peroxidation in Mitochondrial Dysfunctions and Aging. Oxid. Med. Cell. Longev. 2020, 2020, 1323028. [Google Scholar] [CrossRef]
- He, Y.; Jia, K.; Li, L.; Wang, Q.; Zhang, S.; Du, J.; Du, H. Salvianolic acid B attenuates mitochondrial stress against Aβ toxicity in primary cultured mouse neurons. Biochem. Biophys. Res. Commun. 2018, 498, 1066–1072. [Google Scholar] [CrossRef] [PubMed]
- Ruffoli, R.; Bartalucci, A.; Frati, A.; Fornai, F. Ultrastructural studies of ALS mitochondria connect altered function and permeability with defects of mitophagy and mitochondriogenesis. Front. Cell. Neurosci. 2015, 9, 341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palikaras, K.; Tavernarakis, N. Mitochondrial homeostasis: The interplay between mitophagy and mitochondrial biogenesis. Exp. Gerontol. 2014, 56, 182–188. [Google Scholar] [CrossRef] [PubMed]
- Ferese, R.; Lenzi, P.; Fulceri, F.; Biagioni, F.; Fabrizi, C.; Gambardella, S.; Familiari, P.; Frati, A.; Limanaqi, F.; Fornai, F. Quantitative Ultrastructural Morphometry and Gene Expression of mTOR-Related Mitochondriogenesis within Glioblastoma Cells. Int. J. Mol. Sci. 2020, 21, 4570. [Google Scholar] [CrossRef]
- Valero, T. Mitochondrial biogenesis: Pharmacological approaches. Curr. Pharm. Des. 2014, 20, 5507–5509. [Google Scholar] [CrossRef]
- Chin, L.S.; Olzmann, J.A.; Li, L. Parkin-mediated ubiquitin signalling in aggresome formation and autophagy. Biochem. Soc. Trans. 2010, 38, 144–149. [Google Scholar] [CrossRef] [Green Version]
- Nichols, M.; Zhang, J.; Polster, B.M.; Elustondo, P.A.; Thirumaran, A.; Pavlov, E.V.; Robertson, G.S. Synergistic neuroprotection by epicatechin and quercetin: Activation of convergent mitochondrial signaling pathways. Neuroscience 2015, 308, 75–94. [Google Scholar] [CrossRef]
- Cakir, Z.; Funk, K.; Lauterwasser, J.; Todt, F.; Zerbes, R.M.; Oelgeklaus, A.; Tanaka, A.; van der Laan, M.; Edlich, F. Parkin promotes proteasomal degradation of misregulated BAX. J. Cell Sci. 2017, 130, 2903–2913. [Google Scholar] [CrossRef] [Green Version]
- Li, S.; Sun, X.; Xu, L.; Sun, R.; Ma, Z.; Deng, X.; Liu, B.; Fu, Q.; Qu, R.; Ma, S. Baicalin attenuates in vivo and in vitro hyperglycemia-exacerbated ischemia/reperfusion injury by regulating mitochondrial function in a manner dependent on AMPK. Eur. J. Pharmacol. 2017, 815, 118–126. [Google Scholar] [CrossRef]
- Wang, W.; Xu, J. Curcumin Attenuates Cerebral Ischemia-reperfusion Injury through Regulating Mitophagy and Preserving Mitochondrial Function. Curr. Neurovasc. Res. 2020, 17, 113–122. [Google Scholar] [CrossRef]
- Chen, Y.; Chen, J.; Sun, X.; Shi, X.; Wang, L.; Huang, L.; Zhou, W. Evaluation of the neuroprotective effect of EGCG: A potential mechanism of mitochondrial dysfunction and mitochondrial dynamics after subarachnoid hemorrhage. Food Funct. 2018, 9, 6349–6359. [Google Scholar] [CrossRef]
- Yang, X.; Zhang, M.; Dai, Y.; Sun, Y.; Aman, Y.; Xu, Y.; Yu, P.; Zheng, Y.; Yang, J.; Zhu, X. Spermidine inhibits neurodegeneration and delays aging via the PINK1-PDR1-dependent mitophagy pathway in C. elegans. Aging (Albany NY) 2020, 12, 16852–16866. [Google Scholar] [CrossRef]
- Hang, W.; He, B.; Chen, J.; Xia, L.; Wen, B.; Liang, T.; Wang, X.; Zhang, Q.; Wu, Y.; Chen, Q.; et al. Berberine Ameliorates High Glucose-Induced Cardiomyocyte Injury via AMPK Signaling Activation to Stimulate Mitochondrial Biogenesis and Restore Autophagic Flux. Front. Pharmacol. 2018, 9, 1121. [Google Scholar] [CrossRef] [PubMed]
- Toti, L.; Bartalucci, A.; Ferrucci, M.; Fulceri, F.; Lazzeri, G.; Lenzi, P.; Soldani, P.; Gobbi, P.; La Torre, A.; Gesi, M. High-intensity exercise training induces morphological and biochemical changes in skeletal muscles. Biol. Sport. 2013, 30, 301–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davinelli, S.; De Stefani, D.; De Vivo, I.; Scapagnini, G. Polyphenols as Caloric Restriction Mimetics Regulating Mitochondrial Biogenesis and Mitophagy. Trends Endocrinol. Metab. 2020, 31, 536–550. [Google Scholar] [CrossRef] [PubMed]
- Jellinger, K.A. Interaction between pathogenic proteins in neurodegenerative disorders. J. Cell. Mol. Med. 2012, 16, 1166–1183. [Google Scholar] [CrossRef]
- Higashi, S.; Iseki, E.; Yamamoto, R.; Minegishi, M.; Hino, H.; Fujisawa, K.; Togo, T.; Katsuse, O.; Uchikado, H.; Furukawa, Y.; et al. Concurrence of TDP-43, tau and alpha-synuclein pathology in brains of Alzheimer’s disease and dementia with Lewy bodies. Brain Res. 2007, 1184, 284–294. [Google Scholar] [CrossRef]
- Colom-Cadena, M.; Gelpi, E.; Charif, S.; Belbin, O.; Blesa, R.; Martí, M.J.; Clarimón, J.; Lleó, A. Confluence of α-synuclein, tau, and β-amyloid pathologies in dementia with Lewy bodies. J. Neuropathol. Exp. Neurol. 2013, 72, 1203–1212. [Google Scholar] [CrossRef] [Green Version]
- Charles, V.; Mezey, E.; Reddy, P.H.; Dehejia, A.; Young, T.A.; Polymeropoulos, M.H.; Brownstein, M.J.; Tagle, D.A. α-synuclein immunoreactivity of huntingtin polyglutamine aggregates in striatum and cortex of Huntington’s disease patients and transgenic mouse models. Neurosci. Lett. 2000, 289, 29–32. [Google Scholar] [CrossRef]
- Takei, Y.; Oguchi, K.; Koshihara, H.; Hineno, A.; Nakamura, A.; Ohara, S. α-Synuclein coaggregation in familial amyotrophic lateral sclerosis with SOD1 gene mutation. Hum. Pathol. 2013, 44, 1171–1176. [Google Scholar] [CrossRef]
- Ling, S.C.; Polymenidou, M.; Cleveland, D.W. Converging mechanisms in ALS and FTD: Disrupted RNA and protein homeostasis. Neuron 2013, 79, 416–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Trist, B.G.; Davies, K.M.; Cottam, V.; Genoud, S.; Ortega, R.; Roudeau, S.; Carmona, A.; De Silva, K.; Wasinger, V.; Lewis, S.J.G.; et al. Amyotrophic lateral sclerosis-like superoxide dismutase 1 proteinopathy is associated with neuronal loss in Parkinson’s disease brain. Acta Neuropathol 2017, 134, 113–127. [Google Scholar] [CrossRef] [PubMed]
- Ferrucci, M.; Ryskalin, L.; Biagioni, F.; Gambardella, S.; Busceti, C.L.; Falleni, A.; Lazzeri, G.; Fornai, F. Methamphetamine increases Prion Protein and induces dopamine-dependent expression of protease resistant PrPsc. Arch. Ital. Biol. 2017, 155, 81–97. [Google Scholar] [CrossRef] [PubMed]
- da Luz, M.H.; Peres, I.T.; Santos, T.G.; Martins, V.R.; Icimoto, M.Y.; Lee, K.S. Dopamine induces the accumulation of insoluble prion protein and affects autophagic flux. Front. Cell. Neurosci. 2015, 9, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martins, D.; English, A.M. SOD1 oxidation and formation of soluble aggregates in yeast: Relevance to sporadic ALS development. Redox Biol. 2014, 2, 632–639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shamoto-Nagai, M.; Hisaka, S.; Naoi, M.; Maruyama, W. Modification of α-synuclein by lipid peroxidation products derived from polyunsaturated fatty acids promotes toxic oligomerization: Its relevance to Parkinson disease. J. Clin. Biochem. Nutr. 2018, 62, 207–212. [Google Scholar] [CrossRef] [Green Version]
- Vicente, M.H.; Outeiro, T.F. The sour side of neurodegenerative disorders: The effects of protein glycation. J. Pathol. 2010, 221, 13–25. [Google Scholar]
- Adrover, M.; Mariño, L.; Sanchis, P.; Pauwels, K.; Kraan, Y.; Lebrun, P.; Vilanova, B.; Muñoz, F.; Broersen, K.; Donoso, J. Mechanistic insights in glycation-induced protein aggregation. Biomacromolecules 2014, 15, 3449–3462. [Google Scholar] [CrossRef]
- Zhu, M.; Han, S.; Fink, A.L. Oxidized quercetin inhibits α-synuclein fibrillization. Biochim. Biophys. Acta 2013, 1830, 2872–2881. [Google Scholar] [CrossRef]
- Medvedeva, M.; Barinova, K.; Melnikova, A.; Semenyuk, P.; Kolmogorov, V.; Gorelkin, P.; Erofeev, A.; Muronetz, V. Naturally occurring cinnamic acid derivatives prevent amyloid transformation of alpha-synuclein. Biochimie 2020, 170, 128–139. [Google Scholar] [CrossRef]
- Taebnia, N.; Morshedi, D.; Yaghmaei, S.; Aliakbari, F.; Rahimi, F.; Arpanaei, A. Curcumin-loaded amine-functionalized mesoporous silica nanoparticles inhibit α-synuclein fibrillation and reduce its cytotoxicity-associated effects. Langmuir 2016, 32, 13394–13402. [Google Scholar] [CrossRef] [PubMed]
- Šneideris, T.; Baranauskiene, L.; Cannon, J.G.; Rutkiene, R.; Meškys, R.; Smirnovas, V. Looking for a generic inhibitor of amyloid-like fibril formation among flavone derivatives. Peer J. 2015, 3, e1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Carver, J.A.; Calabrese, A.N.; Pukala, T.L. Gallic acid interacts with α-synuclein to prevent the structural collapse necessary for its aggregation. Biochim. Biophys. Acta 2014, 1844, 1481–1485. [Google Scholar] [CrossRef] [PubMed]
- Bieschke, J.; Russ, J.; Friedrich, R.P.; Ehrnhoefer, D.E.; Wobst, H.; Neugebauer, K.; Wanker, E.E. EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and reduces cellular toxicity. Proc. Natl. Acad. Sci. USA 2010, 107, 7710–7715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caughey, B.; Raymond, L.D.; Raymond, G.J.; Maxson, L.; Silveira, J.; Baron, G.S. Inhibition of protease-resistant prion protein accumulation in vitro by curcumin. J. Virol. 2003, 77, 5499–5502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hafner-Bratkovic, I.; Gaspersic, J.; Smid, L.M.; Bresjanac, M.; Jerala, R. Curcumin binds to the alpha-helical intermediate and to the amyloid form of prion protein—A new mechanism for the inhibition of PrPSc accumulation. J. Neurochem. 2008, 104, 1553–1564. [Google Scholar] [CrossRef] [PubMed]
- Lin, C.F.; Yu, K.H.; Jheng, C.P.; Chung, R.; Lee, C.I. Curcumin reduces amyloid fibrillation of prion protein and decreases reactive oxidative stress. Pathogens 2013, 2, 506–519. [Google Scholar] [CrossRef]
- Ladner-Keay, C.L.; Ross, L.; Perez-Pineiro, R.; Zhang, L.; Bjorndahl, T.C.; Cashman, N.; Wishart, D.S. A simple in vitro assay for assessing the efficacy, mechanisms and kinetics of anti-prion fibril compounds. Prion 2018, 12, 280–300. [Google Scholar] [CrossRef]
- Ono, K.; Hirohata, M.; Yamada, M. Ferulic acid destabilizes preformed beta-amyloid fibrils in vitro. Biochem. Biophys. Res. Commun. 2005, 336, 444–449. [Google Scholar] [CrossRef]
- Ono, K.; Yoshiike, Y.; Takashima, A.; Hasegawa, K.; Naiki, H.; Yamada, M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: Implications for the prevention and therapeutics of Alzheimer’s disease. J. Neurochem. 2003, 87, 172–181. [Google Scholar] [CrossRef]
- Hamaguchi, T.; Ono, K.; Murase, A.; Yamada, M. Phenolic compounds prevent Alzheimer’s pathology through different effects on the amyloid-beta aggregation pathway. Am. J. Pathol. 2009, 175, 2557–2565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ladiwala, A.R.A.; Lin, J.C.; Bale, S.S.; Marcelino-Cruz, A.M.; Bhattacharya, M.; Dordick, J.S.; Tessier, P.M. Resveratrol selectively remodels soluble oligomers and fibrils of amyloid a beta into off-pathway conformers. J. Biol. Chem. 2010, 285, 24228–24237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karuppagounder, S.S.; Pinto, J.T.; Xu, H.; Chen, H.L.; Beal, M.F.; Gibson, G.E. Dietary supplementation with resveratrol reduces plaque pathology in a transgenic model of Alzheimer’s disease. Neurochem. Int. 2009, 54, 111–118. [Google Scholar] [CrossRef] [Green Version]
- Hou, T.T.; Yang, H.Y.; Wang, W.; Wu, Q.Q.; Tian, Y.R.; Jia, J.P. Sulforaphane Inhibits the Generation of Amyloid-β Oligomer and Promotes Spatial Learning and Memory in Alzheimer’s Disease (PS1V97L) Transgenic Mice. J. Alzheimers. Dis. 2018, 62, 1803–1813. [Google Scholar] [CrossRef] [PubMed]
- Phan, H.T.T.; Samarat, K.; Takamura, Y.; Azo-Oussou, A.F.; Nakazono, Y.; Vestergaard, M.C. Polyphenols Modulate Alzheimer’s Amyloid Beta Aggregation in a Structure-Dependent Manner. Nutrients 2019, 11, 756. [Google Scholar] [CrossRef] [Green Version]
- Habtemariam, S. Molecular Pharmacology of Rosmarinic and Salvianolic Acids: Potential Seeds for Alzheimer’s and Vascular Dementia Drugs. Int. J. Mol. Sci. 2018, 19, 458. [Google Scholar] [CrossRef] [Green Version]
- Dariya, B.; Nagaraju, G.P. Advanced glycation end products in diabetes, cancer and phytochemical therapy. Drug Discov. Today 2020, 25, 1614–1623. [Google Scholar] [CrossRef]
- Rebollo-Hernanz, M.; Fernández-Gómez, B.; Herrero, M.; Aguilera, Y.; Martín-Cabrejas, M.A.; Uribarri, J.; Del Castillo, M.D. Inhibition of the Maillard Reaction by Phytochemicals Composing an Aqueous Coffee Silverskin Extract via a Mixed Mechanism of Action. Foods 2019, 8, 438. [Google Scholar] [CrossRef] [Green Version]
- Jiang, T.F.; Zhang, Y.J.; Zhou, H.Y.; Wang, H.M.; Tian, L.P.; Liu, J.; Ding, J.Q.; Chen, S.D. Curcumin ameliorates the neurodegenerative pathology in A53T α-synuclein cell model of Parkinson’s disease through the downregulation of mTOR/p70S6K signaling and the recovery of macroautophagy. J. Neuroimmune Pharmacol. 2013, 8, 356–369. [Google Scholar] [CrossRef]
- Buratta, S.; Chiaradia, E.; Tognoloni, A.; Gambelunghe, A.; Meschini, C.; Palmieri, L.; Muzi, G.; Urbanelli, L.; Emiliani, C.; Tancini, B. Effect of Curcumin on Protein Damage Induced by Rotenone in Dopaminergic PC12 Cells. Int. J. Mol. Sci. 2020, 21, 2761. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Maiti, P.; Rossignol, J.; Dunbar, G.L. Curcumin Modulates Molecular Chaperones and Autophagy-Lysosomal Pathways In Vitro after Exposure to Aβ42. J. Alzheimers Dis. Parkinsonism. 2017, 7, 299. [Google Scholar] [CrossRef] [Green Version]
- Sharma, N.; Nehru, B. Curcumin affords neuroprotection and inhibits α-synuclein aggregation in lipopolysaccharide-induced Parkinson’s disease model. Inflammopharmacology 2018, 26, 349–360. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Wei, W.; Gaertig, M.A.; Li, S.; Li, X.-J. Therapeutic Effect of Berberine on Huntington’s Disease Transgenic Mouse Model. PLoS ONE 2015, 10, e0134142. [Google Scholar] [CrossRef]
- Deng, J.; Koutras, C.; Donnelier, J.; Alshehri, M.; Fotouhi, M.; Girard, M.; Casha, S.; McPherson, P.S.; Robbins, S.M.; Braun, J.E.A. Neurons Export Extracellular Vesicles Enriched in Cysteine String Protein and Misfolded Protein Cargo. Sci. Rep. 2017, 7, 956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lyman, M.; Lloyd, D.G.; Ji, X.; Vizcaychipi, M.P.; Ma, D. Neuroinflammation: The role and consequences. Neurosci. Res. 2014, 79, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Ugras, S.; Daniels, M.J.; Fazelinia, H.; Gould, N.S.; Yocum, A.K.; Luk, K.C.; Luna, E.; Ding, H.; McKennan, C.; Seeholzer, S.; et al. Induction of the immunoproteasome subunit Lmp7 links proteostasis and immunity in α-synuclein aggregation disorders. EBioMedicine 2018, 31, 307–319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cebrián, C.; Zucca, F.A.; Mauri, P.; Steinbeck, J.A.; Studer, L.; Scherzer, C.R.; Kanter, E.; Budhu, S.; Mandelbaum, J.; Vonsattel, J.P.; et al. MHC-I expression renders catecholaminergic neurons susceptible to T-cell-mediated degeneration. Nat. Commun. 2014, 5, 3633. [Google Scholar] [CrossRef]
- Wagner, L.K.; Gilling, K.E.; Schormann, E.; Kloetzel, P.M.; Heppner, F.L.; Krüger, E.; Prokop, S. Immunoproteasome deficiency alters microglial cytokine response and improves cognitive deficits in Alzheimer’s disease-like APPPS1 mice. Acta Neuropathol. Commun. 2017, 5, 52. [Google Scholar] [CrossRef]
- Xie, X.; Bi, H.L.; Lai, S.; Zhang, Y.L.; Li, N.; Cao, H.J.; Han, L.; Wang, H.X.; Li, H.H. The immunoproteasome catalytic β5i subunit regulates cardiac hypertrophy by targeting the autophagy protein ATG5 for degradation. Sci. Adv. 2019, 5, eaau0495. [Google Scholar] [CrossRef] [Green Version]
- Chen, C.; Zou, L.X.; Lin, Q.Y.; Yan, X.; Bi, H.L.; Xie, X.; Wang, S.; Wang, Q.S.; Zhang, Y.L.; Li, H.H. Resveratrol as a new inhibitor of immunoproteasome prevents PTEN degradation and attenuates cardiac hypertrophy after pressure overload. Redox Biol. 2019, 20, 390–401. [Google Scholar] [CrossRef] [PubMed]
- Deretic, V.; Levine, B. Autophagy balances inflammation in innate immunity. Autophagy 2018, 14, 243–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xue, Z.; Zhang, Z.; Liu, H.; Li, W.; Guo, X.; Zhang, Z.; Liu, Y.; Jia, L.; Li, Y.; Ren, Y.; et al. lincRNA-Cox2 regulates NLRP3 inflammasome and autophagy mediated neuroinflammation. Cell Death Differ. 2019, 26, 130–145. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Jamwal, S.; Kumar, P. Neuroprotective potential of Quercetin in combination with piperine against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity. Neural Regen. Res. 2017, 12, 1137–1144. [Google Scholar] [CrossRef]
- Cordaro, M.; Siracusa, R.; Crupi, R.; Impellizzeri, D.; Peritore, A.F.; D’Amico, R.; Gugliandolo, E.; di Paola, R.; Cuzzocrea, S. 2-Pentadecyl-2-Oxazoline Reduces Neuroinflammatory Environment in the MPTP Model of Parkinson Disease. Mol. Neurobiol. 2018, 55, 9251–9266. [Google Scholar] [CrossRef]
- Du. Y.: Luo. M.: Du. Y.: Xu, M.; Yao, Q.; Wang, K.; He, G. Liquiritigenin Decreases Aβ Levels and Ameliorates Cognitive Decline by Regulating Microglia M1/M2 Transformation in AD Mice. Neurotox. Res. 2020. Epub ahead of print. [Google Scholar] [CrossRef]
- Chen, L.; Pan, H.; Bai, Y.; Li, H.; Yang, W.; Lin, Z.X.; Cui, W.; Xian, Y.F. Gelsemine, a natural alkaloid extracted from Gelsemium elegans Benth. alleviates neuroinflammation and cognitive impairments in Aβ oligomer-treated mice. Psychopharmacology (Berl). 2020, 237, 2111–2124. [Google Scholar] [CrossRef]
- Yang, E.J.; Mahmood, U.; Kim, H.; Choi, M.; Choi, Y.; Lee, J.P.; Cho, J.Y.; Hyun, J.W.; Kim, Y.S.; Chang, M.J.; et al. Phloroglucinol ameliorates cognitive impairments by reducing the amyloid β peptide burden and pro-inflammatory cytokines in the hippocampus of 5XFAD mice. Free Radic. Biol. Med. 2018, 126, 221–234. [Google Scholar] [CrossRef]
- Mattioli, R.; Francioso, A.; d’Erme, M.; Trovato, M.; Mancini, P.; Piacentini, L.; Casale, A.M.; Wessjohann, L.; Gazzino, R.; Costantino, P.; et al. Anti-Inflammatory Activity of A Polyphenolic Extract from Arabidopsis thaliana in In Vitro and In Vivo Models of Alzheimer’s Disease. Int. J. Mol. Sci. 2019, 20, 708. [Google Scholar] [CrossRef] [Green Version]
- Impellizzeri, D.; Cordaro, M.; Bruschetta, G.; Siracusa, R.; Crupi, R.; Esposito, E.; Cuzzocrea, S. N-Palmitoylethanolamine-Oxazoline as a New Therapeutic Strategy to Control Neuroinflammation: Neuroprotective Effects in Experimental Models of Spinal Cord and Brain Injury. J. Neurotrauma 2017, 34, 2609–2623. [Google Scholar] [CrossRef]
- Gugliandolo, E.; D’Amico, R.; Cordaro, M.; Fusco, R.; Siracusa, R.; Crupi, R.; Impellizzeri, D.; Cuzzocrea, S.; Di Paola, R. Neuroprotective Effect of Artesunate in Experimental Model of Traumatic Brain Injury. Front. Neurol. 2018, 9, 590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gao, Y.; Zhuang, Z.; Lu, Y.; Tao, T.; Zhou, Y.; Liu, G.; Wang, H.; Zhang, D.; Wu, L.; Dai, H.; et al. Curcumin Mitigates Neuro-Inflammation by Modulating Microglia Polarization Through Inhibiting TLR4 Axis Signaling Pathway Following Experimental Subarachnoid Hemorrhage. Front. Neurosci. 2019, 13, 1223. [Google Scholar] [CrossRef] [PubMed]
- Fusco, R.; Scuto, M.; Cordaro, M.; D’Amico, R.; Gugliandolo, E.; Siracusa, R.; Peritore, A.F.; Crupi, R.; Impellizzeri, D.; Cuzzocrea, S.; et al. N-Palmitoylethanolamide-Oxazoline Protects against Middle Cerebral Artery Occlusion Injury in Diabetic Rats by Regulating the SIRT1 Pathway. Int. J. Mol. Sci. 2019, 20, 4845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caltagirone, C.; Cisari, C.; Schievano, C.; Di Paola, R.; Cordaro, M.; Bruschetta, G.; Esposito, E.; Cuzzocrea, S.; Stroke Study Group. Co-ultramicronized Palmitoylethanolamide/Luteolin in the Treatment of Cerebral Ischemia: From Rodent to Man. Transl. Stroke Res. 2016, 7, 54–69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bozkurt, A.A.; Mustafa, G.; Tarık, A.; Adile, O.; Murat, S.H.; Mesut, K.; Yıldıray, K.; Coskun, S.; Murat, C. Syringaldehyde exerts neuroprotective effect on cerebral ischemia injury in rats through anti-oxidative and anti-apoptotic properties. Neural Regen. Res. 2014, 9, 1884–1890. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.; Ali, T.; Rehman, S.U.; Khan, M.S.; Alam, S.I.; Ikram, M.; Muhammad, T.; Saeed, K.; Badshah, H.; Kim, M.O. Neuroprotective Effect of Quercetin Against the Detrimental Effects of LPS in the Adult Mouse Brain. Front. Pharmacol. 2018, 9, 1383. [Google Scholar] [CrossRef]
- Koza, L.A.; Winter, A.N.; Holsopple, J.; Baybayon-Grandgeorge, A.N.; Pena, C.; Olson, J.R.; Mazzarino, R.C.; Patterson, D.; Linseman, D.A. Protocatechuic Acid Extends Survival, Improves Motor Function, Diminishes Gliosis, and Sustains Neuromuscular Junctions in the hSOD1G93A Mouse Model of Amyotrophic Lateral Sclerosis. Nutrients 2020, 12, 1824. [Google Scholar] [CrossRef]
- Winter, A.N.; Ross, E.K.; Wilkins, H.M.; Stankiewicz, T.R.; Wallace, T.; Miller, K.; Linseman, D.A. An anthocyanin-enriched extract from strawberries delays disease onset and extends survival in the hSOD1G93A mouse model of amyotrophic lateral sclerosis. Nutr. Neurosci. 2018, 21, 414–426. [Google Scholar] [CrossRef]
- Huang, Q.; Ye, X.; Wang, L.; Pan, J. Salvianolic acid B abolished chronic mild stress-induced depression through suppressing oxidative stress and neuro-inflammation via regulating NLRP3 inflammasome activation. J. Food Biochem. 2019, 43, e12742. [Google Scholar] [CrossRef]
- Yu, H.; Zhang, F.; Guan, X. Baicalin reverse depressive-like behaviors through regulation SIRT1-NF-kB signalling pathway in olfactory bulbectomized rats. Phytother. Res. 2019, 33, 1480–1489. [Google Scholar] [CrossRef]
- Limanaqi, F.; Biagioni, F.; Busceti, C.L.; Polzella, M.; Fabrizi, C.; Fornai, F. Potential Antidepressant Effects of Scutellaria baicalensis, Hericium erinaceus and Rhodiola rosea. Antioxidants 2020, 9, 234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Testa, G.; Gamba, P.; Badilli, U.; Gargiulo, S.; Maina, M.; Guina, T.; Calfapietra, S.; Biasi, F.; Cavalli, R.; Poli, G.; et al. Loading into nanoparticles improves quercetin’s efficacy in preventing neuroinflammation induced by oxysterols. PLoS ONE 2014, 9, e96795. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Silswal, N.; Reddy, N.S.; Qureshi, A.A.; Qureshi, N. Resveratrol Downregulates Biomarkers of Sepsis Via Inhibition of Proteasome’s Proteases. Shock 2018, 5, 579–588. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.D.; Xie, S.P.; Saw, W.T.; Ho, P.G.H.; Wang, H.; Lei, Z.; Yi, Z.; Tan, E.K. The Therapeutic Implications of Tea Polyphenols Against Dopamine (DA) Neuron Degeneration in Parkinson’s Disease (PD). Cells 2019, 8, 911. [Google Scholar] [CrossRef] [Green Version]
- Farkhondeh, T.; Pourbagher-Shahri, A.M.; Ashrafizadeh, M.; Folgado, S.L.; Rajabpour-Sanati, A.; Khazdair, M.R.; Samarghandian, S. Green tea catechins inhibit microglial activation which prevents the development of neurological disorders. Neural Regen. Res. 2020, 15, 1792–1798. [Google Scholar]
- Bai, D.; Jin, G.; Zhang, D.; Zhao, L.; Wang, M.; Zhu, Q.; Zhu, L.; Sun, Y.; Liu, X.; Chen, X.; et al. Natural silibinin modulates amyloid precursor protein processing and amyloid-β protein clearance in APP/PS1 mice. J. Physiol. Sci. 2019, 69, 643–652. [Google Scholar] [CrossRef] [Green Version]
Models of Neuroinflammation | Anti-Inflammatory Phytochemicals |
---|---|
Parkinson’s disease (PD) SNCA- and LPS-induced neurodegeneration in mice [53] Cells transfected with C-terminal modified α-syn and BAC-α-syn-GFP transgenic mice [54] 6-OHDA-treated rats [95] MPTP-treated rats [235] and mice [236] | Kaempferol [53] Dihydromyricetin and Salvianolic acid [54] Silymarin [95] Quercetin [235] 2-pentadecyl-2-oxazoline [236] |
Alzheimer’s disease (AD) APP/PS1 transgenic mice [237] Aβ oligomer-treated mice [238] 5XFAD mice [239] Aβ25⁻35 peptide-treated BV2 murine microglia cells and human Aβ1⁻42-expressing flies [240] | Liquiritigenin [237] Gelsemine [238] Phloroglucinol [239] Arabidopsis extract (caffeic acid, kaempferol, quercetin, synapic acid, luteolin) [240] |
Traumatic Brain Injury (TBI) | 2-pentadecyl-2-oxazoline [241] Artemisin [242] |
Spinal Cord Injury (SCI) | Berberine [67] Curcumin [57] 2-pentadecyl-2-oxazoline [241] |
Brain hemorrhage Prechiasmatic cistern blood injection [243] and internal carotid rupture [101] and/or Brain ischemia/hypoperfusion Middle Cerebral Artery Occlusion (MCAO) [93,244,245,246] | Curcumin [243] Genistein [93] Resveratrol [101] 2-pentadecyl-2-oxazoline [244] Palmitoylethanolamide-luteolin [245] Syringaldehyde [246] |
Lipopolysaccharide (LPS) | Salvianolic acid [66] Resveratrol [69] Quercetin [247] |
Amyotrophic Lateral Sclerosis (ALS) SOD1G93A mice [112,248,249] | Ashwagandha [112] Protocatechuic acid [248] Anthocyanin-enriched strawberries extract [249] |
Anxiety and depression Chronic mild stress, and olfactory bulbectomized rat model of depression [250,251] | Salvianolic acid [250] Baicalin [251,252] |
Phytochemical Experimental Model Polyphenols | Anti-Oxidant Effects | Mitochondrial Protection | Anti-Apoptotic Effects | Proteostasis | Anti-Inflammatory Effects | Autophagy-Related Effects | |
---|---|---|---|---|---|---|---|
Quercetin | PD [50,81,82,235]; AD [51,222]; Metal neurotoxicity [83]; LPS [247] | ↓LDH ↓ROS ↓MDA ↑GSH [51,81,82,235] | ↑MMP ↓ultrastructural alterations [82,83] | ↓caspase-3 ↓BAX/Bcl-2 ↓CHOP ↓DNA fragmentation [51,158,159,160,161,247] | Aβ [51,211,222] α-syn [200] | ↓Reactive gliosis (GFAP) ↓TLR-4 ↓COX-2 ↓IL-1β, IL-6, TNF-α [235,247] | ↑LC3II ↑Beclin-1 ↓P62 ↑LAMP-2 ↑flux [50,51,81,82,222] |
Kaempferol | PD [52,53,84]; Ischemia [85,132] | ↓ROS ↓SDH ↓MDA ↑NRF2 ↑SOD ↑GSH [52,84,85,132] | ↑MMP ↑ATP ↑mitophagy ↓cyt-c [52,85] | ↓caspase-3/9 ↓JNK/p38MAPK [52] | SOD-1 [84] Aβ [211] | ↓GFAP ↓NLRP3 ↓TNF-α, IL-1β [53,132] | ↑LC3II ↓P62 ↑flux ↑AMPK [52,53,84,85] |
(Dihydro)-Myricetin | PD [54,134]; LPS [86]; Ischemia [135] | ↓ROS ↓MDA, LPO ↑NRF2 ↑SOD ↑GSH ↑CAT [134,135] | ↑MMP ↑ATP ↓cyt-c [134,135] | ↓caspase-3/9 ↓JNK/p38MAPK ↓BAX/Bcl-2 [86,134] | α-syn [54] PrP [209] Aβ [211] | ↓GFAP ↓iNOS, COX-2, ↓PGE2 ↓IL-1β, TNF-α [54,86] | ↑LC3II ↑LAMP-1/2A [54] |
Scutellarin | LPS [88]; Chronic hypoperfusion [89]; H2O2 neurotoxicity [137] | ↓ROS ↓MDA ↓LDH ↑NRF2 ↑SOD ↑GSH [88,137] | ↑MMP [137] | ↓sub-G1 peak in flow cytometry [137] | Aβ [89,203] α-syn [203] | ↓NF-κB ↓ IL-1β, TNF-α ↓IBA-1 microgliosis [88,89] | ↑LC3II ↑Beclin-1 ↓P62 ↓mTOR [88] |
Baicalein | PD [55,87]; SCI [56]; Ischemia [180] | ↓ROS [180] | ↑mitophagy ↑MMP ↓fission [55,180] | ↓caspase-3/9/12 ↓BAX/Bcl-2 [55,56,87,180] | α-syn [87] | ↓Microglial inflammasome ↓IL-1β [87] | ↑PI3K ↑LC3II ↑Beclin-1 ↓P62 ↑AMPK [55,56,180] |
Apigenin | AMD [61]; LPS [90]; | ↑NRF2 ↑HO-1, NQO-1 ↑SOD, GSH-Px ↓ROS, MDA [61] | - | ↓ ERK, STAT3 [90] | - | ↓Astrocyte activation ↓NF-κB ↓IL-31 and IL-33 [90] | ↑LC3II ↓P62 [61] |
Catechins | PD [50,255]; Prion [63]; AD [91]; Chronic stress [92]; Brain hemorrhage [182] | ↓ROS ↑NRF2 ↑PGC-1α [91,182,255] | ↑mitophagy ↓fragmentation ↓mtDNA copy number ↓cyt-c [63,182] | ↓BAX/Bcl-2 ↓TUNEL [63,92] | α-syn [203,205,255] p-tau [91] Aβ [92,203,205,211,216] | ↑M2 microglia polarization ↓NLRP3 ↓NF-κB ↓IL-1β, TNF-α ↓iNOS ↓COX-2 [256] | ↑LC3II ↓P62 ↑SIRT-1 ↓mTOR ↑flux [50,63,92,182] |
Genistein | Ischemia [93]; H2O2 neurotoxicity [94] | ↓LDH, ROS [93,94] | - | ↓BAX/Bcl-2 ↓caspase-1/3/9 ↓JNK, ERK [93,94] | - | ↓NLRP3 in microglia and neurons ↓NF-κB ↓IL-1β, TNF-α [93,94] | - |
Silymarin/ Silibinin | PD [95,96]; AD [257] | ↑SOD, GSH-Px, CAT ↓MDA [95,96,257] | - | ↓BAX/Bcl-2 ↓caspase-3/9 ↓TLR4 [95] | APP and Aβ [257] | - | - |
Resveratrol | PD [50,58,144]; Chronic hypoperfusion [65]; LPS [69]; Ischemia [97,101]; AD [99,214]; Brain hemorrhage [100]; AGEs-induced neuroinflammation [102] | ↓ROS ↓MDA ↑SOD, GSH, CAT [58,59,65,97,99,144] | ↓fragmentation ↑MMP ↑mitophagy ↑Complex-I activity [58,99,144] | ↓TUNEL ↓BAX/Bcl-2 ↓caspase-3/9 ↓ p38 and JNK [65,97,99,100] | polyQ-Htt [59] PrP [209] Aβ [213,214] | ↑M2 microglia polarization ↓STAT3/6 ↑PGC-1α ↓TLR-4/RAGE— NF-κB ↓HMGB1, NLRP3 ↓iNOS, COX-2 [69,101,102] | ↑ATG4 ↑LC3II ↓P62 ↑flux ↓Akt/mTOR ↑SIRT1 [50,58,59,65,99,100,101] |
Syringic/gallic acid | Metabolic syndrome [103]; LPS [104]; Deltamethrin neurotoxicity [105] | ↓ROS ↑SOD, CAT [103,105] | - | ↓caspase-3 [105] | α-syn [104,204] | ↓Reactive gliosis (↓GFAP, ED-1) ↓iNOS, IL-1β, TNF-α [103] | - |
Caffeic and chlorogenic acids | PD [62]; Spinocerebellar ataxia [98]; Glutamate excitotoxicity [138]; AD [139]; Ischemia [140] | ↓ROS ↑NRF2 ↑SOD [98,138,139,140] | ↑MMP [138] | ↓caspase-9 [139] | α-syn [62] polyQ-ataxin3 [98] | ↓Reactive gliosis (↓GFAP, ED-1) ↓IL-2, TNF-α [139,140] | ↑JNK/Bcl-2 ↑LC3II ↓P62 [62,98] |
Rosmarinic and Salvianolic acid | PD [54]; LPS [66]; Brain hemorrhage [141]; AD [172] | ↓ROS ↓MDA ↑SOD, GSH-Px, ↑GSH, CAT ↑NRF2 ↑HO-1, NQO-1 [141,172] | ↓fragmentation ↑MMP ↑ATP [172] | - | α-syn [54] Aβ [212] | ↓GFAP and IBA-1 ↓NLRP3 ↓IL-1β, IL6, and TNF-α [54,66] | ↑LC3II ↑Beclin-1 ↑LAMP-1/2A [54,66] |
Curcumin | PD [50,70,106,142,143,220,224]; SCI [57]; Ischemia [181]; AD [223]; Brain hemorrhage [243] | ↓ROS ↓MDA ↑GSH ↑NRF2 [70,106,142,143,224] | ↑MMP ↑ATP ↑mitophagy [70,181] | ↓BAX/Bcl-2 Caspase-3/9 [106,224] | APP and Aβ [106,223] α-syn [142,202,220,224] PrP [206,207,208,209] | ↓Gliosis ↓iNOS ↓TNF-α, IL-1β, IL-1α ↓TLR4 ↑M2 microglia polarization [57,224,243] | ↑LC3II ↓P62 ↓Akt/mTOR ↑TFEB [50,57,70,106,181,220,223] |
Terpenes | |||||||
Bacosides | Benzo[a]pyrene [71]; PD [110]; H2O2 neurotoxicity [145] | ↓ROS ↑SOD, CAT ↑NRF2 [71,110,145] | ↓ cyt-c ↑mitophagy ↑ATP ↑MMP [71,110,145] | ↓TUNEL, Annexin-V ↓JNK ↓caspase-3 [71,110,145] | - | - | ↑LC3II, Beclin-1, ATG5, ULK1 [71] |
Withanolides | ALS [111]; PD [112]; Diabetes [146] | ↓ROS, LPO ↑GSH, SOD [112,146] | ↑complex I–III and complex II–III activity ↓mitochondrial permeabilization [112,146] | - | SOD-1 [111] | ↓Gliosis (↓GFAP and IBA-1) ↓NF-κB ↓COX-2 [111] | ↑LC3II ↓ P62 [111] |
Carotenoids | AMD [115]; AD [147] | ↓ROS ↑GSH ↑NRF2 [115,147] | ↓mitochondrial uncoupling [147] | ↓JNK and ↓ER stress [115] | - | - | - |
Alkaloids | |||||||
Berberine | SCI [67]; AD [118,119]; ALS/FTD [120]; SBMA [121]; tert-butyl hydroperoxide neurotoxicity [148]; PD [149]; HD [225] | ↓ROS ↑NRF2—HO-1 [148,149] | ↑MMP [148] | ↓caspase-3 ↓BAX/Bcl-2 [67,148,149] | p-tau [118] Aβ, APP [119] TDP-43 [120] ARpolyQ [121] polyQ-Htt [225] | ↓IL-1β, TNF-α [67] | ↑LC3B, ATG16L, and ATG7 ↓P62 ↓GSK3β ↓mTOR ↑ⅢPI3K/Beclin-1 [67,118,119] |
Caffeine | Prion [60] | - | - | ↓JNK ↓DNA strand breakage [60] | PrP [60] | - | ↑LC3II ↑flux [60] |
Other | |||||||
Sulforaphane | Prion [64]; PD [123]; Diabetes [124]; AD [215] | ↓ROS, LDH, MDA ↑NRF-2, GSH, NQO1, THx [64,123,124] | - | ↓TUNEL, Annexin-V ↓caspase-3 [64,123] | PrP [64] AGE in retina [124] Aβ [215] | - | ↑LC3II ↓P62 ↑AMPK ↓mTOR [64,123] |
Spermine/ Spermidine | Ischemia [125]; PD [126,150,183]; AD [183] | ↓MDA ↑GSH [150] | ↑mitophagy ↓ cyt-c [125,183] | ↓caspase-3 [125] | α-syn [126] | ↓TNF-α, IL-1β, IL-6 [150] | ↑LC3II ↑Beclin-1 ↑flux [125,126] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Limanaqi, F.; Biagioni, F.; Mastroiacovo, F.; Polzella, M.; Lazzeri, G.; Fornai, F. Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation. Antioxidants 2020, 9, 1022. https://doi.org/10.3390/antiox9101022
Limanaqi F, Biagioni F, Mastroiacovo F, Polzella M, Lazzeri G, Fornai F. Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation. Antioxidants. 2020; 9(10):1022. https://doi.org/10.3390/antiox9101022
Chicago/Turabian StyleLimanaqi, Fiona, Francesca Biagioni, Federica Mastroiacovo, Maico Polzella, Gloria Lazzeri, and Francesco Fornai. 2020. "Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation" Antioxidants 9, no. 10: 1022. https://doi.org/10.3390/antiox9101022
APA StyleLimanaqi, F., Biagioni, F., Mastroiacovo, F., Polzella, M., Lazzeri, G., & Fornai, F. (2020). Merging the Multi-Target Effects of Phytochemicals in Neurodegeneration: From Oxidative Stress to Protein Aggregation and Inflammation. Antioxidants, 9(10), 1022. https://doi.org/10.3390/antiox9101022