You are viewing a javascript disabled version of the site. Please enable Javascript for this site to function properly.
Go to headerGo to navigationGo to searchGo to contentsGo to footer
In content section. Select this link to jump to navigation

Pro-Oxidants and Antioxidants Imbalance in Alzheimer’s Disease

Alzheimer’s disease (AD) is an irreversible progressive neurological disorder that is manifested by gradual deterioration of cognitive reserve and impairment of activities in daily living. There are 50 million AD patients globally and this number will likely be doubled by 2050 [1]. Two dominant hypotheses, the amyloid-β (Aβ) cascade [2] and the tau propagation [3], were considered causally linked to the progression of AD. However, a disease-modifying drug for AD is not available, although significant number of clinical trials based on Aβ cascade and tau phosphorylation are already being conducted [4, 5]. A recent report based on a meta-analysis on the effect of anti-Aβ drugs on brain revealed that β-secretase inhibitors like verubecestat accelerated hippocampal and whole brain atrophy compared to changes observed after placebo administration [6].

Therapeutic development is urgently required based on potential role of oxidative stress (OS) in AD as early causal process [7–9]. OS is identified as an imbalance in the over production of harmful reactive oxygen species (ROS) by prooxidants and neutralization of these ROS by antioxidants [9]. Recently, significant depletion of master antioxidant, glutathione (GSH), as well as elevation of prooxidant (iron, etc.) in the hippocampus region of AD patients were reported [10–12]. A transgenic AD mice model study also showed that GSH depletion precedes amyloid plaque formation [13]. Significant depletion of plasma GSH from mild cognitive impairment (MCI) subjects has also been reported in a longitudinal study [14, 15].

This supplemental issue of the Journal of Alzheimers Disease highlights OS-based AD research from various laboratories around the globe. The salient extracts of these research works are presented below:

Syed and co-workers have indicated the molecular aspects of aging and age-related disorders, especially in AD. They have emphasized the significance of metal ions, protein ligands, and the oligomerization state of amyloid-β protein precursor (AβPP) in the proteolytic processing of AβPP [16].

Bian and coworkers investigated the potential therapeutic effects of long-term administration of Tocovid, a novel vitamin E mixture, in a transgenic AD mice model. Tocovid treatment significantly improved motor and memory deficits in the AD mice and has potential neuroprotective role in combating AD. Dietary intake of antioxidant supplements is associated with a reduced risk of AD [17]. The meta-analysis by Zhao et al. investigated association between antioxidants and the risk of dementia from seventeen research studies with a total of 98,264 participants. The striking point in this meta-analysis indicated that high antioxidant intake significantly reduced the incidence of AD [18]. Soto-Mercado and coworkers analyzed the therapeutic potential of a combination of epigallocatechin-3-gallate (EGCG) and melatonin (MT) in a three-dimensional in vitro model of familial AD with a mutation in the presenilin-1 gene. The combination of EGCG and MT treatment effectively reduced these pathological markers more efficiently than individual treatments. This study highlights the combined therapy of EGCG and MT which holds a therapeutic promise for familial AD due to inherent antioxidant capability [19].

Majd and coworkers reported the protective effects of coenzyme Q10 (CoQ10) and high-intensity interval training (HIIT), both individually and in combination, on Aβ-induced AD rats. Pretreatment with CoQ10, HIIT, or the combination of both significantly improved cognitive functions and mitigated OS [20].

Yang and coworkers assessed the efficacy of docosapentaenoic acid and/or eicosapentaenoic acid supplements in MCI patients. The meta-analysis revealed that supplementation with n-3 polyunsaturated fatty acids had a positive impact on global cognition compared to a study with placebo. However, no significant differences were observed in language fluency, executive functions, and depression between the intervention and placebo arms [21].

Qaiser and coworkers reported the protective functions of the thioredoxin system [thioredoxin (Trx), thioredoxin reductase (TrxR)], and nicotinamide adenine dinucleotide phosphate in the context of AD-induced damage. Trx shields cells from OS, while TrxR is essential for detoxifying ROS in the brain [22].

Mamelak has presented potential strategies to address early events in AD involving microvasculature deterioration in the hippocampus that precedes Aβ deposition. NADPH oxidase (NOX) activity is elevated with age and contributes to OS and inflammation. Elevated NOX activity in the AD brain correlates inversely with cognitive ability. It is also indicated that apocynin, a NOX inhibitor, and sodium oxybate have potential in mitigating NOX-related damage [23].

Hambali and coworkers investigated the antioxidative and anti-neuroinflammatory effects of Centella asiatica and its triterpene fractions on lipopolysaccharide-induced microglial cells for AD management. Potential therapeutic value of Centella asiatica is highlighted through reduction of OS and neuroinflammation associated with AD [24].

Zhou and coworkers have proposed a computational system pharmacology workflow to uncover the OS triggering AD as well as the potential therapeutic targets and neuroprotective drugs. They have integrated preliminary screening data of pharmacological experiments with drug-targeted interactions. This research group concluded that glucocorticoid receptor gene, sex hormone binding globulin, estrogen receptor 1, etc., are promising therapeutic targets, and several drugs may be repurposed from the perspective of OS in AD [25].

Lazarova and coworkers reported significant improvement of working memory in rats with induced dementia due to Marrubium vulgare extract. This extract also reduce acetylcholinesterase activity in the hippocampus by 20% and alleviate OS in the cortex. These findings suggest that Marrubium vulgare water extract may have a working memory-preserving effect [26].

Timothy Daly emphasized the importance of maintaining diversity in drug development for AD research. The article suggested an option to enhance the flow of information from randomized clinical trials. It focuses on factors like adherence to reporting guidelines, and increased precision in the hippocampus involving GSH–iron tandem biomarker [27].

Accumulating evidence from clinical studies lead us to the converging idea that enrichment of brain GSH is the way forward through candidates with superior bioavailability and efficacy. Recently developed innovative platform, GLUTASCAN will be employed to determine GSH enrichment in the brain non-invasively [28]. It is my hope that multicenter studies from various continents could help in inhibiting the progression of the disease through efficient GSH replenishment in the brain of MCI population. The outcome from singular use of iron chelators (e.g. Deferiprone) in randomized clinical trials (RCT) involving Prodromal and mild AD patients for cognitive preservation is awaited. The efficacy of the RCT can be monitored from the analysis of plasma GSH and serum iron levels from the trial participants [14]. These valuable data can be kept in a dedicated database called SWADESH [29].

ACKNOWLEDGMENTS

I am thankful to Professor George Perry, Editor-in-Chief of the Journal of Alzheimers Disease, for permitting me to be the Guest Editor for this special issue. Thanks to Ms. Beth Kumar, Managing Editor of JAD, for her support in the entire review process. Thanks to all the contributing authors for their latest work and reviewers for giving their valuable time. Additionally, I extend my thanks to Prof. Joseph C. Maroon, MD, Department of Neurosurgery, University of Pittsburgh Medical Center, USA for collaboration. Thanks to Mrs. Ratna Mandal (MA, Psychology) for comment and discussion. Appreciation for Ms. Avantika Samkaria (Researcher, NINS lab) for editorial support. Thanks to Indo-Australian Biotechnology grant (BT/Indo-Aus/10/31/2016/ to PKM).

REFERENCES

[1] 

Behl T , Kaur D , Sehgal A , Singla RK , Makeen HA , Albratty M , Alhazmi HA , Meraya AM , Bungau S ((2022) ) Therapeutic insights elaborating the potential of retinoids in Alzheimer’s disease. Front Pharmacol 13: , 976799.

[2] 

Hardy J ((1997) ) Amyloid, the presenilins and Alzheimer’s disease. Trends Neurosci 20: , 154–159.

[3] 

Kosik KS , Joachim CL , Selkoe DJ ((1986) ) Microtubule-associated protein tau (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proc Natl Acad Sci U S A 83: , 4044–4048.

[4] 

Mandal PK , Samkaria A , Maroon JC ((2021) ) AD hypotheses and suggested clinical trials. ACS Chem Neurosci 12: , 3968–3971.

[5] 

Casey DA , Antimisiaris D , O’Brien J ((2010) ) Drugs for Alzheimer’s disease: Are they effective? P T 35: , 208–211.

[6] 

Alves F , Kalinowski P , Ayton S ((2023) ) Accelerated brain volume loss caused by anti-beta-amyloid drugs: A systematic review and meta-analysis. Neurology 100: ,e2114–e2124.

[7] 

Nunomura A , Perry G , Aliev G , Hirai K , Takeda A , Balraj EK , Jones PK , Ghanbari H , Wataya T , Shimohama S , Chiba S , Atwood CS , Petersen RB , Smith MA ((2001) ) Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol 60: , 759–767.

[8] 

Markesbery WR ((1997) ) Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med 23: , 134–147.

[9] 

Roy RG , Mandal PK , Maroon JC ((2023) ) Oxidative stress occurs prior to amyloid Aβ plaque formation and tau phosphorylation in Alzheimer’s disease: Role of glutathione and metal ions. ACS Chem Neurosci 14: , 2944–2954.

[10] 

Mandal PK , Roy RG , Samkaria A ((2022) ) Oxidative stress: Glutathione and its potential to protect methionine-35 of Aβ peptide from oxidation. ACS Omega 7: , 27052–27061.

[11] 

Mandal PK , Goel A , Bush AI , Punjabi K , Joon S , Mishra R , Tripathi M , Garg A , Kumar NK , Sharma P , Shukla D , Ayton SJ , Fazlollahi A , Maroon JC , Dwivedi D , Samkaria A , Sandal K , Megha K , Shandilya S ((2022) ) Hippocampal glutathione depletion with enhanced iron level in patients with mild cognitive impairment and Alzheimer’s disease compared with healthy elderly participants. Brain Commun 4: , fcac215.

[12] 

Mandal PK , Saharan S , Tripathi M , Murari G ((2015) ) Brain glutathione levels–a novel biomarker for mild cognitive impairment and Alzheimer’s disease. Biol Psychiatry 78: , 702–710.

[13] 

Pontrello CG , McWhirt JM , Glabe CG , Brewer GJ ((2022) ) Age-related oxidative redox and metabolic changes precede intraneuronal amyloid-β accumulation and plaque deposition in a transgenic Alzheimer’s disease mouse model. J Alzheimers Dis 90: , 1501–1521.

[14] 

Mandal PK , Maroon JC , Garg A , Arora NK , Bansal R , Kaushik A , Samkaria A , Kumaran G , Arora Y ((2023) ) Blood biomarkers in Alzheimer’s disease. ACS Chem Neurosci 14: , 3975–3978.

[15] 

Lin CH , Lane HY ((2021) ) Plasma glutathione levels decreased with cognitive decline among people with mild cognitive impairment (MCI): A two-year prospective study. Antioxidants (Basel) 10: , 1839.

[16] 

Syed RA , Hayat M , Qaiser H , Uzair M , Al-Regaiey K , Khallaf R , Kaleem I , Bashir S ((2024) ) Aging-related protein alterations in the brain. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-230801.

[17] 

Bian Z , Yu H , Hu X , Bian Y , Sun H , Tadokoro K , Takemoto M , Yunoki T , Nakano Y , Fukui Y , Morihara R , Abe K , Yamashita T ((2024) ) Tocovid attenuated oxidative stress and cognitive decline by inhibiting amyloid-β-induced NOX2 activation in Alzheimer’s disease mice. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-220761.

[18] 

Zhao R , Han X , Jiang S , Zhao W , Liu J , Zhang H , Mao X , Zhang M , Lei L , You H ((2024) ) Association of dietary and supplement intake of antioxidants with risk of dementia: A meta-analysis of cohort studies. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-220909.

[19] 

Soto-Mercado V , Mendivil-Perez M , Velez-Pardo C , Jimenez-Del-Rio M ((2024) ) Neuroprotective effect of combined treatment with epigallocatechin 3-gallate and melatonin on familial Alzheimer’s disease PSEN1 E280A cerebral spheroids derived from menstrual mesenchymal stromal cells. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-220903.

[20] 

Puoyan-Majd S , Parnow A , Rashno M , Heidarimoghadam R , Komaki A ((2024) ) The protective effects of high-intensity interval training combined with Q10 supplementation on learning and memory impairments in male rats with amyloid-beta-induced Alzheimer’s disease. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-230096.

[21] 

Yang L , Zhao F , Sun Y , Wang Z , Li Q , Wang H , Lu Y ((2024) ) N-3 polyunsaturated fatty acids in elderly with mild cognitive impairment: A systemic review and meta-analysis. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-220863.

[22] 

Qaiser H , Uzair M , Al-Regaiey K , Rafiq S , Arshad M , Yoo WK , Arain OZ , Kaleem I , Abualait T , Wang L , Wang R , Bashir S ((2024) ) Role of thioredoxin system in regulating cellular redox status in Alzheimer’s disease. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-230394.

[23] 

Mamelak M ((2024) ) The Alzheimer’s disease brain, its microvasculature, and NADPH oxidase. J Alzheimers Dis 99: (s1):10.3233/JAD-230415.

[24] 

Hambali A , Jusril NA , Md Hashim NF , Abd Manan N , Adam SK , Mehat MZ , Adenan MI , Stanslas J , Abdul Hamid H ((2024) ) The standardized extract of Centella asiatica and its fractions exert antioxidative and anti-neuroinflammatory effects on microglial cells and regulate the Nrf2/HO-1 signaling pathway. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-230875.

[25] 

Zhou M , Jiao Q , Wu Z , Li W , Liu G , Wang R , Tang Y ((2024) ) Uncovering the oxidative stress mechanisms and targets in Alzheimer’s disease by integrating phenotypic screening data and polypharmacology networks. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-220727.

[26] 

Lazarova MI , Tsvetanova FR , Georgieva AP , Stefanovaa MO , Uzunovaa DN , Denevb PN , Tashevac KN ((2024) ) Marrubium vulgare extract improves spatial working memory, oxidative stress damage and cholinergic dysfunction in scopolamine-treated rats. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-231011.

[27] 

Daly T ((2024) ) Improving clinical trials of antioxidants in Alzheimer’s disease. J Alzheimers Dis 99: (s1)doi:10.3233/JAD-230308.

[28] 

Arora Y , Samkaria A , Maroon JC , Mandal PK A non-invasive imaging platform for longitudinal monitoring and quantification of brain glutathione for clinical trial. Program No. NANO03.08. 2023 Neuroscience Meeting Planner. Society for Neuroscience, Washington, D.C.

[29] 

Mandal PK , Jindal K , Roy S , Arora Y , Sharma S , Joon S , Goel A , Ahasan Z , Maroon JC , Singh K , Sandal K , Tripathi M , Sharma P , Samkaria A , Gaur S , Shandilya S ((2023) ) SWADESH: A multimodal multi-disease brain imaging and neuropsychological database and data analytics platform. Front Neurol 14: , 1258116.