Neurons are especially vulnerable because of their high metabolic activity and limited ability to repair damaged DNA. Oxidative genotoxic stress (OGS), which arises from the buildup of short-lived, highly reactive molecules called reactive oxygen species (ROS), can damage neuronal DNA and compromise antioxidant defense mechanisms in neurons. OGS induces considerable forms of DNA damage, including genomic instability, DNA strand breaks (single or double), DNA base modifications such as 8-oxoguanine, and epigenetic changes, leading to compromised neuronal functions. Moreover, OGS is a silent player in mitochondrial DNA damage and mitochondrial dysfunction. Therefore, ROS-mediated OGS is pivotal for initiating and advancing several neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinsonism (PD), and Huntington's disease (HD). However, there is a significant gap in deciphering the molecular pathways involved in OGS-mediated development of neurodegenerative diseases. Hence, this study focused on the molecular mechanisms by which OGS causes neurodegeneration, with a focus on the contributions of neuroinflammation, mitochondrial dysfunction, and defective DNA repair pathways. Additionally, new therapeutic approaches, such as mitochondrial-targeted medications, antioxidant therapies, gene editing tools such as CRISPR/Cas9, and biomarkers for the early diagnosis of these oxidative diseases, have been assessed. A thorough comprehension of these processes opens exciting possibilities for focused treatments in neurodegenerative illnesses.
Citation: Taslim Uddin. Oxidative genomic or genotoxic stress in neurodegeneration: Mechanisms and therapeutic avenues[J]. AIMS Neuroscience, 2025, 12(3): 369-390. doi: 10.3934/Neuroscience.2025020
Neurons are especially vulnerable because of their high metabolic activity and limited ability to repair damaged DNA. Oxidative genotoxic stress (OGS), which arises from the buildup of short-lived, highly reactive molecules called reactive oxygen species (ROS), can damage neuronal DNA and compromise antioxidant defense mechanisms in neurons. OGS induces considerable forms of DNA damage, including genomic instability, DNA strand breaks (single or double), DNA base modifications such as 8-oxoguanine, and epigenetic changes, leading to compromised neuronal functions. Moreover, OGS is a silent player in mitochondrial DNA damage and mitochondrial dysfunction. Therefore, ROS-mediated OGS is pivotal for initiating and advancing several neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinsonism (PD), and Huntington's disease (HD). However, there is a significant gap in deciphering the molecular pathways involved in OGS-mediated development of neurodegenerative diseases. Hence, this study focused on the molecular mechanisms by which OGS causes neurodegeneration, with a focus on the contributions of neuroinflammation, mitochondrial dysfunction, and defective DNA repair pathways. Additionally, new therapeutic approaches, such as mitochondrial-targeted medications, antioxidant therapies, gene editing tools such as CRISPR/Cas9, and biomarkers for the early diagnosis of these oxidative diseases, have been assessed. A thorough comprehension of these processes opens exciting possibilities for focused treatments in neurodegenerative illnesses.
| [1] |
Erkkinen MG, Kim MO, Geschwind MD (2018) Clinical Neurology and Epidemiology of the Major Neurodegenerative Diseases. Cold Spring Harb Perspect Biol 10: a033118. https://doi.org/10.1101/cshperspect.a033118
|
| [2] | GBD 2016 Parkinson's Disease Collaborators.Global, regional, and national burden of Parkinson's disease, 1990–2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol (2018) 17: 939-953. http://dx.doi.org/10.1016/S1474-4422(18)30295-3 |
| [3] |
Portaccio E, Magyari M, Havrdova EK, et al. (2024) Multiple sclerosis: emerging epidemiological trends and redefining the clinical course. Lancet Reg Health Eur 44: 100977. https://doi.org/10.1016/j.lanepe.2024.100977
|
| [4] |
Kisby GE, Wilson DM, Spencer PS (2024) Introducing the Role of Genotoxicity in Neurodegenerative Diseases and Neuropsychiatric Disorders. Int J Mol Sci 25: 7221. https://doi.org/10.3390/ijms25137221
|
| [5] |
Penalva-Olcina R, Juan C, Fernández-Franzón M, et al. (2025) Neurotoxic implications of gliotoxin and ochratoxin A in SH-SY5Y cells: ROS-induced apoptosis and genotoxicity. Toxicol Lett 405: 51-58. https://doi.org/10.1016/j.toxlet.2025.02.004
|
| [6] | Kumari S, Begum MY, Chinglenthoiba C, et al. (2025) Deciphering the Neurotoxic Burden of Micro- and Nanoplastics: From Multi-model Experimental Evidence to Therapeutic Innovation. Mol Neurobiol 2025: 1-23. https://doi.org/10.1007/s12035-025-05174-z |
| [7] |
Dey KK, Kamila S, Das T, et al. (2025) Chronic Exposure to Lead Causes Neurotoxicity by Generating Oxidative Stress and Inducing DNA Damages in Zebrafish Brain: Involvement of Nrf2-Keap1 Regulation and DNA Repair Pathways. Biol Trace Elem Res 203: 4863-4875. https://doi.org/10.1007/s12011-025-04534-z
|
| [8] |
Balestrazzi A, Confalonieri M, Macovei A, et al. (2011) Genotoxic stress and DNA repair in plants: emerging functions and tools for improving crop productivity. Plant Cell Rep 30: 287-295. https://doi.org/10.1007/s00299-010-0975-9
|
| [9] |
Uddin T, Srite RP (2025) Crosstalk of the neuronal microenvironment in the pathophysiology of Alzheimer's disease and prospective intervention strategies advancing mitochondrial dynamics: A comprehensive review. Int Neuropsychiatr Dis J 22: 135-148. https://doi.org/10.9734/indj/2025/v22i3486
|
| [10] |
Hakem R (2008) DNA-damage repair; the good, the bad, and the ugly. EMBO J 27: 589-605. https://doi.org/10.1038/emboj.2008.15
|
| [11] |
Ferrer MD, Sureda A, Mestre A, et al. (2010) The double edge of reactive oxygen species as damaging and signaling molecules in HL60 cell culture. Cell Physiol Biochem 25: 241-252. https://doi.org/10.1159/000276558
|
| [12] |
Allgayer J, Kitsera N, von der Lippen C, et al. (2013) Modulation of base excision repair of 8-oxoguanine by the nucleotide sequence. Nucleic Acids Res 41: 8559-8571. https://doi.org/10.1093/nar/gkt620
|
| [13] |
Neeley WL, Essigmann JM (2006) Mechanisms of formation, genotoxicity, and mutation of guanine oxidation products. Chem Res Toxicol 19: 491-505. https://doi.org/10.1021/tx0600043
|
| [14] |
Markkanen E, Dorn J, Hübscher U (2013) MUTYH DNA glycosylase: the rationale for removing undamaged bases from the DNA. Front Genet 4: 18. https://doi.org/10.3389/fgene.2013.00018
|
| [15] |
Saxowsky TT, Meadows KL, Klungland A, et al. (2008) 8-Oxoguanine-mediated transcriptional mutagenesis causes Ras activation in mammalian cells. Proc Natl Acad Sci U S A 105: 18877-18882. https://doi.org/10.1073/pnas.0806464105
|
| [16] |
Kitsera N, Stathis D, Lühnsdorf B, et al. (2011) 8-Oxo-7,8-dihydroguanine in DNA does not constitute a barrier to transcription, but is converted into transcription-blocking damage by OGG1. Nucleic Acids Res 39: 5926-5934. https://doi.org/10.1093/nar/gkr163
|
| [17] |
Caldecott KW (2008) Single-strand break repair and genetic disease. Nat Rev Genet 9: 619-631. https://doi.org/10.1038/nrg2380
|
| [18] |
da Silva PFL, Schumacher B (2019) DNA damage responses in ageing. Open Biol 9: 190168. https://doi.org/10.1098/rsob.190168
|
| [19] |
Markkanen E (2017) Not breathing is not an option: How to deal with oxidative DNA damage. DNA Repair (Amst) 59: 82-105. https://doi.org/10.1016/j.dnarep.2017.09.007
|
| [20] |
You YH, Lee DH, Yoon JH, et al. (2001) Cyclobutane pyrimidine dimers are responsible for the vast majority of mutations induced by UVB irradiation in mammalian cells. J Biol Chem 276: 44688-44694. https://doi.org/10.1074/jbc.M107696200
|
| [21] |
Thadathil N, Hori R, Xiao J, et al. (2019) DNA double-strand breaks: a potential therapeutic target for neurodegenerative diseases. Chromosome Res 27: 345-364. https://doi.org/10.1007/s10577-019-09617-x
|
| [22] |
Ambrosio S, Di Palo G, Napolitano G, et al. (2016) Cell cycle-dependent resolution of DNA double-strand breaks. Oncotarget 7: 4949-4960. https://doi.org/10.18632/oncotarget.6644
|
| [23] |
Zhou BB, Elledge SJ (2000) The DNA damage response: putting checkpoints in perspective. Nature 408: 433-439. https://doi.org/10.1038/35044005
|
| [24] |
Cannan WJ, Pederson DS (2016) Mechanisms and Consequences of Double-Strand DNA Break Formation in Chromatin. J Cell Physiol 231: 3-14. https://doi.org/10.1002/jcp.25048
|
| [25] |
Kannan A, Bhatia K, Branzei D, et al. (2018) Combined deficiency of Senataxin and DNA-PKcs causes DNA damage accumulation and neurodegeneration in spinal muscular atrophy. Nucleic Acids Res 46: 8326-8346. https://doi.org/10.1093/nar/gky641
|
| [26] |
Madabhushi R, Gao F, Pfenning AR, et al. (2015) Activity-Induced DNA Breaks Govern the Expression of Neuronal Early-Response Genes. Cell 161: 1592-1605. https://doi.org/10.1016/j.cell.2015.05.032
|
| [27] |
Cáceres L, Paz ML, Garcés M, et al. (2020) NADPH oxidase and mitochondria are relevant sources of superoxide anion in the oxinflammatory response of macrophages exposed to airborne particulate matter. Ecotoxicol Environ Saf 205: 111186. https://doi.org/10.1016/j.ecoenv.2020.111186
|
| [28] |
Wu Z, Sainz AG, Shadel GS (2021) Mitochondrial DNA: cellular genotoxic stress sentinel. Trends Biochem Sci 46: 812-821. https://doi.org/10.1016/j.tibs.2021.05.004
|
| [29] | Phillips DH, Arlt VM (2009) Genotoxicity: damage to DNA and its consequences. EXS 99: 87-110. https://doi.org/10.1007/978-3-7643-8336-7_4 |
| [30] |
Kuznetsov AV (2025) Modeling of ATP transport in an axon: Effects of spontaneous neuron firing and mitochondrial transfer via tunneling nanotubes. Int J Heat Mass Transf 242: 126790. https://doi.org/10.1016/j.ijheatmasstransfer.2025.126790
|
| [31] |
Watanabe H, Shima S, Kawabata K, et al. (2025) Brain network and energy imbalance in Parkinson's disease: linking ATP reduction and α-synuclein pathology. Front Mol Neurosci 17: 1507033. https://doi.org/10.3389/fnmol.2024.1507033
|
| [32] |
Feofilaktova T, Kushnireva L, Segal M, et al. (2025) Calcium signaling in postsynaptic mitochondria: mechanisms, dynamics, and role in ATP production. Front Mol Neurosci 18: 1621070. https://doi.org/10.3389/fnmol.2025.1621070
|
| [33] |
Peter-Okaka U, Boison D (2025) Neuroglia and brain energy metabolism. Handb Clin Neurol 209: 117-126. https://doi.org/10.1016/B978-0-443-19104-6.00007-3
|
| [34] | Rozich E, Ozkurede U, Pakkiriswami S, et al. (2025) Mitochondrial oxidative stress, calcium and dynamics in cardiac ischaemia-reperfusion injury. J Physiol . https://doi.org/10.1113/JP287770 |
| [35] |
Filipek B, Macieja A, Binda A, et al. (2025) Oxidative DNA Damage and Repair Dynamics in Multiple Sclerosis: Insights from Comet Assay Kinetics, Base Excision Repair Gene Expression, and Genotype Analysis. Biomolecules 15: 756. https://doi.org/10.3390/biom15060756
|
| [36] |
Filipek B, Macieja A, Binda A, et al. (2025) Polymorphisms in Base Excision Repair Genes and Association with Multiple Sclerosis in a Pilot Study on a Central European Population. Int J Mol Sci 26: 6612. https://doi.org/10.3390/ijms26146612
|
| [37] |
Chatgilialoglu C (2025) Biological Models of Oxidative Purine DNA Damage in Neurodegenerative Disorders. Antioxidants (Basel) 14: 578. https://doi.org/10.3390/antiox14050578
|
| [38] |
Arat Çelik HE, Yılmaz S, Akşahin İC, et al. (2024) Oxidatively-induced DNA base damage and base excision repair abnormalities in siblings of individuals with bipolar disorder DNA damage and repair in bipolar disorder. Transl Psychiatry 14: 207. https://doi.org/10.1038/s41398-024-02901-3
|
| [39] |
Jacoby AS, Vinberg M, Poulsen HE, et al. (2016) Increased DNA and RNA damage by oxidation in patients with bipolar I disorder. Transl Psychiatry 6: e867. https://doi.org/10.1038/tp.2016.141
|
| [40] |
Bosshard M, Markkanen E, van Loon B (2012) Base excision repair in physiology and pathology of the central nervous system. Int J Mol Sci 13: 16172-16222. https://doi.org/10.3390/ijms131216172
|
| [41] |
Audebert M, Radicella JP, Dizdaroglu M (2000) Effect of single mutations in the OGG1 gene found in human tumors on the substrate specificity of the Ogg1 protein. Nucleic Acids Res 28: 2672-2678. https://doi.org/10.1093/nar/28.14.2672
|
| [42] |
Jensen A, Calvayrac G, Karahalil B, et al. (2003) Mammalian 8-oxoguanine DNA glycosylase 1 incises 8-oxoadenine opposite cytosine in nuclei and mitochondria, while a different glycosylase incises 8-oxoadenine opposite guanine in nuclei. J Biol Chem 278: 19541-19548. https://doi.org/10.1074/jbc.M301504200
|
| [43] |
Morales-Ruiz T, Birincioglu M, Jaruga P, et al. (2003) Arabidopsis thaliana Ogg1 protein excises 8-hydroxyguanine and 2,6-diamino-4-hydroxy-5-formamidopyrimidine from oxidatively damaged DNA containing multiple lesions. Biochemistry 42: 3089-3095. https://doi.org/10.1021/bi027226u
|
| [44] |
Blackford AN, Jackson SP (2017) ATM, ATR, and DNA-PK: The Trinity at the Heart of the DNA Damage Response. Mol Cell 66: 801-817. https://doi.org/10.1016/j.molcel.2017.05.015
|
| [45] |
van den Bosch M, Bree RT, Lowndes NF (2003) The MRN complex: coordinating and mediating the response to broken chromosomes. EMBO Rep 4: 844-849. https://doi.org/10.1038/sj.embor.embor925
|
| [46] |
Polo SE, Jackson SP (2011) Dynamics of DNA damage response proteins at DNA breaks: a focus on protein modifications. Genes Dev 25: 409-433. https://doi.org/10.1101/gad.2021311
|
| [47] |
Shi L, Oberdoerffer P (2012) Chromatin dynamics in DNA double-strand break repair. Biochim Biophys Acta 1819: 811-819. https://doi.org/10.1016/j.bbagrm.2012.01.002
|
| [48] |
van Attikum H, Gasser SM (2009) Crosstalk between histone modifications during the DNA damage response. Trends Cell Biol 19: 207-217. https://doi.org/10.1016/j.tcb.2009.03.001
|
| [49] |
Chang HHY, Pannunzio NR, Adachi N, et al. (2017) Non-homologous DNA end joining and alternative pathways to double-strand break repair. Nat Rev Mol Cell Biol 18: 495-506. https://doi.org/10.1038/nrm.2017.48
|
| [50] |
Huertas P (2010) DNA resection in eukaryotes: deciding how to fix the break. Nat Struct Mol Biol 17: 11-16. https://doi.org/10.1038/nsmb.1710
|
| [51] |
Vítor AC, Huertas P, Legube G, et al. (2020) Studying DNA Double-Strand Break Repair: An Ever-Growing Toolbox. Front Mol Biosci 7: 24. https://doi.org/10.3389/fmolb.2020.00024
|
| [52] |
Haghi M, Masoudi R, Ataellahi F, et al. (2025) Role of Tau and Amyloid-beta in autophagy gene dysregulation through oxidative stress. Tissue Cell 93: 102765. https://doi.org/10.1016/j.tice.2025.102765
|
| [53] |
Chiang MC, Nicol CJB, Yang YP, et al. (2025) The α-MG exhibits neuroprotective potential by reducing amyloid beta peptide-induced inflammation, oxidative stress, and tau aggregation in human neural stem cells. Brain Res 1852: 149506. https://doi.org/10.1016/j.brainres.2025.149506
|
| [54] |
Alrouji M, Alshammari MS, Tasqeeruddin S, et al. (2025) Interplay Between Aging and Tau Pathology in Alzheimer's Disease: Mechanisms and Translational Perspectives. Antioxidants (Basel) 14: 774. https://doi.org/10.3390/antiox14070774
|
| [55] |
Pala M, Yilmaz SG (2025) Circular RNAs, miRNAs, and Exosomes: Their Roles and Importance in Amyloid-Beta and Tau Pathologies in Alzheimer's Disease. Neural Plast 2025: 9581369. https://doi.org/10.1155/np/9581369
|
| [56] |
Hwang O (2013) Role of oxidative stress in Parkinson's disease. Exp Neurobiol 22: 11-17. https://doi.org/10.5607/en.2013.22.1.11
|
| [57] |
Dorszewska J, Kowalska M, Prendecki M, et al. (2021) Oxidative stress factors in Parkinson's disease. Neural Regen Res 16: 1383-1391. https://doi.org/10.4103/1673-5374.300980
|
| [58] |
Wang J, Liu M, Zhao J, et al. (2025) Oxidative stress and dysregulated long noncoding RNAs in the pathogenesis of Parkinson's disease. Biol Res 58: 7. https://doi.org/10.1186/s40659-025-00585-7
|
| [59] |
Zhang J, Liu T, Wu H, et al. (2025) Target oxidative stress-induced disulfidptosis: novel therapeutic avenues in Parkinson's disease. Mol Brain 18: 29. https://doi.org/10.1186/s13041-025-01200-2
|
| [60] |
Evans MD, Dizdaroglu M, Cooke MS (2004) Oxidative DNA damage and disease: induction, repair and significance. Mutat Res 567: 1-61. https://doi.org/10.1016/j.mrrev.2003.11.001
|
| [61] |
Redza-Dutordoir M, Averill-Bates DA (2016) Activation of apoptosis signalling pathways by reactive oxygen species. Biochim Biophys Acta 1863: 2977-2992. https://doi.org/10.1016/j.bbamcr.2016.09.012
|
| [62] |
Smeyne M, Smeyne RJ (2013) Glutathione metabolism and Parkinson's disease. Free Radic Biol Med 62: 13-25. https://doi.org/10.1016/j.freeradbiomed.2013.05.001
|
| [63] |
Long H, Zhu W, Wei L, et al. (2023) Iron homeostasis imbalance and ferroptosis in brain diseases. MedComm 4: e298. https://doi.org/10.1002/mco2.298
|
| [64] |
Cookson MR (2012) Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and oxidative stress and mitochondrial pathways. Cold Spring Harb Perspect Med 2: a009415. https://doi.org/10.1101/cshperspect.a009415
|
| [65] |
Chakrabarti S, Bisaglia M (2023) Oxidative Stress and Neuroinflammation in Parkinson's Disease: The Role of Dopamine Oxidation Products. Antioxidants (Basel) 12: 955. https://doi.org/10.3390/antiox12040955
|
| [66] |
Texier B, Prime M, Atamena D, et al. (2023) Mortalin/Hspa9 involvement and therapeutic perspective in Parkinson's disease. Neural Regen Res 18: 293-298. https://doi.org/10.4103/1673-5374.346487
|
| [67] |
Bonifati V, Rizzu P, van Baren MJ, et al. (2003) Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 299: 256-259. https://doi.org/10.1126/science.1077209
|
| [68] |
Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, et al. (2025) Oxidative Stress in Huntington's Disease. Biomolecules 15: 527. https://doi.org/10.3390/biom15040527
|
| [69] | Maiuri T, Mocle AJ, Hung CL, et al. (2017) Huntingtin is a scaffolding protein in the ATM oxidative DNA damage response complex. Hum Mol Genet 26: 395-406. https://doi.org/10.1093/hmg/ddw395 |
| [70] |
Paul BD, Snyder SH (2019) Impaired Redox Signaling in Huntington's Disease: Therapeutic Implications. Front Mol Neurosci 12: 68. https://doi.org/10.3389/fnmol.2019.00068
|
| [71] |
Gmitterová K, Heinemann U, Gawinecka J, et al. (2009) 8-OHdG in cerebrospinal fluid as a marker of oxidative stress in various neurodegenerative diseases. Neurodegener Dis 6: 263-269. https://doi.org/10.1159/000237221
|
| [72] |
Spies J, Covarrubias-Pinto A, Carcamo C, et al. (2023) Modulation of Synaptic Plasticity Genes Associated to DNA Damage in a Model of Huntington's Disease. Neurochem Res 48: 2093-2103. https://doi.org/10.1007/s11064-023-03889-w
|
| [73] |
Gao R, Chakraborty A, Geater C, et al. (2019) Mutant huntingtin impairs PNKP and ATXN3, disrupting DNA repair and transcription. Elife 8: e42988. https://doi.org/10.7554/eLife.42988
|
| [74] |
Kim MB, Lee J, Lee JY (2024) Targeting Mitochondrial Dysfunction for the Prevention and Treatment of Metabolic Disease by Bioactive Food Components. J Lipid Atheroscler 13: 306-327. https://doi.org/10.12997/jla.2024.13.3.306
|
| [75] |
Gibson GE, Starkov A, Blass JP, et al. (2010) Cause and consequence: mitochondrial dysfunction initiates and propagates neuronal dysfunction, neuronal death and behavioral abnormalities in age-associated neurodegenerative diseases. Biochim Biophys Acta 1802: 122-134. https://doi.org/10.1016/j.bbadis.2009.08.010
|
| [76] |
Rizzuto R, De Stefani D, Raffaello A, et al. (2012) Mitochondria as sensors and regulators of calcium signalling. Nat Rev Mol Cell Biol 13: 566-578. https://doi.org/10.1038/nrm3412
|
| [77] |
Zheng H, Xu Y, Liehn EA, et al. (2024) Vitamin C as Scavenger of Reactive Oxygen Species during Healing after Myocardial Infarction. Int J Mol Sci 25: 3114. https://doi.org/10.3390/ijms25063114
|
| [78] |
Carr AC, Maggini S (2017) Vitamin C and Immune Function. Nutrients 9: 1211. https://doi.org/10.3390/nu9111211
|
| [79] |
Murphy MP, Hartley RC (2018) Mitochondria as a therapeutic target for common pathologies. Nat Rev Drug Discov 17: 865-886. https://doi.org/10.1038/nrd.2018.174
|
| [80] |
McCarthy S, Somayajulu M, Sikorska M, et al. (2004) Paraquat induces oxidative stress and neuronal cell death; neuroprotection by water-soluble Coenzyme Q10. Toxicol Appl Pharmacol 201: 21-31. https://doi.org/10.1016/j.taap.2004.04.019
|
| [81] |
Singh U, Devaraj S, Jialal I (2007) Coenzyme Q10 supplementation and heart failure. Nutr Rev 65: 286-293. https://doi.org/10.1111/j.1753-4887.2007.tb00306.x
|
| [82] |
Sohet FM, Neyrinck AM, Pachikian BD, et al. (2009) Coenzyme Q10 supplementation lowers hepatic oxidative stress and inflammation associated with diet-induced obesity in mice. Biochem Pharmacol 78: 1391-1400. https://doi.org/10.1016/j.bcp.2009.07.008
|
| [83] |
Wilk A, Hayat F, Cunningham R, et al. (2020) Extracellular NAD+ enhances PARP-dependent DNA repair capacity independently of CD73 activity. Sci Rep 10: 651. https://doi.org/10.1038/s41598-020-57506-9
|
| [84] |
Li T, Yang Y, Qi H, et al. (2023) CRISPR/Cas9 therapeutics: progress and prospects. Signal Transduct Target Ther 8: 36. https://doi.org/10.1038/s41392-023-01309-7
|
| [85] |
Fu YW, Dai XY, Wang WT, et al. (2021) Dynamics and competition of CRISPR-Cas9 ribonucleoproteins and AAV donor-mediated NHEJ, MMEJ and HDR editing. Nucleic Acids Res 49: 969-985. https://doi.org/10.1093/nar/gkaa1251
|
| [86] |
Shen S, Loh TJ, Shen H, et al. (2017) CRISPR as a strong gene editing tool. BMB Rep 50: 20-24. https://doi.org/10.5483/BMBRep.2017.50.1.128
|
| [87] |
Nitti M, Marengo B, Furfaro AL, et al. (2022) Hormesis and Oxidative Distress: Pathophysiology of Reactive Oxygen Species and the Open Question of Antioxidant Modulation and Supplementation. Antioxidants (Basel) 11: 1613. https://doi.org/10.3390/antiox11081613
|
| [88] |
Pradeepkiran JA, Islam MA, Sehar U, et al. (2025) Impact of diet and exercise on mitochondrial quality and mitophagy in Alzheimer's disease. Ageing Res Rev 108: 102734. https://doi.org/10.1016/j.arr.2025.102734
|
| [89] |
Yang H, Ren S, Yu S, et al. (2020) Methods Favoring Homology-Directed Repair Choice in Response to CRISPR/Cas9 Induced-Double Strand Breaks. Int J Mol Sci 21: 6461. https://doi.org/10.3390/ijms21186461
|
| [90] |
Kasai H (2003) A new automated method to analyze urinary 8-hydroxydeoxyguanosine by a high-performance liquid chromatography-electrochemical detector system. J Radiat Res 44: 185-189. https://doi.org/10.1269/jrr.44.185
|
| [91] | Valavanidis A, Vlachogianni T, Fiotakis C (2009) 8-hydroxy-2′-deoxyguanosine (8-OHdG): A critical biomarker of oxidative stress and carcinogenesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 27: 120-139. https://doi.org/10.1080/10590500902885684 |
| [92] |
Deng S, Xie H, Xie B (2025) Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases. Stem Cell Res Ther 16: 167. https://doi.org/10.1186/s13287-025-04285-7
|
| [93] |
Hossain MS, Hussain MH (2025) Multi-Target Drug Design in Alzheimer's Disease Treatment: Emerging Technologies, Advantages, Challenges, and Limitations. Pharmacol Res Perspect 13: e70131. https://doi.org/10.1002/prp2.70131
|
| [94] |
Moretti EH, Lin ALY, Peruzzotti-Jametti L, et al. (2025) Neural Stem Cell-Derived Extracellular Vesicles for Advanced Neural Repair. J Neurochem 169: e70170. https://doi.org/10.1111/jnc.70170
|
| [95] |
Jeong H, Kim OJ, Oh SH, et al. (2021) Extracellular Vesicles Released from Neprilysin Gene-Modified Human Umbilical Cord-Derived Mesenchymal Stem Cell Enhance Therapeutic Effects in an Alzheimer's Disease Animal Model. Stem Cells Int 2021: 5548630. https://doi.org/10.1155/2021/5548630
|
| [96] |
Cummings JL, Teunissen CE, Fiske BK, et al. (2025) Biomarker-guided decision making in clinical drug development for neurodegenerative disorders. Nat Rev Drug Discov 24: 589-609. https://doi.org/10.1038/s41573-025-01165-w
|
| [97] |
Sanadgol N, Abedi M, Hashemzaei M, et al. (2025) Exosomes as nanocarriers for brain-targeted delivery of therapeutic nucleic acids: advances and challenges. J Nanobiotechnology 23: 453. https://doi.org/10.1186/s12951-025-03528-2
|
| [98] |
Yang YP, Nicol CJB, Chiang MC (2025) A Review of the Neuroprotective Properties of Exosomes Derived from Stem Cells and Exosome-Coated Nanoparticles for Treating Neurodegenerative Diseases and Stroke. Int J Mol Sci 26: 3915. https://doi.org/10.3390/ijms26083915
|
| [99] |
Dehghani S, Ocakcı O, Hatipoglu PT, et al. (2025) Exosomes as Biomarkers and Therapeutic Agents in Neurodegenerative Diseases: Current Insights and Future Directions. Mol Neurobiol 62: 9190-9215. https://doi.org/10.1007/s12035-025-04825-5
|
| [100] |
Kumari S, Kamiya A, Karnik SS, et al. (2025) Novel Gene Therapy Approaches for Targeting Neurodegenerative Disorders: Focusing on Delivering Neurotrophic Genes. Mol Neurobiol 62: 386-411. https://doi.org/10.1007/s12035-024-04260-y
|
| [101] |
Allen SJ, Watson JJ, Shoemark DK, et al. (2013) GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacol Ther 138: 155-175. https://doi.org/10.1016/j.pharmthera.2013.01.004
|