Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression?
Abstract
:1. Introduction
2. Evidence Supporting Enhanced Oxidative Stress in PD
2.1. Mitochondrial Dysfunction
2.2. Iron Dysregulation
2.3. Dopamine Quinone
3. Antioxidants
3.1. Endogenous Low-Molecular Weight Molecules
3.1.1. Coenzyme Q
3.1.2. Urate
3.1.3. Glutathione
3.2. Dietary Antioxidants
3.2.1. Vitamin C
3.2.2. Vitamin E
3.2.3. Polyphenols and Flavonoids
3.3. Bioactive Natural Herbal Extracts
3.4. Synthetic Antioxidants
Cyclic Nitroxides
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Wakabayashi, K.; Tanji, K.; Odagiri, S.; Miki, Y.; Mori, F.; Takahashi, H. The Lewy Body in Parkinson’s Disease and Related Neurodegenerative Disorders. Mol. Neurobiol. 2012, 47, 495–508. [Google Scholar] [CrossRef] [PubMed]
- Davie, C. A review of Parkinson’s disease. Br. Med. Bull. 2008, 86, 109–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Korczyn, A. Drug treatment of Parkinson’s disease. Dialogues Clin. Neurosci. 2004, 6, 315–322. [Google Scholar] [CrossRef]
- Nakabeppu, Y.; Tsuchimoto, D.; Yamaguchi, H.; Sakumi, K. Oxidative damage in nucleic acids and Parkinson’s disease. J. Neurosci. Res. 2007, 85, 919–934. [Google Scholar] [CrossRef] [PubMed]
- Bosco, D.; Fowler, D.; Zhang, Q.; Nieva, J.; Powers, E.; Wentworth, P., Jr.; Lerner, R.A.; Kelly, J.W. Elevated levels of oxidized cholesterol metabolites in Lewy body disease brains accelerate α-synuclein fibrilization. Nat. Chem. Biol. 2006, 2, 346. [Google Scholar] [CrossRef]
- Zeevalk, G.; Razmpour, R.; Bernard, L. Glutathione and Parkinson’s disease: Is this the elephant in the room? Biomed. Pharmacother. 2008, 62, 236–249. [Google Scholar] [CrossRef] [PubMed]
- Ott, M.; Gogvadze, V.; Orrenius, S.; Zhivotovsky, B. Mitochondria, oxidative stress and cell death. Apoptosis 2007, 12, 913–922. [Google Scholar] [CrossRef] [PubMed]
- Hunot, S.; Brugg, B.; Ricard, D.; Michel, P.; Muriel, M.; Ruberg, M.; Faucheux, B.A.; Agid, Y.; Hirsch, E.C. Nuclear translocation of NF- B is increased in dopaminergic neurons of patients with Parkinson disease. Proc. Natl. Acad. Sci. USA 1997, 94, 7531–7536. [Google Scholar] [CrossRef] [Green Version]
- Jenner, P. Oxidative stress in Parkinson’s disease. Ann. Neurol. 2003, 53, S26–S38. [Google Scholar] [CrossRef]
- Hwang, O. Role of Oxidative Stress in Parkinson’s Disease. Exp. Neurobiol. 2013, 22, 11–17. [Google Scholar] [CrossRef] [Green Version]
- Betarbet, R.; Sherer, T.; Greenamyre, J. Animal models of Parkinson’s disease. Bioessays 2002, 24, 308–318. [Google Scholar] [CrossRef]
- Devi, L.; Raghavendran, V.; Prabhu, B.; Avadhani, N.; Anandatheerthavarada, H. Mitochondrial Import and Accumulation of α-Synuclein Impair Complex I in Human Dopaminergic Neuronal Cultures and Parkinson Disease Brain. J. Biol. Chem. 2008, 283, 9089–9100. [Google Scholar] [CrossRef] [Green Version]
- Palacino, J.; Sagi, D.; Goldberg, M.; Krauss, S.; Motz, C.; Wacker, M.; Klose, J.; Shen, J. Mitochondrial Dysfunction and Oxidative Damage inparkin-deficient Mice. J. Biol. Chem. 2004, 279, 18614–18622. [Google Scholar] [CrossRef] [Green Version]
- Gandhi, S.; Wood-Kaczmar, A.; Yao, Z.; Plun-Favreau, H.; Deas, E.; Klupsch, K.; Downward, J.; Latchman, D.S.; Tabrizi, S.J.; Wood, N.W.; et al. PINK1-Associated Parkinson’s Disease Is Caused by Neuronal Vulnerability to Calcium-Induced Cell Death. Mol. Cell. 2009, 33, 627–638. [Google Scholar] [CrossRef] [Green Version]
- Irrcher, I.; Aleyasin, H.; Seifert, E.; Hewitt, S.; Chhabra, S.; Phillips, M.; Lutz, A.K.; Rousseaux, M.W.C.; Bevilacqua, L.; Jahani-Asl, A.; et al. Loss of the Parkinson’s disease-linked gene DJ-1 perturbs mitochondrial dynamics. Hum. Mol. Genet. 2010, 19, 3734–3746. [Google Scholar] [CrossRef] [Green Version]
- Parker, W.; Boyson, S.; Parks, J. Abnormalities of the electron transport chain in idiopathic parkinson’s disease. Ann. Neurol. 1989, 26, 719–723. [Google Scholar] [CrossRef]
- Bender, A.; Krishnan, K.; Morris, C.; Taylor, G.; Reeve, A.; Perry, R.; Jaros, E.; Hersheson, J.S.; Betts, J.; Klopstock, T.; et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat. Genet. 2006, 38, 515–517. [Google Scholar] [CrossRef]
- Bilgic, B.; Pfefferbaum, A.; Rohlfing, T.; Sullivan, E.; Adalsteinsson, E. MRI estimates of brain iron concentration in normal aging using quantitative susceptibility mapping. Neuroimage 2012, 59, 2625–2635. [Google Scholar] [CrossRef] [Green Version]
- Hare, D.; Double, K. Iron and dopamine: A toxic couple. Brain 2016, 139, 1026–1035. [Google Scholar] [CrossRef]
- Dexter, D.; Carayon, A.; Javoy-Agid, F.; Agid, Y.; Wells, F.; Daniel, S.E.; Lees, A.J.; Jenner, P.; Marsden, C.D. Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s disease and other neurodegenerative diseases affecting the basal ganglia. Brain 1991, 114, 1953–1975. [Google Scholar] [CrossRef]
- James, S.A.; Roberts, B.R.; Hare, D.J.; de Jonge, M.D.; Birchall, I.E.; Jenkins, N.L.; Cherny, R.A.; Bush, A.I.; McColl, G. Direct in vivo imaging of ferrous iron dyshomeostasis in ageing Caenorhabditis elegans. Chem. Sci. 2015, 6, 2952–2962. [Google Scholar] [CrossRef] [Green Version]
- Ayton, S.; Lei, P.; Hare, D.J.; Duce, J.A.; George, J.L.; Adlard, P.A.; McLean, C.; Rogers, J.T.; Cherny, R.A.; Finkelstein, D.I.; et al. Parkinson’s Disease Iron Deposition Caused by Nitric Oxide-Induced Loss of -Amyloid Precursor Protein. J. Neurosci. 2015, 35, 3591–3597. [Google Scholar] [CrossRef] [Green Version]
- Wong, B.; Tsatsanis, A.; Lim, L.; Adlard, P.; Bush, A.; Duce, J. β-Amyloid Precursor Protein Does Not Possess Ferroxidase Activity but Does Stabilize the Cell Surface Ferrous Iron Exporter Ferroportin. PLoS ONE 2015, 9, e114174. [Google Scholar] [CrossRef] [Green Version]
- Napolitano, A.; Pezzella, A.; Prota, G. New Reaction Pathways of Dopamine under Oxidative Stress Conditions: Nonenzymatic Iron-Assisted Conversion to Norepinephrine and the Neurotoxins 6-Hydroxydopamine and 6,7-Dihydroxytetrahydroisoquinoline. Chem. Res. Toxicol. 1999, 12, 1090–1097. [Google Scholar] [CrossRef]
- Blum, D.; Torch, S.; Lambeng, N.; Nissou, M.; Benabid, A.; Sadoul, R.; Verna, J. Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: Contribution to the apoptotic theory in Parkinson’s disease. Prog. Neurobiol. 2001, 65, 135–172. [Google Scholar] [CrossRef]
- Conway, K.; Rochet, J.; Bieganski, R.; Lansbury, P. Kinetic Stabilization of the alpha -Synuclein Protofibril by a Dopamine-alpha -Synuclein Adduct. Science 2001, 294, 1346–1349. [Google Scholar] [CrossRef]
- Lotharius, J.; Brundin, P. Impaired dopamine storage resulting from alphα-synuclein mutations may contribute to the pathogenesis of Parkinson’s disease. Hum. Mol. Genet. 2002, 11, 2395–2407. [Google Scholar] [CrossRef] [Green Version]
- Berman, S.; Hastings, T. Dopamine Oxidation Alters Mitochondrial Respiration and Induces Permeability Transition in Brain Mitochondria. J. Neurochem. 2001, 73, 1127–1137. [Google Scholar] [CrossRef]
- Zecca, L.; Wilms, H.; Geick, S.; Claasen, J.; Brandenburg, L.; Holzknecht, C.; Panizza, M.L.; Zucca, F.A.; Deuschl, G.; Sievers, J.; et al. Human neuromelanin induces neuroinflammation and neurodegeneration in the rat substantia nigra: Implications for Parkinson’s disease. Acta Neuropathol. 2008, 116, 47–55. [Google Scholar] [CrossRef]
- Hernández-Camacho, J.D.; Bernier, M.; López-Lluch, G.; Navas, P. Coenzyme Q10 Supplementation in Aging and Disease. Front. Physiol. 2018, 5, 9–44. [Google Scholar] [CrossRef] [Green Version]
- Navas, P.; Villalba, J.M.; de Cabo, R. The importance of plasma membrane coenzyme Q in aging and stress responses. Mitochondrion 2007, 7, S34–S40. [Google Scholar] [CrossRef] [PubMed]
- Bentinger, M.; Brismar, K.; Dallner, G. The antioxidant role of coenzyme Q. Mitochondrion 2007, 7, S41–S50. [Google Scholar] [CrossRef] [PubMed]
- Mischley, L.K.; Allen, J.; Bradley, R. Coenzyme Q10 deficiency in patients with Parkinson’s disease. J. Neurol. Sci. 2012, 318, 72–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sohmiya, M.; Tanaka, M.; Tak, N.W.; Yanagisawa, M.; Tanino, Y.; Suzuki, Y.; Okamoto, K.; Yamamoto, Y. Redox status of plasma coenzyme Q10 indicates elevated systemic oxidative stress in Parkinson’s disease. J. Neurol. Sci. 2004, 223, 161–166. [Google Scholar] [CrossRef] [PubMed]
- Hargreaves, I.P.; Lane, A.; Sleiman, P.M. The coenzyme Q10 status of the brain region of Parkinson’s disease patients. Neurosci. Lett. 2008, 447, 17–19. [Google Scholar] [CrossRef] [PubMed]
- Götz, M.E.; Gerstner, A.; Harth, R.; Dirr, A.; Janetzky, B.; Kuhn, W.; Riederer, P.; Gerlach, M. Altered redox state of platelet coenzyme Q10 in Parkinson’s disease. J. Neural Transm. 2000, 107, 41–48. [Google Scholar]
- Shults, C.W.; Haas, R.H.; Passov, D.; Beal, M.F. Coenzyme Q10 levels correlate with the activities of complexes I and II/III in mitochondria from parkinsonian and nonparkinsonian subjects. Ann. Neurol. 1997, 42, 261–264. [Google Scholar] [CrossRef]
- Isobe, C.; Abe, T.; Terayama, Y. Levels of reduced and oxidized coenzyme Q-10 and 8-hydroxy-2’-deoxyguanosine in the cerebrospinal fluid of patients with living Parkinson’s disease demonstrate that mitochondrial oxidative damage and/or oxidative DNA damage contributes to the neurodegenerative process. Neurosci. Lett. 2010, 469, 159–163. [Google Scholar]
- McCarthy, S.; Somayajulu, M.; Sikorska, M.; Borowy-Borowski, H.; Pandey, S. Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble Coenzyme Q10. Toxicol. Appl. Pharmacol. 2004, 201, 21–31. [Google Scholar] [CrossRef]
- Somayajulu, M.; McCarthy, S.; Hung, M.; Sikorska, M.; Borowy-Borowski, H.; Pandey, S. Role of mitochondria in neuronal cell death induced by oxidative stress; neuroprotection by Coenzyme Q10. Neurobiol. Dis. 2005, 18, 618–627. [Google Scholar] [CrossRef]
- Horvath, T.L.; Diano, S.; Leranth, C.; Garcia-Segura, L.M.; Cowley, M.A.; Shanabrough, M.; Elsworth, J.D.; Sotonyi, P.; Roth, R.H.; Dietrich, E.H.; et al. Coenzyme Q induces nigral mitochondrial uncoupling and prevents dopamine cell loss in a primate model of Parkinson’s disease. Endocrinology 2003, 144, 2757–2760. [Google Scholar] [CrossRef] [Green Version]
- Attia, H.N.; Maklad, Y.A. Neuroprotective effects of coenzyme Q10 on paraquat-induced Parkinson’s disease in experimental animals. Behav. Pharmacol. 2018, 29, 79–86. [Google Scholar] [CrossRef]
- Ghasemloo, E.; Mostafavi, H.; Hosseini, M.; Forouzandeh, M.; Eskandari, M.; Mousavi, S.S. Neuroprotective effects of coenzyme Q10 in Parkinson’s model via a novel Q10/miR-149-5p/MMPs pathway. Metab. Brain Dis. 2021, 36, 2089–2100. [Google Scholar] [CrossRef]
- Shults, C.W.; Beal, M.F.; Fontaine, D.; Nakano, K.; Haas, R.H. Absorption, tolerability, and effects on mitochondrial activity of oral coenzyme Q10 in parkinsonian patients. Neurology 1998, 50, 793–795. [Google Scholar] [CrossRef]
- Shults, C.W.; Oakes, D.; Kieburtz, K.; Beal, M.F.; Haas, R.; Plumb, S.; Juncos, J.L.; Nutt, J.; Shoulson, I.; Carter, J.; et al. Effects of coenzyme Q10 in early Parkinson disease: Evidence of slowing of the functional decline. Arch. Neurol. 2002, 59, 1541–1550. [Google Scholar] [CrossRef]
- McGarry, A.; McDermott, M.; Kieburtz, K.; de Blieck, E.A.; Beal, F.; Marder, K.; Ross, C.; Shoulson, I.; Gilbert, P.; Mallonee, W.M.; et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology 2017, 88, 152–159. [Google Scholar] [CrossRef] [Green Version]
- Parkinson Study Group QE3 Investigators; Beal, M.F.; Oakes, D.; Shoulson, I.; Henchcliffe, C.; Galpern, W.R.; Haas, R.; Juncos, J.L.; Nutt, J.G.; Voss, T.S.; et al. A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: No evidence of benefit. JAMA Neurol. 2014, 71, 543–552. [Google Scholar] [CrossRef]
- Fahn, S.; Elton, R. Members of the UPDRS Development Committee. Unified Parkinson’s Disease Rating Scale. In Recent Developments in Parkinson’s Disease; Fahn, S., Marsden, C.D., Calne, D.B., Goldstein, M., Eds.; Macmillan Health Care Information: Florham Park, NJ, USA, 1987; Volume 2, pp. 153–163. [Google Scholar]
- Goetz, C.G.; Tilley, B.C. Movement Disorder Society UPDRS Revision Task Force. Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): Scale presentation and clinimetric testing result. Mov. Disord. 2008, 23, 2129–2170. [Google Scholar] [CrossRef]
- Negida, A.; Menshawy, A.; El Ashal, G.; Elfouly, Y.; Hani, Y.; Hegazy, Y.; El Ghonimy, S.; Fouda, S.; Rashad, Y. Coenzyme Q10 for Patients with Parkinson’s Disease: A Systematic Review and Meta-Analysis. CNS Neurol. Disord.-Drug Targets 2016, 15, 45–53. [Google Scholar] [CrossRef]
- Zhu, Z.G.; Sun, M.X.; Zhang, W.L.; Wang, W.W.; Jin, Y.M.; Xie, C.L. The efficacy and safety of coenzyme Q10 in Parkinson’s disease: A meta-analysis of randomized controlled trials. Neurol. Sci. 2017, 38, 215–224. [Google Scholar] [CrossRef]
- Park, H.W.; Park, C.G.; Park, M.; Lee, S.H.; Park, H.R.; Lim, J.; Paek, S.H.; Choy, Y.B. Intrastriatal administration of coenzyme Q10 enhances neuroprotection in a Parkinson’s disease rat model. Sci. Rep. 2020, 10, 9572. [Google Scholar] [CrossRef]
- Cleren, C.; Yang, L.; Lorenzo, B.; Calingasan, N.Y.; Schomer, A.; Sireci, A.; Wille, E.J.; Beal, M.F. Therapeutic effects of coenzyme Q10 (CoQ10) and reduced CoQ10 in the MPTP model of Parkinsonism. J. Neurochem. 2008, 104, 1613–1621. [Google Scholar] [CrossRef]
- Yoritaka, A.; Kawajiri, S.; Yamamoto, Y.; Nakahara, T.; Ando, M.; Hashimoto, K.; Nagase, M.; Saito, Y.; Hattori, N. Randomized, double-blind, placebo-controlled pilot trial of reduced coenzyme Q10 for Parkinson’s disease. Parkinsonism Relat. Disord. 2015, 21, 911–916. [Google Scholar] [CrossRef]
- Prasuhn, J.; Brüggemannm, N.; Hessler, N.; Berg, D.; Gasser, T.; Brockmann, K.; Olbrich, D.; Ziegler, A.; König, I.R.; Klein, C.; et al. An omics-based strategy using coenzyme Q10 in patients with Parkinson’s disease: Concept evaluation in a double-blind randomized placebo-controlled parallel group trial. Neurol. Res. Pract. 2019, 1, 31. [Google Scholar] [CrossRef] [Green Version]
- Yeum, K.J.; Russell, R.M.; Krinsky, N.I.; Aldini, G. Biomarkers of antioxidant capacity in the hydrophilic and lipophilic compartments of human plasma. Arch. Biochem. Biophys. 2004, 430, 97–103. [Google Scholar] [CrossRef]
- Ames, B.N.; Cathcart, R.; Schwiers, E.; Hochstein, P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: A hypothesis. Proc. Natl. Acad. Sci. USA 1981, 78, 6858–6862. [Google Scholar] [CrossRef] [Green Version]
- Muraoka, S.; Miura, T. Inhibition by uric acid of free radicals that damage biological molecules. Pharm. Toxicol. 2003, 93, 284–289. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Wu, G.; Schwarzschild, M.A. Urate in Parkinson’s disease: More than a biomarker? Curr. Neurol. Neurosci. Rep. 2012, 12, 367–375. [Google Scholar] [CrossRef] [PubMed]
- Ascherio, A.; LeWitt, P.A.; Xu, K.; Eberly, S.; Watts, A.; Matson, W.R.; Marras, C.; Kieburtz, K.; Rudolph, A.; Bogdanov, M.B.; et al. Urate as a predictor of the rate of clinical decline in Parkinson disease. Arch. Neurol. 2009, 66, 1460–1468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, C.; Guo, Y.; Luo, W.; Lin, C.; Ding, M. Serum urate and the risk of Parkinson’s disease: Results from a meta-analysis. Can. J. Neurol. Sci. 2013, 40, 73–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, T.G.; Wang, X.X.; Luo, W.F.; Zhang, Q.L.; Huang, T.T.; Xu, X.S.; Liu, C.F. Protective effects of urate against 6-OHDA-induced cell injury in PC12 cells through antioxidant action. Neurosci. Lett. 2012, 506, 175–179. [Google Scholar] [CrossRef]
- Zhang, N.; Shu, H.Y.; Huang, T.; Zhang, Q.L.; Li, D.; Zhang, G.Q.; Peng, X.Y.; Liu, C.F.; Luo, W.F.; Hu, L.F. Nrf2 signaling contributes to the neuroprotective effects of urate against 6-OHDA toxicity. PLoS ONE 2014, 9, e100286. [Google Scholar]
- Bakshi, R.; Zhang, H.; Logan, R.; Joshi, I.; Xu, Y.; Chen, X.; Schwarzschild, M.A. Neuroprotective effects of urate are mediated by augmenting astrocytic glutathione synthesis and release. Neurobiol. Dis. 2015, 82, 574–579. [Google Scholar] [CrossRef] [Green Version]
- Cipriani, S.; Desjardins, C.A.; Burdett, T.C.; Xu, Y.; Xu, K.; Schwarzschild, M.A. Protection of dopaminergic cells by urate requires its accumulation in astrocytes. J. Neurochem. 2012, 123, 172–181. [Google Scholar] [CrossRef] [Green Version]
- Cipriani, S.; Bakshi, R.; Schwarzschild, M.A. Protection by inosine in a cellular model of Parkinson’s disease. Neuroscience 2014, 274, 242–249. [Google Scholar] [CrossRef] [Green Version]
- Huang, T.T.; Hao, D.L.; Wu, B.N.; Mao, L.L.; Zhang, J. Uric acid demonstrates neuroprotective effect on Parkinson’s disease mice through Nrf2-ARE signaling pathway. Biochem. Biophys. Res. Commun. 2017, 493, 1443–1449. [Google Scholar] [CrossRef]
- Gong, L.; Zhang, Q.L.; Zhang, N.; Hua, W.Y.; Huang, Y.X.; Di, P.W.; Huang, T.; Xu, X.S.; Liu, C.F.; Hu, L.F.; et al. Neuroprotection by urate on 6-OHDA-lesioned rat model of Parkinson’s disease: Linking to Akt/GSK3β signaling pathway. J Neurochem. 2012, 123, 876–885. [Google Scholar] [CrossRef]
- Jin, H.; Kanthasamy, A.; Ghosh, A.; Anantharam, V.; Kalyanaraman, B.; Kanthasamy, A.G. Mitochondria-targeted antioxidants for treatment of Parkinson’s disease: Preclinical and clinical outcomes. Biochim. Biophys. Acta 2014, 1842, 1282–1294. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharyya, S.; Bakshi, R.; Logan, R.; Ascherio, A.; Macklin, E.A.; Schwarzschild, M.A. Oral Inosine Persistently Elevates Plasma antioxidant capacity in Parkinson’s disease. Mov. Disord. 2016, 31, 417–421. [Google Scholar] [CrossRef]
- Iwaki, H.; Ando, R.; Miyaue, N.; Tada, S.; Tsujii, T.; Yabe, H.; Nishikawa, N.; Nagai, M.; Nomoto, M. One year safety and efficacy of inosine to increase the serum urate level for patients with Parkinson’s disease in Japan. J. Neurol. Sci. 2017, 383, 75–78. [Google Scholar] [CrossRef]
- Guttuso, T., Jr.; Andrzejewski, K.L.; Lichter, D.G.; Andersen, J.K. Targeting kinases in Parkinson’ss disease: A mechanism shared by LRRK2, neurotrophins, exenatide, urate, nilotinib and lithium. J. Neurol. Sci. 2019, 402, 121–130. [Google Scholar] [CrossRef] [PubMed]
- Parkinson Study Group SURE-PD3 Investigators; Schwarzschild, M.A.; Ascherio, A.; Casaceli, C.; Curhan, G.C.; Fitzgerald, R.; Kamp, C.; Lungu, C.; Macklin, E.A.; Marek, K.; et al. Effect of Urate-Elevating Inosine on Early Parkinson Disease Progression: The SURE-PD3 Randomized Clinical Trial. JAMA 2021, 326, 926–939. [Google Scholar] [CrossRef] [PubMed]
- Bjørklund, G.; Peana, M.; Maes, M.; Dadar, M.; Severin, B. The glutathione system in Parkinson’s disease and its progression. Neurosci. Biobehav. Rev. 2021, 120, 470–478. [Google Scholar] [CrossRef] [PubMed]
- Asanuma, M.; Miyazaki, I. Glutathione and Related Molecules in Parkinsonism. Int. J. Mol. Sci. 2021, 22, 8689. [Google Scholar] [CrossRef]
- Liddell, J.R.; White, A.R. Nexus between mitochondrial function, iron, copper and glutathione in Parkinson’s disease. Neurochem. Int. 2018, 117, 126–138. [Google Scholar] [CrossRef]
- Sian, J.; Dexter, D.T.; Lees, A.J.; Daniel, S.; Agid, Y.; Javoy-Agid, F.; Jenner, P.; Marsden, C.D. Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia. Ann. Neurol. 1994, 36, 348–355. [Google Scholar] [CrossRef]
- Mischley, L.K.; Standish, L.J.; Weiss, N.S.; Padowski, J.M.; Kavanagh, T.J.; White, C.C.; Rosenfeld, M.E. Glutathione as a Biomarker in Parkinson’s Disease: Associations with Aging and Disease Severity. Oxidative Med. Cell. Longev. 2016, 2016, 9409363. [Google Scholar] [CrossRef] [Green Version]
- Wei, H.J.; Du, M.; Bai, H.Y. Correlations of Melatonin and Glutathione Levels with Oxidative Stress Mechanism in Parkinson’s Disease. Zhongguo Yi Xue Ke Xue Yuan Xue Bao 2019, 41, 183–187. [Google Scholar]
- Duarte-Jurado, A.P.; Gopar-Cuevas, Y.; Saucedo-Cardenas, O.; Loera-Arias, M.J.; Montes-de-Oca-Luna, R.; Garcia-Garcia, A.; Rodriguez-Rocha, H. Antioxidant Therapeutics in Parkinson’s Disease: Current Challenges and Opportunities. Antioxidants 2021, 10, 453. [Google Scholar] [CrossRef]
- Virel, A.; Dudka, I.; Laterveer, R.; af Bjerkén, S. 1H NMR profiling of the 6-OHDA parkinsonian rat brain reveals metabolic alterations and signs of recovery after N-acetylcysteine treatment. Mol. Cell. Neurosci. 2019, 98, 131–139. [Google Scholar] [CrossRef]
- Holmay, M.J.; Terpstra, M.; Coles, L.D.; Mishra, U.; Ahlskog, M.; Öz, G.; Cloyd, J.C.; Tuite, P.J. N-Acetylcysteine boosts brain and blood glutathione in Gaucher and Parkinson diseases. Clin. Neuropharmacol. 2013, 36, 103–106. [Google Scholar] [CrossRef] [Green Version]
- Holdiness, M.R. Clinical pharmacokinetics of N-acetylcysteine. Clin. Pharmacokinet. 1991, 20, 123–134. [Google Scholar] [CrossRef]
- Mursaleen, L.; Noble, B.; Chan, S.H.Y.; Somavarapu, S.; Zariwala, M.G. N-Acetylcysteine Nanocarriers Protect against Oxidative Stress in a Cellular Model of Parkinson’s Disease. Antioxidants 2020, 9, 600. [Google Scholar] [CrossRef]
- Pradhan, P.; Majhi, O.; Biswas, A.; Joshi, V.K.; Sinha, D. Enhanced accumulation of reduced glutathione by Scopoletin improves survivability of dopaminergic neurons in Parkinson’s model. Cell Death Dis. 2020, 11, 739. [Google Scholar] [CrossRef]
- Monti, D.A.; Zabrecky, G.; Kremens, D.; Liang, T.W.; Wintering, N.A.; Bazzan, A.J.; Zhong, L.; Bowens, B.K.; Chervoneva, I.; Intenzo, C.; et al. N-Acetyl Cysteine Is Associated With Dopaminergic Improvement in Parkinson’s Disease. Clin. Pharm. Ther. 2019, 106, 884–890. [Google Scholar] [CrossRef]
- Coles, L.D.; Tuite, P.J.; Öz, G.; Mishra, U.R.; Kartha, R.V.; Sullivan, K.M.; Cloyd, J.C.; Terpstra, M. Repeated-Dose Oral N-Acetylcysteine in Parkinson’s Disease: Pharmacokinetics and Effect on Brain Glutathione and Oxidative Stress. J. Clin. Pharmacol. 2018, 58, 158–167. [Google Scholar] [CrossRef]
- Wang, H.L.; Zhang, J.; Li, Y.P.; Dong, L.; Chen, Y.Z. Potential use of glutathione as a treatment for Parkinson’s disease. Exp. Ther. Med. 2021, 21, 125. [Google Scholar] [CrossRef]
- Figueroa-Méndez, R.; Rivas-Arancibia, S. Vitamin C in Health and Disease: Its Role in the Metabolism of Cells and Redox State in the Brain. Front. Physiol. 2015, 6, 397. [Google Scholar] [CrossRef] [Green Version]
- Njus, D.; Kelley, P.M.; Tu, Y.J.; Schlegel, H.B. Ascorbic acid: The chemistry underlying its antioxidant properties. Free Radic. Biol. Med. 2020, 159, 37–43. [Google Scholar] [CrossRef]
- Spencer, E.S.; Pitcher, T.; Veron, G.; Hannam, T.; MacAskill, M.; Anderson, T.; Dalrymple-Alford, J.; Carr, A.C. Positive Association of Ascorbate and Inverse Association of Urate with Cognitive Function in People with Parkinson’s Disease. Antioxidants 2020, 9, 906. [Google Scholar] [CrossRef]
- Medeiros, M.S.; Schumacher-Schuh, A.; Cardoso, A.M.; Bochi, G.V.; Baldissarelli, J.; Kegler, A.; Santana, D.; Chaves, C.M.; Schetinger, M.R.; Moresco, R.N.; et al. Iron and Oxidative Stress in Parkinson’s Disease: An Observational Study of Injury Biomarkers. PLoS ONE 2016, 11, e0146129. [Google Scholar] [CrossRef] [Green Version]
- Ide, K.; Yamada, H.; Umegaki, K.; Mizuno, K.; Kawakami, N.; Hagiwara, Y.; Matsumoto, M.; Yoshida, H.; Kim, K.; Shiosaki, E.; et al. Lymphocyte vitamin C levels as potential biomarker for progression of Parkinson’s disease. Nutrition 2015, 31, 406–408. [Google Scholar] [CrossRef]
- Man Anh, H.; Linh, D.M.; My Dung, V.; Thi Phuong Thao, D. Evaluating Dose- and Time-Dependent Effects of Vitamin C Treatment on a Parkinson’s Disease Fly Model. Parkinson Dis. 2019, 2019, 9720546. [Google Scholar] [CrossRef] [Green Version]
- Zeng, X.; Xu, K.; Wang, J.; Xu, Y.; Qu, S. Pretreatment of Ascorbic Acid Inhibits MPTP-Induced Astrocytic Oxidative Stress through Suppressing NF-κB Signaling. Neural Plast. 2020, 2020, 8872296. [Google Scholar] [CrossRef]
- Song, M.K.; Lee, J.H.; Kim, J.; Kim, J.H.; Hwang, S.; Kim, Y.S.; Kim, Y.J. Neuroprotective effect of NXP031 in the MPTP-induced Parkinson’s disease model. Neurosci. Lett. 2021, 740, 135425. [Google Scholar] [CrossRef] [PubMed]
- Chang, M.C.; Kwak, S.G.; Kwak, S. Effect of dietary vitamins C and E on the risk of Parkinson’s disease: A meta-analysis. Clin. Nutr. 2021, 40, 3922–3930. [Google Scholar] [CrossRef] [PubMed]
- Nagayama, H.; Hamamoto, M.; Ueda, M.; Nito, C.; Yamaguchi, H.; Katayama, Y. The effect of ascorbic acid on the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clin. Neuropharmacol. 2004, 27, 270–273. [Google Scholar] [CrossRef] [PubMed]
- McDougall, M.; Choi, J.; Magnusson, K.; Truong, L.; Tanguay, R.; Traber, M.G. Chronic vitamin E deficiency impairs cognitive function in adult zebrafish via dysregulation of brain lipids and energy metabolism. Free Radic. Biol. Med. 2017, 112, 308–317. [Google Scholar] [CrossRef] [PubMed]
- Witting, P.K.; Upston, J.M.; Stocker, R. The molecular action of alpha-tocopherol in lipoprotein lipid peroxidation. Pro- and antioxidant activity of vitamin E in complex heterogeneous lipid emulsions. Subcell Biochem. 1998, 30, 345–390. [Google Scholar]
- Foy, C.J.; Passmore, A.P.; Vahidassr, M.D.; Young, I.S.; Lawson, J.T. Plasma chain-breaking antioxidants in Alzheimer’s disease, vascular dementia and Parkinson’s disease. QJM 1999, 92, 39–45. [Google Scholar] [CrossRef] [Green Version]
- Lopes da Silva, S.; Vellas, B.; Elemans, S.; Luchsinger, J.; Kamphuis, P.; Yaffe, K.; Sijben, J.; Groenendijk, M.; Stijnen, T. Plasma nutrient status of patients with Alzheimer’s disease: Systematic review and meta-analysis. Alzheimer Dement. 2014, 10, 485–502. [Google Scholar] [CrossRef] [Green Version]
- Kontush, K.; Schekatolina, S. Vitamin E in neurodegenerative disorders: Alzheimer’s disease. Ann. N. Y. Acad. Sci. 2004, 1031, 249–262. [Google Scholar] [CrossRef]
- Ueda, S.; Sakakibara, S.; Nakadate, K.; Noda, T.; Shinoda, M.; Joyce, J.N. Degeneration of dopaminergic neurons in the substantia nigra of zitter mutant rat and protection by chronic intake of Vitamin E. Neurosci. Lett. 2005, 380, 252–256. [Google Scholar] [CrossRef]
- Cadet, J.L.; Katz, M.; Jackson-Lewis, V.; Fahn, S. Vitamin E attenuates the toxic effects of intrastriatal injection of 6-hydroxydopamine (6-OHDA) in rats: Behavioral and biochemical evidence. Brain Res. 1989, 476, 10–15. [Google Scholar] [CrossRef]
- Heim, C.; Kolasiewicz, W.; Kurz, T.; Sontag, K.H. Behavioral alterations after unilateral 6-hydroxydopamine lesions of the striatum. Effect of alpha-tocopherol. Pol. J. Pharmacol. 2001, 53, 435–448. [Google Scholar]
- Roghani, M.; Behzadi, G. Neuroprotective effect of vitamin E on the early model of Parkinson’s disease in rat: Behavioral and histochemical evidence. Brain Res. 2001, 892, 211–217. [Google Scholar] [CrossRef]
- Sharma, N.; Nehru, B. Beneficial Effect of Vitamin E in Rotenone Induced Model of PD: Behavioural, Neurochemical and Biochemical Study. Exp. Neurobiol. 2013, 22, 214–223. [Google Scholar] [CrossRef] [Green Version]
- Schirinzi, T.; Martella, G.; Imbriani, P.; Di Lazzaro, G.; Franco, D.; Colona, V.L.; Alwardat, M.; Sinibaldi Salimei, P.; Mercuri, N.B.; Pierantozzi, M.; et al. Dietary Vitamin E as a Protective Factor for Parkinson’s Disease: Clinical and Experimental Evidence. Front. Neurol. 2019, 10, 148. [Google Scholar] [CrossRef] [Green Version]
- Marko, M.G.; Ahmed, T.; Bunnell, S.C.; Wu, D.; Chung, H.; Huber, B.T.; Meydani, S.N. Age-associated decline in effective immune synapse formation of CD4(+) T cells is reversed by vitamin E supplementation. J. Immunol. 2007, 178, 1443–1449. [Google Scholar] [CrossRef] [Green Version]
- Shim, J.H.; Yoon, S.H.; Kim, K.H.; Han, J.Y.; Ha, J.Y.; Hyun, D.H.; Paek, S.H.; Kang, U.J.; Zhuang, X.; Son, J.H. The antioxidant Trolox helps recovery from the familial Parkinson’s disease-specific mitochondrial deficits caused by PINK1- and DJ-1-deficiency in dopaminergic neuronal cells. Mitochondrion 2011, 11, 707–715. [Google Scholar] [CrossRef]
- Wu, D.; Meydani, S.N. Age-associated changes in immune function: Impact of vitamin E intervention and the underlying mechanisms. Endocr. Metab. Immune Disord.-Drug Targets 2014, 14, 283–289. [Google Scholar] [CrossRef]
- Lewis, E.D.; Meydani, S.N.; Wu, D. Regulatory role of vitamin E in the immune system and inflammation. IUBMB Life 2019, 71, 487–494. [Google Scholar] [CrossRef]
- Meydani, S.N.; Barklund, M.P.; Liu, S.; Meydani, M.; Miller, R.A.; Cannon, J.G.; Morrow, F.D.; Rocklin, R.; Blumberg, J.B. Vitamin E supplementation enhances cell-mediated immunity in healthy elderly subjects. Am. J. Clin. Nutr. 1990, 52, 557–563. [Google Scholar] [CrossRef] [Green Version]
- Ricciarelli, R.; Tasinato, A.; Clément, S.; Ozer, N.K.; Boscoboinik, D.; Azzi, A. Alpha-Tocopherol specifically inactivates cellular protein kinase C alpha by changing its phosphorylation state. Biochem. J. 1998, 334 Pt 1, 243–249. [Google Scholar] [CrossRef] [Green Version]
- Wu, D.; Koga, T.; Martin, K.R.; Meydani, M. Effect of Vitamin E on human aortic endothelial cell production of chemokines and adhesion to monocytes. Atherosclerosis 1999, 147, 297–307. [Google Scholar] [CrossRef]
- De Rijk, M.C.; Breteler, M.M.; den Breeijen, J.H.; Launer, L.J.; Grobbee, D.E.; van der Meché, F.G.; Hofman, A. Dietary antioxidants and Parkinson disease. The Rotterdam Study. Arch. Neurol. 1997, 54, 762–765. [Google Scholar] [CrossRef]
- Buhmann, C.; Arlt, S.; Kontush, A.; Möller-Bertram, T.; Sperber, S.; Oechsner, M.; Stuerenburg, H.J.; Beisiegel, U. Plasma and CSF markers of oxidative stress are increased in Parkinson’s disease and influenced by antiparkinsonian medication. Neurobiol. Dis. 2004, 15, 160–170. [Google Scholar] [CrossRef]
- Chen, C.M.; Liu, J.L.; Wu, Y.R.; Chen, Y.C.; Cheng, H.S.; Cheng, M.L.; Chiu, D.T. Increased oxidative damage in peripheral blood correlates with severity of Parkinson’s disease. Neurobiol. Dis. 2009, 33, 429–435. [Google Scholar] [CrossRef] [PubMed]
- Férnandez-Calle, P.; Molina, J.A.; Jiménez-Jiménez, F.J.; Vázquez, A.; Pondal, M.; García-Ruiz, P.J.; Urra, D.G.; Domingo, J.; Codoceo, R. Serum levels of alpha-tocopherol (vitamin E) in Parkinson’s disease. Neurology 1992, 42, 1064–1066. [Google Scholar] [CrossRef] [PubMed]
- Nicoletti, G.; Crescibene, L.; Scornaienchi, M.; Bastone, L.; Bagalà, A.; Napoli, I.D.; Caracciolo, M.; Quattrone, A. Plasma levels of vitamin E in Parkinson’s disease. Arch. Gerontol. Geriatr. 2001, 33, 7–12. [Google Scholar] [CrossRef]
- Sharma, A.; Weber, D.; Raupbach, J.; Dakal, T.C.; Fließbach, K.; Ramirez, A.; Grune, T.; Wüllner, U. Advanced glycation end products and protein carbonyl levels in plasma reveal sex-specific differences in Parkinson’s and Alzheimer’s disease. Redox Biol. 2020, 34, 101546. [Google Scholar] [CrossRef]
- Wei, Z.; Li, X.; Li, X.; Liu, Q.; Cheng, Y. Oxidative Stress in Parkinson’s Disease: A Systematic Review and Meta-Analysis. Front. Mol. Neurosci. 2018, 11, 236. [Google Scholar] [CrossRef]
- Fahn, S. A pilot trial of high-dose alpha-tocopherol and ascorbate in early Parkinson’s disease. Ann. Neurol. 1992, 32 (Suppl. S1), S128–S132. [Google Scholar] [CrossRef]
- Parkinson Study Group. Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease. N. Engl. J. Med. 1993, 328, 176–183. [Google Scholar] [CrossRef]
- Parkinson Study Group. Mortality in DATATOP: A multicenter trial in early Parkinson’s disease. Ann. Neurol. 1998, 43, 318–325. [Google Scholar] [CrossRef]
- Nakaso, K.; Horikoshi, Y.; Takahashi, T.; Hanaki, T.; Nakasone, M.; Kitagawa, Y.; Koike, T.; Matsura, T. Estrogen receptor-mediated effect of δ-tocotrienol prevents neurotoxicity and motor deficit in the MPTP mouse model of Parkinson’s disease. Neurosci. Lett. 2016, 610, 117–122. [Google Scholar] [CrossRef]
- Kumari, M.; Ramdas, P.; Radhakrishnan, A.K.; Kutty, M.K.; Haleagrahara, N. Tocotrienols Ameliorate Neurodegeneration and Motor Deficits in the 6-OHDA-Induced Rat Model of Parkinsonism: Behavioural and Immunohistochemistry Analysis. Nutrients 2021, 13, 1583. [Google Scholar] [CrossRef]
- McFarthing, K.; Rafaloff, G.; Baptista, M.; Wyse, R.K.; Stott, S. Parkinson’s Disease Drug Therapies in the Clinical Trial Pipeline: 2021 Update. J. Parkinson Dis. 2021, 11, 891–903. [Google Scholar] [CrossRef]
- Imam, M.U.; Zhang, S.; Ma, J.; Wang, H.; Wang, F. Antioxidants Mediate Both Iron Homeostasis and Oxidative Stress. Nutrients 2017, 9, 671. [Google Scholar] [CrossRef]
- Vauzour, D.; Vafeiadou, K.; Rodriguez-Mateos, A.; Rendeiro, C.; Spencer, J.P. The neuroprotective potential of flavonoids: A multiplicity of effects. Genes Nutr. 2008, 3, 115–126. [Google Scholar] [CrossRef] [Green Version]
- Hou, Z.; Lambert, J.D.; Chin, K.V.; Yang, C.S. Effects of tea polyphenols on signal transduction pathways related to cancer chemoprevention. Mutat. Res. 2004, 555, 3–19. [Google Scholar] [CrossRef]
- Commenges, D.; Scotet, V.; Renaud, S.; Jacqmin-Gadda, H.; Barberger-Gateau, P.; Dartigues, J.F. Intake of flavonoids and risk of dementia. Eur. J. Epidemiol. 2000, 16, 357–363. [Google Scholar] [CrossRef]
- Letenneur, L.; Proust-Lima, C.; Le Gouge, A.; Dartigues, J.F.; Barberger-Gateau, P. Flavonoid intake and cognitive decline over a 10-year period. Am. J. Epidemiol. 2007, 165, 1364–1371. [Google Scholar] [CrossRef]
- Qi, H.; Li, S. Dose-response meta-analysis on coffee, tea and caffeine consumption with risk of Parkinson’s disease. Geriatr. Gerontol. Int. 2014, 14, 430–439. [Google Scholar] [CrossRef]
- Kandinov, B.; Giladi, N.; Korczyn, A.D. Smoking and tea consumption delay onset of Parkinson’s disease. Parkinsonism Relat. Disord. 2009, 15, 41–46. [Google Scholar] [CrossRef]
- Li, F.J.; Ji, H.F.; Shen, L. A meta-analysis of tea drinking and risk of Parkinson’s disease. Sci. World J. 2012, 2012, 923464. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Molsberry, S.A.; Yeh, T.S.; Cassidy, A.; Schwarzschild, M.A.; Ascherio, A.; Gao, X. Intake of Flavonoids and Flavonoid-Rich Foods and Mortality Risk Among Individuals With Parkinson Disease: A Prospective Cohort Study. Neurology 2022, 98, e1064–e1076. [Google Scholar] [CrossRef]
- Yeh, T.S.; Yuan, C.; Ascherio, A.; Rosner, B.A.; Willett, W.C.; Blacker, D. Long-term Dietary Flavonoid Intake and Subjective Cognitive Decline in US Men and Women. Neurology 2021, 97, e1041–e1056. [Google Scholar] [CrossRef]
- Kujawska, M.; Jodynis-Liebert, J. Polyphenols in Parkinson’s Disease: A Systematic Review of In Vivo Studies. Nutrients 2018, 10, 642. [Google Scholar] [CrossRef] [Green Version]
- Williams, R.J.; Spencer, J.P.; Rice-Evans, C. Flavonoids: Antioxidants or signalling molecules? Free Radic. Biol. Med. 2004, 36, 838–849. [Google Scholar] [CrossRef]
- Ikram, M.; Park, T.J.; Ali, T.; Kim, M.O. Antioxidant and Neuroprotective Effects of Caffeine against Alzheimer’s and Parkinson’s Disease: Insight into the Role of Nrf-2 and A2AR Signaling. Antioxidants 2020, 9, 902. [Google Scholar] [CrossRef] [PubMed]
- Fattah, A.; Amiri, F.; Mohammadian, M.; Alipourfard, I.; Valilo, M.; Taheraghdam, A.; Hemmati-Dinarvand, M. Dysregulation of body antioxidant content is related to initiation and progression of Parkinson’s disease. Neurosci. Lett. 2020, 736, 135297. [Google Scholar] [CrossRef] [PubMed]
- Ooi, B.K.; Chan, K.G.; Goh, B.H.; Yap, W.H. The Role of Natural Products in Targeting Cardiovascular Diseases via Nrf2 Pathway: Novel Molecular Mechanisms and Therapeutic Approaches. Front. Pharmacol. 2018, 9, 1308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dzamko, N.; Zhou, J.; Huang, Y.; Halliday, G.M. Parkinson’s disease-implicated kinases in the brain; insights into disease pathogenesis. Front. Mol. Neurosci. 2014, 7, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Zhang, D.Q.; Liao, Z.; Wang, B.; Gong, S.; Wang, C.; Zhang, M.Z.; Wang, G.H.; Cai, H.; Liao, F.F.; et al. Anti-oxidant polydatin (piceid) protects against substantia nigral motor degeneration in multiple rodent models of Parkinson’s disease. Mol. Neurodegener. 2015, 10, 4. [Google Scholar] [CrossRef] [Green Version]
- Guo, S.; Yan, J.; Yang, T.; Yang, X.; Bezard, E.; Zhao, B. Protective effects of green tea polyphenols in the 6-OHDA rat model of Parkinson’s disease through inhibition of ROS-NO pathway. Biol. Psychiatry 2007, 62, 1353–1362. [Google Scholar] [CrossRef]
- Jagatha, B.; Mythri, R.B.; Vali, S.; Bharath, M.M. Curcumin treatment alleviates the effects of glutathione depletion in vitro and in vivo: Therapeutic implications for Parkinson’s disease explained via in silico studies. Free Radic. Biol. Med. 2008, 44, 907–917. [Google Scholar] [CrossRef]
- Tripanichkul, W.; Jaroensuppaperch, E.O. Ameliorating effects of curcumin on 6-OHDA-induced dopaminergic denervation, glial response, and SOD1 reduction in the striatum of hemiparkinsonian mice. Eur. Rev. Med. Pharm. Sci. 2013, 17, 1360–1368. [Google Scholar]
- Karuppagounder, S.S.; Madathil, S.K.; Pandey, M.; Haobam, R.; Rajamma, U.; Mohanakumar, K.P. Quercetin up-regulates mitochondrial complex-I activity to protect against programmed cell death in rotenone model of Parkinson’s disease in rats. Neuroscience 2013, 236, 136–148. [Google Scholar] [CrossRef]
- Kwon, S.H.; Lee, S.R.; Park, Y.J.; Ra, M.; Lee, Y.; Pang, C.; Kim, K.H. Suppression of 6-Hydroxydopamine-Induced Oxidative Stress by Hyperoside Via Activation of Nrf2/HO-1 Signaling in Dopaminergic Neurons. Int. J. Mol. Sci. 2019, 20, 5832. [Google Scholar] [CrossRef] [Green Version]
- Sugumar, M.; Sevanan, M.; Sekar, S. Neuroprotective effect of naringenin against MPTP-induced oxidative stress. Int. J. Neurosci. 2019, 129, 534–539. [Google Scholar] [CrossRef]
- Siddique, Y.H.; Jyoti, S.; Naz, F. Effect of epicatechin gallate dietary supplementation on transgenic Drosophila model of Parkinson’s disease. J. Diet. Suppl. 2014, 11, 121–130. [Google Scholar] [CrossRef]
- Xu, Q.; Langley, M.; Kanthasamy, A.G.; Reddy, M.B. Epigallocatechin Gallate Has a Neurorescue Effect in a Mouse Model of Parkinson Disease. J. Nutr. 2017, 147, 1926–1931. [Google Scholar] [CrossRef] [Green Version]
- Levites, Y.; Youdim, M.B.; Maor, G.; Mandel, S. Attenuation of 6-hydroxydopamine (6-OHDA)-induced nuclear factor-kappaB (NF-kappaB) activation and cell death by tea extracts in neuronal cultures. Biochem. Pharmacol. 2002, 63, 21–29. [Google Scholar] [CrossRef]
- Lou, H.; Jing, X.; Wei, X.; Shi, H.; Ren, D.; Zhang, X. Naringenin protects against 6-OHDA-induced neurotoxicity via activation of the Nrf2/ARE signaling pathway. Neuropharmacology 2014, 79, 380–388. [Google Scholar] [CrossRef]
- Kim, H.D.; Jeong, K.H.; Jung, U.J.; Kim, S.R. Naringin treatment induces neuroprotective effects in a mouse model of Parkinson’s disease in vivo, but not enough to restore the lesioned dopaminergic system. J. Nutr. Biochem. 2016, 28, 140–146. [Google Scholar] [CrossRef]
- Gao, L.; Li, C.; Yang, R.Y.; Lian, W.W.; Fang, J.S.; Pang, X.C.; Qin, X.M.; Liu, A.L.; Du, G.H. Ameliorative effects of baicalein in MPTP-induced mouse model of Parkinson’s disease: A microarray study. Pharmacol. Biochem. Behav. 2015, 133, 155–163. [Google Scholar] [CrossRef]
- Kim, H.G.; Ju, M.S.; Ha, S.K.; Lee, H.; Lee, H.; Kim, S.Y.; Oh, M.S. Acacetin protects dopaminergic cells against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neuroinflammation in vitro and in vivo. Biol. Pharm. Bull. 2012, 35, 1287–1294. [Google Scholar] [CrossRef] [Green Version]
- Johnson, D.A.; Johnson, J.A. Nrf2—A therapeutic target for the treatment of neurodegenerative diseases. Free Radic. Biol. Med. 2015, 88(Part B), 253–267. [Google Scholar] [CrossRef] [Green Version]
- Leonardo, C.C.; Doré, S. Dietary flavonoids are neuroprotective through Nrf2-coordinated induction of endogenous cytoprotective proteins. Nutr. Neurosci. 2011, 14, 226–236. [Google Scholar] [CrossRef] [Green Version]
- Gureev, A.P.; Popov, V.N. Nrf2/ARE Pathway as a Therapeutic Target for the Treatment of Parkinson Diseases. Neurochem. Res. 2019, 44, 2273–2279. [Google Scholar] [CrossRef] [PubMed]
- Stacchiotti, A.; Corsetti, G. Natural Compounds and Autophagy: Allies Against Neurodegeneration. Front. Cell Dev. Biol. 2020, 8, e555409. [Google Scholar] [CrossRef] [PubMed]
- Sun, W.; Zheng, J.; Ma, J.; Wang, Z.; Shi, X.; Li, M.; Huang, S.; Hu, S.; Zhao, Z.; Li, D. Increased Plasma Heme Oxygenase-1 Levels in Patients With Early-Stage Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 621508. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Gao, L.; Chen, J.; Li, Q.; Huo, L.; Wang, Y.; Wang, H.; Du, J. Pharmacological Modulation of Nrf2/HO-1 Signaling Pathway as a Therapeutic Target of Parkinson’s Disease. Front. Pharmacol. 2021, 12, 757161. [Google Scholar] [CrossRef]
- Habtemariam, S. Antioxidant and Anti-inflammatory Mechanisms of Neuroprotection by Ursolic Acid: Addressing Brain Injury, Cerebral Ischemia, Cognition Deficit, Anxiety, and Depression. Oxidative Med. Cell. Longev. 2019, 2019, 8512048. [Google Scholar] [CrossRef]
- Zeng, W.; Zhang, W.; Lu, F.; Gao, L.; Gao, G. Resveratrol attenuates MPP +-induced mitochondrial dysfunction and cell apoptosis via AKT/GSK-3β pathway in SN4741 cells. Neurosci. Lett. 2017, 637, 50–56. [Google Scholar] [CrossRef]
- Guo, Y.J.; Dong, S.Y.; Cui, X.X.; Feng, Y.; Liu, T.; Yin, M.; Kuo, S.H.; Tan, E.K.; Zhao, W.J.; Wu, Y.C. Resveratrol alleviates MPTP-induced motor impairments and pathological changes by autophagic degradation of α-synuclein via SIRT1-deacetylated LC3. Mol. Nutr. Food Res. 2016, 60, 2161–2175. [Google Scholar] [CrossRef]
- Moussa, C.; Hebron, M.; Huang, X.; Ahn, J.; Rissman, R.A.; Aisen, P.S.; Turner, R.S. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflamm. 2017, 14, 1. [Google Scholar] [CrossRef] [Green Version]
- Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S. Alzheimer’s Disease Cooperative Study. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology 2015, 85, 1383–1391. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Wang, X.S.; Zhang, Z.R.; Zhang, M.M.; Sun, M.X.; Wang, W.W.; Xie, C.L. Neuroprotective properties of curcumin in toxin-base animal models of Parkinson’s disease: A systematic experiment literatures review. BMC Complement. Altern. Med. 2017, 17, 412. [Google Scholar] [CrossRef]
- Yu, S.; Zheng, W.; Xin, N.; Chi, Z.H.; Wang, N.Q.; Nie, Y.X.; Feng, W.Y.; Wang, Z.Y. Curcumin prevents dopaminergic neuronal death through inhibition of the c-Jun N-terminal kinase pathway. Rejuvenation Res. 2010, 13, 55–64. [Google Scholar] [CrossRef]
- Goozee, K.G.; Shah, T.M.; Sohrabi, H.R.; Rainey-Smith, S.R.; Brown, B.; Verdile, G.; Martins, R.N. Examining the potential clinical value of curcumin in the prevention and diagnosis of Alzheimer’s disease. Br. J. Nutr. 2016, 115, 449–465. [Google Scholar] [CrossRef]
- Hornedo-Ortega, R.; Cerezo, A.B.; de Pablos, R.M.; Krisa, S.; Richard, T.; García-Parrilla, M.C.; Troncoso, A.M. Phenolic Compounds Characteristic of the Mediterranean Diet in Mitigating Microglia-Mediated Neuroinflammation. Front. Cell. Neurosci. 2018, 12, 373. [Google Scholar] [CrossRef]
- DiSilvestro, R.A.; Joseph, E.; Zhao, S.; Bomser, J. Diverse effects of a low dose supplement of lipidated curcumin in healthy middle aged people. Nutr. J. 2012, 11, 79. [Google Scholar] [CrossRef] [Green Version]
- Yin, R.; Xue, J.; Tan, Y.; Fang, C.; Hu, C.; Yang, Q.; Mei, X.; Qi, D. The Positive Role and Mechanism of Herbal Medicine in Parkinson’s Disease. Oxidative Med. Cell. Longev. 2021, 2021, 9923331. [Google Scholar] [CrossRef]
- Sun, C.-P.; Zhou, J.J.; Yu, Z.L.; Huo, X.K.; Zhang, J.; Morisseau, C.; Hammock, B.D.; Ma, X.C. Kurarinone alleviated Parkinson’s disease via stabilization of epoxyeicosatrienoic acids in animal model. Proc. Natl. Acad. Sci. USA 2022, 119, e2118818119. [Google Scholar] [CrossRef]
- Huang, B.; Liu, J.; Meng, T.; Li, Y.; He, D.; Ran, X.; Chen, G.; Guo, W.; Kan, X.; Fu, S.; et al. Polydatin Prevents Lipopolysaccharide (LPS)-Induced Parkinson’s Disease via Regulation of the AKT/GSK3β-Nrf2/NF-κB Signaling Axis. Front. Immunol. 2018, 9, 2527. [Google Scholar] [CrossRef]
- Xia, M.L.; Xie, X.H.; Ding, J.H.; Du, R.H.; Hu, G. Astragaloside IV inhibits astrocyte senescence: Implication in Parkinson’s disease. J. Neuroinflamm. 2020, 17, 105. [Google Scholar] [CrossRef]
- Baek, S.Y.; Lee, N.R.; Kim, D.H.; Gu, A.; Kim, S.Y.; Song, D.Y.; Kim, D.H.; Choi, H.J.; Park, B.J.; Kim, I.S. Protective effect of a novel herbmedicine, Hepad, on apoptosis of SH-SY5Y cells and a rat model of Parkinson’s disease. Mol. Cell. Toxicol. 2015, 11, 223–230. [Google Scholar] [CrossRef]
- Song, D.; Kim, G.-J.; Lee, K.; Shin, J.; Kim, D.-H.; Park, B.-J.; An, J. Mitigation Effects of a Novel Herbal Medicine, Hepad, on Neuroinflammation, Neuroapoptosis, and Neuro-Oxidation. Molecules 2018, 23, 2920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Won, S.; Ko, J.H.; Jeon, H.; Park, S.-S.; Kim, S.-N. Co-Administration of Gagam-Sipjeondaebo-Tang and Ibuprofen Alleviates the Inflammatory Response in MPTP-Induced Parkinson’s Disease Mouse Model and RAW264.7 Macrophages. Pathogens 2021, 10, 268. [Google Scholar] [CrossRef] [PubMed]
- Jayaraj, R.L.; Beiram, R.; Azimullah, S.; Meeran, M.F.N.; Ojha, S.K.; Adem, A.; Jalal, F.Y. Lycopodium Attenuates Loss of Dopaminergic Neurons by Suppressing Oxidative Stress and Neuroinflammation in a Rat Model of Parkinson’s Disease. Molecules 2019, 24, 2182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lim, H.-S.; Kim, Y.J.; Kim, B.-Y.; Park, G.; Jeong, S.-J. The Anti-neuroinflammatory Activity of Tectorigenin Pretreatment via Downregulated NF-κB and ERK/JNK Pathways in BV-2 Microglial and Microglia Inactivation in Mice With Lipopolysaccharide. Front. Pharmacol. 2018, 9, 462. [Google Scholar] [CrossRef]
- Zhou, J.; Deng, Y.; Li, F.; Yin, C.; Shi, J.; Gong, Q. Icariside II attenuates lipopolysaccharide-induced neuroinflammation through inhibiting TLR4/MyD88/NF-κB pathway in rats. Biomed. Pharmacother. 2019, 111, 315–324. [Google Scholar] [CrossRef]
- Ju, I.G.; Huh, E.; Kim, N.; Lee, S.; Choi, J.G.; Hong, J.; Oh, M.S. Artemisiae Iwayomogii Herba inhibits lipopolysaccharide-induced neuroinflammation by regulating NF-κB and MAPK signaling pathways. Phytomedicine 2021, 84, 153501. [Google Scholar] [CrossRef]
- Pandey, T.; Shukla, A.; Trivedi, M.; Khan, F.; Pandey, R. Swertiamarin from Enicostemma littorale, counteracts PD associated neurotoxicity via enhancement α-synuclein suppressive genes and SKN-1/NRF-2 activation through MAPK pathway. Bioorg. Chem. 2021, 108, 104655. [Google Scholar] [CrossRef]
- Pallàs, M.; Vázquez, S.; Sanfeliu, C.; Galdeano, C.; Griñán-Ferré, C. Soluble Epoxide Hydrolase Inhibition to Face Neuroinflammation in Parkinson’s Disease: A New Therapeutic Strategy. Biomolecules 2020, 10, 703. [Google Scholar] [CrossRef]
- Ren, Q.; Ma, M.; Ishima, T.; Morisseau, C.; Yang, J.; Wagner, K.M.; Zhang, J.; Yang, C.; Yao, W.; Dong, C.; et al. Gene deficiency and pharmacological inhibition of soluble epoxide hydrolase confers resilience to repeated social defeat stress. Proc. Natl. Acad. Sci. USA 2016, 113, E1944–E1952. [Google Scholar] [CrossRef] [Green Version]
- Ren, Q.; Ma, M.; Yang, J.; Nonaka, R.; Yamaguchi, A.; Ishikawa, K.; Kobayashi, K.; Murayama, S.; Hwang, S.H.; Saiki, S.; et al. Soluble epoxide hydrolase plays a key role in the pathogenesis of Parkinson’s disease. Proc. Natl. Acad. Sci. USA 2018, 115, E5815–E5823. [Google Scholar] [CrossRef] [Green Version]
- Chen, S.Y.; Xiao, S.J.; Lin, Y.N.; Li, X.Y.; Xu, Q.; Yang, S.S.; Huang, L.H.; Cai, J. Clinical Efficacy and Transcriptomic Analysis of Congrong Shujing Granules (苁蓉舒痉颗粒) in Patients with Parkinson’s Disease and Syndrome of Shen (Kidney) Essence Deficiency. Chin. J. Integr. Med. 2020, 26, 412–419. [Google Scholar] [CrossRef]
- Liu, M.; Hu, C.; Zhang, Y.; Li, Q.; Zhang, Q.; Fang, Y.; Sun, H.; Hou, X.; Jin, S.; Tao, L.; et al. Effect of Huatan Jieyu granules in treatment of Parkinson’s disease patients with sleep disorder identified as symptom pattern of phlegma-heat-stirring wind. J. Tradit. Chin. Med. 2020, 40, 461–466. [Google Scholar]
- Shi, J.; Tian, J.; Li, T.; Fan, D.; Ni, J.; Wei, M.; Zhang, X.; Liu, N.; Liu, J.; Li, Y.; et al. Efficacy and safety of SQJZ herbal mixtures on nonmotor symptoms in Parkinson disease patients Protocol for a randomized, double-blind, placebo-controlled trial. Medicine 2017, 96, e8824. [Google Scholar] [CrossRef]
- Schwarzschild, M.A.; Macklin, E.A.; Bakshi, R.; Battacharyya, S.; Logan, R.; Espay, A.J.; Hung, A.Y.; Bwala, G.; Goetz, C.G.; Russell, D.S.; et al. Sex differences by design and outcome in the Safety of Urate Elevation in PD (SURE-PD) trial. Neurology 2019, 93, e1328–e1338. [Google Scholar] [CrossRef]
- Goldstein, D.S.; Holmes, C.; Sullivan, P.; Jinsmaa, Y.; Kopin, I.J.; Sharabi, Y. Elevated cerebrospinal fluid ratios of cysteinyl-dopamine/3,4-dihydroxyphenylacetic acid in parkinsonian synucleinopathies. Parkinsonism Relat. Disord. 2016, 31, 79–86. [Google Scholar] [CrossRef] [Green Version]
- Petramfar, P.; Hajari, F.; Yousefi, G.; Azadi, S.; Hamedi, A. Efficacy of oral administration of licorice as an adjunct therapy on improving the symptoms of patients with Parkinson’s disease, A randomized double blinded clinical trial. J. Ethnopharmacol. 2020, 247, 112226. [Google Scholar] [CrossRef]
- Filograna, R.; Godena, V.K.; Sanchez-Martinez, A.; Ferrari, E.; Casella, L.; Beltramini, M.; Bubacco, L.; Whitworth, A.J.; Bisaglia, M. Superoxide Dismutase (SOD)-mimetic M40403 Is Protective in Cell and Fly Models of Paraquat Toxicity: IMPLICATIONS FOR PARKINSON DISEASE. J. Biol. Chem. 2016, 291, 9257–9267. [Google Scholar] [CrossRef] [Green Version]
- Biosa, A.; Sanchez-Martinez, A.; Filograna, R.; Terriente-Felix, A.; Alam, S.M.; Beltramini, M.; Bubacco, L.; Bisaglia, M.; Whitworth, A.J. Superoxide dismutating molecules rescue the toxic effects of PINK1 and parkin loss. Hum. Mol. Genet. 2018, 27, 1618–1629. [Google Scholar] [CrossRef]
- Sandhir, R.; Mahajan, N.; Mehrotra, A.; Aggarwal, A.; Sunkaria, A. 4-hydroxy tempo improves mitochondrial and neurobehavioral deficits in experimental model of Huntington’s disease. Synapse 2015, 69, 128–138. [Google Scholar] [CrossRef]
- Chiarotto, G.B.; Drummond, L.; Cavarretto, G.; Bombeiro, A.L.; de Oliveira, A.L. Neuroprotective effect of tempol (4 hydroxy-tempo) on neuronal death induced by sciatic nerve transection in neonatal rats. Brain Res. Bull. 2014, 106, 1–8. [Google Scholar] [CrossRef]
- Ghosh, M.C.; Tong, W.H.; Zhang, D.; Ollivierre-Wilson, H.; Singh, A.; Krishna, M.C.; Mitchell, J.B.; Rouault, T.A. Tempol-mediated activation of latent iron regulatory protein activity prevents symptoms of neurodegenerative disease in IRP2 knockout mice. Proc. Natl. Acad. Sci. USA 2008, 105, 12028–12033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Samaiya, P.K.; Narayan, G.; Kumar, A.; Krishnamurthy, S. Tempol (4 hydroxy-tempo) inhibits anoxia-induced progression of mitochondrial dysfunction and associated neurobehavioral impairment in neonatal rats. J. Neurol. Sci. 2017, 375, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Neil, S.; Huh, J.; Baronas, V.; Li, X.; McFarland, H.F.; Cherukuri, M.; Mitchell, J.B.; Quandt, J.A. Oral administration of the nitroxide radical TEMPOL exhibits immunomodulatory and therapeutic properties in multiple sclerosis models. Brain Behav. Immun. 2017, 62, 332–343. [Google Scholar] [CrossRef] [PubMed]
- Wilcox, C. Effects of tempol and redox-cycling nitroxides in models of oxidative stress. Pharmacol. Ther. 2010, 126, 119–145. [Google Scholar] [CrossRef] [Green Version]
- Soule, B.; Hyodo, F.; Matsumoto, K.; Simone, N.; Cook, J.; Krishna, M.; Mitchell, J. The chemistry and biology of nitroxide compounds. Free Radic. Biol. Med. 2007, 42, 1632–1650. [Google Scholar] [CrossRef] [Green Version]
- Goralska, M.; Holley, B.; McGahan, M. The effects of Tempol on ferritin synthesis and Fe metabolism in lens epithelial cells. Biochim. Biophys. Acta 2000, 1497, 51–60. [Google Scholar] [CrossRef] [Green Version]
- Rees, M.; Bottle, S.; Fairfull-Smith, K.; Malle, E.; Whitelock, J.; Davies, M. Inhibition of myeloperoxidase-mediated hypochlorous acid production by nitroxides. Biochem. J. 2009, 421, 79–86. [Google Scholar] [CrossRef] [Green Version]
- Sadowska-Bartosz, I.; Gajewska, A.; Skolimowski, J.; Szewczyk, R.; Bartosz, G. Nitroxides protect against peroxynitrite-induced nitration and oxidation. Free Radic. Biol. Med. 2015, 89, 1165–1175. [Google Scholar] [CrossRef]
- Leathem, A.; Simone, M.; Dennis, J.M.; Witting, P.K. The Cyclic Nitroxide TEMPOL Ameliorates Oxidative Stress but Not Inflammation in a Cell Model of Parkinson’s Disease. Antioxidants 2022, 11, 257. [Google Scholar] [CrossRef]
- Liang, Q.; Smith, A.; Pan, S.; Tyurin, V.; Kagan, V.; Hastings, T.; Schor, N. Neuroprotective effects of TEMPOL in central and peripheral nervous system models of Parkinson’s disease. Biochem. Pharmacol. 2005, 70, 1371–1381. [Google Scholar] [CrossRef]
- Perier, C.; Bové, J.; Dehay, B.; Jackson-Lewis, V.; Rabinovitch, P.S.; Przedborski, S.; Vila, M. Apoptosis-inducing factor deficiency sensitizes dopaminergic neurons to parkinsonian neurotoxins. Ann. Neurol. 2010, 68, 184–192. [Google Scholar]
- Sadowska-Bartosz, I.; Bartosz, G. Redox nanoparticles: Synthesis, properties and perspectives of use for treatment of neurodegenerative diseases. J. Nanobiotech. 2018, 16, 87. [Google Scholar] [CrossRef] [Green Version]
- Carroll, R.T.; Bhatia, D.; Geldenhuys, W.; Bhatia, R.; Miladore, N.; Bishayee, A.; Sutariya, V. Brain-targeted delivery of Tempol-loaded nanoparticles for neurological disorders. J. Drug Target. 2010, 18, 665–674. [Google Scholar] [CrossRef]
- Weinberg, A.; Nylander, K.D.; Yan, C.; Ma, L.; Hsia, C.J.C.; Tyurin, V.A.; Kagan, V.E.; Schor, N.F. Prevention of catecholaminergic oxidative toxicity by 4-hydroxy-2,2,6,6-tetramethylpiperidine-1-oxyl and its recycling complex with polynitroxylated albumin, TEMPOL/PNA. Brain Res. 2004, 1012, 13–21. [Google Scholar] [CrossRef]
- Pichla, M.; Pulaski, Ł.; Kania, K.D.; Stefaniuk, I.; Cieniek, B.; Pieńkowska, N.; Bartosz, G.; Sadowska-Bartosz, I. Nitroxide Radical-Containing Redox Nanoparticles Protect Neuroblastoma SH-SY5Y Cells against 6-Hydroxydopamine Toxicity. Oxidative Med. Cell. Longev. 2020, 9260748. [Google Scholar] [CrossRef]
- Zink, A.; Lisowski, P.; Prigione, A. Generation of Human iPSC-derived Neural Progenitor Cells (NPCs) as Drug Discovery Model for Neurological and Mitochondrial Disorders. Bio-protocol 2021, 11, e3939. [Google Scholar] [CrossRef]
- Nguyen, H.N. Generation of iPSC-Derived Brain Organoids for Drug Testing and Toxicological Evaluation. Methods Mol. Biol. 2022, 2474, 93–105. [Google Scholar]
- Bose, A.; Petsko, G.A.; Studer, L. Induced pluripotent stem cells: A tool for modeling Parkinson’s disease. Trends Neurosci. 2022; Epub ahead of print. [Google Scholar] [CrossRef]
- McComish, S.F.; MacMahon Copas, A.N.; Caldwell, M.A. Human Brain-Based Models Provide a Powerful Tool for the Advancement of Parkinson’s Disease Research and Therapeutic Development. Front. Neurosci. 2022, 16, e851058. [Google Scholar] [CrossRef]
- Sabate-Soler, S.; Nickels, S.L.; Saraiva, C.; Berger, E.; Dubonyte, U.; Barmpa, K.; Lan, Y.J.; Kouno, T.; Jarazo, J.; Robertson, G.; et al. Microglia integration into human midbrain organoids leads to increased neuronal maturation and functionality. Glia 2022, 70, 1267–1288. [Google Scholar] [CrossRef]
N Patients | Recruitment Selection Strategy | Study Design | Primary Outcomes | Secondary Effects | Status |
---|---|---|---|---|---|
CoQ10 | |||||
20 | -Stable medication with carbidopa/LD, Selegiline and anticholinergic medication. -Symptoms of PD from 1–17 years. -54 to 70 years; All sexes | 4 groups: CoQ10 provided at doses of 300, 600 or 1200 mg/day plus vitamin E (300 UI) and a placebo. | -↑ of plasma CoQ10 level -Normalize Complex I activity mitochondrial -↓ decrease in the original UPDRS score | Nonsignificant secondary effects | Phase 2 NCT00004731 Ref [45] |
600 | -Without PD medication before 60 days. -Presence of all cardinal signs within 5 years. -Hoehn and Yahr stage of 2.5 or less -30 years and older; All sexes | 3 groups: CoQ10 at doses of 1200 and 2400 mg/day plus vitamin E (1200 UI) and placebo | No change in MDS-UPDRS score, Hoehn and Yahr score, PDQOL and SE-ADL scales. | -Back pain, -Constipation, -Anxiety, -Headache | Phase 3 NCT00740714 Ref [47] |
72 | -Stable PD medication for at least 4 weeks -18 years and older -All sexes | 4 groups: Stratified by their genetic mitochondrial risk burden and randomized 1:1. CoQ10 at dose of 156 mg/day [QuinoMit Q10® Fluid] and placebo | Not reported | Not reported | Phase 2 DRKS00015880 Ref [55] |
Urate/Inosine | |||||
298 | -Without PD medication -Serum urate ≤ 5.7 mg/dL at 1st visit -Presence at least 2 of the cardinal signs -Hoehn and Yahr stage of 2.5 or less -30 years and older; All sexes | 2 groups: Inosine at dose of 500 mg, 1 to 6 capsules per day to achieve serum urate at 7.1 to 8.0 mg/dL and placebo for 24 months | -No change in MDS-UPDRS score and DAT binding -↑ of serum urate level. | -Kidney stones | Phase 3 NCT00833690 Ref [73] |
75 | -Without PD medication -Serum urate ≤ 5.8 mg/dL at 1st visit -Presence at least 2 of the cardinal signs -30 years and older; All sexes | 3 groups: Inosine at dose of 500 mg, 1 to 6 capsules per day to achieve serum urate at 6.1–7.0 mg/dL (mild), 7.1–8.0 mg/dL (moderate) and placebo for 2 years | -↓ significant decrease in the MDS-UPDRS score in moderate group in woman. -↑ plasma antioxidant capacity in woman. -↑ serum urate level and CSF in women | -Kidney stones | Phase 2 NCT02642393 Ref [195] |
Glutathione/N-acetyl-cysteine | |||||
42 | -Stable PD medication for at least 1 month -Hoehn and Yahr score of 1–2 inclusive -30 to 80 years -All sexes | 2 groups: IV NAC at dose of 50 mg in 200 mL at frequency over one hour 1 × per week for 90 days plus Oral NAC at dose of 1200 mg/day and standard of care treatment (control group) | -↓ significant decrease in the MDS-UPDRS score -↑ DAT binding in caudate and putamen | Not reported | Not applicable NCT02445651 Ref [86] |
8 | -Stable PD medication -18 years and older -All sexes | 2 groups: NAC at dose 6000 mg/day for 4 weeks and healthy controls | -↑ significant increase in catalase and GSH/GSSG peripheric -No change in 4–HNE and MDA. -No change in brain levels of GSH | -Gastrointestinal disorder -Nervous system disorders | Phase 2 NCT02212678 https://clinicaltrials.gov accessed on 3 June 2022 |
35 | -Stable medication with monoamine oxidase inhibitor. -Diagnosis within the past 5 years -18 years and older; All sexes | 2 groups: NAC at dose 2 g/day for 3 days and healthy controls | -↑ Cys–DA/DOPAC ratio | Not reported | Phase 1 NCT03104725 Ref [196] |
47 | -Without PD medication -50 to 75 years -All sexes | 3 groups: NAC at dose of 1800 mg/day, 3600 mg daily and placebo for 30 days | Not reported | Not reported | Phase 2 NCT01470027 https://clinicaltrials.gov accessed on 3 June 2022 |
Vitamin C | |||||
67 | -Stable medication with LD -64 years and older -All sexes | 1 group: First time: 100 mg LD + 10 mg carbidopa and Second time (1 week later) 100 mg LD + 10 mg carbidopa + 200 mg AsA. | -↑ LD pharmacokinetics | Not reported | Not applicable Ref [98] |
Vitamin E (Tocotrienols) | |||||
100 | -Stable PD medication -Diagnosis within the past 1 year -PD of more than 1 year duration from diagnosis. -Hoehn and Yahr score ≥ 2 -40 to 90 years -All sexes | 2 groups: Tocotrienols (400 mg/day) and placebo for 12 months | Not reported | Not reported | Phase 2 NCT04491383 https://clinicaltrials.gov accessed on 3 June 2022 |
Polyphenols | |||||
38 | -Stable PD medication for at least 1 month. -Hoehn and Yahr score of 1–3 -30 years and older -All sexes | 3 groups: EGCG at dose of 800 mg, dose of 1200 mg and placebo (mannitol) group for 4 weeks. Then one capsule twice daily for 4 weeks, and then one capsule three times daily for 40 weeks | -No change in UMSARS score | -Hepatotoxicity | Phase 3 NCT00461942 https://clinicaltrials.gov accessed on 3 June 2022 |
Herbal Medicine | |||||
30 | -Stable PD medication -Initiation of PD symptoms in recent 6 years -Hoehn and Yahr score of ≤3 -30 to 80 years | 2 groups: Oral Licorice at dose 10 cc/day and placebo (syrup) for 6 months | -↓ significant decrease in the MDS-UPDRS score -↓ daily activities score -↓ motor test score -No change in tremor, rigidity score and YAHR score. | -Nausea -Diarrhea -Urticaria | Not applicable Ref [197] |
72 | -Stable PD medication -Hoehn-Yahr score ≤4 -50 to 80 years; All sexes | 2 groups: CSGs (Herbal cistanches 6 g, Polygonatum sibiricum 12 g, Salvia miltiorrhiza Bge. 15 g, Radix Paeoniae rubra 12 g, and Cortex Moutan 10 g), 2 capsules per day and placebo (Radix Bupleuri) for 12 weeks. | -↓ significative in the MDS-UPDRS sub-II score, -↓ significative in PDQ-39 score, -Significative improvement in CM syndrome score. | -Internal heat -Thirsty -Dryness heat. | ChiCTR-IOR-16008394 Ref [192] |
107 |
-Basic treatment with prednisone. -Sleep disorder | 2 groups: Huatan Jieyu granule and placebo for 4 weeks. | -↑ significative of total sleep time -↑ significative of NREM 2-3 -↑ significative of REM sleep -↓ significative in NREM1 | Not reported | Not applicable Ref [193] |
240 | -Stable medication with carbidopa/LD, anticholinergics, MAO inhibitors, or amantadine for at least 28 days -Hoehn and Yahr stage score of ≤4 -NMSS score ≥40 -18 to 80 years; All sexes | 2 groups: SQJZ herbal mixtures at dose of 29.375 g, 2 times per day and placebo for 12 weeks | Not reported | Not reported | Phase 2 NCT02616120 https://clinicaltrials.gov accessed on 3 June 2022 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Leathem, A.; Ortiz-Cerda, T.; Dennis, J.M.; Witting, P.K. Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression? Int. J. Mol. Sci. 2022, 23, 6923. https://doi.org/10.3390/ijms23136923
Leathem A, Ortiz-Cerda T, Dennis JM, Witting PK. Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression? International Journal of Molecular Sciences. 2022; 23(13):6923. https://doi.org/10.3390/ijms23136923
Chicago/Turabian StyleLeathem, Alexander, Tamara Ortiz-Cerda, Joanne M. Dennis, and Paul K. Witting. 2022. "Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression?" International Journal of Molecular Sciences 23, no. 13: 6923. https://doi.org/10.3390/ijms23136923
APA StyleLeathem, A., Ortiz-Cerda, T., Dennis, J. M., & Witting, P. K. (2022). Evidence for Oxidative Pathways in the Pathogenesis of PD: Are Antioxidants Candidate Drugs to Ameliorate Disease Progression? International Journal of Molecular Sciences, 23(13), 6923. https://doi.org/10.3390/ijms23136923