CD36 upregulation mediated by intranasal LV-NRF2 treatment mitigates hypoxia-induced progression of Alzheimer's-like pathogenesis

CY Wang, ZY Wang, JW Xie, JH Cai… - Antioxidants & redox …, 2014 - liebertpub.com
CY Wang, ZY Wang, JW Xie, JH Cai, T Wang, Y Xu, X Wang, L An
Antioxidants & redox signaling, 2014liebertpub.com
Aims: There is extensive evidence that oxidative stress induces cellular dysfunction in the
brain and plays a critical role in Alzheimer's disease (AD) pathogenesis. Hypoxia increases
factors involved in oxidative stress injury and contributes to the onset and progression of AD.
Nuclear factor erythroid 2-related factor 2 (NRF2), a major component regulating antioxidant
response, is attenuated in the AD brain. Importantly, NRF2 directly regulates the alternative
first exons of CD36, an important participant in oxidative and inflammatory processes. To …
Abstract
Aims: There is extensive evidence that oxidative stress induces cellular dysfunction in the brain and plays a critical role in Alzheimer's disease (AD) pathogenesis. Hypoxia increases factors involved in oxidative stress injury and contributes to the onset and progression of AD. Nuclear factor erythroid 2-related factor 2 (NRF2), a major component regulating antioxidant response, is attenuated in the AD brain. Importantly, NRF2 directly regulates the alternative first exons of CD36, an important participant in oxidative and inflammatory processes. To explore the effects of hypoxia-induced deterioration of AD-like pathogenesis and investigate the correlation between hypoxia-induced NRF2 signal alterations and CD36 expression, we examined the NRF2 signaling, CD36, and oxidative stress events in hypoxia-treated APPswe/PSEN1dE9 (APP/PS1) mice brain. Results: We observed that hypoxia treatment increased oxidative stress, exacerbated inflammation, and aggravated learning defects in aged APP/PS1 mice. Microglia from hypoxia-treated mice brain exhibited marked reduction in CD36 expression and inhibition of β-amyloid (Aβ) degradation. Accordingly, hypoxia treatment caused a decrease in transactivation of NRF2 target genes in the aging mouse brain. Intranasal administration with a lentiviral vector encoding human NRF2 increased CD36 expression, ameliorated the weak antioxidant response triggered by hypoxia, diminished Aβ deposition, and improved spatial memory defects. Innovation: In this study, we demonstrated for the first time that NRF2 intranasal treatment-induced increases of CD36 could enhance Aβ clearance in AD transgenic mouse. Conclusion: These results suggest that targeting NRF2-mediated CD36 expression might provide a beneficial intervention for cognitive impairment and oxidative stress in AD progression. Antioxid. Redox Signal. 21, 2208–2230.
Mary Ann Liebert