Generic placeholder image

Current Neurovascular Research

Editor-in-Chief

ISSN (Print): 1567-2026
ISSN (Online): 1875-5739

Research Article

MiR-340 Reduces the Accumulation of Amyloid-β Through Targeting BACE1 (β-site Amyloid Precursor Protein Cleaving Enzyme 1) in Alzheimer’s Disease

Author(s): Xianpei Tan*, Yi Luo, Dingfang Pi, Liexin Xia, Zhilian Li and Qiang Tu

Volume 17, Issue 1, 2020

Page: [86 - 92] Pages: 7

DOI: 10.2174/1567202617666200117103931

Price: $65

Abstract

Background: Alzheimer’s disease (AD) is the most common neurodegenerative disease, and the accumulation of amyloid-β is the initial process in AD. MicroRNAs (miRNAs) are widely known as key regulators of the accumulation of amyloid-β in AD. This study analyzed the potential effects and possible internal mechanisms of miR-340 on AD.

Methods: The expression of miR-340 in senescence-accelerated mouse prone-8 (SAMP8) mouse and senescence-accelerated mice/resistant-1 (SAMR1) mouse was evaluated by qRT-PCR (quantitative real-time polymerase chain reaction). The expression of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) was determined by qRT-PCR and western blot. The binding ability between miR-340 and BACE1 was verified by dual-luciferase reporter assay. In vitro cell model of AD was established in human neuroblastoma SH-SY5Y cells transfected with Swedish mutant form of amyloid precursor protein (APPswe). The effect of miR-340 on the accumulation of amyloid- β was investigated by western blot analysis. Flow cytometry was conducted to detect cell apoptosis.

Results: MiR-340 was down-regulated in the hippocampus of AD model SAMP8 mouse compared to SAMR1 mouse, while BACE1 was up-regulated in SAMP8, suggesting a negative correlation between miR-340 and BACE1 in SAMP8 mouse. MiR-340 could directly bind with BACE1, and over-expression of miR-340 decreased expression of BACE1 in SH-SY5Y/APPswe cells. MiR- 340 reduced the accumulation of amyloid-β and suppressed cell apoptosis through targeting BACE1 in SH-SY5Y/APPswe cells.

Conclusion: MiR-340 was downregulated in AD and reduced the accumulation of amyloid-β through targeting BACE1, suggesting a potential therapeutic target for AD.

Keywords: miR-340, BACE1, amyloid-β, Alzheimer's disease, amyloid precursor protein (APPswe), neuronal cell.

« Previous
[1]
Mattson, M.P. Pathways towards and away from Alzheimer’s disease. Nature, 2004, 430(7000), 631-639.
[http://dx.doi.org/10.1038/nature02621] [PMID: 15295589]
[2]
Li, L.; Liu, Z.; Jiang, Y-Y.; Shen, W.X.; Peng, Y.P.; Qiu, Y.H. Acetylcholine suppresses microglial inflammatory response via α7nAChR to protect hippocampal neurons. J. Integr. Neurosci., 2019, 18(1), 51-56.https://www.ncbi.nlm.nih.gov/pubmed/31091848
[PMID: 31091848]
[3]
Peng, Q.; Bakulski, K.M.; Nan, B.; Park, S.K. Cadmium and Alzheimer’s disease mortality in U.S. adults: Updated evidence with a urinary biomarker and extended follow-up time. Environ. Res., 2017, 157, 44-51.
[http://dx.doi.org/10.1016/j.envres.2017.05.011] [PMID: 28511080]
[4]
Zhu, Y.; Peng, L.; Hu, J.; Chen, Y.; Chen, F. Current anti-Alzheimer’s disease effect of natural products and their principal targets. J. Integr. Neurosci., 2019, 18(3), 327-339.
[http://dx.doi.org/10.31083/j.jin.2019.03.1105] [PMID: 31601083]
[5]
Tian, Z.; Zhang, X.; Zhao, Z.; Zhang, F.; Deng, T. The Wnt/β-catenin signaling pathway affects the distribution of cytoskeletal proteins in Aβ treated PC12 cells. J. Integr. Neurosci., 2019, 18(3), 309-312.
[http://dx.doi.org/10.31083/j.jin.2019.03.168] [PMID: 31601081]
[6]
De Strooper, B. Proteases and proteolysis in Alzheimer disease: A multifactorial view on the disease process. Physiol. Rev., 2010, 90(2), 465-494.
[http://dx.doi.org/10.1152/physrev.00023.2009] [PMID: 20393191]
[7]
John, V. Human beta-secretase (BACE) and BACE inhibitors: Progress report. Curr. Top. Med. Chem., 2006, 6(6), 569-578.
[http://dx.doi.org/10.2174/156802606776743084] [PMID: 16712492]
[8]
Che, H.; Sun, L.H.; Guo, F. Expression of amyloid-associated miRNAs in both the forebrain cortex and hippocampus of middle-aged rat. Cell. Physiol. Biochem., 2014, 33(1), 11-22.
[http://dx.doi.org/10.1159/000356646] [PMID: 24401368]
[9]
Roberds, S.L.; Anderson, J.; Basi, G. BACE knockout mice are healthy despite lacking the primary beta-secretase activity in brain: implications for Alzheimer’s disease therapeutics. Hum. Mol. Genet., 2001, 10(12), 1317-1324.
[http://dx.doi.org/10.1093/hmg/10.12.1317] [PMID: 11406613]
[10]
Lourdes, O.; Lucía, B-M.; Jordán, J. Sildenafil decreases BACE1 and Cathepsin B levels and reduces APP amyloidogenic processing in the SAMP8 Mouse. J. Gerontol. A Biol. Sci. Med. Sci., 2014, 70(6), 675-685.https://www.ncbi.nlm.nih.gov/pubmed/25063079
[PMID: 25063079]
[11]
Takahashi, R.; Ma, S.; Deese, A. Elucidating the mechanism of cytochrome P450-mediated pyrimidine ring conversion to pyrazole metabolites with the BACE1 inhibitor GNE-892 in rats. Drug Metab. Dispos., 2014, 42(5), 890-898.
[http://dx.doi.org/10.1124/dmd.114.057141] [PMID: 24595682]
[12]
Kao, S.C.; Krichevsky, A.M.; Kosik, K.S.; Tsai, L.H. BACE1 suppression by RNA interference in primary cortical neurons. J. Biol. Chem., 2004, 279(3), 1942-1949.
[http://dx.doi.org/10.1074/jbc.M309219200] [PMID: 14600149]
[13]
Kim, V.N. MicroRNA biogenesis: Coordinated cropping and dicing. Nat. Rev. Mol. Cell Biol., 2005, 6(5), 376-385.
[http://dx.doi.org/10.1038/nrm1644] [PMID: 15852042]
[14]
Femminella, G.D.; Ferrara, N.; Rengo, G. The emerging role of microRNAs in Alzheimer’s disease. Front. Physiol., 2015, 6, 40.
[http://dx.doi.org/10.3389/fphys.2015.00040] [PMID: 25729367]
[15]
Wang, Y.; Veremeyko, T.; Wong, A.H. Downregulation of miR-132/212 impairs S-nitrosylation balance and induces tau phosphorylation in Alzheimer’s disease. Neurobiol. Aging, 2017, 51, 156-166.
[http://dx.doi.org/10.1016/j.neurobiolaging.2016.12.015] [PMID: 28089352]
[16]
Tang, H.; Ma, M.; Wu, Y. Activation of MT2 receptor ameliorates dendritic abnormalities in Alzheimer’s disease via C/EBPα/miR-125b pathway. Aging Cell, 2019, 18(2)e12902
[http://dx.doi.org/10.1111/acel.12902] [PMID: 30706990]
[17]
Kim, J.; Yoon, H.; Chung, D.E.; Brown, J.L.; Belmonte, K.C.; Kim, J. miR-186 is decreased in aged brain and suppresses BACE1 expression. J. Neurochem., 2016, 137(3), 436-445.
[http://dx.doi.org/10.1111/jnc.13507] [PMID: 26710318]
[18]
Lei, X.; Lei, L.; Zhang, Z.; Zhang, Z.; Cheng, Y. Downregulated miR-29c correlates with increased BACE1 expression in sporadic Alzheimer’s disease. Int. J. Clin. Exp. Pathol., 2015, 8(2), 1565-1574.https://www.ncbi.nlm.nih.gov/pubmed/25973041
[PMID: 25973041]
[19]
Gong, G.; An, F.; Wang, Y.; Bian, M.; Yu, L.J.; Wei, C. miR-15b represses BACE1 expression in sporadic Alzheimer’s disease. Oncotarget, 2017, 8(53), 91551-91557.
[http://dx.doi.org/10.18632/oncotarget.21177] [PMID: 29207665]
[20]
Wang, W.X.; Rajeev, B.W.; Stromberg, A.J. The expression of microRNA miR-107 decreases early in Alzheimer’s disease and may accelerate disease progression through regulation of beta-site amyloid precursor protein-cleaving enzyme 1. J. Neurosci., 2008, 28(5), 1213-1223.
[http://dx.doi.org/10.1523/JNEUROSCI.5065-07.2008] [PMID: 18234899]
[21]
Pereira, P.A.; Tomás, J.F.; Queiroz, J.A.; Figueiras, A.R.; Sousa, F. Recombinant pre-miR-29b for Alzheimer’s disease therapeutics. Sci. Rep., 2016, 6, 19946.
[http://dx.doi.org/10.1038/srep19946] [PMID: 26818210]
[22]
Li, S.; Zhang, R.; Yuan, Y. MiR-340 regulates fibrinolysis and axon regrowth following sciatic nerve injury. Mol. Neurobiol., 2017, 54(6), 4379-4389.
[http://dx.doi.org/10.1007/s12035-016-9965-4] [PMID: 27344331]
[23]
Yoo, H.; Kim, J.; Lee, A.R. Alteration of microRNA 340-5p and Arginase-1 expression in peripheral blood cells during acute ischemic stroke. Mol. Neurobiol., 2019, 56(5), 3211-3221.
[http://dx.doi.org/10.1007/s12035-018-1295-2] [PMID: 30112629]
[24]
Wang, W.; Kwon, E.J.; Tsai, L.H. MicroRNAs in learning, memory, and neurological diseases. Learn. Mem., 2012, 19(9), 359-368.
[http://dx.doi.org/10.1101/lm.026492.112] [PMID: 22904366]
[25]
Mushtaq, G.; Greig, N.H.; Anwar, F. miRNAs as circulating biomarkers for alzheimer’s disease and parkinson’s disease. Med. Chem., 2016, 12(3), 217-225.
[http://dx.doi.org/10.2174/1573406411666151030112140] [PMID: 26527155]
[26]
Hu, Y.B.; Li, C.B.; Song, N. Diagnostic value of microRNA for Alzheimer’s disease: A systematic review and meta-analysis. Front. Aging Neurosci., 2016, 8, 13.
[http://dx.doi.org/10.3389/fnagi.2016.00013] [PMID: 26903857]
[27]
Wang, W.X.; Huang, Q.; Hu, Y.; Stromberg, A.J.; Nelson, P.T. Patterns of microRNA expression in normal and early Alzheimer’s disease human temporal cortex: white matter versus gray matter. Acta Neuropathol., 2011, 121(2), 193-205.
[http://dx.doi.org/10.1007/s00401-010-0756-0] [PMID: 20936480]
[28]
Wang, H.; Lian, K.; Han, B. Age-related alterations in the metabolic profile in the hippocampus of the senescence-accelerated mouse prone 8: A spontaneous Alzheimer’s disease mouse model. J. Alzheimers Dis., 2014, 39(4), 841-848.
[http://dx.doi.org/10.3233/JAD-131463] [PMID: 24284365]
[29]
Karuppagounder, V.; Arumugam, S.; Babu, S.S. The senescence accelerated mouse prone 8 (SAMP8): A novel murine model for cardiac aging. Ageing Res. Rev., 2017, 35, 291-296.
[http://dx.doi.org/10.1016/j.arr.2016.10.006] [PMID: 27825897]
[30]
Takeda, T.; Hosokawa, M.; Higuchi, K. Senescence-accelerated mouse (SAM): A novel murine model of senescence. Exp. Gerontol., 1997, 32(1-2), 105-109.
[http://dx.doi.org/10.1016/S0531-5565(96)00036-8] [PMID: 9088907]
[31]
Zhang, R.; Zhang, Q.; Niu, J. Screening of microRNAs associated with Alzheimer’s disease using oxidative stress cell model and different strains of senescence accelerated mice. J. Neurol. Sci., 2014, 338(1-2), 57-64.
[http://dx.doi.org/10.1016/j.jns.2013.12.017] [PMID: 24423585]
[32]
Meyers, K.T.; Marballi, K.K.; Brunwasser, S.J. The immediate early gene Egr3 is required for hippocampal induction of Bdnf by electroconvulsive stimulation. Front. Behav. Neurosci., 2018, 12, 92.
[http://dx.doi.org/10.3389/fnbeh.2018.00092] [PMID: 29867393]
[33]
Salta, E.; Sierksma, A.; Vanden Eynden, E.; De Strooper, B. miR-132 loss de-represses ITPKB and aggravates amyloid and TAU pathology in Alzheimer’s brain. EMBO Mol. Med., 2016, 8(9), 1005-1018.
[http://dx.doi.org/10.15252/emmm.201606520] [PMID: 27485122]
[34]
Jack, C.R., Jr; Knopman, D.S.; Jagust, W.J. Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol., 2010, 9(1), 119-128.
[http://dx.doi.org/10.1016/S1474-4422(09)70299-6] [PMID: 20083042]
[35]
Niikura, T.; Tajima, H.; Kita, Y. Neuronal cell death in Alzheimer’s disease and a neuroprotective factor, humanin. Curr. Neuropharmacol., 2006, 4(2), 139-147.
[http://dx.doi.org/10.2174/157015906776359577] [PMID: 18615127]
[36]
Norfray, J.F.; Provenzale, J.M. Alzheimer’s disease: Neuropathologic findings and recent advances in imaging. AJR Am. J. Roentgenol., 2004, 182(1), 3-13.
[http://dx.doi.org/10.2214/ajr.182.1.1820003] [PMID: 14684506]

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