Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases
Abstract
:1. Introduction
1.1. Endogenous Oxidants and Antioxidants
1.2. ROS Generation
1.3. Oxidative Damage
2. The Vulnerability of the CNS to Oxidative Stress
3. Crosstalk Between Oxidative Stress and Inflammation
3.1. Roles of Microglia
3.2. Roles of Astrocytes
3.3. Cell Signaling Pathways
3.4. Feedback on Inflammation
3.5. Summary
4. Role of Oxidative Stress in Alzheimer’s Disease
4.1. ROS Generation in AD
4.2. Risk Factors for AD and Oxidative Stress
4.3. Oxidative Stress and Extracellular Plaques
4.4. Oxidative Stress and Intracellular Neurofibrillary Tangles
4.5. Oxidative Stress and Neuronal Dysfunction
Proteins | Function | Role in AD | References |
---|---|---|---|
ATP synthase | ATP synthase is mitochondrial complex V of the electron transport chain that plays a key role in ATP production. | The oxidation of its alpha-subunit has been found in the AD brain. Decreased activity of ATP synthase was observed in the cortex of an AD mouse and patients. | [142,143,144] |
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) | GAPDH is a key enzyme in glycolysis that metabolizes glucose to ATP. | Oxidative stress can induce GAPDH to denature and aggregate, decreasing its activity. Aggregated GADPH promotes amyloidogenesis. Oxidative stress also causes the interaction between GADPH and c-Jun N-terminal kinase (JNK) and activates JNK. | [145,146,147] |
Enolase 1 (ENO1, alpha-enolase) | ENO1 is a glycolytic enzyme catalyzing the conversion of 2-phosphoglycerate to phosphoenolpyruvate. | ENO1 is upregulated in AD, reflecting a defense mechanism. ENO1 also interacts with heat shock protein 70 (HSP70) to protect neurons against oxidative stress. The ENO1 oxidation and decreased activity were found in AD brains. | [136,137,147,148] |
Phosphoglucomutase 1 (PGM1) | Glycolytic enzyme catalyzes the conversion between glucose-1-phosphate and glucose-6-phosphate | The oxidized PGM1 and decreased activity were found in late AD patients. | [149] |
Fructose bisphosphate aldolase (FBA) | Converts fructose 1,6-bisphosphate to glyceraldehyde 3-phosphate and dihydroxyacetone phosphate in glycolysis. It also plays a role in DNA repair. | Oxidative stress can inactivate FBA in AD brains, disrupting glucose metabolism, reducing ATP production, impairing DNA repair, and causing neuronal dysfunction. | [150,151] |
Creatine kinase (CK) | Facilitates the transfer of phosphate between ATP and creatine, increasing ATP generation from ADP | Cytosolic brain-type creatine kinase (BB-CK) was significantly inactivated by oxidation in AD patients, and carbonylated BB-CK was identified in AD brains. | [152] |
Phosphoglycerate mutase 1 (PGAM1) | A crucial enzyme in glycolysis, which catalyzes the interconversion of 3-phosphoglycerate and 2-phosphoglycerate | The oxidation of PGAM1 was observed in AD, altering its structure and function and impairing its role in energy metabolism. | [153] |
Pyruvate kinase | Glycolytic enzyme, catalyzing the transfer of phosphate from phosphoenolpyruvate to ADP, producing pyruvate and ATP. | Pyruvate kinase M2 (PKM2) is an activator of γ-secretase, which is involved in the production of Aβ. ROS inhibits PKM2, potentially increasing Aβ production and impairing energy production. | [154,155] |
Lactate dehydrogenase (LDH) | Catalyzes the interconversion of pyruvate and lactate, playing a role in glycolysis and ATP production. | Increased lactate levels and the activity of LDH were considered a promoting factor in Aβ accumulation and plaque formation. Oxidative stress may induce cells to shift more to glycolysis and increase LDH. | [156,157] |
Malate dehydrogenase (MDH) | Catalyzes the oxidation of malate to oxaloacetate using the reduction in NAD+ to NADH in the tricarboxylic acid cycle and the malate-aspartate shuttle. | Oxidative stress can increase mitochondrial MDH activity and mRNA levels in hippocampal cells, possibly as a compensatory response. | [158] |
Triose-phosphate isomerase (TPI) | Catalyzes the conversion of dihydroxyacetone phosphate to glyceraldehyde-3-phosphate in glycolysis. | In the AD brains, TPI is oxidized by carbonyl and nitrotyrosinated modification, disrupting its enzymatic activity. Nitrotyrosinated TPI interacts with Tau to promote intraneuronal aggregation and increases methylglyoxal toxic proteins. | [142,159] |
Ubiquitin Carboxyl-terminal Hydrolase L1 (UCH-L1) | Deubiquitinates proteins, influencing their degradation pathways. | Oxidative stress induces the oxidation of UCH-L1, decreasing its activity and impairing protein quality control, which is implicated in AD pathology. In AD, a reduction in UCH-L1 levels is observed. | [160,161] |
Glucose-regulated protein precursor (GRP) | GRP78 and GRP94 are primarily expressed in the ER lumen to regulate the proper folding of proteins synthesized within the endoplasmic reticulum (ER). | The ER stress and oxidation of GRP78 and GRP94 have been observed in AD brains, leading to misfolding of proteins and aggregation. | [162,163] |
Superoxide dismutase (SOD) | Catalyzes the dismutation of the superoxide anion radical into normal molecular oxygen and hydrogen peroxide. | Increased expression of SOD1 and SOD2 within senile plaques, but decreased activities of SOD were found in AD. Another study indicates that levels of SOD1 were significantly decreased in human AD patients. | [164,165] |
Glutathione peroxidase (GPX) | Catalyzes the reduction in hydrogen peroxide and lipid peroxides. | Knockout of GPX1 aggravates Aβ-induced neurotoxicity in cortical neurons. Increasing the expression and activity of GPX4 ameliorates cognitive function in AD. | [166,167] |
Heme oxygenase-1 (HO-1) | Catalyzes heme into carbon monoxide (CO), free iron, and biliverdin, then to bilirubin. | Protects against oxidative stress and inflammation through its catalyzed products and also inhibits Aβ-induced neurotoxicity by CO. | [168] |
Heat shock proteins (HSPs) | Functions in protein folding, degradation, and cellular stress responses. | HSPs may function to protect against oxidative stress and improve the clearance of Aβ plaques. | [169] |
Glutamine Synthetase | Synthesizes glutamine from glutamate and ammonia, playing a role in ammonia detoxification and neurotransmitter regulation in the brain | Reduced expression and activity in AD brains have been observed. | [170,171] |
4.6. Antioxidants in AD
5. Role of Oxidative Stress in Parkinson’s Disease
5.1. Mitochondria and Oxidative Stress in PD
5.2. NADPH Oxidase and Oxidative Stress in PD
5.3. Nrf2-ARE-NLRP3 Inflammasomes
5.4. Antioxidants in PD
6. Role of Oxidative Stress in Huntington’s Disease
7. Role of Oxidative Stress in Amyotrophic Lateral Sclerosis
7.1. Gene Mutations, Mitochondria, and Oxidative Stress in ALS
7.1.1. SOD1
7.1.2. TARDBP
7.1.3. C9orf72
7.1.4. FUS
7.1.5. ANG
7.1.6. CHCHD10
7.2. Oxidative and Inflammation Crosstalk in ALS
7.3. Toxin Exposure and Oxidative Stress in ALS
7.4. Antioxidants in ALS
8. Conclusions
Funding
Acknowledgments
Conflicts of Interest
References
- Rotariu, D.; Babes, E.E.; Tit, D.M.; Moisi, M.; Bustea, C.; Stoicescu, M.; Radu, A.F.; Vesa, C.M.; Behl, T.; Bungau, A.F.; et al. Oxidative stress—Complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed. Pharmacother. 2022, 152, 113238. [Google Scholar] [CrossRef] [PubMed]
- Chong, Z.Z.; Li, F.; Maiese, K. Oxidative stress in the brain: Novel cellular targets that govern survival during neurodegenerative disease. Prog. Neurobiol. 2005, 75, 207–246. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. Oxidative Stress: Concept and Some Practical Aspects. Antioxidants 2020, 9, 852. [Google Scholar] [CrossRef]
- Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free radicals: Properties, sources, targets, and their implication in various diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [PubMed]
- Younus, H. Therapeutic potentials of superoxide dismutase. Int. J. Health Sci. (Qassim) 2018, 12, 88–93. [Google Scholar] [PubMed]
- Lubos, E.; Loscalzo, J.; Handy, D.E. Glutathione peroxidase-1 in health and disease: From molecular mechanisms to therapeutic opportunities. Antioxid. Redox Signal. 2011, 15, 1957–1997. [Google Scholar] [CrossRef]
- Okoye, C.N.; Koren, S.A.; Wojtovich, A.P. Mitochondrial complex I ROS production and redox signaling in hypoxia. Redox Biol. 2023, 67, 102926. [Google Scholar] [CrossRef]
- Srivastava, S.P.; Shi, S.; Koya, D.; Kanasaki, K. Lipid mediators in diabetic nephropathy. Fibrogenesis Tissue Repair 2014, 7, 12. [Google Scholar] [CrossRef]
- Inoguchi, T.; Sonta, T.; Tsubouchi, H.; Etoh, T.; Kakimoto, M.; Sonoda, N.; Sato, N.; Sekiguchi, N.; Kobayashi, K.; Sumimoto, H.; et al. Protein kinase C-dependent increase in reactive oxygen species (ROS) production in vascular tissues of diabetes: Role of vascular NAD(P)H oxidase. J. Am. Soc. Nephrol. 2003, 14, S227–S232. [Google Scholar] [CrossRef]
- Cho, K.J.; Seo, J.M.; Kim, J.H. Bioactive lipoxygenase metabolites stimulation of NADPH oxidases and reactive oxygen species. Mol. Cells 2011, 32, 1–5. [Google Scholar] [CrossRef]
- Kim, H.R.; Lee, H.N.; Lim, K.; Surh, Y.J.; Na, H.K. 15-Deoxy-Delta12,14-prostaglandin J2 induces expression of 15-hydroxyprostaglandin dehydrogenase through Elk-1 activation in human breast cancer MDA-MB-231 cells. Mutat. Res. 2014, 768, 6–15. [Google Scholar] [CrossRef]
- Sadok, A.; Bourgarel-Rey, V.; Gattacceca, F.; Penel, C.; Lehmann, M.; Kovacic, H. Nox1-dependent superoxide production controls colon adenocarcinoma cell migration. Biochim. Biophys. Acta 2008, 1783, 23–33. [Google Scholar] [CrossRef]
- Zhang, Z.; Blake, D.R.; Stevens, C.R.; Kanczler, J.M.; Winyard, P.G.; Symons, M.C.; Benboubetra, M.; Harrison, R. A reappraisal of xanthine dehydrogenase and oxidase in hypoxic reperfusion injury: The role of NADH as an electron donor. Free Radic. Res. 1998, 28, 151–164. [Google Scholar] [CrossRef] [PubMed]
- Veith, A.; Moorthy, B. Role of Cytochrome P450s in the Generation and Metabolism of Reactive Oxygen Species. Curr. Opin. Toxicol. 2018, 7, 44–51. [Google Scholar] [CrossRef]
- Su, L.J.; Zhang, J.H.; Gomez, H.; Murugan, R.; Hong, X.; Xu, D.; Jiang, F.; Peng, Z.Y. Reactive Oxygen Species-Induced Lipid Peroxidation in Apoptosis, Autophagy, and Ferroptosis. Oxid. Med. Cell. Longev. 2019, 2019, 5080843. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.; Sun, J.; Luo, Z.; Li, Y.; Huang, Y. Emerging mechanisms of lipid peroxidation in regulated cell death and its physiological implications. Cell Death Dis. 2024, 15, 859. [Google Scholar] [CrossRef] [PubMed]
- Juan, C.A.; Perez de la Lastra, J.M.; Plou, F.J.; Perez-Lebena, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
- Del Rio, D.; Stewart, A.J.; Pellegrini, N. A review of recent studies on malondialdehyde as toxic molecule and biological marker of oxidative stress. Nutr. Metab. Cardiovasc. Dis. 2005, 15, 316–328. [Google Scholar] [CrossRef]
- Cai, J.; Chen, J.; He, H.; Yin, Z.; Zhu, Z.; Yin, D. Carbonyl stress: Malondialdehyde induces damage on rat hippocampal neurons by disturbance of Ca2+ homeostasis. Cell Biol. Toxicol. 2009, 25, 435–445. [Google Scholar] [CrossRef]
- Li, F.; Yang, Z.; Lu, Y.; Wei, Y.; Wang, J.; Yin, D.; He, R. Malondialdehyde suppresses cerebral function by breaking homeostasis between excitation and inhibition in turtle Trachemys scripta. PLoS ONE 2010, 5, e15325. [Google Scholar] [CrossRef]
- Sultana, R.; Perluigi, M.; Butterfield, D.A. Lipid peroxidation triggers neurodegeneration: A redox proteomics view into the Alzheimer disease brain. Free Radic. Biol. Med. 2013, 62, 157–169. [Google Scholar] [CrossRef]
- Abarikwu, S.O.; Pant, A.B.; Farombi, E.O. 4-Hydroxynonenal induces mitochondrial-mediated apoptosis and oxidative stress in SH-SY5Y human neuronal cells. Basic Clin. Pharmacol. Toxicol. 2012, 110, 441–448. [Google Scholar] [CrossRef] [PubMed]
- Neely, M.D.; Boutte, A.; Milatovic, D.; Montine, T.J. Mechanisms of 4-hydroxynonenal-induced neuronal microtubule dysfunction. Brain Res. 2005, 1037, 90–98. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Kukal, S.; Kanojia, N.; Singh, M.; Saso, L.; Kukreti, S.; Kukreti, R. Lipid mediated brain disorders: A perspective. Prostaglandins Other Lipid Mediat. 2023, 167, 106737. [Google Scholar] [CrossRef]
- Chang, X.; Wang, Y.; Zheng, B.; Chen, Y.; Xie, J.; Song, Y.; Ding, X.; Hu, X.; Hu, X.; Yu, Q. The Role of Acrolein in Neurodegenerative Diseases and Its Protective Strategy. Foods 2022, 11, 3203. [Google Scholar] [CrossRef] [PubMed]
- McMinimy, R.; Manford, A.G.; Gee, C.L.; Chandrasekhar, S.; Mousa, G.A.; Chuang, J.; Phu, L.; Shih, K.Y.; Rose, C.M.; Kuriyan, J.; et al. Reactive oxygen species control protein degradation at the mitochondrial import gate. Mol. Cell 2024, 84, 4612–4628.e13. [Google Scholar] [CrossRef]
- Gonos, E.S.; Kapetanou, M.; Sereikaite, J.; Bartosz, G.; Naparlo, K.; Grzesik, M.; Sadowska-Bartosz, I. Origin and pathophysiology of protein carbonylation, nitration and chlorination in age-related brain diseases and aging. Aging 2018, 10, 868–901. [Google Scholar] [CrossRef]
- Chong, Z.Z.; Souayah, N. Crumbling Pathogenesis and Biomarkers for Diabetic Peripheral Neuropathy. Biomedicines 2025, 13, 413. [Google Scholar] [CrossRef]
- Moldogazieva, N.T.; Mokhosoev, I.M.; Mel’nikova, T.I.; Porozov, Y.B.; Terentiev, A.A. Oxidative Stress and Advanced Lipoxidation and Glycation End Products (ALEs and AGEs) in Aging and Age-Related Diseases. Oxid. Med. Cell. Longev. 2019, 2019, 3085756. [Google Scholar] [CrossRef]
- Poetsch, A.R. The genomics of oxidative DNA damage, repair, and resulting mutagenesis. Comput. Struct. Biotechnol. J. 2020, 18, 207–219. [Google Scholar] [CrossRef]
- Areti, A.; Yerra, V.G.; Naidu, V.; Kumar, A. Oxidative stress and nerve damage: Role in chemotherapy induced peripheral neuropathy. Redox Biol. 2014, 2, 289–295. [Google Scholar] [CrossRef] [PubMed]
- Salim, S. Oxidative Stress and the Central Nervous System. J. Pharmacol. Exp. Ther. 2017, 360, 201–205. [Google Scholar] [CrossRef]
- Cobley, J.N.; Fiorello, M.L.; Bailey, D.M. 13 reasons why the brain is susceptible to oxidative stress. Redox Biol. 2018, 15, 490–503. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Michaelis, E.K. Selective neuronal vulnerability to oxidative stress in the brain. Front. Aging Neurosci. 2010, 2, 12. [Google Scholar] [CrossRef] [PubMed]
- Yakunin, E.; Kisos, H.; Kulik, W.; Grigoletto, J.; Wanders, R.J.; Sharon, R. The regulation of catalase activity by PPAR gamma is affected by alpha-synuclein. Ann. Clin. Transl. Neurol. 2014, 1, 145–159. [Google Scholar] [CrossRef]
- Vinokurov, A.Y.; Stelmashuk, O.A.; Ukolova, P.A.; Zherebtsov, E.A.; Abramov, A.Y. Brain region specificity in reactive oxygen species production and maintenance of redox balance. Free Radic. Biol. Med. 2021, 174, 195–201. [Google Scholar] [CrossRef]
- Wilkinson, B.L.; Landreth, G.E. The microglial NADPH oxidase complex as a source of oxidative stress in Alzheimer’s disease. J. Neuroinflamm. 2006, 3, 30. [Google Scholar] [CrossRef]
- Cipriano, A.; Viviano, M.; Feoli, A.; Milite, C.; Sarno, G.; Castellano, S.; Sbardella, G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J. Med. Chem. 2023, 66, 11632–11655. [Google Scholar] [CrossRef]
- Geng, L.; Fan, L.M.; Liu, F.; Smith, C.; Li, J. Nox2 dependent redox-regulation of microglial response to amyloid-beta stimulation and microgliosis in aging. Sci. Rep. 2020, 10, 1582. [Google Scholar] [CrossRef]
- Tu, D.Z.; Velagapudi, R.; Gao, Y.; Hong, J.S.; Zhou, H.; Gao, H.M. Activation of neuronal NADPH oxidase NOX2 promotes inflammatory neurodegeneration. Free Radic. Biol. Med. 2023, 200, 47–58. [Google Scholar] [CrossRef]
- Choi, S.H.; Aid, S.; Kim, H.W.; Jackson, S.H.; Bosetti, F. Inhibition of NADPH oxidase promotes alternative and anti-inflammatory microglial activation during neuroinflammation. J. Neurochem. 2012, 120, 292–301. [Google Scholar] [CrossRef] [PubMed]
- Hu, Z.; Yu, F.; Gong, P.; Qiu, Y.; Zhou, W.; Cui, Y.; Li, J.; Chen, H. Subneurotoxic copper(II)-induced NF-kappaB-dependent microglial activation is associated with mitochondrial ROS. Toxicol. Appl. Pharmacol. 2014, 276, 95–103. [Google Scholar] [CrossRef] [PubMed]
- Peruzzotti-Jametti, L.; Willis, C.M.; Krzak, G.; Hamel, R.; Pirvan, L.; Ionescu, R.B.; Reisz, J.A.; Prag, H.A.; Garcia-Segura, M.E.; Wu, V.; et al. Mitochondrial complex I activity in microglia sustains neuroinflammation. Nature 2024, 628, 195–203. [Google Scholar] [CrossRef]
- Pehar, M.; Vargas, M.R.; Robinson, K.M.; Cassina, P.; Diaz-Amarilla, P.J.; Hagen, T.M.; Radi, R.; Barbeito, L.; Beckman, J.S. Mitochondrial superoxide production and nuclear factor erythroid 2-related factor 2 activation in p75 neurotrophin receptor-induced motor neuron apoptosis. J. Neurosci. 2007, 27, 7777–7785. [Google Scholar] [CrossRef]
- Liu, R.; Li, B.; Flanagan, S.W.; Oberley, L.W.; Gozal, D.; Qiu, M. Increased mitochondrial antioxidative activity or decreased oxygen free radical propagation prevent mutant SOD1-mediated motor neuron cell death and increase amyotrophic lateral sclerosis-like transgenic mouse survival. J. Neurochem. 2002, 80, 488–500. [Google Scholar] [CrossRef]
- Bellaver, B.; Souza, D.G.; Souza, D.O.; Quincozes-Santos, A. Hippocampal Astrocyte Cultures from Adult and Aged Rats Reproduce Changes in Glial Functionality Observed in the Aging Brain. Mol. Neurobiol. 2017, 54, 2969–2985. [Google Scholar] [CrossRef]
- Bellaver, B.; Dos Santos, J.P.; Leffa, D.T.; Bobermin, L.D.; Roppa, P.H.A.; da Silva Torres, I.L.; Goncalves, C.A.; Souza, D.O.; Quincozes-Santos, A. Systemic Inflammation as a Driver of Brain Injury: The Astrocyte as an Emerging Player. Mol. Neurobiol. 2018, 55, 2685–2695. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Hagemann, T.L.; Kalwa, H.; Michel, T.; Messing, A.; Feany, M.B. Nitric oxide mediates glial-induced neurodegeneration in Alexander disease. Nat. Commun. 2015, 6, 8966. [Google Scholar] [CrossRef]
- Simpson, D.S.A.; Oliver, P.L. ROS Generation in Microglia: Understanding Oxidative Stress and Inflammation in Neurodegenerative Disease. Antioxidants 2020, 9, 743. [Google Scholar] [CrossRef]
- Tschopp, J.; Schroder, K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production? Nat. Rev. Immunol. 2010, 10, 210–215. [Google Scholar] [CrossRef]
- Morgan, M.J.; Liu, Z.G. Crosstalk of reactive oxygen species and NF-kappaB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed]
- Hou, L.; Wang, K.; Zhang, C.; Sun, F.; Che, Y.; Zhao, X.; Zhang, D.; Li, H.; Wang, Q. Complement receptor 3 mediates NADPH oxidase activation and dopaminergic neurodegeneration through a Src-Erk-dependent pathway. Redox Biol. 2018, 14, 250–260. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.; Fan, L.; Xia, S.; Yang, Q.; Zhang, Z.; Chen, H.; Zeng, H.; Fu, X.; Peng, Y.; Xu, C.; et al. Role of complement C1q/C3-CR3 signaling in brain injury after experimental intracerebral hemorrhage and the effect of minocycline treatment. Front. Immunol. 2022, 13, 919444. [Google Scholar] [CrossRef]
- Doz-Deblauwe, E.; Carreras, F.; Arbues, A.; Remot, A.; Epardaud, M.; Malaga, W.; Mayau, V.; Prandi, J.; Astarie-Dequeker, C.; Guilhot, C.; et al. CR3 Engaged by PGL-I Triggers Syk-Calcineurin-NFATc to Rewire the Innate Immune Response in Leprosy. Front. Immunol. 2019, 10, 2913. [Google Scholar] [CrossRef]
- Pinkus, R.; Weiner, L.M.; Daniel, V. Role of oxidants and antioxidants in the induction of AP-1, NF-kappaB, and glutathione S-transferase gene expression. J. Biol. Chem. 1996, 271, 13422–13429. [Google Scholar] [CrossRef]
- Qiao, Y.; He, H.; Jonsson, P.; Sinha, I.; Zhao, C.; Dahlman-Wright, K. AP-1 Is a Key Regulator of Proinflammatory Cytokine TNFalpha-mediated Triple-negative Breast Cancer Progression. J. Biol. Chem. 2016, 291, 5068–5079. [Google Scholar] [CrossRef]
- Ranneh, Y.; Ali, F.; Akim, A.M.; Hamid, H.A.; Khazaai, H.; Fadel, A. Crosstalk between reactive oxygen species and pro-inflammatory markers in developing various chronic diseases: A review. Appl. Biol. Chem. 2017, 60, 327–338. [Google Scholar] [CrossRef]
- Bhosale, P.B.; Kim, H.H.; Abusaliya, A.; Vetrivel, P.; Ha, S.E.; Park, M.Y.; Lee, H.J.; Kim, G.S. Structural and Functional Properties of Activator Protein-1 in Cancer and Inflammation. Evid.-Based Complement. Altern. Med. 2022, 2022, 9797929. [Google Scholar] [CrossRef] [PubMed]
- Fujioka, S.; Niu, J.; Schmidt, C.; Sclabas, G.M.; Peng, B.; Uwagawa, T.; Li, Z.; Evans, D.B.; Abbruzzese, J.L.; Chiao, P.J. NF-kappaB and AP-1 connection: Mechanism of NF-kappaB-dependent regulation of AP-1 activity. Mol. Cell. Biol. 2004, 24, 7806–7819. [Google Scholar] [CrossRef]
- Chen, B.; Lu, Y.R.; Chen, Y.N.; Cheng, J.Q. The role of Nrf2 in oxidative stress-induced endothelial injuries. J. Endocrinol. 2015, 225, R83–R99. [Google Scholar] [CrossRef]
- Reichard, J.F.; Motz, G.T.; Puga, A. Heme oxygenase-1 induction by NRF2 requires inactivation of the transcriptional repressor BACH1. Nucleic Acids Res. 2007, 35, 7074–7086. [Google Scholar] [CrossRef] [PubMed]
- Vijayan, V.; Wagener, F.; Immenschuh, S. The macrophage heme-heme oxygenase-1 system and its role in inflammation. Biochem. Pharmacol. 2018, 153, 159–167. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Zhang, X.; Ding, Y.; Zhou, W.; Tao, L.; Lu, P.; Wang, Y.; Hu, R. Nuclear Factor E2-Related Factor-2 Negatively Regulates NLRP3 Inflammasome Activity by Inhibiting Reactive Oxygen Species-Induced NLRP3 Priming. Antioxid. Redox Signal. 2017, 26, 28–43. [Google Scholar] [CrossRef] [PubMed]
- Thimmulappa, R.K.; Scollick, C.; Traore, K.; Yates, M.; Trush, M.A.; Liby, K.T.; Sporn, M.B.; Yamamoto, M.; Kensler, T.W.; Biswal, S. Nrf2-dependent protection from LPS induced inflammatory response and mortality by CDDO-Imidazolide. Biochem. Biophys. Res. Commun. 2006, 351, 883–889. [Google Scholar] [CrossRef]
- Angeloni, C.; Prata, C.; Dalla Sega, F.V.; Piperno, R.; Hrelia, S. Traumatic brain injury and NADPH oxidase: A deep relationship. Oxid. Med. Cell. Longev. 2015, 2015, 370312. [Google Scholar] [CrossRef]
- Lawrence, T. The nuclear factor NF-kappaB pathway in inflammation. Cold Spring Harb. Perspect. Biol. 2009, 1, a001651. [Google Scholar] [CrossRef]
- Jena, A.B.; Samal, R.R.; Bhol, N.K.; Duttaroy, A.K. Cellular Red-Ox system in health and disease: The latest update. Biomed. Pharmacother. 2023, 162, 114606. [Google Scholar] [CrossRef]
- Solleiro-Villavicencio, H.; Rivas-Arancibia, S. Effect of Chronic Oxidative Stress on Neuroinflammatory Response Mediated by CD4+T Cells in Neurodegenerative Diseases. Front. Cell. Neurosci. 2018, 12, 114. [Google Scholar] [CrossRef]
- Ansari, M.A.; Scheff, S.W. NADPH-oxidase activation and cognition in Alzheimer disease progression. Free Radic. Biol. Med. 2011, 51, 171–178. [Google Scholar] [CrossRef]
- Han, B.H.; Zhou, M.L.; Johnson, A.W.; Singh, I.; Liao, F.; Vellimana, A.K.; Nelson, J.W.; Milner, E.; Cirrito, J.R.; Basak, J.; et al. Contribution of reactive oxygen species to cerebral amyloid angiopathy, vasomotor dysfunction, and microhemorrhage in aged Tg2576 mice. Proc. Natl. Acad. Sci. USA 2015, 112, E881–E890. [Google Scholar] [CrossRef]
- Park, L.; Anrather, J.; Zhou, P.; Frys, K.; Pitstick, R.; Younkin, S.; Carlson, G.A.; Iadecola, C. NADPH-oxidase-derived reactive oxygen species mediate the cerebrovascular dysfunction induced by the amyloid beta peptide. J. Neurosci. 2005, 25, 1769–1777. [Google Scholar] [CrossRef] [PubMed]
- Reed, T.; Perluigi, M.; Sultana, R.; Pierce, W.M.; Klein, J.B.; Turner, D.M.; Coccia, R.; Markesbery, W.R.; Butterfield, D.A. Redox proteomic identification of 4-hydroxy-2-nonenal-modified brain proteins in amnestic mild cognitive impairment: Insight into the role of lipid peroxidation in the progression and pathogenesis of Alzheimer’s disease. Neurobiol. Dis. 2008, 30, 107–120. [Google Scholar] [CrossRef]
- Misrani, A.; Tabassum, S.; Yang, L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer’s Disease. Front. Aging Neurosci. 2021, 13, 617588. [Google Scholar] [CrossRef]
- Hung, C.H.L.; Cheng, S.S.Y.; Cheung, Y.T.; Wuwongse, S.; Zhang, N.Q.; Ho, Y.S.; Lee, S.M.Y.; Chang, R.C.C. A reciprocal relationship between reactive oxygen species and mitochondrial dynamics in neurodegeneration. Redox Biol. 2018, 14, 7–19. [Google Scholar] [CrossRef] [PubMed]
- Leuner, K.; Schutt, T.; Kurz, C.; Eckert, S.H.; Schiller, C.; Occhipinti, A.; Mai, S.; Jendrach, M.; Eckert, G.P.; Kruse, S.E.; et al. Mitochondrion-derived reactive oxygen species lead to enhanced amyloid beta formation. Antioxid. Redox Signal. 2012, 16, 1421–1433. [Google Scholar] [CrossRef]
- Jurcau, M.C.; Andronie-Cioara, F.L.; Jurcau, A.; Marcu, F.; Tit, D.M.; Pascalau, N.; Nistor-Cseppento, D.C. The Link between Oxidative Stress, Mitochondrial Dysfunction and Neuroinflammation in the Pathophysiology of Alzheimer’s Disease: Therapeutic Implications and Future Perspectives. Antioxidants 2022, 11, 2167. [Google Scholar] [CrossRef]
- Manczak, M.; Anekonda, T.S.; Henson, E.; Park, B.S.; Quinn, J.; Reddy, P.H. Mitochondria are a direct site of A beta accumulation in Alzheimer’s disease neurons: Implications for free radical generation and oxidative damage in disease progression. Hum. Mol. Genet. 2006, 15, 1437–1449. [Google Scholar] [CrossRef] [PubMed]
- Maldonado, E.; Morales-Pison, S.; Urbina, F.; Solari, A. Aging Hallmarks and the Role of Oxidative Stress. Antioxidants 2023, 12, 651. [Google Scholar] [CrossRef]
- Sahoo, S.; Meijles, D.N.; Pagano, P.J. NADPH oxidases: Key modulators in aging and age-related cardiovascular diseases? Clin. Sci. 2016, 130, 317–335. [Google Scholar] [CrossRef]
- Fernandez, C.G.; Hamby, M.E.; McReynolds, M.L.; Ray, W.J. The Role of APOE4 in Disrupting the Homeostatic Functions of Astrocytes and Microglia in Aging and Alzheimer’s Disease. Front. Aging Neurosci. 2019, 11, 14. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Mattson, M.P. Apolipoprotein E and oxidative stress in brain with relevance to Alzheimer’s disease. Neurobiol. Dis. 2020, 138, 104795. [Google Scholar] [CrossRef] [PubMed]
- Manna, P.; Jain, S.K. Obesity, Oxidative Stress, Adipose Tissue Dysfunction, and the Associated Health Risks: Causes and Therapeutic Strategies. Metab. Syndr. Relat. Disord. 2015, 13, 423–444. [Google Scholar] [CrossRef] [PubMed]
- Khanna, D.; Khanna, S.; Khanna, P.; Kahar, P.; Patel, B.M. Obesity: A Chronic Low-Grade Inflammation and Its Markers. Cureus 2022, 14, e22711. [Google Scholar] [CrossRef] [PubMed]
- de Mello, A.H.; Costa, A.B.; Engel, J.D.G.; Rezin, G.T. Mitochondrial dysfunction in obesity. Life Sci. 2018, 192, 26–32. [Google Scholar] [CrossRef] [PubMed]
- Carnevale, D.; Mascio, G.; D’Andrea, I.; Fardella, V.; Bell, R.D.; Branchi, I.; Pallante, F.; Zlokovic, B.; Yan, S.S.; Lembo, G. Hypertension induces brain beta-amyloid accumulation, cognitive impairment, and memory deterioration through activation of receptor for advanced glycation end products in brain vasculature. Hypertension 2012, 60, 188–197. [Google Scholar] [CrossRef]
- Minatel, I.O.; Francisqueti, F.V.; Correa, C.R.; Lima, G.P. Antioxidant Activity of gamma-Oryzanol: A Complex Network of Interactions. Int. J. Mol. Sci. 2016, 17, 1107. [Google Scholar] [CrossRef]
- Perona, R.; Montaner, S.; Saniger, L.; Sanchez-Perez, I.; Bravo, R.; Lacal, J.C. Activation of the nuclear factor-kappaB by Rho, CDC42, and Rac-1 proteins. Genes Dev. 1997, 11, 463–475. [Google Scholar] [CrossRef]
- Bierhaus, A.; Schiekofer, S.; Schwaninger, M.; Andrassy, M.; Humpert, P.M.; Chen, J.; Hong, M.; Luther, T.; Henle, T.; Kloting, I.; et al. Diabetes-associated sustained activation of the transcription factor nuclear factor-kappaB. Diabetes 2001, 50, 2792–2808. [Google Scholar] [CrossRef]
- Amponsah-Offeh, M.; Diaba-Nuhoho, P.; Speier, S.; Morawietz, H. Oxidative Stress, Antioxidants and Hypertension. Antioxidants 2023, 12, 281. [Google Scholar] [CrossRef]
- Chong, Z.Z.; Menkes, D.L.; Souayah, N. Targeting neuroinflammation in distal symmetrical polyneuropathy in diabetes. Drug Discov. Today 2024, 29, 104087. [Google Scholar] [CrossRef]
- Mudyanselage, A.W.; Wijamunige, B.C.; Kocon, A.; Turner, R.; McLean, D.; Morentin, B.; Callado, L.F.; Carter, W.G. Alcohol Triggers the Accumulation of Oxidatively Damaged Proteins in Neuronal Cells and Tissues. Antioxidants 2024, 13, 580. [Google Scholar] [CrossRef] [PubMed]
- Tsermpini, E.E.; Plemenitas Iljes, A.; Dolzan, V. Alcohol-Induced Oxidative Stress and the Role of Antioxidants in Alcohol Use Disorder: A Systematic Review. Antioxidants 2022, 11, 1374. [Google Scholar] [CrossRef] [PubMed]
- Durazzo, T.C.; Mattsson, N.; Weiner, M.W.; Alzheimer’s Disease Neuroimaging, I. Smoking and increased Alzheimer’s disease risk: A review of potential mechanisms. Alzheimers Dement. 2014, 10, S122–S145. [Google Scholar] [CrossRef]
- Cataldo, J.K.; Prochaska, J.J.; Glantz, S.A. Cigarette smoking is a risk factor for Alzheimer’s Disease: An analysis controlling for tobacco industry affiliation. J. Alzheimers Dis. 2010, 19, 465–480. [Google Scholar] [CrossRef] [PubMed]
- Valavanidis, A.; Vlachogianni, T.; Fiotakis, K. Tobacco smoke: Involvement of reactive oxygen species and stable free radicals in mechanisms of oxidative damage, carcinogenesis and synergistic effects with other respirable particles. Int. J. Environ. Res. Public Health 2009, 6, 445–462. [Google Scholar] [CrossRef]
- Ahmadkhaniha, R.; Yousefian, F.; Rastkari, N. Impact of smoking on oxidant/antioxidant status and oxidative stress index levels in serum of the university students. J. Environ. Health Sci. Eng. 2021, 19, 1043–1046. [Google Scholar] [CrossRef]
- Caliri, A.W.; Tommasi, S.; Besaratinia, A. Relationships among smoking, oxidative stress, inflammation, macromolecular damage, and cancer. Mutat. Res. Rev. Mutat. Res. 2021, 787, 108365. [Google Scholar] [CrossRef]
- Cheignon, C.; Tomas, M.; Bonnefont-Rousselot, D.; Faller, P.; Hureau, C.; Collin, F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol. 2018, 14, 450–464. [Google Scholar] [CrossRef]
- Mouton-Liger, F.; Paquet, C.; Dumurgier, J.; Bouras, C.; Pradier, L.; Gray, F.; Hugon, J. Oxidative stress increases BACE1 protein levels through activation of the PKR-eIF2α pathway. Biochim. Biophys. Acta-Mol. Basis Dis. 2012, 1822, 885–896. [Google Scholar] [CrossRef]
- Hampel, H.; Vassar, R.; De Strooper, B.; Hardy, J.; Willem, M.; Singh, N.; Zhou, J.; Yan, R.; Vanmechelen, E.; De Vos, A.; et al. The beta-Secretase BACE1 in Alzheimer’s Disease. Biol. Psychiatry 2021, 89, 745–756. [Google Scholar] [CrossRef]
- Borghi, R.; Patriarca, S.; Traverso, N.; Piccini, A.; Storace, D.; Garuti, A.; Gabriella, C.; Patrizio, O.; Massimo, T. The increased activity of BACE1 correlates with oxidative stress in Alzheimer’s disease. Neurobiol. Aging 2007, 28, 1009–1014. [Google Scholar] [CrossRef] [PubMed]
- Tamagno, E.; Parola, M.; Bardini, P.; Piccini, A.; Borghi, R.; Guglielmotto, M.; Santoro, G.; Davit, A.; Danni, O.; Smith, M.A.; et al. Beta-site APP cleaving enzyme up-regulation induced by 4-hydroxynonenal is mediated by stress-activated protein kinases pathways. J. Neurochem. 2005, 92, 628–636. [Google Scholar] [CrossRef] [PubMed]
- Thonda, S.; Puttapaka, S.N.; Kona, S.V.; Kalivendi, S.V. Extracellular-Signal-Regulated Kinase Inhibition Switches APP Processing from beta- to alpha-Secretase under Oxidative Stress: Modulation of ADAM10 by SIRT1/NF-kappaB Signaling. ACS Chem. Neurosci. 2021, 12, 4175–4186. [Google Scholar] [CrossRef]
- Cheignon, C.; Hureau, C.; Collin, F. Real-time evolution of Aβ metal-catalyzed oxidation reveals Asp1 as the main target and a dependence on metal binding site. Inorganica Chim. Acta 2018, 472, 111–118. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Galvan, V.; Lange, M.B.; Tang, H.; Sowell, R.A.; Spilman, P.; Fombonne, J.; Gorostiza, O.; Zhang, J.; Sultana, R.; et al. In vivo oxidative stress in brain of Alzheimer disease transgenic mice: Requirement for methionine 35 in amyloid beta-peptide of APP. Free Radic. Biol. Med. 2010, 48, 136–144. [Google Scholar] [CrossRef]
- Moskovitz, J.; Du, F.; Bowman, C.F.; Yan, S.S. Methionine sulfoxide reductase A affects beta-amyloid solubility and mitochondrial function in a mouse model of Alzheimer’s disease. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E388–E393. [Google Scholar] [CrossRef]
- Cioffi, F.; Adam, R.H.I.; Broersen, K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer’s Disease. J. Alzheimers Dis. 2019, 72, 981–1017. [Google Scholar] [CrossRef]
- Friedemann, M.; Helk, E.; Tiiman, A.; Zovo, K.; Palumaa, P.; Tougu, V. Effect of methionine-35 oxidation on the aggregation of amyloid-beta peptide. Biochem. Biophys. Rep. 2015, 3, 94–99. [Google Scholar]
- Murray, I.V.J.; Liu, L.; Komatsu, H.; Uryu, K.; Xiao, G.; Lawson, J.A.; Axelsen, P.H. Membrane-mediated amyloidogenesis and the promotion of oxidative lipid damage by amyloid beta proteins. J. Biol. Chem. 2007, 282, 9335–9345. [Google Scholar] [CrossRef]
- Barnham, K.J.; Haeffner, F.; Ciccotosto, G.D.; Curtain, C.C.; Tew, D.; Mavros, C.; Beyreuther, K.; Carrington, D.; Masters, C.L.; Cherny, R.A.; et al. Tyrosine gated electron transfer is key to the toxic mechanism of Alzheimer’s disease beta-amyloid. FASEB J. 2004, 18, 1427–1429. [Google Scholar] [CrossRef]
- Sagare, A.; Deane, R.; Bell, R.D.; Johnson, B.; Hamm, K.; Pendu, R.; Marky, A.; Lenting, P.J.; Wu, Z.; Zarcone, T.; et al. Clearance of amyloid-beta by circulating lipoprotein receptors. Nat. Med. 2007, 13, 1029–1031. [Google Scholar] [CrossRef] [PubMed]
- Owen, J.B.; Sultana, R.; Aluise, C.D.; Erickson, M.A.; Price, T.O.; Bu, G.J.; Banks, W.A.; Butterfield, D.A. Oxidative modification to LDL receptor-related protein 1 in hippocampus from subjects with Alzheimer disease Implications for Aβ accumulation in AD brain. Free Radic. Biol. Med. 2010, 49, 1798–1803. [Google Scholar] [CrossRef]
- Ando, K.; Maruko-Otake, A.; Ohtake, Y.; Hayashishita, M.; Sekiya, M.; Iijima, K.M. Stabilization of Microtubule-Unbound Tau via Tau Phosphorylation at Ser262/356 by Par-1/MARK Contributes to Augmentation of AD-Related Phosphorylation and Abeta42-Induced Tau Toxicity. PLoS Genet. 2016, 12, e1005917. [Google Scholar] [CrossRef]
- Liu, Y.; Chen, Y.; Fukui, K. Oxidative stress induces tau hyperphosphorylation via MARK activation in neuroblastoma N1E-115 cells. J. Clin. Biochem. Nutr. 2023, 73, 24–33. [Google Scholar] [CrossRef] [PubMed]
- Mondragon-Rodriguez, S.; Perry, G.; Luna-Munoz, J.; Acevedo-Aquino, M.C.; Williams, S. Phosphorylation of tau protein at sites Ser(396-404) is one of the earliest events in Alzheimer’s disease and Down syndrome. Neuropathol. Appl. Neurobiol. 2014, 40, 121–135. [Google Scholar] [CrossRef]
- Feng, Y.; Xia, Y.; Yu, G.; Shu, X.; Ge, H.; Zeng, K.; Wang, J.; Wang, X. Cleavage of GSK-3beta by calpain counteracts the inhibitory effect of Ser9 phosphorylation on GSK-3beta activity induced by H2O2. J. Neurochem. 2013, 126, 234–242. [Google Scholar] [CrossRef] [PubMed]
- Maiese, K.; Chong, Z.Z.; Shang, Y.C.; Hou, J. Rogue proliferation versus restorative protection: Where do we draw the line for Wnt and forkhead signaling? Expert Opin. Ther. Targets 2008, 12, 905–916. [Google Scholar] [CrossRef]
- Law, S.M.; Zheng, J.J. Premise and peril of Wnt signaling activation through GSK-3β inhibition. Iscience 2022, 25, 104159. [Google Scholar] [CrossRef]
- Culbreth, M.; Aschner, M. GSK-3beta, a double-edged sword in Nrf2 regulation: Implications for neurological dysfunction and disease. F1000Research 2018, 7, 1043. [Google Scholar] [CrossRef]
- Expression of Concern: Lithium Inhibits Aβ-Induced Stress in Endoplasmic Reticulum of Rabbit Hippocampus but Does Not Prevent Oxidative Damage and Tau Phosphorylation. J. Neurosci. Res. 2025, 103, e70031. [CrossRef]
- Rogers, C.; Erkes, D.A.; Nardone, A.; Aplin, A.E.; Fernandes-Alnemri, T.; Alnemri, E.S. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 2019, 10, 1689. [Google Scholar] [CrossRef]
- Giraldo, E.; Lloret, A.; Fuchsberger, T.; Vina, J. Abeta and tau toxicities in Alzheimer’s are linked via oxidative stress-induced p38 activation: Protective role of vitamin E. Redox Biol. 2014, 2, 873–877. [Google Scholar] [CrossRef]
- Saito, T.; Chiku, T.; Oka, M.; Wada-Kakuda, S.; Nobuhara, M.; Oba, T.; Shinno, K.; Abe, S.; Asada, A.; Sumioka, A.; et al. Disulfide bond formation in microtubule-associated tau protein promotes tau accumulation and toxicity in vivo. Hum. Mol. Genet. 2021, 30, 1955–1967. [Google Scholar] [CrossRef] [PubMed]
- Song, H.; Kim, W.; Choi, J.H.; Kim, S.H.; Lee, D.; Park, C.H.; Kim, S.; Kim, D.Y.; Kim, K.T. Stress-induced nuclear translocation of CDK5 suppresses neuronal death by downregulating ERK activation via VRK3 phosphorylation. Sci. Rep. 2016, 6, 28634. [Google Scholar] [CrossRef]
- Guo, D.; Xie, W.; Xiong, P.; Li, H.; Wang, S.; Chen, G.; Gao, Y.; Zhou, J.; Zhang, Y.; Bu, G.; et al. Cyclin-dependent kinase 5-mediated phosphorylation of chloride intracellular channel 4 promotes oxidative stress-induced neuronal death. Cell Death Dis. 2018, 9, 951. [Google Scholar] [CrossRef] [PubMed]
- Kimura, T.; Ishiguro, K.; Hisanaga, S. Physiological and pathological phosphorylation of tau by Cdk5. Front. Mol. Neurosci. 2014, 7, 65. [Google Scholar] [CrossRef] [PubMed]
- Maitra, S.; Vincent, B. Cdk5-p25 as a key element linking amyloid and tau pathologies in Alzheimer’s disease: Mechanisms and possible therapeutic interventions. Life Sci. 2022, 308, 120986. [Google Scholar] [CrossRef]
- Ji, R.; Jia, F.Y.; Chen, X.; Wang, Z.H.; Jin, W.Y.; Yang, J. Salidroside alleviates oxidative stress and apoptosis via AMPK/Nrf2 pathway in DHT-induced human granulosa cell line KGN. Arch. Biochem. Biophys. 2022, 715, 109094. [Google Scholar] [CrossRef]
- Wang, X.; Zimmermann, H.R.; Ma, T. Therapeutic Potential of AMP-Activated Protein Kinase in Alzheimer’s Disease. J. Alzheimers Dis. 2019, 68, 33–38. [Google Scholar] [CrossRef]
- Roth, A.; Sander, A.; Oswald, M.S.; Gartner, F.; Knippschild, U.; Bischof, J. Comprehensive Characterization of CK1delta-Mediated Tau Phosphorylation in Alzheimer’s Disease. Front. Mol. Biosci. 2022, 9, 872171. [Google Scholar] [CrossRef]
- Arif, M.; Wei, J.S.; Zhang, Q.; Liu, F.; Basurto-Islas, G.; Grundke-Iqbal, I.; Iqbal, K. Cytoplasmic Retention of Protein Phosphatase 2A Inhibitor 2 (I2PP2A) Induces Alzheimer-like Abnormal Hyperphosphorylation of Tau. J. Biol. Chem. 2014, 289, 27677–27691. [Google Scholar] [CrossRef]
- Wang, L.; Zhou, Y.; Chen, D.; Lee, T.H. Peptidyl-Prolyl Cis/Trans Isomerase Pin1 and Alzheimer’s Disease. Front. Cell Dev. Biol. 2020, 8, 355. [Google Scholar] [CrossRef] [PubMed]
- Subramaniam, R.; Roediger, F.; Jordan, B.; Mattson, M.P.; Keller, J.N.; Waeg, G.; Butterfield, D.A. The lipid peroxidation product, 4-hydroxy-2-trans-nonenal, alters the conformation of cortical synaptosomal membrane proteins. J. Neurochem. 1997, 69, 1161–1169. [Google Scholar] [CrossRef] [PubMed]
- Castegna, A.; Lauderback, C.M.; Mohmmad-Abdul, H.; Butterfield, D.A. Modulation of phospholipid asymmetry in synaptosomal membranes by the lipid peroxidation products, 4-hydroxynonenal and acrolein: Implications for Alzheimer’s disease. Brain Res. 2004, 1004, 193–197. [Google Scholar] [CrossRef]
- Foret, M.K.; Orciani, C.; Welikovitch, L.A.; Huang, C.; Cuello, A.C.; Do Carmo, S. Early oxidative stress and DNA damage in Abeta-burdened hippocampal neurons in an Alzheimer’s-like transgenic rat model. Commun. Biol. 2024, 7, 861. [Google Scholar] [CrossRef]
- Perluigi, M.; Sultana, R.; Cenini, G.; Di Domenico, F.; Memo, M.; Pierce, W.M.; Coccia, R.; Butterfield, D.A. Redox proteomics identification of 4-hydroxynonenal-modified brain proteins in Alzheimer’s disease: Role of lipid peroxidation in Alzheimer’s disease pathogenesis. Proteom. Clin. Appl. 2009, 3, 682–693. [Google Scholar] [CrossRef]
- Tramutola, A.; Lanzillotta, C.; Perluigi, M.; Butterfield, D.A. Oxidative stress, protein modification and Alzheimer disease. Brain Res. Bull. 2017, 133, 88–96. [Google Scholar] [CrossRef]
- Wang, W.Y.; Tan, M.S.; Yu, J.T.; Tan, L. Role of pro-inflammatory cytokines released from microglia in Alzheimer’s disease. Ann. Transl. Med. 2015, 3, 136. [Google Scholar]
- Welikovitch, L.A.; Do Carmo, S.; Magloczky, Z.; Malcolm, J.C.; Loke, J.; Klein, W.L.; Freund, T.; Cuello, A.C. Early intraneuronal amyloid triggers neuron-derived inflammatory signaling in APP transgenic rats and human brain. Proc. Natl. Acad. Sci. USA 2020, 117, 6844–6854. [Google Scholar] [CrossRef]
- Mary, A.; Eysert, F.; Checler, F.; Chami, M. Mitophagy in Alzheimer’s disease: Molecular defects and therapeutic approaches. Mol. Psychiatry 2023, 28, 202–216. [Google Scholar] [CrossRef]
- Bhatia, S.; Rawal, R.; Sharma, P.; Singh, T.; Singh, M.; Singh, V. Mitochondrial Dysfunction in Alzheimer’s Disease: Opportunities for Drug Development. Curr. Neuropharmacol. 2022, 20, 675–692. [Google Scholar] [CrossRef] [PubMed]
- Sultana, R.; Poon, H.F.; Cai, J.; Pierce, W.M.; Merchant, M.; Klein, J.B.; Markesbery, W.R.; Butterfield, D.A. Identification of nitrated proteins in Alzheimer’s disease brain using a redox proteomics approach. Neurobiol. Dis. 2006, 22, 76–87. [Google Scholar] [CrossRef] [PubMed]
- Hartl, D.; Schuldt, V.; Forler, S.; Zabel, C.; Klose, J.; Rohe, M. Presymptomatic Alterations in Energy Metabolism and Oxidative Stress in the APP23 Mouse Model of Alzheimer Disease. J. Proteome Res. 2012, 11, 3295–3304. [Google Scholar] [CrossRef]
- Terni, B.; Boada, J.; Portero-Otin, M.; Pamplona, R.; Ferrer, I. Mitochondrial ATP-synthase in the entorhinal cortex is a target of oxidative stress at stages I/II of Alzheimer’s disease pathology. Brain Pathol. 2010, 20, 222–233. [Google Scholar] [CrossRef]
- Itakura, M.; Kubo, T.; Kaneshige, A.; Nakajima, H. Glyceraldehyde-3-phosphate dehydrogenase regulates activation of c-Jun N-terminal kinase under oxidative stress. Biochem. Biophys. Res. Commun. 2023, 657, 1–7. [Google Scholar] [CrossRef]
- Gerszon, J.; Rodacka, A. Oxidatively modified glyceraldehyde-3-phosphate dehydrogenase in neurodegenerative processes and the role of low molecular weight compounds in counteracting its aggregation and nuclear translocation. Ageing Res. Rev. 2018, 48, 21–31. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Hardas, S.S.; Lange, M.L. Oxidatively modified glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and Alzheimer’s disease: Many pathways to neurodegeneration. J. Alzheimers Dis. 2010, 20, 369–393. [Google Scholar] [CrossRef]
- Butterfield, D.A.; Gnjec, A.; Poon, H.F.; Castegna, A.; Pierce, W.M.; Klein, J.B.; Martins, R.N. Redox proteomics identification of oxidatively modified brain proteins in inherited Alzheimer’s disease: An initial assessment. J. Alzheimers Dis. 2006, 10, 391–397. [Google Scholar] [CrossRef]
- Perluigi, M.; Di Domenico, F.; Butterfield, D.A. Oxidative damage in neurodegeneration: Roles in the pathogenesis and progression of Alzheimer disease. Physiol. Rev. 2024, 104, 103–197. [Google Scholar] [CrossRef]
- Sobanski, T.; Suraweera, A.; Burgess, J.T.; Richard, I.; Cheong, C.M.; Dave, K.; Rose, M.; Adams, M.N.; O’Byrne, K.J.; Richard, D.J.; et al. The fructose-bisphosphate, Aldolase A (ALDOA), facilitates DNA-PKcs and ATM kinase activity to regulate DNA double-strand break repair. Sci. Rep. 2023, 13, 15171. [Google Scholar] [CrossRef]
- Rummel, N.G.; Butterfield, D.A. Altered Metabolism in Alzheimer Disease Brain: Role of Oxidative Stress. Antioxid. Redox Signal. 2022, 36, 1289–1305. [Google Scholar] [CrossRef] [PubMed]
- Aksenov, M.; Aksenova, M.; Butterfield, D.A.; Markesbery, W.R. Oxidative modification of creatine kinase BB in Alzheimer’s disease brain. J. Neurochem. 2000, 74, 2520–2527. [Google Scholar] [CrossRef] [PubMed]
- Sultana, R.; Butterfield, D.A. Protein Oxidation in Aging and Alzheimer’s Disease Brain. Antioxidants 2024, 13, 574. [Google Scholar] [CrossRef]
- Han, J.; Hyun, J.; Park, J.; Jung, S.M.; Oh, Y.; Kim, Y.; Ryu, S.H.; Kim, S.H.; Jeong, E.I.; Jo, D.G.; et al. Aberrant role of pyruvate kinase M2 in the regulation of gamma-secretase and memory deficits in Alzheimer’s disease. Cell Rep. 2021, 37, 110102. [Google Scholar] [CrossRef]
- Anastasiou, D.; Poulogiannis, G.; Asara, J.M.; Boxer, M.B.; Jiang, J.K.; Shen, M.; Bellinger, G.; Sasaki, A.T.; Locasale, J.W.; Auld, D.S.; et al. Inhibition of pyruvate kinase M2 by reactive oxygen species contributes to cellular antioxidant responses. Science 2011, 334, 1278–1283. [Google Scholar] [CrossRef]
- Long, D.M.; Frame, A.K.; Reardon, P.N.; Cumming, R.C.; Hendrix, D.A.; Kretzschmar, D.; Giebultowicz, J.M. Lactate dehydrogenase expression modulates longevity and neurodegeneration in Drosophila melanogaster. Aging 2020, 12, 10041–10058. [Google Scholar] [CrossRef] [PubMed]
- Jovanovic, P.; Zoric, L.; Stefanovic, I.; Dzunic, B.; Djordjevic-Jocic, J.; Radenkovic, M.; Jovanovic, M. Lactate dehydrogenase and oxidative stress activity in primary open-angle glaucoma aqueous humour. Bosn. J. Basic Med. Sci. 2010, 10, 83–88. [Google Scholar] [CrossRef] [PubMed]
- Shi, Q.; Gibson, G.E. Up-regulation of the mitochondrial malate dehydrogenase by oxidative stress is mediated by miR-743a. J. Neurochem. 2011, 118, 440–448. [Google Scholar] [CrossRef]
- Tajes, M.; Guivernau, B.; Ramos-Fernandez, E.; Bosch-Morato, M.; Palomer, E.; Guix, F.X.; Munoz, F.J. The pathophysiology of triose phosphate isomerase dysfunction in Alzheimer’s disease. Histol. Histopathol. 2013, 28, 43–51. [Google Scholar]
- Butterfield, D.A. Ubiquitin carboxyl-terminal hydrolase L-1 in brain: Focus on its oxidative/nitrosative modification and role in brains of subjects with Alzheimer disease and mild cognitive impairment. Free Radic. Biol. Med. 2021, 177, 278–286. [Google Scholar] [CrossRef]
- Reichelt, J.; Sachs, W.; Frombling, S.; Fehlert, J.; Studencka-Turski, M.; Betz, A.; Loreth, D.; Blume, L.; Witt, S.; Pohl, S.; et al. Non-functional ubiquitin C-terminal hydrolase L1 drives podocyte injury through impairing proteasomes in autoimmune glomerulonephritis. Nat. Commun. 2023, 14, 2114. [Google Scholar] [CrossRef] [PubMed]
- Akinyemi, A.O.; Simpson, K.E.; Oyelere, S.F.; Nur, M.; Ngule, C.M.; Owoyemi, B.C.D.; Ayarick, V.A.; Oyelami, F.F.; Obaleye, O.; Esoe, D.P.; et al. Unveiling the dark side of glucose-regulated protein 78 (GRP78) in cancers and other human pathology: A systematic review. Mol. Med. 2023, 29, 112. [Google Scholar] [CrossRef] [PubMed]
- Sultana, R.; Perluigi, M.; Butterfield, D.A. Oxidatively modified proteins in Alzheimer’s disease (AD), mild cognitive impairment and animal models of AD: Role of Abeta in pathogenesis. Acta Neuropathol. 2009, 118, 131–150. [Google Scholar] [CrossRef] [PubMed]
- Omar, R.A.; Chyan, Y.J.; Andorn, A.C.; Poeggeler, B.; Robakis, N.K.; Pappolla, M.A. Increased Expression but Reduced Activity of Antioxidant Enzymes in Alzheimer’s Disease. J. Alzheimers Dis. 1999, 1, 139–145. [Google Scholar] [CrossRef]
- Murakami, K.; Murata, N.; Noda, Y.; Tahara, S.; Kaneko, T.; Kinoshita, N.; Hatsuta, H.; Murayama, S.; Barnham, K.J.; Irie, K.; et al. SOD1 (Copper/Zinc Superoxide Dismutase) Deficiency Drives Amyloid β Protein Oligomerization and Memory Loss in Mouse Model of Alzheimer Disease. J. Biol. Chem. 2011, 286, 44557–44568. [Google Scholar] [CrossRef]
- Crack, P.J.; Cimdins, K.; Ali, U.; Hertzog, P.J.; Iannello, R.C. Lack of glutathione peroxidase-1 exacerbates Abeta-mediated neurotoxicity in cortical neurons. J. Neural Transm. 2006, 113, 645–657. [Google Scholar] [CrossRef]
- Shen, Y.; Zhang, G.; Wei, C.; Zhao, P.; Wang, Y.; Li, M.; Sun, L. Potential role and therapeutic implications of glutathione peroxidase 4 in the treatment of Alzheimer’s disease. Neural Regen. Res. 2025, 20, 613–631. [Google Scholar] [CrossRef]
- Hettiarachchi, N.T.; Dallas, M.L.; Al-Owais, M.M.; Griffiths, H.H.; Hooper, N.M.; Scragg, J.L.; Boyle, J.P.; Peers, C. Heme oxygenase-1 protects against Alzheimer’s amyloid-β-induced toxicity carbon monoxide production. Cell Death Dis. 2014, 5, e1569. [Google Scholar] [CrossRef]
- Ou, J.R.; Tan, M.S.; Xie, A.M.; Yu, J.T.; Tan, L. Heat shock protein 90 in Alzheimer’s disease. Biomed. Res. Int. 2014, 2014, 796869. [Google Scholar] [CrossRef]
- Rose, C.F.; Verkhratsky, A.; Parpura, V. Astrocyte glutamine synthetase: Pivotal in health and disease. Biochem. Soc. Trans. 2013, 41, 1518–1524. [Google Scholar] [CrossRef]
- Gunnersen, D.; Haley, B. Detection of Glutamine-Synthetase in the Cerebrospinal-Fluid of Alzheimer Diseased Patients—A Potential Diagnostic Biochemical Marker. Proc. Natl. Acad. Sci. USA 1992, 89, 11949–11953. [Google Scholar] [CrossRef] [PubMed]
- Knight, E.; Geetha, T.; Broderick, T.L.; Babu, J.R. The Role of Dietary Antioxidants and Their Potential Mechanisms in Alzheimer’s Disease Treatment. Metabolites 2023, 13, 438. [Google Scholar] [CrossRef] [PubMed]
- Figueira, I.; Garcia, G.; Pimpao, R.C.; Terrasso, A.P.; Costa, I.; Almeida, A.F.; Tavares, L.; Pais, T.F.; Pinto, P.; Ventura, M.R.; et al. Correction to: Polyphenols journey through blood-brain barrier towards neuronal protection. Sci. Rep. 2021, 11, 17112. [Google Scholar] [CrossRef]
- Delaidelli, A.; Richner, M.; Jiang, L.; van der Laan, A.; Bergholdt Jul Christiansen, I.; Ferreira, N.; Nyengaard, J.R.; Vaegter, C.B.; Jensen, P.H.; Mackenzie, I.R.; et al. alpha-Synuclein pathology in Parkinson disease activates homeostatic NRF2 anti-oxidant response. Acta Neuropathol. Commun. 2021, 9, 105. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Rodriguez, P.; Zampese, E.; Stout, K.A.; Guzman, J.N.; Ilijic, E.; Yang, B.; Tkatch, T.; Stavarache, M.A.; Wokosin, D.L.; Gao, L.; et al. Disruption of mitochondrial complex I induces progressive parkinsonism. Nature 2021, 599, 650–656. [Google Scholar] [CrossRef]
- Ren, H.; Fu, K.; Wang, D.; Mu, C.; Wang, G. Oxidized DJ-1 interacts with the mitochondrial protein BCL-XL. J. Biol. Chem. 2011, 286, 35308–35317. [Google Scholar] [CrossRef]
- Skou, L.D.; Johansen, S.K.; Okarmus, J.; Meyer, M. Pathogenesis of DJ-1/PARK7-Mediated Parkinson’s Disease. Cells 2024, 13, 296. [Google Scholar] [CrossRef]
- Oh, S.E.; Mouradian, M.M. Cytoprotective mechanisms of DJ-1 against oxidative stress through modulating ERK1/2 and ASK1 signal transduction. Redox Biol. 2018, 14, 211–217. [Google Scholar] [CrossRef]
- Waak, J.; Weber, S.S.; Gorner, K.; Schall, C.; Ichijo, H.; Stehle, T.; Kahle, P.J. Oxidizable residues mediating protein stability and cytoprotective interaction of DJ-1 with apoptosis signal-regulating kinase 1. J. Biol. Chem. 2009, 284, 14245–14257. [Google Scholar] [CrossRef]
- Rui, Q.; Ni, H.; Li, D.; Gao, R.; Chen, G. The Role of LRRK2 in Neurodegeneration of Parkinson Disease. Curr. Neuropharmacol. 2018, 16, 1348–1357. [Google Scholar] [CrossRef]
- Wang, X.; Yan, M.H.; Fujioka, H.; Liu, J.; Wilson-Delfosse, A.; Chen, S.G.; Perry, G.; Casadesus, G.; Zhu, X. LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum. Mol. Genet. 2012, 21, 1931–1944. [Google Scholar] [CrossRef] [PubMed]
- Iaccarino, C.; Crosio, C.; Vitale, C.; Sanna, G.; Carri, M.T.; Barone, P. Apoptotic mechanisms in mutant LRRK2-mediated cell death. Hum. Mol. Genet. 2007, 16, 1319–1326. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, H.N.; Byers, B.; Cord, B.; Shcheglovitov, A.; Byrne, J.; Gujar, P.; Kee, K.; Schule, B.; Dolmetsch, R.E.; Langston, W.; et al. LRRK2 mutant iPSC-derived DA neurons demonstrate increased susceptibility to oxidative stress. Cell Stem Cell 2011, 8, 267–280. [Google Scholar] [CrossRef]
- Oun, A.; Sabogal-Guaqueta, A.M.; Galuh, S.; Alexander, A.; Kortholt, A.; Dolga, A.M. The multifaceted role of LRRK2 in Parkinson’s disease: From human iPSC to organoids. Neurobiol. Dis. 2022, 173, 105837. [Google Scholar] [CrossRef]
- Pena, N.; Richbourg, T.; Gonzalez-Hunt, C.P.; Qi, R.; Wren, P.; Barlow, C.; Shanks, N.F.; Carlisle, H.J.; Sanders, L.H. G2019S selective LRRK2 kinase inhibitor abrogates mitochondrial DNA damage. npj Park. Dis. 2024, 10, 49. [Google Scholar] [CrossRef]
- Al Rawi, S.; Simpson, L.; Agnarsdottir, G.; McDonald, N.Q.; Chernuha, V.; Elpeleg, O.; Zeviani, M.; Barker, R.A.; Spiegel, R.; Laman, H. Study of an FBXO7 patient mutation reveals Fbxo7 and PI31 co-regulate proteasomes and mitochondria. FEBS J. 2024, 291, 2565–2589. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.D.; Xie, S.P.; Sathiyamoorthy, S.; Saw, W.T.; Sing, T.Y.; Ng, S.H.; Chua, H.P.; Tang, A.M.; Shaffra, F.; Li, Z.; et al. F-box protein 7 mutations promote protein aggregation in mitochondria and inhibit mitophagy. Hum. Mol. Genet. 2015, 24, 6314–6330. [Google Scholar] [CrossRef]
- Vrijsen, S.; Besora-Casals, L.; van Veen, S.; Zielich, J.; Van den Haute, C.; Hamouda, N.N.; Fischer, C.; Ghesquiere, B.; Tournev, I.; Agostinis, P.; et al. ATP13A2-mediated endo-lysosomal polyamine export counters mitochondrial oxidative stress. Proc. Natl. Acad. Sci. USA 2020, 117, 31198–31207. [Google Scholar] [CrossRef]
- Croucher, K.M.; Fleming, S.M. ATP13A2 (PARK9) and basal ganglia function. Front. Neurol. 2023, 14, 1252400. [Google Scholar] [CrossRef]
- Zhang, F.; Wu, Z.; Long, F.; Tan, J.; Gong, N.; Li, X.; Lin, C. The Roles of ATP13A2 Gene Mutations Leading to Abnormal Aggregation of alpha-Synuclein in Parkinson’s Disease. Front. Cell. Neurosci. 2022, 16, 927682. [Google Scholar]
- Gusdon, A.M.; Zhu, J.; Van Houten, B.; Chu, C.T. ATP13A2 regulates mitochondrial bioenergetics through macroautophagy. Neurobiol. Dis. 2012, 45, 962–972. [Google Scholar] [CrossRef] [PubMed]
- Reeve, A.K.; Ludtmann, M.H.R.; Angelova, P.R.; Simcox, E.M.; Horrocks, M.H.; Klenerman, D.; Gandhi, S.; Turnbull, D.M.; Abramov, A.Y. Aggregated α-synuclein and complex I deficiency: Exploration of their relationship in differentiated neurons. Cell Death Dis. 2015, 6, e1820. [Google Scholar] [CrossRef]
- Kim, C.; Ho, D.H.; Suk, J.E.; You, S.; Michael, S.; Kang, J.; Lee, S.J.; Masliah, E.; Hwang, D.; Lee, H.J.; et al. Neuron-released oligomeric α-synuclein is an endogenous agonist of TLR2 for paracrine activation of microglia. Nat. Commun. 2013, 4, 1562. [Google Scholar] [CrossRef] [PubMed]
- Chedid, J.; Labrador-Garrido, A.; Zhong, S.; Gao, J.; Zhao, Y.; Perera, G.; Kim, W.S.; Halliday, G.M.; Dzamko, N. A small molecule toll-like receptor antagonist rescues alpha-synuclein fibril pathology. J. Biol. Chem. 2022, 298, 102260. [Google Scholar] [CrossRef]
- Zawada, W.M.; Banninger, G.P.; Thornton, J.; Marriott, B.; Cantu, D.; Rachubinski, A.L.; Das, M.; Griffin, W.S.; Jones, S.M. Generation of reactive oxygen species in 1-methyl-4-phenylpyridinium (MPP+) treated dopaminergic neurons occurs as an NADPH oxidase-dependent two-wave cascade. J. Neuroinflamm. 2011, 8, 129. [Google Scholar] [CrossRef]
- Choi, D.H.; Cristovao, A.C.; Guhathakurta, S.; Lee, J.; Joh, T.H.; Beal, M.F.; Kim, Y.S. NADPH oxidase 1-mediated oxidative stress leads to dopamine neuron death in Parkinson’s disease. Antioxid. Redox Signal. 2012, 16, 1033–1045. [Google Scholar] [CrossRef]
- Cristovao, A.C.; Guhathakurta, S.; Bok, E.; Je, G.; Yoo, S.D.; Choi, D.H.; Kim, Y.S. NADPH oxidase 1 mediates alpha-synucleinopathy in Parkinson’s disease. J. Neurosci. 2012, 32, 14465–14477. [Google Scholar] [CrossRef]
- Keeney, M.T.; Hoffman, E.K.; Farmer, K.; Bodle, C.R.; Fazzari, M.; Zharikov, A.; Castro, S.L.; Hu, X.; Mortimer, A.; Kofler, J.K.; et al. NADPH oxidase 2 activity in Parkinson’s disease. Neurobiol. Dis. 2022, 170, 105754. [Google Scholar] [CrossRef]
- Pullara, F.; Forsmann, M.C.; General, I.J.; Ayoob, J.C.; Furbee, E.; Castro, S.L.; Hu, X.; Greenamyre, J.T.; Di Maio, R. NADPH oxidase 2 activity disrupts Calmodulin/CaMKIIalpha complex via redox modifications of CaMKIIalpha-contained Cys30 and Cys289: Implications in Parkinson’s disease. Redox Biol. 2024, 75, 103254. [Google Scholar] [CrossRef]
- Keeney, M.T.; Rocha, E.M.; Hoffman, E.K.; Farmer, K.; Di Maio, R.; Weir, J.; Wagner, W.G.; Hu, X.; Clark, C.L.; Castro, S.L.; et al. LRRK2 regulates production of reactive oxygen species in cell and animal models of Parkinson’s disease. Sci. Transl. Med. 2024, 16, eadl3438. [Google Scholar] [CrossRef]
- Lin, Z.; Ying, C.; Si, X.; Xue, N.; Liu, Y.; Zheng, R.; Chen, Y.; Pu, J.; Zhang, B. NOX4 exacerbates Parkinson’s disease pathology by promoting neuronal ferroptosis and neuroinflammation. Neural Regen. Res. 2025, 20, 2038–2052. [Google Scholar] [CrossRef]
- Tan, Q.; Zhang, C.; Rao, X.; Wan, W.; Lin, W.; Huang, S.; Ying, J.; Lin, Y.; Hua, F. The interaction of lipocalin-2 and astrocytes in neuroinflammation: Mechanisms and therapeutic application. Front. Immunol. 2024, 15, 1358719. [Google Scholar] [CrossRef]
- Sharma, V.; Sharma, P.; Singh, T.G. Emerging role of Nrf2 in Parkinson’s disease therapy: A critical reassessment. Metab. Brain Dis. 2024, 40, 70. [Google Scholar] [CrossRef] [PubMed]
- Rajan, S.; Tryphena, K.P.; Khan, S.; Vora, L.; Srivastava, S.; Singh, S.B.; Khatri, D.K. Understanding the involvement of innate immunity and the Nrf2-NLRP3 axis on mitochondrial health in Parkinson’s disease. Ageing Res. Rev. 2023, 87, 101915. [Google Scholar] [CrossRef]
- Ramsey, C.P.; Glass, C.A.; Montgomery, M.B.; Lindl, K.A.; Ritson, G.P.; Chia, L.A.; Hamilton, R.L.; Chu, C.T.; Jordan-Sciutto, K.L. Expression of Nrf2 in neurodegenerative diseases. J. Neuropathol. Exp. Neurol. 2007, 66, 75–85. [Google Scholar] [CrossRef] [PubMed]
- van Muiswinkel, F.L.; de Vos, R.A.; Bol, J.G.; Andringa, G.; Jansen Steur, E.N.; Ross, D.; Siegel, D.; Drukarch, B. Expression of NAD(P)H:quinone oxidoreductase in the normal and Parkinsonian substantia nigra. Neurobiol. Aging 2004, 25, 1253–1262. [Google Scholar] [CrossRef]
- Niu, Y.; Zhang, J.; Dong, M. Nrf2 as a potential target for Parkinson’s disease therapy. J. Mol. Med. 2021, 99, 917–931. [Google Scholar] [CrossRef]
- Johri, A.; Chandra, A.; Flint Beal, M. PGC-1alpha, mitochondrial dysfunction, and Huntington’s disease. Free Radic. Biol. Med. 2013, 62, 37–46. [Google Scholar] [CrossRef]
- Bono-Yagüe, J.; Gómez-Escribano, A.P.; Millán, J.M.; Vázquez-Manrique, R.P. Reactive Species in Huntington Disease: Are They Really the Radicals You Want to Catch? Antioxidants 2020, 9, 577. [Google Scholar] [CrossRef]
- Kolobkova, Y.A.; Vigont, V.A.; Shalygin, A.V.; Kaznacheyeva, E.V. Huntington’s Disease: Calcium Dyshomeostasis and Pathology Models. Acta Nat. 2017, 9, 34–46. [Google Scholar] [CrossRef]
- Valle, I.; Alvarez-Barrientos, A.; Arza, E.; Lamas, S.; Monsalve, M. PGC-1α regulates the mitochondrial antioxidant defense system in vascular endothelial cells. Cardiovasc. Res. 2005, 66, 562–573. [Google Scholar] [CrossRef] [PubMed]
- Okamoto, S.; Pouladi, M.A.; Talantova, M.; Yao, D.; Xia, P.; Ehrnhoefer, D.E.; Zaidi, R.; Clemente, A.; Kaul, M.; Graham, R.K.; et al. Balance between synaptic versus extrasynaptic NMDA receptor activity influences inclusions and neurotoxicity of mutant huntingtin. Nat. Med. 2009, 15, 1407–1413. [Google Scholar] [CrossRef]
- Reddy, P.H.; Shirendeb, U.P. Mutant huntingtin, abnormal mitochondrial dynamics, defective axonal transport of mitochondria, and selective synaptic degeneration in Huntington’s disease. Biochim. Biophys. Acta 2012, 1822, 101–110. [Google Scholar] [CrossRef]
- Carmo, C.; Naia, L.; Lopes, C.; Rego, A.C. Mitochondrial Dysfunction in Huntington’s Disease. Adv. Exp. Med. Biol. 2018, 1049, 59–83. [Google Scholar]
- Shirendeb, U.; Reddy, A.P.; Manczak, M.; Calkins, M.J.; Mao, P.; Tagle, D.A.; Reddy, P.H. Abnormal mitochondrial dynamics, mitochondrial loss and mutant huntingtin oligomers in Huntington’s disease: Implications for selective neuronal damage. Hum. Mol. Genet. 2011, 20, 1438–1455. [Google Scholar] [CrossRef] [PubMed]
- Machiela, E.; Rudich, P.D.; Traa, A.; Anglas, U.; Soo, S.K.; Senchuk, M.M.; Van Raamsdonk, J.M. Targeting Mitochondrial Network Disorganization is Protective in Models of Huntington’s Disease. Aging Dis. 2021, 12, 1753–1772. [Google Scholar] [CrossRef] [PubMed]
- Yablonska, S.; Ganesan, V.; Ferrando, L.M.; Kim, J.; Pyzel, A.; Baranova, O.V.; Khattar, N.K.; Larkin, T.M.; Baranov, S.V.; Chen, N.; et al. Mutant huntingtin disrupts mitochondrial proteostasis by interacting with TIM23. Proc. Natl. Acad. Sci. USA 2019, 116, 16593–16602. [Google Scholar] [CrossRef]
- Yablonska, S.; Strohlein, C.E.; Baranov, S.V.; Yeh, S.M.; Patel, A.; Singh, T.; Jauhari, A.; Kim, J.; Khattar, N.K.; Li, F.; et al. Regulation of Mutant Huntingtin Mitochondrial Toxicity by Phosphomimetic Mutations within Its N-Terminal Region. J. Neurosci. 2025, 45, e1254242024. [Google Scholar] [CrossRef]
- Maity, S.; Komal, P.; Kumar, V.; Saxena, A.; Tungekar, A.; Chandrasekar, V. Impact of ER Stress and ER-Mitochondrial Crosstalk in Huntington’s Disease. Int. J. Mol. Sci. 2022, 23, 780. [Google Scholar] [CrossRef]
- Chong, W.C.; Shastri, M.D.; Eri, R. Endoplasmic Reticulum Stress and Oxidative Stress: A Vicious Nexus Implicated in Bowel Disease Pathophysiology. Int. J. Mol. Sci. 2017, 18, 771. [Google Scholar] [CrossRef]
- D’Egidio, F.; Qosja, E.; Ammannito, F.; Topi, S.; d’Angelo, M.; Cimini, A.; Castelli, V. Antioxidant and Anti-Inflammatory Defenses in Huntington’s Disease: Roles of NRF2 and PGC-1alpha, and Therapeutic Strategies. Life 2025, 15, 577. [Google Scholar] [CrossRef] [PubMed]
- Khan, S.; Bano, N.; Ahamad, S.; Dar, N.J.; Nazir, A.; Bhat, S.A. Advances in nanotherapeutic strategies for Huntington’s disease: Design, delivery, and neuroprotective mechanisms. Coord. Chem. Rev. 2025, 522, 216206. [Google Scholar] [CrossRef]
- Byun, S.; Lee, M.; Kim, M. Gene Therapy for Huntington’s Disease: The Final Strategy for a Cure? J. Mov. Disord. 2022, 15, 15–20. [Google Scholar] [CrossRef]
- Spuch, C.; Navarro, C. Liposomes for Targeted Delivery of Active Agents against Neurodegenerative Diseases (Alzheimer’s Disease and Parkinson’s Disease). J. Drug Deliv. 2011, 2011, 469679. [Google Scholar] [CrossRef]
- Motataianu, A.; Serban, G.; Barcutean, L.; Balasa, R. Oxidative Stress in Amyotrophic Lateral Sclerosis: Synergy of Genetic and Environmental Factors. Int. J. Mol. Sci. 2022, 23, 9339. [Google Scholar] [CrossRef] [PubMed]
- Barber, S.C.; Mead, R.J.; Shaw, P.J. Oxidative stress in ALS: A mechanism of neurodegeneration and a therapeutic target. Biochim. Biophys. Acta 2006, 1762, 1051–1067. [Google Scholar] [CrossRef] [PubMed]
- Park, H.R.; Yang, E.J. Oxidative Stress as a Therapeutic Target in Amyotrophic Lateral Sclerosis: Opportunities and Limitations. Diagnostics 2021, 11, 1546. [Google Scholar] [CrossRef]
- Pollari, E.; Goldsteins, G.; Bart, G.; Koistinaho, J.; Giniatullin, R. The role of oxidative stress in degeneration of the neuromuscular junction in amyotrophic lateral sclerosis. Front. Cell. Neurosci. 2014, 8, 131. [Google Scholar] [CrossRef]
- Rosen, D.R.; Siddique, T.; Patterson, D.; Figlewicz, D.A.; Sapp, P.; Hentati, A.; Donaldson, D.; Goto, J.; O’Regan, J.P.; Deng, H.X.; et al. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 1993, 362, 59–62. [Google Scholar] [CrossRef]
- Gagliardi, D.; Ripellino, P.; Meneri, M.; Del Bo, R.; Antognozzi, S.; Comi, G.P.; Gobbi, C.; Ratti, A.; Ticozzi, N.; Silani, V.; et al. Clinical and molecular features of patients with amyotrophic lateral sclerosis and SOD1 mutations: A monocentric study. Front. Neurol. 2023, 14, 1169689. [Google Scholar] [CrossRef]
- Roe, J.A.; Wiedau-Pazos, M.; Moy, V.N.; Goto, J.J.; Gralla, E.B.; Valentine, J.S. In vivo peroxidative activity of FALS-mutant human CuZnSODs expressed in yeast. Free Radic. Biol. Med. 2002, 32, 169–174. [Google Scholar] [CrossRef] [PubMed]
- Liochev, S.I.; Chen, L.L.; Hallewell, R.A.; Fridovich, I. The familial amyotrophic lateral sclerosis-associated amino acid substitutions E100G, G93A, and G93R do not influence the rate of inactivation of copper- and zinc-containing superoxide dismutase by H2O2. Arch. Biochem. Biophys. 1998, 352, 237–239. [Google Scholar] [CrossRef] [PubMed]
- Browne, S.E.; Bowling, A.C.; Baik, M.J.; Gurney, M.; Brown, R.H., Jr.; Beal, M.F. Metabolic dysfunction in familial, but not sporadic, amyotrophic lateral sclerosis. J. Neurochem. 1998, 71, 281–287. [Google Scholar] [CrossRef]
- Richardson, K.; Allen, S.P.; Mortiboys, H.; Grierson, A.J.; Wharton, S.B.; Ince, P.G.; Shaw, P.J.; Heath, P.R. The Effect of SOD1 Mutation on Cellular Bioenergetic Profile and Viability in Response to Oxidative Stress and Influence of Mutation-Type. PLoS ONE 2013, 8, e68256. [Google Scholar] [CrossRef] [PubMed]
- Cunha-Oliveira, T.; Montezinho, L.; Mendes, C.; Firuzi, O.; Saso, L.; Oliveira, P.J.; Silva, F.S.G. Oxidative Stress in Amyotrophic Lateral Sclerosis: Pathophysiology and Opportunities for Pharmacological Intervention. Oxid. Med. Cell. Longev. 2020, 2020, 5021694. [Google Scholar] [CrossRef]
- Chong, Z.Z.; Souayah, N. Pathogenic TDP-43 in amyotrophic lateral sclerosis. Drug Discov. Today 2025, 30, 104351. [Google Scholar] [CrossRef]
- Zuo, X.; Zhou, J.; Li, Y.; Wu, K.; Chen, Z.; Luo, Z.; Zhang, X.; Liang, Y.; Esteban, M.A.; Zhou, Y.; et al. TDP-43 aggregation induced by oxidative stress causes global mitochondrial imbalance in ALS. Nat. Struct. Mol. Biol. 2021, 28, 132–142. [Google Scholar] [CrossRef]
- Lee, Y.B.; Scotter, E.L.; Lee, D.Y.; Troakes, C.; Mitchell, J.; Rogelj, B.; Gallo, J.M.; Shaw, C.E. Cytoplasmic TDP-43 is involved in cell fate during stress recovery. Hum. Mol. Genet. 2021, 31, 166–175. [Google Scholar] [CrossRef]
- Au, W.H.; Miller-Fleming, L.; Sanchez-Martinez, A.; Lee, J.A.; Twyning, M.J.; Prag, H.A.; Raik, L.; Allen, S.P.; Shaw, P.J.; Ferraiuolo, L.; et al. Activation of the Keap1/Nrf2 pathway suppresses mitochondrial dysfunction, oxidative stress, and motor phenotypes in C9orf72 ALS/FTD models. Life Sci. Alliance 2024, 7, e202402853. [Google Scholar] [CrossRef]
- Chong, Z.Z.; Menkes, D.L.; Souayah, N. Pathogenesis underlying hexanucleotide repeat expansions in C9orf72 gene in amyotrophic lateral sclerosis. Rev. Neurosci. 2024, 35, 85–97. [Google Scholar] [CrossRef]
- Dafinca, R.; Scaber, J.; Ababneh, N.; Lalic, T.; Weir, G.; Christian, H.; Vowles, J.; Douglas, A.G.; Fletcher-Jones, A.; Browne, C.; et al. C9orf72 Hexanucleotide Expansions Are Associated with Altered Endoplasmic Reticulum Calcium Homeostasis and Stress Granule Formation in Induced Pluripotent Stem Cell-Derived Neurons from Patients with Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Stem Cells 2016, 34, 2063–2078. [Google Scholar] [PubMed]
- Riemslagh, F.W.; Verhagen, R.F.M.; van der Toorn, E.C.; Smits, D.J.; Quint, W.H.; van der Linde, H.C.; van Ham, T.J.; Willemsen, R. Reduction of oxidative stress suppresses poly-GR-mediated toxicity in zebrafish embryos. Dis. Model. Mech. 2021, 14, dmm049092. [Google Scholar] [CrossRef] [PubMed]
- Cicardi, M.E.; Hallgren, J.H.; Mawrie, D.; Krishnamurthy, K.; Markandaiah, S.S.; Nelson, A.T.; Kankate, V.; Anderson, E.N.; Pasinelli, P.; Pandey, U.B.; et al. C9orf72 poly(PR) mediated neurodegeneration is associated with nucleolar stress. iScience 2023, 26, 107505. [Google Scholar] [CrossRef] [PubMed]
- Jimenez-Villegas, J.; Kirby, J.; Mata, A.; Cadenas, S.; Turner, M.R.; Malaspina, A.; Shaw, P.J.; Cuadrado, A.; Rojo, A.I. Dipeptide Repeat Pathology in C9orf72-ALS Is Associated with Redox, Mitochondrial and NRF2 Pathway Imbalance. Antioxidants 2022, 11, 1897. [Google Scholar] [CrossRef]
- Sama, R.R.; Ward, C.L.; Kaushansky, L.J.; Lemay, N.; Ishigaki, S.; Urano, F.; Bosco, D.A. FUS/TLS assembles into stress granules and is a prosurvival factor during hyperosmolar stress. J. Cell. Physiol. 2013, 228, 2222–2231. [Google Scholar] [CrossRef]
- Wang, H.; Guo, W.; Mitra, J.; Hegde, P.M.; Vandoorne, T.; Eckelmann, B.J.; Mitra, S.; Tomkinson, A.E.; Van Den Bosch, L.; Hegde, M.L. Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in Amyotrophic Lateral Sclerosis. Nat. Commun. 2018, 9, 3683. [Google Scholar] [CrossRef]
- Kodavati, M.; Wang, H.; Guo, W.; Mitra, J.; Hegde, P.M.; Provasek, V.; Rao, V.H.M.; Vedula, I.; Zhang, A.; Mitra, S.; et al. FUS unveiled in mitochondrial DNA repair and targeted ligase-1 expression rescues repair-defects in FUS-linked motor neuron disease. Nat. Commun. 2024, 15, 2156. [Google Scholar] [CrossRef]
- Hoang, T.T.; Johnson, D.A.; Raines, R.T.; Johnson, J.A. Angiogenin activates the astrocytic Nrf2/antioxidant-response element pathway and thereby protects murine neurons from oxidative stress. J. Biol. Chem. 2019, 294, 15095–15103. [Google Scholar] [CrossRef]
- Aluri, K.C.; Salisbury, J.P.; Prehn, J.H.M.; Agar, J.N. Loss of angiogenin function is related to earlier ALS onset and a paradoxical increase in ALS duration. Sci. Rep. 2020, 10, 3715. [Google Scholar] [CrossRef]
- Anderson, C.J.; Bredvik, K.; Burstein, S.R.; Davis, C.; Meadows, S.M.; Dash, J.; Case, L.; Milner, T.A.; Kawamata, H.; Zuberi, A.; et al. ALS/FTD mutant CHCHD10 mice reveal a tissue-specific toxic gain-of-function and mitochondrial stress response. Acta Neuropathol. 2019, 138, 103–121. [Google Scholar] [CrossRef]
- Straub, I.R.; Janer, A.; Weraarpachai, W.; Zinman, L.; Robertson, J.; Rogaeva, E.; Shoubridge, E.A. Loss of CHCHD10-CHCHD2 complexes required for respiration underlies the pathogenicity of a CHCHD10 mutation in ALS. Hum. Mol. Genet. 2018, 27, 178–189. [Google Scholar] [CrossRef]
- Genin, E.C.; Plutino, M.; Bannwarth, S.; Villa, E.; Cisneros-Barroso, E.; Roy, M.; Ortega-Vila, B.; Fragaki, K.; Lespinasse, F.; Pinero-Martos, E.; et al. CHCHD10 mutations promote loss of mitochondrial cristae junctions with impaired mitochondrial genome maintenance and inhibition of apoptosis. EMBO Mol. Med. 2016, 8, 58–72. [Google Scholar] [CrossRef] [PubMed]
- Ikeda, A.; Meng, H.; Taniguchi, D.; Mio, M.; Funayama, M.; Nishioka, K.; Yoshida, M.; Li, Y.; Yoshino, H.; Inoshita, T.; et al. CHCHD2 P14L, found in amyotrophic lateral sclerosis, exhibits cytoplasmic mislocalization and alters Ca2+ homeostasis. PNAS Nexus 2024, 3, 319. [Google Scholar] [CrossRef] [PubMed]
- D’Ambrosi, N.; Cozzolino, M.; Carrì, M.T. Neuroinflammation in Amyotrophic Lateral Sclerosis: Role of Redox (dys)Regulation. Antioxid. Redox Signal. 2018, 29, 15–36. [Google Scholar] [CrossRef]
- Xiong, L.Y.; McCoy, M.; Komuro, H.; West, X.Z.; Yakubenko, V.; Gao, D.T.; Dudiki, T.; Milo, A.; Chen, J.; Podrez, E.A.; et al. Inflammation-dependent oxidative stress metabolites as a hallmark of amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2022, 178, 125–133. [Google Scholar] [CrossRef]
- D’Amico, E.; Factor-Litvak, P.; Santella, R.M.; Mitsumoto, H. Clinical perspective on oxidative stress in sporadic amyotrophic lateral sclerosis. Free Radic. Biol. Med. 2013, 65, 509–527. [Google Scholar] [CrossRef]
- Orrell, R.W.; Lane, R.J.; Ross, M. Antioxidant treatment for amyotrophic lateral sclerosis/motor neuron disease. Cochrane Database Syst. Rev. 2007, 2007, CD002829. [Google Scholar] [CrossRef]
- Sala, G.; Arosio, A.; Conti, E.; Beretta, S.; Lunetta, C.; Riva, N.; Ferrarese, C.; Tremolizzo, L. Riluzole Selective Antioxidant Effects in Cell Models Expressing Amyotrophic Lateral Sclerosis Endophenotypes. Clin. Psychopharmacol. Neurosci. 2019, 17, 438–442. [Google Scholar] [CrossRef]
- Hogg, M.C.; Halang, L.; Woods, I.; Coughlan, K.S.; Prehn, J.H.M. Riluzole does not improve lifespan or motor function in three ALS mouse models. Amyotroph. Lateral Scler. Front. Degener. 2018, 19, 438–445. [Google Scholar] [CrossRef] [PubMed]
- Cho, H.; Shukla, S. Role of Edaravone as a Treatment Option for Patients with Amyotrophic Lateral Sclerosis. Pharmaceuticals 2020, 14, 29. [Google Scholar] [CrossRef]
- Yoshino, H. Edaravone for the treatment of amyotrophic lateral sclerosis. Expert. Rev. Neurother. 2019, 19, 185–193. [Google Scholar] [CrossRef] [PubMed]
- Ortiz, J.F.; Khan, S.A.; Salem, A.; Lin, Z.; Iqbal, Z.; Jahan, N. Post-Marketing Experience of Edaravone in Amyotrophic Lateral Sclerosis: A Clinical Perspective and Comparison With the Clinical Trials of the Drug. Cureus 2020, 12, e10818. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 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
Chong, Z.Z.; Souayah, N. Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases. Antioxidants 2025, 14, 696. https://doi.org/10.3390/antiox14060696
Chong ZZ, Souayah N. Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases. Antioxidants. 2025; 14(6):696. https://doi.org/10.3390/antiox14060696
Chicago/Turabian StyleChong, Zhao Zhong, and Nizar Souayah. 2025. "Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases" Antioxidants 14, no. 6: 696. https://doi.org/10.3390/antiox14060696
APA StyleChong, Z. Z., & Souayah, N. (2025). Oxidative Stress: Pathological Driver in Chronic Neurodegenerative Diseases. Antioxidants, 14(6), 696. https://doi.org/10.3390/antiox14060696