Oxidative Stress and Antioxidants in Neurodegenerative Disorders

Antioxidants - Tập 12 Số 2 - Trang 517
Edward O. Olufunmilayo1,2, Michelle B. Gerke-Duncan3, R. M. Damian Holsinger2
1Department of Medicine, University College Hospital, Queen Elizabeth Road, Oritamefa, Ibadan 5116, PMB, Nigeria
2Laboratory of Molecular Neuroscience and Dementia, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
3Education Innovation, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia

Tóm tắt

Neurodegenerative disorders constitute a substantial proportion of neurological diseases with significant public health importance. The pathophysiology of neurodegenerative diseases is characterized by a complex interplay of various general and disease-specific factors that lead to the end point of neuronal degeneration and loss, and the eventual clinical manifestations. Oxidative stress is the result of an imbalance between pro-oxidant species and antioxidant systems, characterized by an elevation in the levels of reactive oxygen and reactive nitrogen species, and a reduction in the levels of endogenous antioxidants. Recent studies have increasingly highlighted oxidative stress and associated mitochondrial dysfunction to be important players in the pathophysiologic processes involved in neurodegenerative conditions. In this article, we review the current knowledge of the general effects of oxidative stress on the central nervous system, the different specific routes by which oxidative stress influences the pathophysiologic processes involved in Alzheimer’s disease, Parkinson’s disease, Amyotrophic Lateral Sclerosis and Huntington’s disease, and how oxidative stress may be therapeutically reversed/mitigated in order to stall the pathological progression of these neurodegenerative disorders to bring about clinical benefits.

Từ khóa


Tài liệu tham khảo

Li, 2013, Oxidative stress and neurodegenerative disorders, Int. J. Mol. Sci., 14, 24438, 10.3390/ijms141224438

Allen, 2009, Oxidative stress and its role in the pathogenesis of ischaemic stroke, Int. J. Stroke, 4, 461, 10.1111/j.1747-4949.2009.00387.x

2014, Oxidative stress in traumatic brain injury, Curr. Med. Chem., 21, 1201, 10.2174/0929867321666131217153310

Anandhan, 2014, Antioxidant gene therapy against neuronal cell death, Pharmacol. Ther., 142, 206, 10.1016/j.pharmthera.2013.12.007

Hansen, 2018, Microglia in Alzheimer’s disease, J. Cell Biol., 217, 459, 10.1083/jcb.201709069

Deponte, 2013, Glutathione catalysis and the reaction mechanisms of glutathione-dependent enzymes, Biochim. Biophys. Acta, 1830, 3217, 10.1016/j.bbagen.2012.09.018

Franklin, 2011, Redox regulation of the intrinsic pathway in neuronal apoptosis, Antioxid. Redox Signal., 14, 1437, 10.1089/ars.2010.3596

Shibata, 2010, Paraquat-induced oxidative stress represses phosphatidylinositol 3-kinase activities leading to impaired glucose uptake in 3T3-L1 adipocytes, J. Biol. Chem., 285, 20915, 10.1074/jbc.M110.126482

Shinohara, 2010, Reactive oxygen generated by NADPH oxidase 1 (Nox1) contributes to cell invasion by regulating matrix metalloprotease-9 production and cell migration, J. Biol. Chem., 285, 4481, 10.1074/jbc.M109.071779

Patten, 2010, Reactive oxygen species: Stuck in the middle of neurodegeneration, J. Alzheimers Dis., 20, S357, 10.3233/JAD-2010-100498

Paiva, 2014, Are reactive oxygen species always detrimental to pathogens?, Antioxid. Redox Signal, 20, 1000, 10.1089/ars.2013.5447

Niedzielska, 2016, Oxidative Stress in Neurodegenerative Diseases, Mol. Neurobiol., 53, 4094, 10.1007/s12035-015-9337-5

Metodiewa, 2000, Reactive oxygen species and reactive nitrogen species: Relevance to cyto(neuro)toxic events and neurologic disorders. An overview, Neurotox. Res., 1, 197, 10.1007/BF03033290

Mitran, 2013, ROS and brain diseases: The good, the bad, and the ugly, Oxid. Med. Cell Longev., 2013, 963520

Lambert, 2009, Reactive oxygen species production by mitochondria, Methods Mol. Biol., 554, 165, 10.1007/978-1-59745-521-3_11

Braganza, 2020, Mitochondrial electron transport chain: Oxidative phosphorylation, oxidant production, and methods of measurement, Redox Biol., 37, 101674, 10.1016/j.redox.2020.101674

Cogliati, 2021, Regulation and functional role of the electron transport chain supercomplexes, Biochem. Soc. Trans., 49, 2655, 10.1042/BST20210460

Mailloux, 2013, Unearthing the secrets of mitochondrial ROS and glutathione in bioenergetics, Trends Biochem. Sci., 38, 592, 10.1016/j.tibs.2013.09.001

Pizzino, 2017, Oxidative Stress: Harms and Benefits for Human Health, Oxid. Med. Cell Longev., 2017, 8416763, 10.1155/2017/8416763

2002, Free radicals in the physiological control of cell function, Physiol Rev., 82, 47, 10.1152/physrev.00018.2001

Valko, 2004, Role of oxygen radicals in DNA damage and cancer incidence, Mol. Cell Biochem., 266, 37, 10.1023/B:MCBI.0000049134.69131.89

Valko, 2007, Free radicals and antioxidants in normal physiological functions and human disease, Int. J. Biochem. Cell Biol., 39, 44, 10.1016/j.biocel.2006.07.001

Sessa, 2012, Nitric oxide synthases: Regulation and function, Eur. Heart J., 33, 829, 10.1093/eurheartj/ehr304

Zhou, 2009, Neuronal nitric oxide synthase: Structure, subcellular localization, regulation, and clinical implications, Nitric Oxide, 20, 223, 10.1016/j.niox.2009.03.001

Nathan, 1991, Role of nitric oxide synthesis in macrophage antimicrobial activity, Curr. Opin. Immunol., 3, 65, 10.1016/0952-7915(91)90079-G

Augusto, 2002, Nitrogen dioxide and carbonate radical anion: Two emerging radicals in biology, Free Radic. Biol. Med., 32, 841, 10.1016/S0891-5849(02)00786-4

(2023, February 09). Nitronium Ion. Available online: https://www.chemeurope.com/en/encyclopedia/Nitronium_ion.html.

Bedard, 2007, The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology, Physiol. Rev., 87, 245, 10.1152/physrev.00044.2005

Frei, 1994, Reactive oxygen species and antioxidant vitamins: Mechanisms of action, Am. J. Med., 97, 5S, 10.1016/0002-9343(94)90292-5

Bozek, 2015, Organization and evolution of brain lipidome revealed by large-scale analysis of human, chimpanzee, macaque, and mouse tissues, Neuron, 85, 695, 10.1016/j.neuron.2015.01.003

Fritz, 2013, An overview of the chemistry and biology of reactive aldehydes, Free Radic. Biol. Med., 59, 85, 10.1016/j.freeradbiomed.2012.06.025

Halliwell, 2007, Biochemistry of oxidative stress, BioChem. Soc. Trans., 35, 1147, 10.1042/BST0351147

Vistoli, 2013, Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): An overview of their mechanisms of formation, Free Radic. Res., 47, 3, 10.3109/10715762.2013.815348

Nishida, 2013, Reactive oxygen species induce epigenetic instability through the formation of 8-hydroxydeoxyguanosine in human hepatocarcinogenesis, Dig. Dis., 31, 459, 10.1159/000355245

Mitsumoto, 2008, Oxidative stress biomarkers in sporadic ALS, Amyotroph. Lateral. Scler., 9, 177, 10.1080/17482960801933942

Valavanidis, 2013, Pulmonary oxidative stress, inflammation and cancer: Respirable particulate matter, fibrous dusts and ozone as major causes of lung carcinogenesis through reactive oxygen species mechanisms, Int. J. Environ. Res. Public Health, 10, 3886, 10.3390/ijerph10093886

Pinchuk, 2012, Evaluation of antioxidants: Scope, limitations and relevance of assays, Chem. Phys. Lipids, 165, 638, 10.1016/j.chemphyslip.2012.05.003

Ho, 2013, Biological markers of oxidative stress: Applications to cardiovascular research and practice, Redox Biol., 1, 483, 10.1016/j.redox.2013.07.006

Miller, 2014, Isoprostanes and neuroprostanes as biomarkers of oxidative stress in neurodegenerative diseases, Oxid. Med. Cell Longev., 2014, 572491, 10.1155/2014/572491

Wang, 2010, Selective neuronal vulnerability to oxidative stress in the brain, Front. Aging Neurosci., 2, 12

Braak, 1991, Neuropathological stageing of Alzheimer-related changes, Acta Neuropathol., 82, 239, 10.1007/BF00308809

Eichenbaum, 2001, The hippocampus and declarative memory: Cognitive mechanisms and neural codes, Behav. Brain Res., 127, 199, 10.1016/S0166-4328(01)00365-5

Nieuwenhuis, 2011, The role of the ventromedial prefrontal cortex in memory consolidation, Behav. Brain Res., 218, 325, 10.1016/j.bbr.2010.12.009

Talamini, 2012, Aging memories: Differential decay of episodic memory components, Learn. Mem., 19, 239, 10.1101/lm.024281.111

Carney, 1991, Reversal of age-related increase in brain protein oxidation, decrease in enzyme activity, and loss in temporal and spatial memory by chronic administration of the spin-trapping compound N-tert-butyl-alpha-phenylnitrone, Proc. Natl. Acad. Sci. USA, 88, 3633, 10.1073/pnas.88.9.3633

Fukui, 2001, Impairment of learning and memory in rats caused by oxidative stress and aging, and changes in antioxidative defense systems, Ann. N. Y. Acad. Sci., 928, 168, 10.1111/j.1749-6632.2001.tb05646.x

Sousa, 2000, Reorganization of the morphology of hippocampal neurites and synapses after stress-induced damage correlates with behavioral improvement, Neuroscience, 97, 253, 10.1016/S0306-4522(00)00050-6

McEwen, 2008, Understanding the potency of stressful early life experiences on brain and body function, Metabolism, 57, S11, 10.1016/j.metabol.2008.07.006

Sarnowska, 2002, Application of organotypic hippocampal culture for study of selective neuronal death, Folia Neuropathol., 40, 101

Wang, 2005, High intrinsic oxidative stress may underlie selective vulnerability of the hippocampal CA1 region, Brain Res. Mol. Brain Res., 140, 120, 10.1016/j.molbrainres.2005.07.018

Bearden, 2009, Altered hippocampal morphology in unmedicated patients with major depressive illness, ASN Neuro, 1, e00020, 10.1042/AN20090026

Ortega, 2010, Oxidative stress in Alzheimer’s disease hippocampus: A topographical study, J. Neurol. Sci., 299, 163, 10.1016/j.jns.2010.08.029

Brown, 2005, Mild, short-term stress alters dendritic morphology in rat medial prefrontal cortex, Cereb. Cortex, 15, 1714, 10.1093/cercor/bhi048

Radley, 2006, Repeated stress induces dendritic spine loss in the rat medial prefrontal cortex, Cereb. Cortex, 16, 313, 10.1093/cercor/bhi104

Liston, 2009, Psychosocial stress reversibly disrupts prefrontal processing and attentional control, Proc. Natl. Acad. Sci. USA, 106, 912, 10.1073/pnas.0807041106

Luethi, 2008, Stress effects on working memory, explicit memory, and implicit memory for neutral and emotional stimuli in healthy men, Front. Behav. Neurosci., 2, 5, 10.3389/neuro.08.005.2008

Cohen, 1999, The ubiquitin/proteasome pathway: Friend or foe in zinc-, cadmium-, and H2O2-induced neuronal oxidative stress, Mol. Biol. Rep., 26, 65, 10.1023/A:1006909918866

Wang, 2016, Protein misfolding in the endoplasmic reticulum as a conduit to human disease, Nature, 529, 326, 10.1038/nature17041

Preston, 2017, The evolving role of ubiquitin modification in endoplasmic reticulum-associated degradation, Biochem. J., 474, 445, 10.1042/BCJ20160582

Rizor, A., Pajarillo, E., Johnson, J., Aschner, M., and Lee, E. (2019). Astrocytic Oxidative/Nitrosative Stress Contributes to Parkinson’s Disease Pathogenesis: The Dual Role of Reactive Astrocytes. Antioxidants, 8.

Sofroniew, 2009, Molecular dissection of reactive astrogliosis and glial scar formation, Trends Neurosci., 32, 638, 10.1016/j.tins.2009.08.002

Pekny, 2016, Astrocytes: A central element in neurological diseases, Acta Neuropathol., 131, 323, 10.1007/s00401-015-1513-1

Jensen, 2013, Immune players in the CNS: The astrocyte, J. Neuroimmune Pharmacol., 8, 824, 10.1007/s11481-013-9480-6

Strowig, 2012, Inflammasomes in health and disease, Nature, 481, 278, 10.1038/nature10759

Zhou, 2011, A role for mitochondria in NLRP3 inflammasome activation, Nature, 469, 221, 10.1038/nature09663

Choi, 2007, Hydrogen peroxide induces the death of astrocytes through the down-regulation of the constitutive nuclear factor-kappaB activity, Free Radic. Res., 41, 555, 10.1080/10715760601173010

Jha, 2019, Microglia-Astrocyte Crosstalk: An Intimate Molecular Conversation, Neuroscientist, 25, 227, 10.1177/1073858418783959

Zuo, L., Prather, E.R., Stetskiv, M., Garrison, D.E., Meade, J.R., Peace, T.I., and Zhou, T. (2019). Inflammaging and Oxidative Stress in Human Diseases: From Molecular Mechanisms to Novel Treatments. Int. J. Mol. Sci., 20.

Ehrlich, 1998, Cytokine regulation of human microglial cell IL-8 production, J. Immunol., 160, 1944, 10.4049/jimmunol.160.4.1944

Olufunmilayo, 2022, Variant TREM2 Signaling in Alzheimer’s Disease, J. Mol. Biol., 434, 167470, 10.1016/j.jmb.2022.167470

Almazan, 2000, Exposure of developing oligodendrocytes to cadmium causes HSP72 induction, free radical generation, reduction in glutathione levels, and cell death, Free Radic. Biol. Med., 29, 858, 10.1016/S0891-5849(00)00384-1

Lassmann, 2016, Oxidative stress and its impact on neurons and glia in multiple sclerosis lesions, Biochim. Biophys. Acta, 1862, 506, 10.1016/j.bbadis.2015.09.018

Giacci, 2018, Oligodendroglia Are Particularly Vulnerable to Oxidative Damage after Neurotrauma In Vivo, J. Neurosci., 38, 6491, 10.1523/JNEUROSCI.1898-17.2018

Long, 2019, Alzheimer Disease: An Update on Pathobiology and Treatment Strategies, Cell, 179, 312, 10.1016/j.cell.2019.09.001

Nelson, 2009, Neuropathology and cognitive impairment in Alzheimer disease: A complex but coherent relationship, J. Neuropathol. Exp. Neurol., 68, 1, 10.1097/NEN.0b013e3181919a48

Butterfield, 2019, Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease, Nat Rev. Neurosci., 20, 148, 10.1038/s41583-019-0132-6

Grimm, 2017, Brain aging and neurodegeneration: From a mitochondrial point of view, J. Neurochem., 143, 418, 10.1111/jnc.14037

Mecocci, 2018, A Long Journey into Aging, Brain Aging, and Alzheimer’s Disease Following the Oxidative Stress Tracks, J. Alzheimers Dis., 62, 1319, 10.3233/JAD-170732

Swerdlow, 2014, The Alzheimer’s disease mitochondrial cascade hypothesis: Progress and perspectives, Biochim. Biophys. Acta, 1842, 1219, 10.1016/j.bbadis.2013.09.010

Cheignon, 2018, Oxidative stress and the amyloid beta peptide in Alzheimer’s disease, Redox Biol., 14, 450, 10.1016/j.redox.2017.10.014

Hensley, 1994, A model for beta-amyloid aggregation and neurotoxicity based on free radical generation by the peptide: Relevance to Alzheimer disease, Proc. Natl. Acad. Sci. USA, 91, 3270, 10.1073/pnas.91.8.3270

Pike, 1992, beta-Amyloid induces neuritic dystrophy in vitro: Similarities with Alzheimer pathology, Neuroreport, 3, 769, 10.1097/00001756-199209000-00012

Elangovan, 2020, Cyclical amyloid beta-astrocyte activity induces oxidative stress in Alzheimer’s disease, Biochimie, 171–172, 38, 10.1016/j.biochi.2020.02.003

Mark, 1997, A role for 4-hydroxynonenal, an aldehydic product of lipid peroxidation, in disruption of ion homeostasis and neuronal death induced by amyloid beta-peptide, J. Neurochem., 68, 255, 10.1046/j.1471-4159.1997.68010255.x

Liu, 1999, F4-isoprostanes as specific marker of docosahexaenoic acid peroxidation in Alzheimer’s disease, J. Neurochem., 72, 734, 10.1046/j.1471-4159.1999.0720734.x

Tramutola, 2017, Role of 4-hydroxy-2-nonenal (HNE) in the pathogenesis of alzheimer disease and other selected age-related neurodegenerative disorders, Free Radic. Biol. Med., 111, 253, 10.1016/j.freeradbiomed.2016.10.490

Smith, 1997, Widespread peroxynitrite-mediated damage in Alzheimer’s disease, J. Neurosci., 17, 2653, 10.1523/JNEUROSCI.17-08-02653.1997

Lyras, 1997, An assessment of oxidative damage to proteins, lipids, and DNA in brain from patients with Alzheimer’s disease, J. Neurochem., 68, 2061, 10.1046/j.1471-4159.1997.68052061.x

Nunomura, 2009, RNA oxidation in Alzheimer disease and related neurodegenerative disorders, Acta Neuropathol., 118, 151, 10.1007/s00401-009-0508-1

Butterfield, 2020, Brain lipid peroxidation and alzheimer disease: Synergy between the Butterfield and Mattson laboratories, Ageing Res. Rev., 64, 101049, 10.1016/j.arr.2020.101049

Keller, 1997, 4-Hydroxynonenal, an aldehydic product of membrane lipid peroxidation, impairs glutamate transport and mitochondrial function in synaptosomes, Neuroscience, 80, 685, 10.1016/S0306-4522(97)00065-1

Press, O.U. (2015). Free Radicals in Biology and Medicine, Oxford University Press. [5th ed.].

Cotman, 2021, Emerging roles of oxidative stress in brain aging and Alzheimer’s disease, Neurobiol. Aging, 107, 86, 10.1016/j.neurobiolaging.2021.07.014

Leuner, 2012, Mitochondrion-derived reactive oxygen species lead to enhanced amyloid beta formation, Antioxid. Redox Signal., 16, 1421, 10.1089/ars.2011.4173

Shelat, 2008, Amyloid beta peptide and NMDA induce ROS from NADPH oxidase and AA release from cytosolic phospholipase A2 in cortical neurons, J. Neurochem., 106, 45, 10.1111/j.1471-4159.2008.05347.x

Nanetti, 2005, Peroxynitrite production and NOS expression in astrocytes U373MG incubated with lipoproteins from Alzheimer patients, Brain Res., 1054, 38, 10.1016/j.brainres.2005.06.025

Fahn, 2004, Neurodegeneration and neuroprotection in Parkinson disease, NeuroRx, 1, 139, 10.1602/neurorx.1.1.139

Jenner, 2003, Oxidative stress in Parkinson’s disease, Ann. Neurol., 53, S26, 10.1002/ana.10483

Zhu, 2010, Mitochondrial dysfunction in Parkinson’s disease, J. Alzheimers Dis., 20, S325, 10.3233/JAD-2010-100363

Yoritaka, 1996, Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease, Proc. Natl. Acad. Sci. USA, 93, 2696, 10.1073/pnas.93.7.2696

Alam, 1997, Oxidative DNA damage in the parkinsonian brain: An apparent selective increase in 8-hydroxyguanine levels in substantia nigra, J. Neurochem., 69, 1196, 10.1046/j.1471-4159.1997.69031196.x

Richardson, 2005, Paraquat neurotoxicity is distinct from that of MPTP and rotenone, Toxicol. Sci., 88, 193, 10.1093/toxsci/kfi304

Callio, 2005, Manganese superoxide dismutase protects against 6-hydroxydopamine injury in mouse brains, J. Biol. Chem., 280, 18536, 10.1074/jbc.M413224200

Fowler, 1997, Age-related increases in brain monoamine oxidase B in living healthy human subjects, Neurobiol. Aging, 18, 431, 10.1016/S0197-4580(97)00037-7

Saura, 1997, Biphasic and region-specific MAO-B response to aging in normal human brain, Neurobiol. Aging, 18, 497, 10.1016/S0197-4580(97)00113-9

Kumar, 2004, Perspectives on MAO-B in aging and neurological disease: Where do we go from here?, Mol. Neurobiol., 30, 77, 10.1385/MN:30:1:077

Mallajosyula, J.K., Kaur, D., Chinta, S.J., Rajagopalan, S., Rane, A., Nicholls, D.G., Di Monte, D.A., Macarthur, H., and Andersen, J.K. (2008). MAO-B elevation in mouse brain astrocytes results in Parkinson’s pathology. PLoS ONE, 3.

Huenchuguala, 2012, Dopamine oxidation and autophagy, Park. Dis., 2012, 920953

Hastings, 2009, The role of dopamine oxidation in mitochondrial dysfunction: Implications for Parkinson’s disease, J. Bioenerg. Biomembr., 41, 469, 10.1007/s10863-009-9257-z

Yim, 1994, On the protective mechanism of the thiol-specific antioxidant enzyme against the oxidative damage of biomacromolecules, J. Biol. Chem., 269, 1621, 10.1016/S0021-9258(17)42072-2

Sian, 1994, Alterations in glutathione levels in Parkinson’s disease and other neurodegenerative disorders affecting basal ganglia, Ann. Neurol., 36, 348, 10.1002/ana.410360305

Sofic, 1992, Reduced and oxidized glutathione in the substantia nigra of patients with Parkinson’s disease, Neurosci. Lett., 142, 128, 10.1016/0304-3940(92)90355-B

Pearce, 1997, Alterations in the distribution of glutathione in the substantia nigra in Parkinson’s disease, J. Neural Transm., 104, 661, 10.1007/BF01291884

Ndayisaba, 2019, Iron in Neurodegeneration—Cause or Consequence?, Front. Neurosci., 13, 180, 10.3389/fnins.2019.00180

Dexter, 1991, Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s disease and other neurodegenerative diseases affecting the basal ganglia, Brain, 114, 1953, 10.1093/brain/114.4.1953

Urrutia, 2012, Iron toxicity in neurodegeneration, Biometals, 25, 761, 10.1007/s10534-012-9523-0

Ma, 2021, Parkinson’s disease: Alterations in iron and redox biology as a key to unlock therapeutic strategies, Redox Biol., 41, 101896, 10.1016/j.redox.2021.101896

Berg, 2001, Brain iron pathways and their relevance to Parkinson’s disease, J. Neurochem., 79, 225, 10.1046/j.1471-4159.2001.00608.x

Kaur, 2004, Does cellular iron dysregulation play a causative role in Parkinson’s disease?, Ageing Res. Rev., 3, 327, 10.1016/j.arr.2004.01.003

Langston, 1983, Chronic Parkinsonism in humans due to a product of meperidine-analog synthesis, Science, 219, 979, 10.1126/science.6823561

Martinez, 2012, Toxin models of mitochondrial dysfunction in Parkinson’s disease, Antioxid. Redox Signal., 16, 920, 10.1089/ars.2011.4033

Schapira, 1989, Mitochondrial complex I deficiency in Parkinson’s disease, Lancet, 1, 1269, 10.1016/S0140-6736(89)92366-0

Keeney, 2006, Parkinson’s disease brain mitochondrial complex I has oxidatively damaged subunits and is functionally impaired and misassembled, J. Neurosci., 26, 5256, 10.1523/JNEUROSCI.0984-06.2006

Elstner, 2011, Expression analysis of dopaminergic neurons in Parkinson’s disease and aging links transcriptional dysregulation of energy metabolism to cell death, Acta Neuropathol., 122, 75, 10.1007/s00401-011-0828-9

Spillantini, 1997, Alpha-synuclein in Lewy bodies, Nature, 388, 839, 10.1038/42166

Paxinou, 2001, Induction of alpha-synuclein aggregation by intracellular nitrative insult, J. Neurosci., 21, 8053, 10.1523/JNEUROSCI.21-20-08053.2001

Castellani, 2000, Sequestration of iron by Lewy bodies in Parkinson’s disease, Acta Neuropathol, 100, 111, 10.1007/s004010050001

Winklhofer, 2010, Mitochondrial dysfunction in Parkinson’s disease, Biochim. Biophys. Acta, 1802, 29, 10.1016/j.bbadis.2009.08.013

Chinta, 2010, Mitochondrial α-synuclein accumulation impairs complex I function in dopaminergic neurons and results in increased mitophagy in vivo, Neurosci. Lett., 486, 235, 10.1016/j.neulet.2010.09.061

Devi, 2008, Mitochondrial import and accumulation of alpha-synuclein impair complex I in human dopaminergic neuronal cultures and Parkinson disease brain, J. Biol. Chem., 283, 9089, 10.1074/jbc.M710012200

Hyun, 2002, Effect of wild-type or mutant Parkin on oxidative damage, nitric oxide, antioxidant defenses, and the proteasome, J. Biol. Chem., 277, 28572, 10.1074/jbc.M200666200

Iaccarino, 2007, Apoptotic mechanisms in mutant LRRK2-mediated cell death, Hum. Mol. Genet., 16, 1319, 10.1093/hmg/ddm080

Paterna, 2007, DJ-1 and Parkin modulate dopamine-dependent behavior and inhibit MPTP-induced nigral dopamine neuron loss in mice, Mol. Ther., 15, 698, 10.1038/sj.mt.6300067

Ziviani, 2010, Drosophila parkin requires PINK1 for mitochondrial translocation and ubiquitinates mitofusin, Proc. Natl. Acad. Sci. USA, 107, 5018, 10.1073/pnas.0913485107

Zarei, 2015, A comprehensive review of amyotrophic lateral sclerosis, Surg. Neurol. Int., 6, 171, 10.4103/2152-7806.169561

Tanaka, 2016, A 24-Week, Phase III, Double-Blind, Parallel-Group Study of Edaravone (MCI-186) for Treatment of Amyotrophic Lateral Sclerosis (ALS) (P3.189), Neurology, 86, P3.189

Paganoni, 2020, Trial of Sodium Phenylbutyrate-Taurursodiol for Amyotrophic Lateral Sclerosis, N. Engl. J. Med., 383, 919, 10.1056/NEJMoa1916945

Valdmanis, 2008, Genetics of familial amyotrophic lateral sclerosis, Neurology, 70, 144, 10.1212/01.wnl.0000296811.19811.db

Renton, 2011, A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD, Neuron, 72, 257, 10.1016/j.neuron.2011.09.010

Deng, 1993, Amyotrophic lateral sclerosis and structural defects in Cu,Zn superoxide dismutase, Science, 261, 1047, 10.1126/science.8351519

Kaur, 2016, Mutant SOD1 mediated pathogenesis of Amyotrophic Lateral Sclerosis, Gene, 577, 109, 10.1016/j.gene.2015.11.049

Sreedharan, 2008, TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis, Science, 319, 1668, 10.1126/science.1154584

Vance, 2009, Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6, Science, 323, 1208, 10.1126/science.1165942

Smith, 2019, The role of mitochondria in amyotrophic lateral sclerosis, Neurosci. Lett., 710, 132933, 10.1016/j.neulet.2017.06.052

Shaw, 1995, Oxidative damage to protein in sporadic motor neuron disease spinal cord, Ann Neurol, 38, 691, 10.1002/ana.410380424

Ferrante, 1997, Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis, J. Neurochem., 69, 2064, 10.1046/j.1471-4159.1997.69052064.x

Pedersen, 1998, Protein modification by the lipid peroxidation product 4-hydroxynonenal in the spinal cords of amyotrophic lateral sclerosis patients, Ann. Neurol., 44, 819, 10.1002/ana.410440518

Shibata, 2001, Morphological evidence for lipid peroxidation and protein glycoxidation in spinal cords from sporadic amyotrophic lateral sclerosis patients, Brain Res., 917, 97, 10.1016/S0006-8993(01)02926-2

Beal, 1997, Increased 3-nitrotyrosine in both sporadic and familial amyotrophic lateral sclerosis, Ann. Neurol., 42, 644, 10.1002/ana.410420416

Babu, 2008, Oxidant-antioxidant imbalance in the erythrocytes of sporadic amyotrophic lateral sclerosis patients correlates with the progression of disease, Neurochem. Int., 52, 1284, 10.1016/j.neuint.2008.01.009

Ferri, 2006, Familial ALS-superoxide dismutases associate with mitochondria and shift their redox potentials, Proc. Natl. Acad. Sci. USA, 103, 13860, 10.1073/pnas.0605814103

Haenen, 2003, Superoxide dismutase: The balance between prevention and induction of oxidative damage, Chem. Biol. Interact., 145, 33, 10.1016/S0009-2797(02)00160-6

Ihara, 2005, Oxidative stress and metal content in blood and cerebrospinal fluid of amyotrophic lateral sclerosis patients with and without a Cu, Zn-superoxide dismutase mutation, Neurol. Res., 27, 105, 10.1179/016164105X18430

Sau, 2007, Mutation of SOD1 in ALS: A gain of a loss of function, Hum. Mol. Genet., 16, 1604, 10.1093/hmg/ddm110

Menzies, 2002, Mitochondrial involvement in amyotrophic lateral sclerosis, Neurochem. Int., 40, 543, 10.1016/S0197-0186(01)00125-5

Bakavayev, 2019, Cu/Zn-superoxide dismutase and wild-type like fALS SOD1 mutants produce cytotoxic quantities of H(2)O(2) via cysteine-dependent redox short-circuit, Sci. Rep., 9, 10826, 10.1038/s41598-019-47326-x

Beckman, 1993, ALS, SOD and peroxynitrite, Nature, 364, 584, 10.1038/364584a0

Sheng, 2012, SOD1 aggregation and ALS: Role of metallation states and disulfide status, Curr. Top. Med. Chem., 12, 2560, 10.2174/1568026611212220010

Sato, 1992, Hydroxyl radical production by H2O2 plus Cu,Zn-superoxide dismutase reflects the activity of free copper released from the oxidatively damaged enzyme, J. Biol. Chem., 267, 25371, 10.1016/S0021-9258(19)74050-2

Petrov, 2016, Effect of Oxidative Damage on the Stability and Dimerization of Superoxide Dismutase 1, Biophys. J., 110, 1499, 10.1016/j.bpj.2016.02.037

Pickles, 2013, Mitochondrial damage revealed by immunoselection for ALS-linked misfolded SOD1, Hum. Mol. Genet., 22, 3947, 10.1093/hmg/ddt249

Wang, 2018, Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in Amyotrophic Lateral Sclerosis, Nat. Commun., 9, 3683, 10.1038/s41467-018-06111-6

Lai, 2019, C9ORF72 protein function and immune dysregulation in amyotrophic lateral sclerosis, Neurosci. Lett., 713, 134523, 10.1016/j.neulet.2019.134523

Lu, 2016, Poly(GR) in C9ORF72-Related ALS/FTD Compromises Mitochondrial Function and Increases Oxidative Stress and DNA Damage in iPSC-Derived Motor Neurons, Neuron, 92, 383, 10.1016/j.neuron.2016.09.015

The Huntington’s Disease Collaborative Research Group (1993). A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell, 72, 971–983.

DiFiglia, 1997, Aggregation of huntingtin in neuronal intranuclear inclusions and dystrophic neurites in brain, Science, 277, 1990, 10.1126/science.277.5334.1990

Penney, 1997, CAG repeat number governs the development rate of pathology in Huntington’s disease, Ann. Neurol., 41, 689, 10.1002/ana.410410521

Langbehn, 2004, A new model for prediction of the age of onset and penetrance for Huntington’s disease based on CAG length, Clin. Genet., 65, 267, 10.1111/j.1399-0004.2004.00241.x

Vonsattel, 1998, Huntington disease, J. Neuropathol. Exp. Neurol., 57, 369, 10.1097/00005072-199805000-00001

Browne, 1999, Oxidative stress in Huntington’s disease, Brain Pathol., 9, 147, 10.1111/j.1750-3639.1999.tb00216.x

Stack, 2008, Evidence of oxidant damage in Huntington’s disease: Translational strategies using antioxidants, Ann. N. Y. Acad. Sci., 1147, 79, 10.1196/annals.1427.008

Sorolla, 2008, Proteomic and oxidative stress analysis in human brain samples of Huntington disease, Free Radic. Biol. Med., 45, 667, 10.1016/j.freeradbiomed.2008.05.014

Long, 2012, 8OHdG as a marker for Huntington disease progression, Neurobiol. Dis., 46, 625, 10.1016/j.nbd.2012.02.012

Bandookwala, 2020, 3-Nitrotyrosine: A versatile oxidative stress biomarker for major neurodegenerative diseases, Int. J. Neurosci., 130, 1047, 10.1080/00207454.2020.1713776

Lee, 2011, Modulation of lipid peroxidation and mitochondrial function improves neuropathology in Huntington’s disease mice, Acta Neuropathol., 121, 487, 10.1007/s00401-010-0788-5

Enokido, 2010, Mutant huntingtin impairs Ku70-mediated DNA repair, J. Cell Biol., 189, 425, 10.1083/jcb.200905138

Hands, 2011, In vitro and in vivo aggregation of a fragment of huntingtin protein directly causes free radical production, J. Biol. Chem., 286, 44512, 10.1074/jbc.M111.307587

Giorgini, 2005, A genomic screen in yeast implicates kynurenine 3-monooxygenase as a therapeutic target for Huntington disease, Nat. Genet., 37, 526, 10.1038/ng1542

Sorolla, 2010, Protein oxidation in Huntington disease affects energy production and vitamin B6 metabolism, Free Radic. Biol. Med., 49, 612, 10.1016/j.freeradbiomed.2010.05.016

Brennan, 1985, Regional mitochondrial respiratory activity in Huntington’s disease brain, J. Neurochem., 44, 1948, 10.1111/j.1471-4159.1985.tb07192.x

Liot, 2009, Complex II inhibition by 3-NP causes mitochondrial fragmentation and neuronal cell death via an NMDA- and ROS-dependent pathway, Cell Death Differ., 16, 899, 10.1038/cdd.2009.22

Yang, 2008, Mitochondrial DNA damage and repair in neurodegenerative disorders, DNA Repair, 7, 1110, 10.1016/j.dnarep.2008.03.012

Siddiqui, 2012, Mitochondrial DNA damage is associated with reduced mitochondrial bioenergetics in Huntington’s disease, Free Radic. Biol. Med., 53, 1478, 10.1016/j.freeradbiomed.2012.06.008

Brustovetsky, 2016, Mutant Huntingtin and Elusive Defects in Oxidative Metabolism and Mitochondrial Calcium Handling, Mol. Neurobiol., 53, 2944, 10.1007/s12035-015-9188-0

Giacomello, 2007, Mitochondrial Ca2+ as a key regulator of cell life and death, Cell Death Differ., 14, 1267, 10.1038/sj.cdd.4402147

Suzuki, 2012, Calcium leak through ryanodine receptor is involved in neuronal death induced by mutant huntingtin, Biochem. Biophys. Res. Commun., 429, 18, 10.1016/j.bbrc.2012.10.107

Halestrap, 2006, Calcium, mitochondria and reperfusion injury: A pore way to die, Biochem. Soc. Trans., 34, 232, 10.1042/BST0340232

Rasola, 2010, Signal transduction to the permeability transition pore, FEBS Lett., 584, 1989, 10.1016/j.febslet.2010.02.022

Murphy, 1988, Calcium-dependent glutamate cytotoxicity in a neuronal cell line, Brain Res., 444, 325, 10.1016/0006-8993(88)90941-9

Miyamoto, 1989, Antioxidants protect against glutamate-induced cytotoxicity in a neuronal cell line, J. Pharmacol. Exp. Ther., 250, 1132

Bannai, 1980, Transport interaction of L-cystine and L-glutamate in human diploid fibroblasts in culture, J. Biol. Chem., 255, 2372, 10.1016/S0021-9258(19)85901-X

Tan, 2001, Oxytosis: A novel form of programmed cell death, Curr. Top. Med. Chem., 1, 497, 10.2174/1568026013394741

Dolma, 2003, Identification of genotype-selective antitumor agents using synthetic lethal chemical screening in engineered human tumor cells, Cancer Cell, 3, 285, 10.1016/S1535-6108(03)00050-3

Yagoda, 2007, RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels, Nature, 447, 864, 10.1038/nature05859

Yang, 2008, Synthetic lethal screening identifies compounds activating iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells, Chem. Biol., 15, 234, 10.1016/j.chembiol.2008.02.010

Dixon, 2012, Ferroptosis: An iron-dependent form of nonapoptotic cell death, Cell, 149, 1060, 10.1016/j.cell.2012.03.042

Yang, 2014, Regulation of ferroptotic cancer cell death by GPX4, Cell, 156, 317, 10.1016/j.cell.2013.12.010

Dixon, 2014, Pharmacological inhibition of cystine-glutamate exchange induces endoplasmic reticulum stress and ferroptosis, eLife, 3, e02523, 10.7554/eLife.02523

Tan, 1998, The regulation of reactive oxygen species production during programmed cell death, J. Cell Biol., 141, 1423, 10.1083/jcb.141.6.1423

Liu, 2009, The specificity of neuroprotection by antioxidants, J. Biomed. Sci., 16, 98, 10.1186/1423-0127-16-98

Neitemeier, 2017, BID links ferroptosis to mitochondrial cell death pathways, Redox Biol., 12, 558, 10.1016/j.redox.2017.03.007

Ansari, 2010, Oxidative stress in the progression of Alzheimer disease in the frontal cortex, J. Neuropathol. Exp. Neurol., 69, 155, 10.1097/NEN.0b013e3181cb5af4

Schulz, 2000, Glutathione, oxidative stress and neurodegeneration, Eur. J. Biochem., 267, 4904, 10.1046/j.1432-1327.2000.01595.x

Maher, 2020, Using the Oxytosis/Ferroptosis Pathway to Understand and Treat Age-Associated Neurodegenerative Diseases, Cell Chem. Biol., 27, 1456, 10.1016/j.chembiol.2020.10.010

Currais, 2014, Modulation of p25 and inflammatory pathways by fisetin maintains cognitive function in Alzheimer’s disease transgenic mice, Aging Cell, 13, 379, 10.1111/acel.12185

Currais, 2018, Fisetin Reduces the Impact of Aging on Behavior and Physiology in the Rapidly Aging SAMP8 Mouse, J. Gerontol. Ser. A Biol. Sci. Med. Sci., 73, 299, 10.1093/gerona/glx104

Hassan, 2022, The neuroprotective effects of fisetin, a natural flavonoid in neurodegenerative diseases: Focus on the role of oxidative stress, Front. Pharmacol., 13, 1015835, 10.3389/fphar.2022.1015835

Liu, 2008, A broadly neuroprotective derivative of curcumin, J. Neurochem., 105, 1336, 10.1111/j.1471-4159.2008.05236.x

Valera, 2013, Modulation of 5-lipoxygenase in proteotoxicity and Alzheimer’s disease, J. Neurosci., 33, 10512, 10.1523/JNEUROSCI.5183-12.2013

Jayaraj, 2014, CNB-001, a novel pyrazole derivative mitigates motor impairments associated with neurodegeneration via suppression of neuroinflammatory and apoptotic response in experimental Parkinson’s disease mice, Chem. Biol. Interact., 220, 149, 10.1016/j.cbi.2014.06.022

Kadiiska, 2013, Biomarkers of oxidative stress study V: Ozone exposure of rats and its effect on lipids, proteins, and DNA in plasma and urine, Free Radic. Biol. Med., 61, 408, 10.1016/j.freeradbiomed.2013.04.023

Nandi, 2019, Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases, Oxid. Med. Cell Longev., 2019, 9613090, 10.1155/2019/9613090

Sarıkaya, E., and Doğan, S. (2020). Glutathione System and Oxidative Stress in Health and Disease, IntechOpen.

Couto, 2016, The role of glutathione reductase and related enzymes on cellular redox homoeostasis network, Free Radic. Biol. Med., 95, 27, 10.1016/j.freeradbiomed.2016.02.028

Holmgren, 2000, Physiological functions of thioredoxin and thioredoxin reductase, Eur. J. Biochem., 267, 6102, 10.1046/j.1432-1327.2000.01701.x

Ren, 1997, The crystal structure of seleno-glutathione peroxidase from human plasma at 2.9 A resolution, J. Mol. Biol., 268, 869, 10.1006/jmbi.1997.1005

Gokul, 2014, Oral supplements of aqueous extract of tomato seeds alleviate motor abnormality, oxidative impairments and neurotoxicity induced by rotenone in mice: Relevance to Parkinson’s disease, Neurochem. Res, 39, 1382, 10.1007/s11064-014-1323-1

Budzynska, 2015, Effects of imperatorin on scopolamine-induced cognitive impairment and oxidative stress in mice, Psychopharmacology, 232, 931, 10.1007/s00213-014-3728-6

Tanaka, K., Kanno, T., Yanagisawa, Y., Yasutake, K., Inoue, S., Hirayama, N., and Ikeda, J.E. (2014). A novel acylaminoimidazole derivative, WN1316, alleviates disease progression via suppression of glial inflammation in ALS mouse model. PLoS ONE, 9.

Suganya, 2017, Effect of rutin against a mitochondrial toxin, 3-nitropropionicacid induced biochemical, behavioral and histological alterations-a pilot study on Huntington’s disease model in rats, Metab. Brain Dis., 32, 471, 10.1007/s11011-016-9929-4

Feng, 2012, Antioxidant therapies for Alzheimer’s disease, Oxid. Med. Cell Longev., 2012, 472932, 10.1155/2012/472932

Lloret, A., Esteve, D., Monllor, P., Cervera-Ferri, A., and Lloret, A. (2019). The Effectiveness of Vitamin E Treatment in Alzheimer’s Disease. Int. J. Mol. Sci., 20.

Dong, 2018, Do low-serum vitamin E levels increase the risk of Alzheimer disease in older people? Evidence from a meta-analysis of case-control studies, Int. J. Geriatr. Psychiatry, 33, e257, 10.1002/gps.4780

Casati, 2020, Vitamin E and Alzheimer’s disease: The mediating role of cellular aging, Aging Clin. Exp. Res., 32, 459, 10.1007/s40520-019-01209-3

Kandiah, 2019, Treatment of dementia and mild cognitive impairment with or without cerebrovascular disease: Expert consensus on the use of Ginkgo biloba extract, EGb 761(®), CNS Neurosci. Ther., 25, 288, 10.1111/cns.13095

Cowan, 2021, Suppression of tau-induced phenotypes by vitamin E demonstrates the dissociation of oxidative stress and phosphorylation in mechanisms of tau toxicity, J. Neurochem., 157, 684, 10.1111/jnc.15253

Marí, M., de Gregorio, E., de Dios, C., Roca-Agujetas, V., Cucarull, B., Tutusaus, A., Morales, A., and Colell, A. (2020). Mitochondrial Glutathione: Recent Insights and Role in Disease. Antioxidants, 9.

Fernandez, 2013, APP/PS1 mice overexpressing SREBP-2 exhibit combined Abeta accumulation and tau pathology underlying Alzheimer’s disease, Hum. Mol. Genet., 22, 3460, 10.1093/hmg/ddt201

Nishida, 2005, Metabolic effects of melatonin on oxidative stress and diabetes mellitus, Endocrine, 27, 131, 10.1385/ENDO:27:2:131

Deng, 2005, Effects of melatonin on wortmannin-induced tau hyperphosphorylation, Acta Pharmacol. Sin., 26, 519, 10.1111/j.1745-7254.2005.00102.x

Zhou, 2008, Melatonin impairs NADPH oxidase assembly and decreases superoxide anion production in microglia exposed to amyloid-beta1-42, J. Pineal. Res., 45, 157, 10.1111/j.1600-079X.2008.00570.x

Baum, 2004, Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer’s disease animal models, J. Alzheimers Dis., 6, 367, 10.3233/JAD-2004-6403

Nishinaka, 2007, Curcumin activates human glutathione S-transferase P1 expression through antioxidant response element, Toxicol. Lett., 170, 238, 10.1016/j.toxlet.2007.03.011

Frank, 2005, A review of antioxidants and Alzheimer’s disease, Ann. Clin. Psychiatry, 17, 269, 10.1080/10401230500296428

Sheykhhasan, 2022, Neuroprotective effects of coenzyme Q10-loaded exosomes obtained from adipose-derived stem cells in a rat model of Alzheimer’s disease, Biomed. Pharmacother., 152, 113224, 10.1016/j.biopha.2022.113224

Kaur, 2021, Decrypting the potential role of α-lipoic acid in Alzheimer’s disease, Life Sci., 284, 119899, 10.1016/j.lfs.2021.119899

Gomes, 2018, Neuroprotective Mechanisms of Resveratrol in Alzheimer’s Disease: Role of SIRT1, Oxid. Med. Cell Longev., 2018, 8152373, 10.1155/2018/8152373

Duan, 2015, Silibinin inhibits acetylcholinesterase activity and amyloid β peptide aggregation: A dual-target drug for the treatment of Alzheimer’s disease, Neurobiol. Aging, 36, 1792, 10.1016/j.neurobiolaging.2015.02.002

Jakaria, 2021, Biological evidence of gintonin efficacy in memory disorders, Pharmacol. Res., 163, 105221, 10.1016/j.phrs.2020.105221

Tripathi, 2020, Ameliorative effects of apple cider vinegar on neurological complications via regulation of oxidative stress markers, J. Food Biochem., 44, e13504, 10.1111/jfbc.13504

Burgener, 2008, Evidence supporting nutritional interventions for persons in early stage Alzheimer’s disease (AD), J. Nutr. Health Aging, 12, 18, 10.1007/BF02982159

Fahn, 1992, A pilot trial of high-dose alpha-tocopherol and ascorbate in early Parkinson’s disease, Ann. Neurol., 32, S128, 10.1002/ana.410320722

Hantikainen, 2021, Dietary Antioxidants and the Risk of Parkinson Disease: The Swedish National March Cohort, Neurology, 96, e895, 10.1212/WNL.0000000000011373

Zhang, 2002, Intakes of vitamins E and C, carotenoids, vitamin supplements, and PD risk, Neurology, 59, 1161, 10.1212/01.WNL.0000028688.75881.12

Group, 1993, Effects of tocopherol and deprenyl on the progression of disability in early Parkinson’s disease, N. Engl. J. Med., 328, 176, 10.1056/NEJM199301213280305

Group, 1998, Mortality in DATATOP: A multicenter trial in early Parkinson’s disease. Parkinson Study Group, Ann. Neurol., 43, 318, 10.1002/ana.410430309

Shults, 2002, Effects of coenzyme Q10 in early Parkinson disease: Evidence of slowing of the functional decline, Arch. Neurol., 59, 1541, 10.1001/archneur.59.10.1541

Beal, 2014, A randomized clinical trial of high-dosage coenzyme Q10 in early Parkinson disease: No evidence of benefit, JAMA Neurol., 71, 543, 10.1001/jamaneurol.2014.131

Lan, 1997, Desferrioxamine and vitamin E protect against iron and MPTP-induced neurodegeneration in mice, J. Neural Transm., 104, 469, 10.1007/BF01277665

Dexter, 2011, Clinically available iron chelators induce neuroprotection in the 6-OHDA model of Parkinson’s disease after peripheral administration, J. Neural Transm., 118, 223, 10.1007/s00702-010-0531-3

Thomas, 2004, Melatonin protects against oxidative stress caused by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in the mouse nigrostriatum, J. Pineal. Res., 36, 25, 10.1046/j.1600-079X.2003.00096.x

Su, 2015, Melatonin attenuates MPTP-induced neurotoxicity via preventing CDK5-mediated autophagy and SNCA/α-synuclein aggregation, Autophagy, 11, 1745, 10.1080/15548627.2015.1082020

Ozsoy, 2015, Melatonin is protective against 6-hydroxydopamine-induced oxidative stress in a hemiparkinsonian rat model, Free Radic. Res., 49, 1004, 10.3109/10715762.2015.1027198

Saravanan, 2007, Melatonin protects against rotenone-induced oxidative stress in a hemiparkinsonian rat model, J. Pineal. Res., 42, 247, 10.1111/j.1600-079X.2006.00412.x

Morgan, 2001, Chronic administration of pharmacological levels of melatonin does not ameliorate the MPTP-induced degeneration of the nigrostriatal pathway, Brain Res., 921, 115, 10.1016/S0006-8993(01)03106-7

Beal, 2011, Neuroprotective effects of creatine, Amino Acids, 40, 1305, 10.1007/s00726-011-0851-0

Mo, J.J., Liu, L.Y., Peng, W.B., Rao, J., Liu, Z., and Cui, L.L. (2017). The effectiveness of creatine treatment for Parkinson’s disease: An updated meta-analysis of randomized controlled trials. BMC Neurol, 17.

Khan, 2018, Role of free radicals and certain antioxidants in the management of huntington’s disease: A review, J. Anal. Pharm. Res., 7, 386, 10.15406/japlr.2018.07.00256

Chabrier, 2007, Pharmacological properties of BN82451: A novel multitargeting neuroprotective agent, CNS Drug Rev., 13, 317, 10.1111/j.1527-3458.2007.00018.x

Xun, 2012, Targeting of XJB-5-131 to mitochondria suppresses oxidative DNA damage and motor decline in a mouse model of Huntington’s disease, Cell Rep., 2, 1137, 10.1016/j.celrep.2012.10.001

Polyzos, 2016, Mitochondrial targeting of XJB-5-131 attenuates or improves pathophysiology in HdhQ150 animals with well-developed disease phenotypes, Hum. Mol. Genet., 25, 1792, 10.1093/hmg/ddw051

Kumar, 2010, Protective effect of hesperidin and naringin against 3-nitropropionic acid induced Huntington’s like symptoms in rats: Possible role of nitric oxide, Behav. Brain Res., 206, 38, 10.1016/j.bbr.2009.08.028

Richetti, 2011, Quercetin and rutin prevent scopolamine-induced memory impairment in zebrafish, Behav. Brain Res., 217, 10, 10.1016/j.bbr.2010.09.027

Menze, 2012, Potential neuroprotective effects of hesperidin on 3-nitropropionic acid-induced neurotoxicity in rats, Neurotoxicology, 33, 1265, 10.1016/j.neuro.2012.07.007

Petri, 2012, Nrf2/ARE Signaling Pathway: Key Mediator in Oxidative Stress and Potential Therapeutic Target in ALS, Neurol. Res. Int., 2012, 878030, 10.1155/2012/878030

Wang, 2011, Vitamin E intake and risk of amyotrophic lateral sclerosis: A pooled analysis of data from 5 prospective cohort studies, Am. J. Epidemiol., 173, 595, 10.1093/aje/kwq416

Freedman, 2013, Comment on “Intakes of vitamin C and carotenoids and risk of amyotrophic lateral sclerosis: Pooled results from 5 cohort studies”, Ann. Neurol., 74, 307, 10.1002/ana.23952

Desnuelle, 2001, A double-blind, placebo-controlled randomized clinical trial of alpha-tocopherol (vitamin E) in the treatment of amyotrophic lateral sclerosis. ALS riluzole-tocopherol Study Group, Amyotroph. Lateral. Scler. OTher. Motor Neuron Disord, 2, 9, 10.1080/146608201300079364

Noh, 2000, A novel neuroprotective mechanism of riluzole: Direct inhibition of protein kinase C, Neurobiol. Dis., 7, 375, 10.1006/nbdi.2000.0297

Deng, 2012, Riluzole-triggered GSH synthesis via activation of glutamate transporters to antagonize methylmercury-induced oxidative stress in rat cerebral cortex, Oxid. Med. Cell Longev., 2012, 534705, 10.1155/2012/534705

Beretta, 2003, Mitochondrial dysfunction due to mutant copper/zinc superoxide dismutase associated with amyotrophic lateral sclerosis is reversed by N-acetylcysteine, Neurobiol. Dis., 13, 213, 10.1016/S0969-9961(03)00043-3

Andreassen, 2000, N-acetyl-L-cysteine improves survival and preserves motor performance in an animal model of familial amyotrophic lateral sclerosis, Neuroreport, 11, 2491, 10.1097/00001756-200008030-00029

Kurano, T., Kanazawa, T., Iioka, S., Kondo, H., Kosuge, Y., and Suzuki, T. (2022). Intranasal Administration of N-acetyl-L-cysteine Combined with Cell-Penetrating Peptide-Modified Polymer Nanomicelles as a Potential Therapeutic Approach for Amyotrophic Lateral Sclerosis. Pharmaceutics, 14.

Louwerse, 1995, Randomized, double-blind, controlled trial of acetylcysteine in amyotrophic lateral sclerosis, Arch. Neurol., 52, 559, 10.1001/archneur.1995.00540300031009

Matthews, 1998, Coenzyme Q10 administration increases brain mitochondrial concentrations and exerts neuroprotective effects, Proc. Natl. Acad. Sci. USA, 95, 8892, 10.1073/pnas.95.15.8892

Kawasaki, 2012, Effects of Coenzyme Q10 Administration in Amyotrophic Lateral Sclerosis (ALS). Report of a Case and Review, Open Nutraceuticals J., 5, 187, 10.2174/1876396001205010187

Lucchetti, J., Marino, M., Papa, S., Tortarolo, M., Guiso, G., Pozzi, S., Bonetto, V., Caccia, S., Beghi, E., and Bendotti, C. (2013). A mouse model of familial ALS has increased CNS levels of endogenous ubiquinol9/10 and does not benefit from exogenous administration of ubiquinol10. PLoS ONE, 8.

Weishaupt, 2006, Reduced oxidative damage in ALS by high-dose enteral melatonin treatment, J. Pineal. Res., 41, 313, 10.1111/j.1600-079X.2006.00377.x

Zhang, 2013, Melatonin inhibits the caspase-1/cytochrome c/caspase-3 cell death pathway, inhibits MT1 receptor loss and delays disease progression in a mouse model of amyotrophic lateral sclerosis, Neurobiol. Dis., 55, 26, 10.1016/j.nbd.2013.03.008

Dardiotis, 2013, Intraperitoneal melatonin is not neuroprotective in the G93ASOD1 transgenic mouse model of familial ALS and may exacerbate neurodegeneration, Neurosci. Lett., 548, 170, 10.1016/j.neulet.2013.05.058

Barua, 2019, The role of NOX inhibitors in neurodegenerative diseases, IBRO Rep., 7, 59, 10.1016/j.ibror.2019.07.1721

Wu, 2006, The inflammatory NADPH oxidase enzyme modulates motor neuron degeneration in amyotrophic lateral sclerosis mice, Proc. Natl. Acad. Sci. USA, 103, 12132, 10.1073/pnas.0603670103

Harraz, 2008, SOD1 mutations disrupt redox-sensitive Rac regulation of NADPH oxidase in a familial ALS model, J. Clin. Investig., 118, 659

Trumbull, 2012, Diapocynin and apocynin administration fails to significantly extend survival in G93A SOD1 ALS mice, Neurobiol. Dis., 45, 137, 10.1016/j.nbd.2011.07.015

Salminen, 2013, Crosstalk between Oxidative Stress and SIRT1: Impact on the Aging Process, Int. J. Mol. Sci., 14, 3834, 10.3390/ijms14023834

Thau, 2013, Differential sirtuin expression patterns in amyotrophic lateral sclerosis (ALS) postmortem tissue: Neuroprotective or neurotoxic properties of sirtuins in ALS?, Neurodegener. Dis., 11, 141, 10.1159/000338048

Han, 2012, Resveratrol upregulated heat shock proteins and extended the survival of G93A-SOD1 mice, Brain Res., 1483, 112, 10.1016/j.brainres.2012.09.022

Song, 2014, Resveratrol ameliorates motor neuron degeneration and improves survival in SOD1(G93A) mouse model of amyotrophic lateral sclerosis, Biomed. Res Int., 2014, 483501, 10.1155/2014/483501

Yoshino, 2006, Investigation of the therapeutic effects of edaravone, a free radical scavenger, on amyotrophic lateral sclerosis (Phase II study), Amyotroph. Lateral. Scler., 7, 241, 10.1080/17482960600881870

Kaya, 2013, Beneficial effect of melatonin treatment on substantia nigra in an experimental model of Parkinson’s disease, J. Neurol. Sci., 30, 142

Sharma, 2007, Attenuation of 1-methyl-4-phenyl-1, 2,3,6-tetrahydropyridine induced nigrostriatal toxicity in mice by N-acetyl cysteine, Cell Mol. Biol., 53, 48

Galasko, 2012, Antioxidants for Alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures, Arch. Neurol., 69, 836, 10.1001/archneurol.2012.85