Generic placeholder image

Current Alzheimer Research

Editor-in-Chief

ISSN (Print): 1567-2050
ISSN (Online): 1875-5828

Research Article

Hypoxic Preconditioning Ameliorates Amyloid-β Pathology and Longterm Cognitive Decline in AβPP/PS1 Transgenic Mice

Author(s): Jian Zhang, Ji Zhang, Xiao-Jia Li, Jun Xiao and Fang Ye*

Volume 17, Issue 7, 2020

Page: [626 - 634] Pages: 9

DOI: 10.2174/1567205017666201007121730

Price: $65

Abstract

Background and Objective: Hypoxic Preconditioning (HPC) has been well established to trigger endogenous mechanisms of neuroprotection basing on models of hypoxic and ischemic diseases in the Central Nervous System (CNS). However, its effects against Alzheimer's Disease (AD) still lack substantial evidence and in-depth exploration. The present study aimed to investigate the impacts of HPC on AD-related memory decline and amyloid-β (Aβ) pathology in AβPP/PS1 transgenic mice.

Methods: Seven-week-old AβPP/PS1 transgenic mice were randomized into HPC and non-HPC groups. The HPC groups were treated with early and repetitive HPC for four weeks, while the non-HPC group was raised under normoxia condition. All the animals were then raised until the age of 28 weeks when Morris water maze tests were conducted to examine the animals’ spatial memory. Indicators for Aβ pathology (soluble Aβ levels and numbers of Aβ plaques) and the expression of relevant proteins were measured to explore potential mechanisms.

Results: The results showed that HPC ameliorated memory decline and Aβ pathology in AβPP/PS1 mice. The protein levels of Amyloid-β Precursor Protein (AβPP) and β-site APP Cleaving Enzyme 1 (BACE1) were reduced while that of Hypoxic inducible factor 1α (HIF-1α) was elevated in HPC groups.

Conclusion: HPC might be a promising strategy for AD intervention. Its potential protection might be realized via downregulating the expressions of AβPP and BACE1 and hence inhibiting Aβ pathology. Notably, HIF-1α might play a key role in mediating subsequent neuroadaptive changes following HPC.

Keywords: Hypoxic preconditioning, Alzheimer's disease, Aβ pathology, hypoxic inducible factor 1α, dementia, protective factors.

[1]
American Psychiatric Association Diagnostic and statistical manual of mental disorders Fifth Edition (DSM-5) Arlington (VA): American Psychiatric Publishing . 2013; pp. 602-11.
[2]
Shah H, Albanese E, Duggan C, et al. Research priorities to reduce the global burden of dementia by 2025. Lancet Neurol 2016; 15(12): 1285-94.
[http://dx.doi.org/10.1016/S1474-4422(16)30235-6] [PMID: 27751558]
[3]
Jack CR Jr, Bennett DA, Blennow K, et al. Contributors. NIA-AA research framework: Toward a biological definition of Alzheimer’s disease. Alzheimers Dement 2018; 14(4): 535-62.
[http://dx.doi.org/10.1016/j.jalz.2018.02.018] [PMID: 29653606]
[4]
Magalingam KB, Radhakrishnan A, Ping NS, Haleagrahara N. Current concepts of neurodegenerative mechanisms in Alzheimer’s disease. BioMed Res Int 2018; 20183740461
[http://dx.doi.org/10.1155/2018/3740461] [PMID: 29707568]
[5]
Li S, Hafeez A, Noorulla F, et al. Preconditioning in neuroprotection: From hypoxia to ischemia. Prog Neurobiol 2017; 157: 79-91.
[http://dx.doi.org/10.1016/j.pneurobio.2017.01.001] [PMID: 28110083]
[6]
Baillieul S, Chacaroun S, Doutreleau S, Detante O, Pépin JL, Verges S. Hypoxic conditioning and the central nervous system: A new therapeutic opportunity for brain and spinal cord injuries? Exp Biol Med (Maywood) 2017; 242(11): 1198-206.
[http://dx.doi.org/10.1177/1535370217712691] [PMID: 28585890]
[7]
Stetler RA, Leak RK, Gan Y, et al. Preconditioning provides neuroprotection in models of CNS disease: Paradigms and clinical significance. Prog Neurobiol 2014; 114: 58-83.
[http://dx.doi.org/10.1016/j.pneurobio.2013.11.005] [PMID: 24389580]
[8]
Gonzalez-Rothi EJ, Lee KZ, Dale EA, Reier PJ, Mitchell GS, Fuller DD. Intermittent hypoxia and neurorehabilitation. J Appl Physiol 2015; 119(12): 1455-65.
[http://dx.doi.org/10.1152/japplphysiol.00235.2015] [PMID: 25997947]
[9]
Schega L, Peter B, Brigadski T, et al. Effect of intermittent normobaric hypoxia on aerobic capacity and cognitive function in older people. J Sci Med Sport 2016; 19(11): 941-5.
[http://dx.doi.org/10.1016/j.jsams.2016.02.012] [PMID: 27134133]
[10]
Sardeli AV, Ferreira MLV. Low-load resistance exercise improves cognitive function in older adults. Rev Bras Med Esporte 2018; 24: 125-9.
[http://dx.doi.org/10.1590/1517-869220182402179200]
[11]
Mecocci P, Boccardi V, Cecchetti R, et al. A long journey into aging, brain aging, and Alzheimer’s disease following the oxidative stress tracks. J Alzheimers Dis 2018; 62(3): 1319-35.
[http://dx.doi.org/10.3233/JAD-170732] [PMID: 29562533]
[12]
Serebrovska ZO, Serebrovska TV, Kholin VA, et al. Intermittent hypoxia-hyperoxia training improves cognitive function and decreases circulating biomarkers of Alzheimer’s disease in patients with mild cognitive impairment: A pilot study. Int J Mol Sci 2019; 20(21): 5405.
[http://dx.doi.org/10.3390/ijms20215405] [PMID: 31671598]
[13]
Manukhina EB, Goryacheva AV, Barskov IV, et al. Prevention of neurodegenerative damage to the brain in rats in experimental Alzheimer’s disease by adaptation to hypoxia. Neurosci Behav Physiol 2010; 40(7): 737-43.
[http://dx.doi.org/10.1007/s11055-010-9320-6] [PMID: 20635216]
[14]
Cui GH, Wu J, Mou FF, et al. Exosomes derived from hypoxia-preconditioned mesenchymal stromal cells ameliorate cognitive decline by rescuing synaptic dysfunction and regulating inflammatory responses in APP/PS1 mice. FASEB J 2018; 32(2): 654-68.
[http://dx.doi.org/10.1096/fj.201700600R] [PMID: 28970251]
[15]
Meng SX, Wang B, Li WT. Intermittent hypoxia improves cognition and reduces anxiety-related behavior in APP/PS1 mice. Brain Behav 2020; 10(2)e01513
[http://dx.doi.org/10.1002/brb3.1513] [PMID: 31877583]
[16]
Koelsch G. BACE1 Function and inhibition: Implications of intervention in the amyloid pathway of Alzheimer’s disease pathology. Molecules 2017; 22(10)E1723
[http://dx.doi.org/10.3390/molecules22101723] [PMID: 29027981]
[17]
Selkoe DJ, Hardy J. The amyloid hypothesis of Alzheimer’s disease at 25 years. EMBO Mol Med 2016; 8(6): 595-608.
[http://dx.doi.org/10.15252/emmm.201606210] [PMID: 27025652]
[18]
Nalivaeva NN, Turner AJ. Targeting amyloid clearance in Alzheimer’s disease as a therapeutic strategy. Br J Pharmacol 2019; 176(18): 3447-63.
[http://dx.doi.org/10.1111/bph.14593] [PMID: 30710367]
[19]
Zhang F, Niu L, Li S, Le W. Pathological impacts of chronic hypoxia on Alzheimer’s disease. ACS Chem Neurosci 2019; 10(2): 902-9.
[http://dx.doi.org/10.1021/acschemneuro.8b00442] [PMID: 30412668]
[20]
Sun X, He G, Qing H, et al. Hypoxia facilitates Alzheimer’s disease pathogenesis by up-regulating BACE1 gene expression. Proc Natl Acad Sci USA 2006; 103(49): 18727-32.
[http://dx.doi.org/10.1073/pnas.0606298103] [PMID: 17121991]
[21]
Almendros I, Wang Y, Gozal D. The polymorphic and contradictory aspects of intermittent hypoxia. Am J Physiol Lung Cell Mol Physiol 2014; 307(2): L129-40.
[http://dx.doi.org/10.1152/ajplung.00089.2014] [PMID: 24838748]
[22]
Verges S, Chacaroun S, Godin-Ribuot D, Baillieul S. hypoxic conditioning as a new therapeutic modality. Front Pediatr 2015; 3: 58.
[http://dx.doi.org/10.3389/fped.2015.00058] [PMID: 26157787]
[23]
Zhang F, Zhong R, Qi H, et al. Impacts of acute hypoxia on Alzheimer’s disease-like pathologies in APPswe/PS1dE9 mice and their wild type littermates. Front Neurosci 2018; 12: 314.
[http://dx.doi.org/10.3389/fnins.2018.00314] [PMID: 29867325]
[24]
Stowe AM, Altay T, Freie AB, Gidday JM. Repetitive hypoxia extends endogenous neurovascular protection for stroke. Ann Neurol 2011; 69(6): 975-85.
[http://dx.doi.org/10.1002/ana.22367] [PMID: 21437933]
[25]
Gustavsson M, Anderson MF, Mallard C, Hagberg H. Hypoxic preconditioning confers long-term reduction of brain injury and improvement of neurological ability in immature rats. Pediatr Res 2005; 57(2): 305-9.
[http://dx.doi.org/10.1203/01.PDR.0000151122.58665.70] [PMID: 15611346]
[26]
Martin N, Bossenmeyer-Pourié C, Koziel V, et al. Non-injurious neonatal hypoxia confers resistance to brain senescence in aged male rats. PLoS One 2012; 7(11)e48828
[http://dx.doi.org/10.1371/journal.pone.0048828] [PMID: 23173039]
[27]
Bickler PE, Fahlman CS, Gray JJ. Hypoxic preconditioning failure in aging hippocampal neurons: Impaired gene expression and rescue with intracellular calcium chelation. J Neurosci Res 2010; 88(16): 3520-9.
[http://dx.doi.org/10.1002/jnr.22508] [PMID: 20936695]
[28]
Ashok BS, Ajith TA, Sivanesan S. Hypoxia-inducible factors as neuroprotective agent in Alzheimer’s disease. Clin Exp Pharmacol Physiol 2017; 44(3): 327-34.
[http://dx.doi.org/10.1111/1440-1681.12717] [PMID: 28004401]
[29]
Guo C, Wang T, Zheng W, Shan ZY, Teng WP, Wang ZY. Intranasal deferoxamine reverses iron-induced memory deficits and inhibits amyloidogenic APP processing in a transgenic mouse model of Alzheimer’s disease. Neurobiol Aging 2013; 34(2): 562-75.
[http://dx.doi.org/10.1016/j.neurobiolaging.2012.05.009] [PMID: 22717236]
[30]
Guo C, Zhang YX, Wang T, et al. Intranasal deferoxamine attenuates synapse loss via up-regulating the P38/HIF-1α pathway on the brain of APP/PS1 transgenic mice. Front Aging Neurosci 2015; 7: 104-4.
[http://dx.doi.org/10.3389/fnagi.2015.00104] [PMID: 26082716]
[31]
Lu N, Li X, Tan R, et al. HIF-1α/beclin1-mediated autophagy is involved in neuroprotection induced by hypoxic preconditioning. J Mol Neurosci 2018; 66(2): 238-50.
[http://dx.doi.org/10.1007/s12031-018-1162-7] [PMID: 30203298]
[32]
Zhang H, Bosch-Marce M, Shimoda LA, et al. Mitochondrial autophagy is an HIF-1-dependent adaptive metabolic response to hypoxia. J Biol Chem 2008; 283(16): 10892-903.
[http://dx.doi.org/10.1074/jbc.M800102200] [PMID: 18281291]
[33]
Gong G, Hu L, Liu Y, et al. Upregulation of HIF-1α protein induces mitochondrial autophagy in primary cortical cell cultures through the inhibition of the mTOR pathway. Int J Mol Med 2014; 34(4): 1133-40.
[http://dx.doi.org/10.3892/ijmm.2014.1850] [PMID: 25017576]
[34]
Li Q, Liu Y, Sun M. Autophagy and Alzheimer’s disease. Cell Mol Neurobiol 2017; 37(3): 377-88.
[http://dx.doi.org/10.1007/s10571-016-0386-8] [PMID: 27260250]
[35]
Schubert D, Soucek T, Blouw B. The induction of HIF-1 reduces astrocyte activation by amyloid beta peptide. Eur J Neurosci 2009; 29(7): 1323-34.
[http://dx.doi.org/10.1111/j.1460-9568.2009.06712.x] [PMID: 19519624]
[36]
Chai X, Kong W, Liu L, Yu W, Zhang Z, Sun Y. A viral vector expressing hypoxia-inducible factor 1 alpha inhibits hippocampal neuronal apoptosis. Neural Regen Res 2014; 9(11): 1145-53.
[http://dx.doi.org/10.4103/1673-5374.135317] [PMID: 25206774]
[37]
Echeverria V, Barreto GE, Avila-Rodriguezc M, Tarasov VV, Aliev G. Is VEGF a key target of cotinine and other potential therapies against Alzheimer disease? Curr Alzheimer Res 2017; 14(11): 1155-63.
[http://dx.doi.org/10.2174/1567205014666170329113007] [PMID: 28356047]
[38]
Bassil F, Fernagut PO, Bezard E, Meissner WG. Insulin, IGF-1 and GLP-1 signaling in neurodegenerative disorders: Targets for disease modification? Prog Neurobiol 2014; 118: 1-18.
[http://dx.doi.org/10.1016/j.pneurobio.2014.02.005] [PMID: 24582776]
[39]
Merelli A, Rodríguez JCG, Folch J, Regueiro MR, Camins A, Lazarowski A. Understanding the role of hypoxia inducible factor during neurodegeneration for new therapeutics opportunities. Curr Neuropharmacol 2018; 16(10): 1484-98.
[http://dx.doi.org/10.2174/1570159X16666180110130253] [PMID: 29318974]
[40]
Reiserer RS, Harrison FE, Syverud DC, Mcdonald MP. Impaired spatial learning in the APPSwe+ PSEN1ΔE9 bigenic mouse model of Alzheimer’s disease genes brain. Behaviour 2007; 6(1): 54-65.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy