Nrf2-Mediated Signaling as a Therapeutic Target in Alzheimer’s Disease
REVIEW ARTICLE

Nrf2-Mediated Signaling as a Therapeutic Target in Alzheimer’s Disease

The Open Neurology Journal 21 Jun 2024 REVIEW ARTICLE DOI: 10.2174/011874205X319474240611070113

Abstract

Nrf2 is a major transcriptional factor that controls gene expression in normal health and pathological conditions. It regulates and controls the manifestation of various major elements of oxidative stress, neuro-inflammation, autophagy, and mitochondrial bioenergetics in the centre and periphery. Besides, Nrf2 activity is also controlled at various stages, such as protein degradation, transcription, and post-translation. Growing evidence suggests changes in the levels of Nrf2 in degenerative disorders, such as Alzheimer's disease (AD). AD is characterised by elevated oxidative stress, neuro-inflammation, synaptic dysfunction, and proteinopathies, which lead to the progressive loss of memory. A decrease in the expression of Nrf2 and its downstream target genes was identified in AD. Recent studies have shown that Nrf2 interferes with various main pathogenic processes in AD, including amyloid and tau pathologies. The current review focuses on brief in the regulation of Nrf2 and the association of Nrf2 with AD, along with the currently available Nrf2 activators.

Keywords: Nrf2, Alzheimer’s disease, Keap1, Reactive oxygen species, Amyloid, DNA.

1. INTRODUCTION

Alzheimer’s disease (AD) is a progressive neurode- generative disorder that displays amyloid [1] and tau proteinopathies [2]. Apart from these proteinopathies, the classic theme in various diseases, including AD, is oxidative stress. The disparity between the generation of free radicals and the body's natural antioxidant defense mechanism is known as oxidative stress [3, 4]. In mitochondria, peroxisomes, and the endoplasmic reti- culum, Reactive Oxygen Species (ROS) are essential for various endogenous metabolic reactions and molecular signaling pathways [3, 5], and the excess production of ROS damages biological elements, such as DNA, RNA, and proteins [2, 6, 7].

The unregulated redox balance is the collective result of a variety of biological processes, including metal homeostasis and endogenous and exogenous stress [3, 4]. CNS is more prone to oxidative stress due to its high metabolic rate and high ascorbate levels and transition metals, which act as pro-oxidants [8, 9]. Depletion of antioxidants increases the vulnerability to lipid peroxidation, genotoxicity, protein oxidation, DNA and RNA oxidation, and mitochondrial depolarization. Oxidative stress is also produced by excitotoxicity and plasticity [8, 9], which leads to the activation of various transcription factors [10]. The antioxidant system in the body generates various enzymes that scavenge these free radicals and offer cytoprotection. One such defence system is the Nrf2 signaling system [11, 12].

2. STRUCTURE AND FUNCTION OF THE Nrf2

Nrf2 (Nuclear Factor-Erythroid Factor 2) is a 66 KDa protein molecule. Based on molecular architecture, the Nrf2 is categorised into 7 conserved Neh (Nrf2-ECH) domains, namely Neh1-Neh7 [13]. Neh1 region consists of a regular leucine zipper motif, and the Neh2 domain interrelates with Keap1 via ETGE and DLG motifs [14]. This Neh2 domain is required for the stability of Nrf2. While Neh3, Neh4, and Neh5 are essential for the transcription of Nrf2 target genes [15]. Furthermore, Neh6 allows Keap1-independent regulation, caused by GSK3β phosphorylation [16] and the Neh7 domain, associated with the retinoic X receptor a, which aids in the repression of gene transcription of Nrf2 [17].

Nrf2 acts as a master regulator of antioxidant response via increasing antioxidant enzymes, metabolism of xenobiotics, elimination of damaged proteins, suppression of inflammation and metabolic damage, and influencing various growth regulatory factors [18].

3. REGULATION OF Nrf2

There are three major regulatory pathways, where Nrf2 gets degraded via ubiquitin. The first important one is the use of Keap1; second, the use of glycogen synthase (GSK3); and lastly, the β-TrCP-dependent cul-1-dependent ligase complex [18].

3.1. Keap-1 Dependent Regulation

Kelch, like ECH-associated protein 1 (Keap 1), is known to be the new Nrf2-binding protein. It consists of three main domains that interact with the proteins: the BTB domain in the N-terminal region, the IV region in the centre, and Kelch repeats in the C terminal. BTB mediates binding to Cul3 of Nrf2 ubiquitin ligase [19]. The activation of Keap 1 is required for homeostasis, and the depletion of Keap 1 induces cell death [18]. Under physiological conditions, the activity of Nrf2 is inhibited in the cytoplasm by sequestration and degradation of Nrf2 via Keap1. The Nrf2 Neh2 region binds to Keap1 at the area between the BTB and Kelch repeat domains [19]. This interaction acts as an adaptor for the Cul3/Rbx1 E3 ubiquitin ligase complex, which causes the ubiquitination of Nrf2 [20, 21]. Moreover, Keap1 sequesters Nrf2 to the mitochondria via a ternary complex along with the PGAM5 protein [22, 23]. This sequestration is known to increase the activation of Nrf2 in response to stressors [22].

Fig. (1).

Schematic illustration of Nrf2-mediated regulation in neuronal cells (negative and positive regulation). The image is adapted from Tejo and Quintanilla, 2021. CC BY 4.0 [https://creativecommons.org/licenses/by/4.0/].

3.2. Regulation of Nrf2 via GSK-3

GSK-3β regulates a number of targets in many cell signalling pathways, and it is not astonishing that this enzyme is also involved in the control of Nrf-2 (Fig. 1). It is a constitutively active kinase whose action is nullified by insulin [24]. Increased insulin signaling triggers protein kinase B/Akt through phosphorylation. Consecutively, Akt inhibits GSK-3β via phosphorylation of Ser9. This kinase exerts negative regulation on Nrf-2 by modulating its distribution. Increased oxidative stress conditions cause down-regulation of Akt, stimulation of GSK-3β, and translocation of Nrf-2 from the nucleus to cytoplasm, thus, restricting the antioxidant response [25, 26]. The GSK-3β inhibition results in the elevation of transcriptional activity of Nrf-2 because of nuclear accumulation, thus signifying the fundamental role of the GSK-3β in Nrf-2-ARE down-regulation after oxidative injury [22, 25]. The main mechanism by which GSK-3β mediates Nrf-2 is known to comprise Fyn, a tyrosine kinase that is phosphorylated by activated GSK-3β and causes the nuclear localisation of Fyn. This activated Fyn further causes phosphorylation of tyrosine 568 in nuclear Nrf-2, triggering the inhibition of Nrf-2 protective gene transcription [26, 27].

3.3. Keap 1 Independent Regulation via β-TrCP

Apart from the β-TrCP, the PI3K/Akt pathway also plays a role in Nrf2 induction, independent of KEAP1 [28]. The PI3K/Akt signaling stimulates Nrf2 through the GSk-3β inhibition, which causes phosphorylation of Nrf2, thus causing β-TrCP to consecutively ubiquitinate Nrf2 for consequent proteasomal degradation [25, 29]. The β-TrCP serves as a substratum receptor inside the SCF-ubiquitin ligase β-TrCP and takes part in the ubiquitination of Nrf2 [30]. β-TrCP recognises its substrates by directly attaching to phosphorylated destructed motifs, and this pattern represents the GSK-3β target elements; thus, a variety of its target proteins are known to be processed by β-TrCP.

4. Nrf2 AND ITS ROLE IN AD

AD is a neurodegenerative disorder predominantly exemplified by cognitive dysfunction. Pathologically, the disease exhibits β-amyloid plaques [31] and neurofibrillary tangles [32] as pathological hallmarks. Besides, it also causes oxidative stress [33, 34], neuroinflammation [35, 36], microglial activation [36], astrogliosis [37], and synaptic loss [33, 38]. The amyloid-β proteins of the amyloid plaques and the neurofibrillary tangles disrupt homeostasis and lead to neuronal death and neurodegeneration. The prevalence of AD is known to increase logarithmically with age. The amyloid pathogenesis and tauopathies alter the mitochondrial biogenesis and bioenergetics, which eventually lead to oxidative stress [18]. Post-mortem AD brain studies revealed the presence of high levels of lipid oxidation products, oxidised DNA bases, and carbonyls in proteins [39], which suggests the presence of oxidation of macromolecules in AD brains.

Nrf2 is uniformly found in a different range of tissues and cells, with varying amounts of protein present in particular cell types that are primarily in charge of homeostatic adjustments. Astrocytes and microglia, for instance, play a particularly important role in Nrf2 in the brain since their expression levels are higher than those of neurons [40]. Astrocytes are highly responsive to Nrf2 activation, while the Nrf2 modulates microglial dynamics, thereby reducing the inflammatory mediators and increasing the anti-inflammatory markers [41].

Nrf2 signaling has been widely studied in AD animal models over the last few decades, indicating its downregulation. Nrf2 serves as a main mediator in protecting against oxidative insults. Previous research has shown that the Nrf2/ARE pathway was modulated in AD [34], and the levels of Nrf2 were decreased in the human AD hippocampus [12]. The absence of Keap 1 activated Nrf2 and induced enzymes in phase II reactions in neuronal cells, thereby amplifying resistance to oxidative insults [42]. It has been observed that Nrf2 can hinder AD, and the agents that stimulate Nrf2 have shown neuroprotective activities in preclinical models [43], while the deficiency of Nrf2 exacerbates memory deficits [44]. Furthermore, in the rat model of AD, the Nrf2/ARE levels were found to be downregulated, and the oxidative stress markers, such as the SOD and catalase levels, were decreased. Also, the MDA levels were increased in the cortex and hippocampus [34, 35].

In another study, GSK-3β inhibition in mice was known to raise nuclear Nrf2 in the cortex [45]. Glial activation was found in AD, leading to elevated inflammatory markers, such as IL-1β, TNF-α, and COX2 [34]. Besides, NF-κB was found to be associated near the amyloid plaques. It was also found that the discrepancy between Nrf2 and NF-κB could be a key part of AD pathology. While the NF-κB activation causes the production of pro-inflammatory markers like various cytokines and chemokines that further increase the amyloid-β production and microglia activation [46, 47], NF-κB is associated with β-secretase enzyme expression, as this enzyme possesses a binding site with NF-κB, thus, associating with the inflammatory response [40]. In another study, the molecular association between Nrf2 and inflammation was studied. It was found that Nrf2 restricted lipopoly- saccharide-induced transcriptional stimulation of proinflammatory cytokines like IL-6 and IL-1β. It was also demonstrated that Nrf2-mediated suppression is independent of oxidative stress and Nrf2-binding pattern. Contrary to the widely accepted view, these studies reported that Nrf2 is the upstream controller of cytokines and the basis for the Nrf2-mediated anti-inflammatory approach [48].

4.1. Targeting Nrf2 for AD Pathology

Multi-drug targeted therapy in AD came into existence after the failure of many clinical trials for AD. Target-specific options can be achieved by targeting multiple pathological pathways. One such pathway is the Nrf2 signaling pathway. As discussed above, Nrf2 can significantly offer neuroprotection in AD. So, treatment with Nrf2 activating agents can mitigate AD. Various Nrf2 agents/compounds are listed in Table 1.

Table 1.
Literature representing the role of Nrf2 in various preclinical and clinical models.
S.No. Compound/molecule Models Uses Reference
1. Benincasa hispida Rat model of AD Antioxidant, anti-inflammatory [34]
2. Carnosic acid Transgenic mice model Antioxidant, anti-microbial, anti-inflammatory [49]
3. Astaxanthin 3xTg mice model Antioxidant, blood pressure regulation,
Anti-hyperlipidaemic
[50]
4. DMF Transgenic model Anti-inflammatory [41]
5. Lycopene Transgenic mice model Antioxidant, anti-apoptotic [51]
6. Phenols (resveratrol, curcumin, gastrodin) Clinical trials and Rodent models Antioxidant, anti-inflammatory NCT00678431,
NCT00164749 [52].
7. Ebselen 3xTg mice Antioxidant, suppression of mitogen-activated kinases [53]
8. Tideglusib Clinical trial Antioxidant,
anti-inflammatory
NCT01350362
9. Flavonoids (Fisetin, Hesperidin anthocyanins, Apigenin) Mice models Antioxidant,
anti-inflammatory
[54-57]
.
10. Sulforaphane Clinical trial Antioxidant,
anti-inflammatory, and
anti-apoptotic properties
NCT04213391

CONCLUSION

In this review, we discussed various studies that highlighted the neuroprotective role of the Nrf2 pathway in AD. Nrf2 is a conventional example of targeting oxidative stress, neuro-inflammation, and associated underlying disease pathology. Furthermore, Nrf2 is crucial to redox balance and the control of inflammation, and an enhancement in the effect of Nrf2 could be a possible therapeutic approach for AD. Evidence from animal models revealed that a pharmacological therapy to slow the progression of AD might be possible through Nrf2 activators. Nrf2 activating compounds/molecules are currently approved by the FDA for various neurodegenerative conditions, and some molecules, such as resveratrol, curcumin, and tideglusib (Table 1), are under clinical trials. In summary, the upregulation of Nrf2 leads to the synthesis of antioxidant and anti-inflammatory elements, which improves AD pathogenesis.

AUTHORS’ CONTRIBUTION

All authors have accepted responsibility for the manuscript's content and consented to its submission. They have meticulously reviewed all results and unanimously approved the final version of the manuscript.

LIST OF ABBREVIATIONS

COX-2 = Cyclooxygenase-2
IL-1 = Interleukin 1
NF-kB = Nuclear factor kappa-light-chain- enhancer of activated B cells
PI3K/AKT = Phosphatidylinositol 3-kinase/protein kinase B
TNF = Tumor Necrosis Factor

CONSENT FOR PUBLICATION

Not applicable.

FUNDING

None.

CONFLICT OF INTEREST

The authors declare no conflict of interest, financial or otherwise.

ACKNOWLEDGEMENTS

The authors would like to thank the University of Botswana for providing the required facilities.

REFERENCES

1
Schweighauser M, Arseni D, Bacioglu M, et al. Age-dependent formation of TMEM106B amyloid filaments in human brains. Nature 2022; 605(7909): 310-4.
2
Bitra VR, Challa SR, Adiukwu PC, Rapaka D. Tau trajectory in Alzheimer’s disease: Evidence from the connectome-based computational models. Brain Res Bull 2023; 203: 110777.
3
Wang H, Liu K, Geng M, et al. RXRα inhibits the NRF2-ARE signaling pathway through a direct interaction with the Neh7 domain of NRF2. Cancer Res 2013; 73(10): 3097-108.
4
Alkadi H. A review on free radicals and antioxidants. Infect Disord Drug Targets 2020; 20(1): 16-26.
5
Zhang B, Pan C, Feng C, et al. Role of mitochondrial reactive oxygen species in homeostasis regulation. Redox Rep 2022; 27(1): 45-52.
6
Jomova K, Raptova R, Alomar SY, et al. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: Chronic diseases and aging. Arch Toxicol 2023; 97(10): 2499-574.
7
Peña-Bautista C, Baquero M, Vento M, Cháfer-Pericás C. Free radicals in Alzheimer’s disease: Lipid peroxidation biomarkers. Clin Chim Acta 2019; 491: 85-90.
8
Ashleigh T, Swerdlow RH, Beal MF. The role of mitochondrial dysfunction in Alzheimer’s disease pathogenesis. Alzheimers Dement 2023; 19(1): 333-42.
9
Rahal A, Kumar A, Singh V, et al. Oxidative stress, prooxidants, and antioxidants: the interplay. BioMed Res Int 2014; 2014: 1-19.
10
Priya Dharshini LC, Vishnupriya S, Sakthivel KM, Rasmi RR. Oxidative stress responsive transcription factors in cellular signalling transduction mechanisms. Cell Signal 2020; 72: 109670.
11
Prasad KN. Simultaneous activation of Nrf2 and elevation of antioxidant compounds for reducing oxidative stress and chronic inflammation in human Alzheimer’s disease. Mech Ageing Dev 2016; 153(153): 41-7.
12
Zgorzynska E, Dziedzic B, Walczewska A. An Overview of the Nrf2/ARE pathway and its role in neurodegenerative diseases. Int J Mol Sci 2021; 22(17): 9592.
13
Moi P, Chan K, Asunis I, Cao A, Kan YW. Isolation of NF-E2-related factor 2 (Nrf2), a NF-E2-like basic leucine zipper transcriptional activator that binds to the tandem NF-E2/AP1 repeat of the beta-globin locus control region. Proc Natl Acad Sci 1994; 91(21): 9926-30.
14
Ulasov AV, Rosenkranz AA, Georgiev GP, Sobolev AS. Nrf2/Keap1/ARE signaling: Towards specific regulation. Life Sci 2022; 291: 120111.
15
Nioi P, Nguyen T, Sherratt PJ, Pickett CB. The carboxy-terminal Neh3 domain of Nrf2 is required for transcriptional activation. Mol Cell Biol 2005; 25(24): 10895-906.
16
Cuadrado A. Structural and functional characterization of Nrf2 degradation by glycogen synthase kinase 3/β-TrCP. Free Radic Biol Med 2015; 88(Pt B): 147-57.
17
Wang CH, Wu SB, Wu YT, Wei YH. Oxidative stress response elicited by mitochondrial dysfunction: Implication in the pathophysiology of aging. Exp Biol Med 2013; 238(5): 450-60.
18
George M, Tharakan M, Culberson J, Reddy AP, Reddy PH. Role of Nrf2 in aging, Alzheimer’s and other neurodegenerative diseases. Ageing Res Rev 2022; 82: 101756.
19
Tu W, Wang H, Li S, Liu Q, Sha H. The anti-inflammatory and anti-oxidant mechanisms of the Keap1/Nrf2/ARE signaling pathway in chronic diseases. Aging Dis 2019; 10(3): 637-51.
20
Kaspar JW, Niture SK, Jaiswal AK. Nrf2:INrf2 (Keap1) signaling in oxidative stress. Free Radic Biol Med 2009; 47(9): 1304-9.
21
Zhang DD. Mechanistic studies of the Nrf2-Keap1 signaling pathway. Drug Metab Rev 2006; 38(4): 769-89.
22
Vomhof-DeKrey EE, Picklo MJ Sr. The Nrf2-antioxidant response element pathway: A target for regulating energy metabolism. J Nutr Biochem 2012; 23(10): 1201-6.
23
Saha S, Buttari B, Panieri E, Profumo E, Saso L. An overview of Nrf2 signaling pathway and its role in inflammation. Molecules 2020; 25(22): 5474.
24
Rapaka D, Bitra VR, Akula A. Prediabetes and alzheimer’s disease. Indian J Pharm Sci 2015; 77(5): 511-4.
25
Hayes JD, Chowdhry S, Dinkova-Kostova AT, Sutherland C. Dual regulation of transcription factor Nrf2 by Keap1 and by the combined actions of β-TrCP and GSK-3. Biochem Soc Trans 2015; 43(4): 611-20.
26
Armagan G, Sevgili E, Gürkan FT, et al. Regulation of the Nrf2 pathway by glycogen synthase kinase-3β in MPP+-induced cell damage. Molecules 2019; 24(7): 1377.
27
Jain AK, Jaiswal AK. GSK-3beta acts upstream of Fyn kinase in regulation of nuclear export and degradation of NF-E2 related factor 2. J Biol Chem 2007; 282(22): 16502-10.
28
Qiao J, Zhao Y, Liu Y, et al. Neuroprotective effect of Ginsenoside Re against neurotoxin‑induced Parkinson’s disease models via induction of Nrf2. Mol Med Rep 2022; 25(6): 215.
29
Rada P, Rojo AI, Chowdhry S, McMahon M, Hayes JD, Cuadrado A. SCF/beta-TrCP promotes glycogen synthase kinase 3-dependent degradation of the Nrf2 transcription factor in a Keap1-independent manner. Mol Cell Biol 2011; 31(6): 1121-33.
30
Cores Á, Piquero M, Villacampa M, León R, Menéndez JC. NRF2 regulation processes as a source of potential drug targets against neurodegenerative diseases. Biomolecules 2020; 10(6): 904.
31
Hardy JA, Higgins GA. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992; 256(5054): 184-5.
32
Mudher A, Lovestone S. Alzheimer’s disease : Do tauists and baptists finally shake hands? Trends Neurosci 2002; 25(1): 22-6.
33
Tönnies E, Trushina E. Oxidative stress, synaptic dysfunction, and alzheimer’s disease. J Alzheimers Dis 2017; 57(4): 1105-21.
34
Rapaka D, Bitra VR, Ummidi R, Akula A. Benincasa hispida alleviates amyloid pathology by inhibition of Keap1/Nrf2-axis: Emphasis on oxidative and inflammatory stress involved in Alzheimer’s disease model. Neuropeptides 2021; 88: 102151. a
35
Rapaka D, Bitra VR, Challa SR, Adiukwu PC. Potentiation of microglial endocannabinoid signaling alleviates neuroinflammation in Alzheimer’s disease. Neuropeptides 2021; 90: 102196. b
36
Rapaka D, Bitra VR, Vishala TC, Akula A. Vitis vinifera acts as anti-Alzheimer’s agent by modulating biochemical parameters implicated in cognition and memory. J Ayurveda Integr Med 2019; 10(4): 241-7.
37
Frost GR, Li YM. The role of astrocytes in amyloid production and Alzheimer’s disease. Open Biol 2017; 7(12): 170228.
38
Pereira JB, Janelidze S, Ossenkoppele R, et al. Untangling the association of amyloid-β and tau with synaptic and axonal loss in Alzheimer’s disease. Brain 2021; 144(1): 310-24.
39
Sultana R, Perluigi M, Butterfield DA. Protein oxidation and lipid peroxidation in brain of subjects with Alzheimer’s disease: insights into mechanism of neurodegeneration from redox proteomics. Antioxid Redox Signal 2006; 8(11-12): 2021-37.
40
Qu Z, Sun J, Zhang W, Yu J, Zhuang C. Transcription factor NRF2 as a promising therapeutic target for Alzheimer’s disease. Free Radic Biol Med 2020; 159: 87-102.
41
Rojo AI, Pajares M, García-Yagüe AJ, et al. Deficiency in the transcription factor NRF2 worsens inflammatory parameters in a mouse model with combined tauopathy and amyloidopathy. Redox Biol 2018; 18: 173-80.
42
Satoh T, Harada N, Hosoya T, Tohyama K, Yamamoto M, Itoh K. Keap1/Nrf2 system regulates neuronal survival as revealed through study of keap1 gene-knockout mice. Biochem Biophys Res Commun 2009; 380(2): 298-302.
43
Zhou Y, Xie N, Li L, Zou Y, Zhang X, Dong M. Puerarin alleviates cognitive impairment and oxidative stress in APP/PS1 transgenic mice. Int J Neuropsychopharmacol 2014; 17(4): 635-44.
44
Branca C, Ferreira E, Nguyen TV, Doyle K, Caccamo A, Oddo S. Genetic reduction of Nrf2 exacerbates cognitive deficits in a mouse model of Alzheimer’s disease. Hum Mol Genet 2017; 26(24): 4823-35.
45
Farr SA, Ripley JL, Sultana R, et al. Antisense oligonucleotide against GSK-3β in brain of SAMP8 mice improves learning and memory and decreases oxidative stress: Involvement of transcription factor Nrf2 and implications for Alzheimer disease. Free Radic Biol Med 2014; 67: 387-95.
46
Granic I, Dolga AM, Nijholt IM, van Dijk G, Eisel ULM. Inflammation and NF-kappaB in Alzheimer’s disease and diabetes. J Alzheimers Dis 2009; 16(4): 809-21.
47
Seo EJ, Fischer N, Efferth T. Phytochemicals as inhibitors of NF-κB for treatment of Alzheimer’s disease. Pharmacol Res 2018; 129: 262-73.
48
Kobayashi EH, Suzuki T, Funayama R, et al. Nrf2 suppresses macrophage inflammatory response by blocking proinflammatory cytokine transcription. Nat Commun 2016; 7(1): 11624.
49
Lipton SA, Rezaie T, Nutter A, et al. Therapeutic advantage of pro-electrophilic drugs to activate the Nrf2/ARE pathway in Alzheimer’s disease models. Cell Death Dis 2016; 7(12): e2499.
50
Fanaee-Danesh E, Gali CC, Tadic J, et al. Astaxanthin exerts protective effects similar to bexarotene in Alzheimer’s disease by modulating amyloid-beta and cholesterol homeostasis in blood-brain barrier endothelial cells. Biochim Biophys Acta Mol Basis Dis 2019; 1865(9): 2224-45.
51
Yu L, Wang W, Pang W, Xiao Z, Jiang Y, Hong Y. Dietary lycopene supplementation improves cognitive performances in tau transgenic mice expressing P301L mutation via inhibiting oxidative stress and tau hyperphosphorylation. J Alzheimers Dis 2017; 57(2): 475-82.
52
Hu Y, Li C, Shen W. Gastrodin alleviates memory deficits and reduces neuropathology in a mouse model of Alzheimer’s disease. Neuropathology 2014; 34(4): 370-7.
53
Xie Y, Tan Y, Zheng Y, Du X, Liu Q. Ebselen ameliorates β-amyloid pathology, tau pathology, and cognitive impairment in triple-transgenic Alzheimer’s disease mice. J Biol Inorg Chem 2017; 22(6): 851-65.
54
Currais A, Farrokhi C, Dargusch R, et al. Fisetin reduces the impact of aging on behavior and physiology in the rapidly aging SAMP8 mouse. J Gerontol A Biol Sci Med Sci 2018; 73(3): 299-307.
55
Hong Y, An Z. Hesperidin attenuates learning and memory deficits in APP/PS1 mice through activation of Akt/Nrf2 signaling and inhibition of RAGE/NF-κB signaling. Arch Pharm Res 2018; 41(6): 655-63.
56
Ali T, Kim T, Rehman SU, et al. Natural dietary supplementation of anthocyanins via PI3K/Akt/Nrf2/HO-1 pathways mitigate oxidative stress, neurodegeneration, and memory impairment in a mouse model of alzheimer’s disease. Mol Neurobiol 2018; 55(7): 6076-93.
57
Zhao L, Wang JL, Liu R, Li XX, Li JF, Zhang L. Neuroprotective, anti-amyloidogenic and neurotrophic effects of apigenin in an Alzheimer’s disease mouse model. Molecules 2013; 18(8): 9949-65.