Korean J Physiol Pharmacol.  2020 Jul;24(4):299-310. 10.4196/kjpp.2020.24.4.299.

Combination therapy with cilostazol, aripiprazole, and donepezil protects neuronal cells from β-amyloid neurotoxicity through synergistically enhanced SIRT1 expression

Affiliations
  • 1Department of Pharmacology, Pusan National University School of Medicine, Korea
  • 2Gene & Cell Therapy Research Center for Vessel-associated Diseases, Pusan National University, Korea
  • 3Department of Korean Medical Science, Pusan National University School of Korean Medicine, Yangsan 50612, Korea

Abstract

Alzheimer’s disease (AD) is a multi-faceted neurodegenerative disease. Thus, current therapeutic strategies require multitarget-drug combinations to treat or prevent the disease. At the present time, single drugs have proven to be inadequate in terms of addressing the multifactorial pathology of AD, and multitarget-directed drug design has not been successful. Based on these points of views, it is judged that combinatorial drug therapies that target several pathogenic factors may offer more attractive therapeutic options. Thus, we explored that the combination therapy with lower doses of cilostazol and aripiprazole with add-on donepezil (CAD) might have potential in the pathogenesis of AD. In the present study, we found the superior efficacies of donepezil add-on with combinatorial mixture of cilostazol plus aripiprazole in modulation of expression of AD-relevant genes: Aβ accumulation, GSK-3β, P300, acetylated tau, phosphorylated-tau levels, and activation of α-secretase/ADAM 10 through SIRT1 activation in the N2a Swe cells expressing human APP Swedish mutation (N2a Swe cells). We also assessed that CAD synergistically raised acetylcholine release and choline acetyltransferase (CHAT) expression that were declined by increased β-amyloid level in the activated N2a Swe cells. Consequently, CAD treatment synergistically increased neurite elongation and improved cell viability through activations of PI3K, BDNF, β-catenin and a7-nicotinic cholinergic receptors in neuronal cells in the presence of Aβ1-42. This work endorses the possibility for efficient treatment of AD by supporting the synergistic therapeutic potential of donepezil add-on therapy in combination with lower doses of cilostazol and aripiprazole.

Keyword

Alzheimer’s disease; Aripiprazole; β-amyloid; Cilostazol; Donepezil

Figure

  • Fig. 1 Time-dependent increases in Aβ production and phosphorylation. (A) Time-dependent increases in Aβ production in mouse neuroblastoma neuro2A (N2a) Swe cells as determined by ELISA, represented as percentages of N2a cells cultured for 24 h as 100%. (B) Time-dependent increases in Aβ phosphorylated Tau (P-tau), acetylated tau (Ac-tau), P300, and GSK-3β P-Tyr-216 levels as determined by Western blot. Cells were cultured in 1% FBS for 3, 12, 24, or 48 h, respectively. (C) Quantitative results are expressed as the mean percentages ± SEMs with respect to N2a cells cultured for 24 h (control = 100%) from five experiments. *p < 0.05, **p < 0.01; ***p < 0.001 vs. N2a controls.

  • Fig. 2 Effects of donepezil (DNP), cilostazol + aripiprazole (CA), and CA + DNP (CAD). (A) Suppressive effects of 3 μM donepezil (DNP), 3 μM cilostazol + 3 μM aripiprazole (CA), and CA + 3 μM DNP (CAD) on Aβ production in neuro2A (N2a) Swe cells as determined by ELISA, are represented as percentages of N2a cells cultured for 24 h. (B) Representative Western blots of GSK-3β P-Tyr-216, phosphorylated tau (P-tau), P300, and acetylated tau (Ac-tau) expression levels in activated N2a Swe cells cultured for 24 h. Quantitative analysis of GSK-3β P-Tyr 216 (C), P-tau (D), P300 (E), and Ac-tau expression levels (F). Results are expressed as the means ± SEMs of percentages of vehicle values (100%) of activated N2a Swe cells from four experiments. #p < 0.05, ##p < 0.01, ###p < 0.001 vs. vehicle (veh); $p < 0.05, $$$p < 0.001 vs. DNP.

  • Fig. 3 Decreased ADAM10 expression in the activated neuro2A (N2a) Swe cells and its recovery. (A) Time-dependent decreases in ADAM10 protein levels when N2a Swe cells were cultured in 1% FBS-containing medium for 3, 12, 24, or 48 h. Means ± standard error of the mean (SEMs) are expressed as percentages of values of N2a cell cultured for 24 h (100%) from four experiments. **p < 0.01; ***p < 0.001 vs. values of N2a cells cultured for 24 h. (B) Synergistic upregulation of decreased ADAM10 expression in the N2a Swe cells (at 24 h after FBS depletion [control] = 100%) by 3 μM donepezil (DNP), 3 μM cilostazol + 3 μM aripiprazole (CA) and CA + DNP (CAD). Inhibition of CAD-stimulated ADAM10 expression by 10 μM TIMP (an ADAM10 inhibitor). Means ± SEMs are expressed as percentages of vehicle values (100%) of activated N2a Swe cells from four experiments. ##p < 0.01, ###p < 0.001 vs. vehicle (veh); $p < 0.05, $$$p < 0.001 vs. DNP; †††p < 0.001 vs. CAD alone.

  • Fig. 4 Significantly decreased α-secretase activity at 24 h after FBS depletion and concentration-dependent recovery by cilostazol. (A), 3 μM cilostazol (C3) + 1, 3, 10 μM donepezil (D1, D3, D10) (B), 3 μM cilostazol (C3) + 1, 3, 10 μM aripiprazole (A1, A3, A10) (C). (D) Synergistic upregulation of α-secretase activity in the neuro2A (N2a) Swe cells by 3 μM donepezil (DNP) alone, 3 μM cilostazol + 3 μM aripiprazole (CA), CAD, and blocking of CAD-stimulated α-secretase activity by 10 μM Rp-cAMPS, 20 μM sirtinol and 10 μM TIMP, respectively. Means ± SEMs are expressed as percentages of control values (100%) of N2a cells (with 10% FBS) from 4–8 experiments. **p < 0.01, ***p < 0.001 vs. control; #p < 0.01, ###p < 0.001 vs. vehicle (veh); $$p < 0.01, $$$p < 0.001 vs. DNP; †††p < 0.001 vs. CAD alone. ϕϕϕp < 0.001 vs. CA.

  • Fig. 5 Drug effects on SIRT1 expression and deacetylase activity in the activated neuro2A (N2a) Swe cells. (A) Upregulation of SIRT1 expression in the N2a Swe cells by 3 μM donepezil (DNP), 3 μM cilostazol + 3 μM aripiprazole (CA) and CA + 3 μM DNP (CAD) and blocking of CAD-stimulated SIRT1 expression by 20 μM Sirtinol. Means ± standard error of the mean (SEMs) are expressed as percentages of vehicle (veh = 100%) of activated N2a Swe cells from 5 experiments. (B) Significantly decreased SIRT1 deacetylase activity at 24 h after 1% FBS medium and synergistic upregulation over the control level by drugs and blocking of CAD-stimulated SIRT1 expression by 20 μM sirtinol. (C) Synergistic upregulation of nuclear P-CREB Ser133 levels by DNP, CA, CAD, and blocking of CAD-stimulated intranuclear P-CREB Ser133 levels by 30 μM of LY294002. Means ± SEMs are expressed as percentages of control (100%) of N2a Swe cells (10% FBS) from 4 experiments. *p < 0.05 vs. control; ##p < 0.01, ###p < 0.001 vs. vehicle (veh); $p < 0.05, $$p < 0.01, $$$p < 0.001 vs. DNP; Ψp < 0.05 vs. CA; †p < 0.05, ††p < 0.01, †††p < 0.001 vs. CAD alone.

  • Fig. 6 Drug effects on acetylcholine (ACh) release and expression of choline acetyltransferase (CHAT) in the activated neuro2A (N2a) Swe cells. (A) Significant upregulation of decreased ACh release into the culture medium containing 1% FBS with N2a Swe cells (24 h) by 3 μM donepezil (DNP), 3 μM cilostazol + 3 μM aripiprazole (CA) and CA + 3 μM DNP (CAD). (B) Significant upregulation of CHAT expression by CAD, which was blocked by 10 nM MLA (methyllycaconitine). Means ± SEMs are expressed as percentages of control (ACh; 0.15 ± 0.03 pg/ml = 100%) of N2a Swe cells (10% FBS) from 5 experiments. *p < 0.05, ***p < 0.001 vs. control; #p < 0.05, ##p < 0.01, ###p < 0.001 vs. vehicle; $$p < 0.01 vs. DNP; Ψp < 0.05 vs. CA; †††p < 0.001 vs. CAD alone.

  • Fig. 7 Representative microscopic features (A) and their quantitative analyses (B). The neurite elongation of the HT22 cells was markedly decreased by 3 μM Aβ1-42, which was recovered over the control level by treatment with CA and synergistically increased by CAD, but not by 3 μm donepezil (DNP) alone. Blocking of CAD-stimulated neurite lengths by LY294002 (30 μM), K252A (100 nM), imatinib (10 μM) and methyllycaconitine (MLA) (10 nM), respectively. Results are expressed as means ± SEMs of neurite length (μm) in HT22 cells from 7–14 experiments. ***p < 0.001 vs. control (con); ###p < 0.001 vs. vehicle (veh); $$$p < 0.001 vs. DNP; ΨΨΨp < 0.001 vs. CA; †††p < 0.001 vs. CAD alone.

  • Fig. 8 Effects of drugs on the cell viability of neuro2A (N2a) cells. Cells were pretreated with CAD (3 μM donepezil + 3 μM cilostazol + 3 μM aripiprazole) for 2 h before addition of Aβ1-42 (20 μM) and then cultured forward for 24 h in the absence and presence of 30 μM of LY294002, 100 nM of K252A, 10 μM of imatinib and 10 nM of methyllycaconitine (MLA), respectively. Results are expressed as means ± SEMs of cell viability from 5 experiments. ***p < 0.001 vs. control; #p < 0.001 vs. vehicle; ††p < 0.01, †††p < 0.001 vs. CAD alone.


Reference

1. Lue LF, Kuo YM, Roher AE, Brachova L, Shen Y, Sue L, Beach T, Kurth JH, Rydel RE, Rogers J. 1999; Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease. Am J Pathol. 155:853–862. DOI: 10.1016/S0002-9440(10)65184-X. PMID: 10487842. PMCID: PMC1866907.
2. Nunan J, Small DH. 2000; Regulation of APP cleavage by alpha-, beta- and gamma-secretases. FEBS Lett. 483:6–10. DOI: 10.1016/S0014-5793(00)02076-7. PMID: 11033346.
3. Postina R, Schroeder A, Dewachter I, Bohl J, Schmitt U, Kojro E, Prinzen C, Endres K, Hiemke C, Blessing M, Flamez P, Dequenne A, Godaux E, van Leuven F, Fahrenholz F. 2004; A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest. 113:1456–1464. DOI: 10.1172/JCI20864. PMID: 15146243. PMCID: PMC406531.
Article
4. Kojro E, Fahrenholz F. 2005; The non-amyloidogenic pathway: structure and function of alpha-secretases. Subcell Biochem. 38:105–127. DOI: 10.1007/0-387-23226-5_5. PMID: 15709475.
5. Rojo AI, Sagarra MR, Cuadrado A. 2008; GSK-3beta down-regulates the transcription factor Nrf2 after oxidant damage: relevance to exposure of neuronal cells to oxidative stress. J Neurochem. 105:192–202. DOI: 10.1111/j.1471-4159.2007.05124.x. PMID: 18005231.
6. Ballatore C, Lee VM, Trojanowski JQ. 2007; Tau-mediated neurodegeneration in Alzheimer's disease and related disorders. Nat Rev Neurosci. 8:663–672. DOI: 10.1038/nrn2194. PMID: 17684513.
Article
7. Hooper C, Killick R, Lovestone S. 2008; The GSK3 hypothesis of Alzheimer's disease. J Neurochem. 104:1433–1439. DOI: 10.1111/j.1471-4159.2007.05194.x. PMID: 18088381. PMCID: PMC3073119.
Article
8. Jämsä A, Hasslund K, Cowburn RF, Bäckström A, Vasänge M. 2004; The retinoic acid and brain-derived neurotrophic factor differentiated SH-SY5Y cell line as a model for Alzheimer's disease-like tau phosphorylation. Biochem Biophys Res Commun. 319:993–1000. DOI: 10.1016/j.bbrc.2004.05.075. PMID: 15184080.
Article
9. Lee HR, Park SY, Kim HY, Shin HK, Lee WS, Rhim BY, Hong KW, Kim CD. 2012; Protection by cilostazol against amyloid-β1-40-induced suppression of viability and neurite elongation through activation of CK2α in HT22 mouse hippocampal cells. J Neurosci Res. 90:1566–1576. DOI: 10.1002/jnr.23037. PMID: 22422579.
Article
10. Counts SE, Mufson EJ. 2005; The role of nerve growth factor receptors in cholinergic basal forebrain degeneration in prodromal Alzheimer disease. J Neuropathol Exp Neurol. 64:263–272. DOI: 10.1093/jnen/64.4.263. PMID: 15835262.
Article
11. Niewiadomska G, Baksalerska-Pazera M, Riedel G. 2009; The septo-hippocampal system, learning and recovery of function. Prog Neuropsychopharmacol Biol Psychiatry. 33:791–805. DOI: 10.1016/j.pnpbp.2009.03.039. PMID: 19389457.
Article
12. Francis PT, Palmer AM, Snape M, Wilcock GK. 1999; The cholinergic hypothesis of Alzheimer's disease: a review of progress. J Neurol Neurosurg Psychiatry. 66:137–147. DOI: 10.1136/jnnp.66.2.137. PMID: 10071091. PMCID: PMC1736202.
Article
13. Sanabria-Castro A, Alvarado-Echeverría I, Monge-Bonilla C. 2017; Molecular pathogenesis of Alzheimer's disease: an update. Ann Neurosci. 24:46–54. DOI: 10.1159/000464422. PMID: 28588356. PMCID: PMC5448443.
Article
14. Anekonda TS, Reddy PH. 2006; Neuronal protection by sirtuins in Alzheimer's disease. J Neurochem. 96:305–313. DOI: 10.1111/j.1471-4159.2005.03492.x. PMID: 16219030.
Article
15. Qin W, Yang T, Ho L, Zhao Z, Wang J, Chen L, Zhao W, Thiyagarajan M, MacGrogan D, Rodgers JT, Puigserver P, Sadoshima J, Deng H, Pedrini S, Gandy S, Sauve AA, Pasinetti GM. 2006; Neuronal SIRT1 activation as a novel mechanism underlying the prevention of Alzheimer disease amyloid neuropathology by calorie restriction. J Biol Chem. 281:21745–21754. DOI: 10.1074/jbc.M602909200. PMID: 16751189.
Article
16. Michán S, Li Y, Chou MM, Parrella E, Ge H, Long JM, Allard JS, Lewis K, Miller M, Xu W, Mervis RF, Chen J, Guerin KI, Smith LE, McBurney MW, Sinclair DA, Baudry M, de Cabo R, Longo VD. 2010; SIRT1 is essential for normal cognitive function and synaptic plasticity. J Neurosci. 30:9695–9707. DOI: 10.1523/JNEUROSCI.0027-10.2010. PMID: 20660252. PMCID: PMC2921958.
17. Julien C, Tremblay C, Emond V, Lebbadi M, Salem N Jr, Bennett DA, Calon F. 2009; Sirtuin 1 reduction parallels the accumulation of tau in Alzheimer disease. J Neuropathol Exp Neurol. 68:48–58. DOI: 10.1097/NEN.0b013e3181922348. PMID: 19104446. PMCID: PMC2813570.
Article
18. Tippmann F, Hundt J, Schneider A, Endres K, Fahrenholz F. 2009; Up-regulation of the alpha-secretase ADAM10 by retinoic acid receptors and acitretin. FASEB J. 23:1643–1654. DOI: 10.1096/fj.08-121392. PMID: 19144697.
19. Lee HR, Shin HK, Park SY, Kim HY, Lee WS, Rhim BY, Hong KW, Kim CD. 2014; Cilostazol suppresses β-amyloid production by activating a disintegrin and metalloproteinase 10 via the upregulation of SIRT1-coupled retinoic acid receptor-β. J Neurosci Res. 92:1581–1590. DOI: 10.1002/jnr.23421. PMID: 24903973.
Article
20. Lee JH, Kim KY, Lee YK, Park SY, Kim CD, Lee WS, Rhim BY, Hong KW. 2004; Cilostazol prevents focal cerebral ischemic injury by enhancing casein kinase 2 phosphorylation and suppression of phosphatase and tensin homolog deleted from chromosome 10 phosphorylation in rats. J Pharmacol Exp Ther. 308:896–903. DOI: 10.1124/jpet.103.061853. PMID: 14634032.
Article
21. Hong KW, Lee JH, Kima KY, Park SY, Lee WS. 2006; Cilostazol: therapeutic potential against focal cerebral ischemic damage. Curr Pharm Des. 12:565–573. DOI: 10.2174/138161206775474323. PMID: 16472148.
Article
22. Park SH, Kim JH, Bae SS, Hong KW, Lee DS, Leem JY, Choi BT, Shin HK. 2011; Protective effect of the phosphodiesterase III inhibitor cilostazol on amyloid β-induced cognitive deficits associated with decreased amyloid β accumulation. Biochem Biophys Res Commun. 408:602–608. DOI: 10.1016/j.bbrc.2011.04.068. PMID: 21530492.
Article
23. Burris KD, Molski TF, Xu C, Ryan E, Tottori K, Kikuchi T, Yocca FD, Molinoff PB. 2002; Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. J Pharmacol Exp Ther. 302:381–389. DOI: 10.1124/jpet.102.033175. PMID: 12065741.
Article
24. Croxtall JD. 2012; Aripiprazole: a review of its use in the management of schizophrenia in adults. CNS Drugs. 26:155–183. DOI: 10.2165/11208400-000000000-00000. PMID: 22296317.
25. Pae CU, Forbes A, Patkar AA. 2011; Aripiprazole as adjunctive therapy for patients with major depressive disorder: overview and implications of clinical trial data. CNS Drugs. 25:109–127. DOI: 10.2165/11538980-000000000-00000. PMID: 21254788.
26. Paulsen JS, Salmon DP, Thal LJ, Romero R, Weisstein-Jenkins C, Galasko D, Hofstetter CR, Thomas R, Grant I, Jeste DV. 2000; Incidence of and risk factors for hallucinations and delusions in patients with probable AD. Neurology. 54:1965–1971. DOI: 10.1212/WNL.54.10.1965. PMID: 10822438.
Article
27. Eustace A, Coen R, Walsh C, Cunningham CJ, Walsh JB, Coakley D, Lawlor BA. 2002; A longitudinal evaluation of behavioural and psychological symptoms of probable Alzheimer's disease. Int J Geriatr Psychiatry. 17:968–973. DOI: 10.1002/gps.736. PMID: 12325059.
Article
28. De Deyn P, Jeste DV, Swanink R, Kostic D, Breder C, Carson WH, Iwamoto T. 2005; Aripiprazole for the treatment of psychosis in patients with Alzheimer's disease: a randomized, placebo-controlled study. J Clin Psychopharmacol. 25:463–467. DOI: 10.1097/01.jcp.0000178415.22309.8f. PMID: 16160622.
29. Park SY, Shin HK, Lee WS, Bae SS, Kim K, Hong KW, Kim CD. 2017; Neuroprotection by aripiprazole against β-amyloid-induced toxicity by P-CK2α activation via inhibition of GSK-3β. Oncotarget. 8:110380–110391. DOI: 10.18632/oncotarget.22777. PMID: 29299155. PMCID: PMC5746390.
Article
30. Guzior N, Wieckowska A, Panek D, Malawska B. 2015; Recent development of multifunctional agents as potential drug candidates for the treatment of Alzheimer's disease. Curr Med Chem. 22:373–404. DOI: 10.2174/0929867321666141106122628. PMID: 25386820. PMCID: PMC4435057.
Article
31. Whitehouse PJ, Price DL, Struble RG, Clark AW, Coyle JT, Delon MR. 1982; Alzheimer's disease and senile dementia: loss of neurons in the basal forebrain. Science. 215:1237–1239. DOI: 10.1126/science.7058341. PMID: 7058341.
Article
32. Takada-Takatori Y, Kume T, Izumi Y, Ohgi Y, Niidome T, Fujii T, Sugimoto H, Akaike A. 2009; Roles of nicotinic receptors in acetylcholinesterase inhibitor-induced neuroprotection and nicotinic receptor up-regulation. Biol Pharm Bull. 32:318–324. DOI: 10.1248/bpb.32.318. PMID: 19252271.
Article
33. Sugimoto H, Yamanishi Y, Iimura Y, Kawakami Y. 2000; Donepezil hydrochloride (E2020) and other acetylcholinesterase inhibitors. Curr Med Chem. 7:303–339. DOI: 10.2174/0929867003375191. PMID: 10637367.
Article
34. Giacobini E. 2000; Cholinesterase inhibitors stabilize Alzheimer's disease. Ann N Y Acad Sci. 920:321–327. DOI: 10.1111/j.1749-6632.2000.tb06942.x. PMID: 11193171.
35. Lee HR, Shin HK, Park SY, Kim HY, Lee WS, Rhim BY, Hong KW, Kim CD. 2014; Attenuation of β-amyloid-induced tauopathy via activation of CK2α/SIRT1: targeting for cilostazol. J Neurosci Res. 92:206–217. DOI: 10.1002/jnr.23310. PMID: 24254769.
Article
36. Leroy K, Yilmaz Z, Brion JP. 2007; Increased level of active GSK-3beta in Alzheimer's disease and accumulation in argyrophilic grains and in neurones at different stages of neurofibrillary degeneration. Neuropathol Appl Neurobiol. 33:43–55. DOI: 10.1111/j.1365-2990.2006.00795.x. PMID: 17239007.
Article
37. Hoshi M, Sato M, Matsumoto S, Noguchi A, Yasutake K, Yoshida N, Sato K. 2003; Spherical aggregates of beta-amyloid (amylospheroid) show high neurotoxicity and activate tau protein kinase I/glycogen synthase kinase-3beta. Proc Natl Acad Sci U S A. 100:6370–6375. DOI: 10.1073/pnas.1237107100. PMID: 12750461. PMCID: PMC164453.
38. Bannister AJ, Kouzarides T. 1996; The CBP co-activator is a histone acetyltransferase. Nature. 384:641–643. DOI: 10.1038/384641a0. PMID: 8967953.
Article
39. Min SW, Cho SH, Zhou Y, Schroeder S, Haroutunian V, Seeley WW, Huang EJ, Shen Y, Masliah E, Mukherjee C, Meyers D, Cole PA, Ott M, Gan L. 2010; Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron. 67:953–966. DOI: 10.1016/j.neuron.2010.08.044. PMID: 20869593. PMCID: PMC3035103.
Article
40. Amour A, Knight CG, Webster A, Slocombe PM, Stephens PE, Knäuper V, Docherty AJ, Murphy G. 2000; The in vitro activity of ADAM-10 is inhibited by TIMP-1 and TIMP-3. FEBS Lett. 473:275–279. DOI: 10.1016/S0014-5793(00)01528-3. PMID: 10818225.
Article
41. Araki T, Sasaki Y, Milbrandt J. 2004; Increased nuclear NAD biosynthesis and SIRT1 activation prevent axonal degeneration. Science. 305:1010–1013. DOI: 10.1126/science.1098014. PMID: 15310905.
Article
42. Fusco S, Ripoli C, Podda MV, Ranieri SC, Leone L, Toietta G, McBurney MW, Schütz G, Riccio A, Grassi C, Galeotti T, Pani G. 2012; A role for neuronal cAMP responsive-element binding (CREB)-1 in brain responses to calorie restriction. Proc Natl Acad Sci U S A. 109:621–626. DOI: 10.1073/pnas.1109237109. PMID: 22190495. PMCID: PMC3258594.
Article
43. Auld DS, Kar S, Quirion R. 1998; Beta-amyloid peptides as direct cholinergic neuromodulators: a missing link? Trends Neurosci. 21:43–49. DOI: 10.1016/S0166-2236(97)01144-2. PMID: 9464686.
44. Potter PE, Rauschkolb PK, Pandya Y, Sue LI, Sabbagh MN, Walker DG, Beach TG. 2011; Pre- and post-synaptic cortical cholinergic deficits are proportional to amyloid plaque presence and density at preclinical stages of Alzheimer's disease. Acta Neuropathol. 122:49–60. DOI: 10.1007/s00401-011-0831-1. PMID: 21533854. PMCID: PMC3362487.
Article
45. Takada-Takatori Y, Kume T, Ohgi Y, Fujii T, Niidome T, Sugimoto H, Akaike A. 2008; Mechanisms of alpha7-nicotinic receptor up-regulation and sensitization to donepezil induced by chronic donepezil treatment. Eur J Pharmacol. 590:150–156. DOI: 10.1016/j.ejphar.2008.06.027. PMID: 18585378.
46. Votin V, Nelson WJ, Barth AI. 2005; Neurite outgrowth involves adenomatous polyposis coli protein and beta-catenin. J Cell Sci. 118(Pt 24):5699–5708. DOI: 10.1242/jcs.02679. PMID: 16303851. PMCID: PMC3373789.
47. Rössler OG, Giehl KM, Thiel G. 2004; Neuroprotection of immortalized hippocampal neurones by brain-derived neurotrophic factor and Raf-1 protein kinase: role of extracellular signal-regulated protein kinase and phosphatidylinositol 3-kinase. J Neurochem. 88:1240–1252. DOI: 10.1046/j.1471-4159.2003.02255.x. PMID: 15009680.
Article
48. Tapley P, Lamballe F, Barbacid M. 1992; K252a is a selective inhibitor of the tyrosine protein kinase activity of the trk family of oncogenes and neurotrophin receptors. Oncogene. 7:371–381. PMID: 1312698.
49. Torii K, Nishizawa K, Kawasaki A, Yamashita Y, Katada M, Ito M, Nishimoto I, Terashita K, Aiso S, Matsuoka M. 2008; Anti-apoptotic action of Wnt5a in dermal fibroblasts is mediated by the PKA signaling pathways. Cell Signal. 20:1256–1266. DOI: 10.1016/j.cellsig.2008.02.013. PMID: 18407462.
Article
50. Takada-Takatori Y, Kume T, Ohgi Y, Fujii T, Niidome T, Sugimoto H, Akaike A. 2008; Mechanisms of alpha7-nicotinic receptor up-regulation and sensitization to donepezil induced by chronic donepezil treatment. Eur J Pharmacol. 590:150–156. DOI: 10.1016/j.ejphar.2008.06.027. PMID: 18585378.
51. Mantamadiotis T, Lemberger T, Bleckmann SC, Kern H, Kretz O, Martin Villalba A, Tronche F, Kellendonk C, Gau D, Kapfhammer J, Otto C, Schmid W, Schütz G. 2002; Disruption of CREB function in brain leads to neurodegeneration. Nat Genet. 31:47–54. DOI: 10.1038/ng882. PMID: 11967539.
Article
52. Noriega LG, Feige JN, Canto C, Yamamoto H, Yu J, Herman MA, Mataki C, Kahn BB, Auwerx J. 2011; CREB and ChREBP oppositely regulate SIRT1 expression in response to energy availability. EMBO Rep. 12:1069–1076. DOI: 10.1038/embor.2011.151. PMID: 21836635. PMCID: PMC3185337.
Article
53. Kosasa T, Kuriya Y, Matsui K, Yamanishi Y. 1999; Effect of donepezil hydrochloride (E2020) on basal concentration of extracellular acetylcholine in the hippocampus of rats. Eur J Pharmacol. 380:101–107. DOI: 10.1016/S0014-2999(99)00545-2. PMID: 10513568.
54. Kihara T, Shimohama S, Sawada H, Kimura J, Kume T, Kochiyama H, Maeda T, Akaike A. 1997; Nicotinic receptor stimulation protects neurons against beta-amyloid toxicity. Ann Neurol. 42:159–163. DOI: 10.1002/ana.410420205. PMID: 9266724.
55. Hashimoto M, Kazui H, Matsumoto K, Nakano Y, Yasuda M, Mori E. 2005; Does donepezil treatment slow the progression of hippocampal atrophy in patients with Alzheimer's disease? Am J Psychiatry. 162:676–682. DOI: 10.1176/appi.ajp.162.4.676. PMID: 15800138.
Article
56. Kimura M, Akasofu S, Ogura H, Sawada K. 2005; Protective effect of donepezil against Abeta(1-40) neurotoxicity in rat septal neurons. Brain Res. 1047:72–84. DOI: 10.1016/j.brainres.2005.04.014. PMID: 15893738.
57. Zimmermann M, Gardoni F, Marcello E, Colciaghi F, Borroni B, Padovani A, Cattabeni F, Di Luca M. 2004; Acetylcholinesterase inhibitors increase ADAM10 activity by promoting its trafficking in neuroblastoma cell lines. J Neurochem. 90:1489–1499. DOI: 10.1111/j.1471-4159.2004.02680.x. PMID: 15341532.
Article
58. Zhang SJ, Xu TT, Li L, Xu YM, Qu ZL, Wang XC, Huang SQ, Luo Y, Luo NC, Lu P, Shi YF, Yang X, Wang Q. 2017; Bushen-Yizhi formula ameliorates cognitive dysfunction through SIRT1/ER stress pathway in SAMP8 mice. Oncotarget. 8:49338–49350. DOI: 10.18632/oncotarget.17638. PMID: 28521305. PMCID: PMC5564772.
Article
Full Text Links
  • KJPP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr