Generic placeholder image

Current Reviews in Clinical and Experimental Pharmacology

Editor-in-Chief

ISSN (Print): 2772-4328
ISSN (Online): 2772-4336

Research Article

Berberine Nanoencapsulation Attenuates Hallmarks of Scoplomine Induced Alzheimer's-Like Disease in Rats

Author(s): Samar R. Saleh, Mariam M. Abady, Mohammed Nofal, Nashwa W. Yassa, Mohamed S. Abdel-Latif, Mohamed Ismail Nounou*, Doaa A. Ghareeb and Nihad Abdel-Monaem

Volume 16, Issue 2, 2021

Published on: 28 June, 2020

Page: [139 - 154] Pages: 16

DOI: 10.2174/1574884715666200628112844

Price: $65

Abstract

Background: Berberine (BBR), an isoquinoline alkaloid, acts as a multipotent active pharmaceutical ingredient to counteract several types of dementia based on its numerous pharmacological actions including antioxidant, anti-inflammatory, cholesterol-lowering effect, and inhibition of Aβ production and AChE. However, BBR suffers from poor absorption, bioavailability and brain drug uptake. The present study is directed for the formulation and characterization of Chitosan BBR-Nanoparticles (BBR-NPs) as well as the estimation of its neuroprotective effects against scopolamine induced cognitive impairments.

Methods: BBR-NPs were formulated using the ionic gelation method, and tripolyphosphate was chosen as a crosslinker. Nanoparticles size, zeta potential, encapsulation efficiency and releasing profile were estimated. To investigate the neuroprotective effects, adult fifty-six Wistar male rats were randomly distributed into three control groups, received saline, polyethylene glycol or Chitosan- NPs, respectively; induced group, received scopolamine (2 mg/ kg, i.p.) and three treated groups were orally administrated BBR (50 mg/ kg), BBR- NP (7 mg/ kg) and donepezil (2.25 mg/ kg, as positive control) followed by scopolamine injection after 40 min, daily for 4 weeks. Morris water maze test, oxidative stress parameters, cholinergic and amyloid-β processing intermediates, as well as neuroplasticity markers and histopathological examination were assessed.

Results: Our results showed that BBR- NPs were better than BBR and donepezil as BBR- NPs were powerful inhibitory ligands towards AChE and Aβ42 formation and significantly down-regulated Tau, iNOS and BACE gene expression in rats’ hippocampus. BBR-NPs administration, at 1/6 of BBR therapeutic recommended dose, significantly improved learning and memory function. This could be accredited to the diminution of oxidative stress and amyloid-β toxicity in addition to the improvement of the neuroplasticity markers.

Conclusion: The enhancing effect of BBR- NPs could be related to the enhancing of its bioavailability, absorption and brain drug uptake, which need more investigation in future work.

Keywords: Scopolamine, acetylcholinesterase, berberine, oxidative stress, amyloid-β, morris water maze test, neuroplasticity, chitosan.

Graphical Abstract
[1]
Huang M, Jiang X, Liang Y, Liu Q, Chen S, Guo Y. Berberine improves cognitive impairment by promoting autophagic clearance and inhibiting production of β-amyloid in APP/tau/PS1 mouse model of Alzheimer’s disease. Exp Gerontol 2017; 91: 25-33.
[http://dx.doi.org/10.1016/j.exger.2017.02.004] [PMID: 28223223]
[2]
Soudi SA, Nounou MI, Sheweita SA, Ghareeb DA, Younis LK, El-Khordagui LK. Protective effect of surface-modified berberine nanoparticles against LPS-induced neurodegenerative changes: A preclinical study. Drug Deliv Transl Res 2019; 9(5): 906-19.
[http://dx.doi.org/10.1007/s13346-019-00626-1] [PMID: 30868509]
[3]
Huang M, Chen S, Liang Y, Guo Y. The role of berberine in the multi-target treatment of senile dementia. Curr Top Med Chem 2016; 16(8): 867-73.
[http://dx.doi.org/10.2174/1568026615666150827095433] [PMID: 26311424]
[4]
Cai Z, Wang C, Yang W. Role of berberine in Alzheimer’s disease. Neuropsychiatr Dis Treat 2016; 12: 2509-20.
[http://dx.doi.org/10.2147/NDT.S114846] [PMID: 27757035]
[5]
Ji HF, Shen L. Berberine: A potential multipotent natural product to combat Alzheimer’s disease. Molecules 2011; 16(8): 6732-40.
[http://dx.doi.org/10.3390/molecules16086732] [PMID: 21829148]
[6]
Liu X, Zhou J, Abid MD, et al. Berberine attenuates axonal transport impairment and axonopathy induced by Calyculin A in N2a cells. PLoS One 2014; 9(4): e93974.
[http://dx.doi.org/10.1371/journal.pone.0093974] [PMID: 24713870]
[7]
Wang X, Wang R, Xing D, et al. Kinetic difference of berberine between hippocampus and plasma in rat after intravenous administration of Coptidis rhizoma extract. Life Sci 2005; 77(24): 3058-67.
[http://dx.doi.org/10.1016/j.lfs.2005.02.033] [PMID: 15996686]
[8]
Amat-Ur-Rasool H, Ahmed M. Designing second generation anti-alzheimer compounds as inhibitors of human acetylcholinesterase: Computational screening of synthetic molecules and dietary phytochemicals. PLoS One 2015; 10(9): e0136509.
[http://dx.doi.org/10.1371/journal.pone.0136509] [PMID: 26325402]
[9]
Tan X-S, Ma J-Y, Feng R, et al. Tissue distribution of berberine and its metabolites after oral administration in rats. PLoS One 2013; 8(10): e77969.
[http://dx.doi.org/10.1371/journal.pone.0077969] [PMID: 24205048]
[10]
Rad SZK, Rameshrad M, Hosseinzadeh H. Toxicology effects of Berberis vulgaris (barberry) and its active constituent, berberine: A review. Iran J Basic Med Sci 2017; 20(5): 516-29.
[PMID: 28656087]
[11]
Tan W, Li Y, Chen M, Wang Y. Berberine hydrochloride: Anticancer activity and nanoparticulate delivery system. Int J Nanomedicine 2011; 6: 1773-7.
[http://dx.doi.org/10.2147/IJN.S22683] [PMID: 21931477]
[12]
Gunasekaran T, Haile T, Nigusse T, Dhanaraju MD. Nanotechnology: An effective tool for enhancing bioavailability and bioactivity of phytomedicine. Asian Pac J Trop Biomed 2014; 4(Suppl. 1): S1-7.
[http://dx.doi.org/10.12980/APJTB.4.2014C980] [PMID: 25183064]
[13]
Wang Z, Li Y, Zhou Q, Wang Y, Chen T. Berberine-loaded solid lipid nanoparticles enhance hypoglycemic efficacy on streptozotocin-induced diabetic C57BL/6 mice. Nanomedicine (Lond) 2016; 12(2): 483-4.
[http://dx.doi.org/10.1016/j.nano.2015.12.108] [PMID: 27771431]
[14]
Xue M, Yang MX, Zhang W, et al. Characterization, pharmacokinetics, and hypoglycemic effect of berberine loaded solid lipid nanoparticles. Int J Nanomedicine 2013; 8: 4677-87.
[http://dx.doi.org/10.2147/IJN.S51262] [PMID: 24353417]
[15]
Li J, Yang L, Shen R, et al. Self-nanoemulsifying system improves oral absorption and enhances anti-acute myeloid leukemia activity of berberine. J Nanobiotechnology 2018; 16(1): 76-6.
[http://dx.doi.org/10.1186/s12951-018-0402-x] [PMID: 30290822]
[16]
Wu SJ, Don TM, Lin CW, Mi FL. Delivery of berberine using chitosan/fucoidan-taurine conjugate nanoparticles for treatment of defective intestinal epithelial tight junction barrier. Mar Drugs 2014; 12(11): 5677-97.
[http://dx.doi.org/10.3390/md12115677] [PMID: 25421323]
[17]
Zhang Y, Sun T, Jiang C. Biomacromolecules as carriers in drug delivery and tissue engineering. Acta Pharm Sin B 2017.
[PMID: 29872621]
[18]
Yu S, Xu X, Feng J, Liu M, Hu K. Chitosan and chitosan coating nanoparticles for the treatment of brain disease. Int J Pharm 2019; 560: 282-93.
[http://dx.doi.org/10.1016/j.ijpharm.2019.02.012] [PMID: 30772458]
[19]
Shimoda J, Onishi H, Machida Y. Bioadhesive characteristics of chitosan microspheres to the mucosa of rat small intestine. Drug Dev Ind Pharm 2001; 27(6): 567-76.
[http://dx.doi.org/10.1081/DDC-100105182] [PMID: 11548864]
[20]
Pan Y, Li YJ, Zhao HY, et al. Bioadhesive polysaccharide in protein delivery system: Chitosan nanoparticles improve the intestinal absorption of insulin in vivo. Int J Pharm 2002; 249(1-2): 139-47.
[http://dx.doi.org/10.1016/S0378-5173(02)00486-6] [PMID: 12433442]
[21]
Zhang H, Huang X, Sun Y, Xing J, Yamamoto A, Gao Y. Absorption-improving effects of chitosan oligomers based on their mucoadhesive properties: A comparative study on the oral and pulmonary delivery of calcitonin. Drug Deliv 2016; 23(7): 2419-27.
[PMID: 25594571]
[22]
Chen B, Li J, Borgens RB. Neuroprotection by chitosan nanoparticles in oxidative stress-mediated injury. BMC Res Notes 2018; 11(1): 49-9.
[http://dx.doi.org/10.1186/s13104-018-3162-7] [PMID: 29351805]
[23]
Sahin A, Yoyen-Ermis D, Caban-Toktas S, et al. Evaluation of brain-targeted chitosan nanoparticles through blood-brain barrier cerebral microvessel endothelial cells. J Microencapsul 2017; 34(7): 659-66.
[http://dx.doi.org/10.1080/02652048.2017.1375039] [PMID: 28862080]
[24]
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BioMed Res Int 2014; 2014: 869269.
[http://dx.doi.org/10.1155/2014/869269] [PMID: 25136634]
[25]
Songjiang Z, Lixiang W. Amyloid-beta associated with chitosan nano-carrier has favorable immunogenicity and permeates the BBB. AAPS PharmSciTech 2009; 10(3): 900-5.
[http://dx.doi.org/10.1208/s12249-009-9279-1] [PMID: 19609682]
[26]
Fazil M, Md S, Haque S, et al. Development and evaluation of rivastigmine loaded chitosan nanoparticles for brain targeting. Eur J Pharm Sci 2012; 47(1): 6-15.
[http://dx.doi.org/10.1016/j.ejps.2012.04.013] [PMID: 22561106]
[27]
Wilson B, Samanta MK, Santhi K, Kumar KPS, Ramasamy M, Suresh B. Chitosan nanoparticles as a new delivery system for the anti-Alzheimer drug tacrine. Nanomedicine (Lond) 2010; 6(1): 144-52.
[http://dx.doi.org/10.1016/j.nano.2009.04.001] [PMID: 19446656]
[28]
Wang X, Chi N, Tang X. Preparation of estradiol chitosan nanoparticles for improving nasal absorption and brain targeting Eur J pharmaceut biopharmaceut 2008; 70(3): 735-40.
[http://dx.doi.org/10.1016/j.ejpb.2008.07.005]
[29]
Fàbregas A, Miñarro M, García-Montoya E, et al. Impact of physical parameters on particle size and reaction yield when using the ionic gelation method to obtain cationic polymeric chitosan-tripolyphosphate nanoparticles. Int J Pharm 2013; 446(1-2): 199-204.
[http://dx.doi.org/10.1016/j.ijpharm.2013.02.015] [PMID: 23434543]
[30]
Stoica R, Şomoghi R, Ion RM. Preparation of chitosan – tripolyphosphate nanoparticles for the encapsulation of polyphenols extracted from rose hips. Dig J Nanomater Biostruct 2013; 8(3): 955-63.
[31]
Lee B, Sur B, Shim I, Lee H, Hahm DH. Phellodendron amurense and its major alkaloid compound, berberine ameliorates scopolamine-induced neuronal impairment and memory dysfunction in rats. Korean J Physiol Pharmacol 2012; 16(2): 79-89.
[http://dx.doi.org/10.4196/kjpp.2012.16.2.79] [PMID: 22563252]
[32]
Ghareeb DA, Khalil S, Hafez HS, et al. Berberine reduces neurotoxicity related to nonalcoholic steatohepatitis in rats. Evid Based Complement Alternat Med 2015; 2015: 361847.
[http://dx.doi.org/10.1155/2015/361847] [PMID: 26576191]
[33]
Hafez HS, Ghareeb DA, Saleh SR, et al. Neuroprotective effect of ipriflavone against scopolamine-induced memory impairment in rats. Psychopharmacology (Berl) 2017; 234(20): 3037-53.
[http://dx.doi.org/10.1007/s00213-017-4690-x] [PMID: 28733814]
[34]
Hussien HM, Abd-Elmegied A, Ghareeb DA, Hafez HS, Ahmed HEA, El-Moneam NA. Neuroprotective effect of berberine against environmental heavy metals-induced neurotoxicity and Alzheimer’s-like disease in rats. Food Chem Toxicol 2018; 111: 432-44.
[http://dx.doi.org/10.1016/j.fct.2017.11.025] [PMID: 29170048]
[35]
Giese KP, Friedman E, Telliez JB, et al. Hippocampus-dependent learning and memory is impaired in mice lacking the Ras-Guanine-nucleotide Releasing Factor 1 (Ras-GRF1). Neuropharmacology 2001; 41(6): 791-800.
[http://dx.doi.org/10.1016/S0028-3908(01)00096-X] [PMID: 11640934]
[36]
Montgomery H, Dymock JF. "Determination of nitrite in water", Royal Soc Chemistry Thomas Graham House, Science Park, Milton RD, Cambridge CB4 0WF, Cambridge: England, 1961; p. 414.
[37]
Tappel AL, Zalkin H. Inhibition of lipide peroxidation in mitochondria by vitamin E. Arch Biochem Biophys 1959; 80(2): 333-6.
[http://dx.doi.org/10.1016/0003-9861(59)90259-0]
[38]
Ellman GL. Tissue sulfhydryl groups. Arch Biochem Biophys 1959; 82(1): 70-7.
[http://dx.doi.org/10.1016/0003-9861(59)90090-6] [PMID: 13650640]
[39]
Habig WH, Pabst MJ, Jakoby WB. Glutathione S-transferases. The first enzymatic step in mercapturic acid formation. J Biol Chem 1974; 249(22): 7130-9.
[PMID: 4436300]
[40]
Paglia DE, Valentine WN. Studies on the quantitative and qualitative characterization of erythrocyte glutathione peroxidase. J Lab Clin Med 1967; 70(1): 158-69.
[PMID: 6066618]
[41]
Lowry OH, Rosebrough NJ, Farr AL, Randall RJ. Protein measurement with the Folin phenol reagent. J Biol Chem 1951; 193(1): 265-75.
[PMID: 14907713]
[42]
Ellman GL, Courtney KD, Andres V Jr, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961; 7: 88-95.
[http://dx.doi.org/10.1016/0006-2952(61)90145-9] [PMID: 13726518]
[43]
Motulsky H. Intuitive biostatistics: A nonmathematical guide to statistical thinking. New York: Oxford University Press 2010.
[44]
Kole S, Qadiri SSN, Shin SM, Kim WS, Lee J, Jung SJ. Nanoencapsulation of inactivated-viral vaccine using chitosan nanoparticles: Evaluation of its protective efficacy and immune modulatory effects in olive flounder (Paralichthys olivaceus) against Viral Haemorrhagic Septicaemia Virus (VHSV) infection. Fish Shellfish Immunol 2019; 91: 136-47.
[http://dx.doi.org/10.1016/j.fsi.2019.05.017] [PMID: 31096061]
[45]
Hassan EE, Gallo JM. Targeting anticancer drugs to the brain. I: Enhanced brain delivery of oxantrazole following administration in magnetic cationic microspheres. J Drug Target 1993; 1(1): 7-14.
[http://dx.doi.org/10.3109/10611869308998759] [PMID: 8069547]
[46]
Hombach J, Bernkop-Schnürch A. Chitosan solutions and particles: Evaluation of their permeation enhancing potential on MDCK cells used as blood brain barrier model. Int J Pharm 2009; 376(1-2): 104-9.
[http://dx.doi.org/10.1016/j.ijpharm.2009.04.027] [PMID: 19409469]
[47]
Kuo YC, Rajesh R. Targeted delivery of rosmarinic acid across the blood-brain barrier for neuronal rescue using polyacrylamide-chitosan-poly(lactide-co-glycolide) nanoparticles with surface cross-reacting material 197 and apolipoprotein E. Int J Pharm 2017; 528(1-2): 228-41.
[http://dx.doi.org/10.1016/j.ijpharm.2017.05.039] [PMID: 28549973]
[48]
Roy Chowdhury S, Mondal S, Muthuraj B, Balaji SN, Trivedi V, Krishnan Iyer P. Remarkably efficient blood-brain barrier crossing polyfluorene-chitosan nanoparticle selectively tweaks amyloid oligomer in cerebrospinal fluid and Aβ1-40. ACS Omega 2018; 3(7): 8059-66.
[http://dx.doi.org/10.1021/acsomega.8b00764] [PMID: 30087934]
[49]
Piazzini V, Landucci E, D’Ambrosio M, et al. Chitosan coated human serum albumin nanoparticles: A promising strategy for nose-to-brain drug delivery. Int J Biol Macromol 2019; 129: 267-80.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.02.005] [PMID: 30726749]
[50]
Qureshi M, Aqil M, Imam SS, Ahad A, Sultana Y. Formulation and evaluation of neuroactive drug loaded chitosan nanoparticle for nose to brain delivery: In-vitro characterization and in-vivo behavior study. Curr Drug Deliv 2019; 16(2): 123-35.
[http://dx.doi.org/10.2174/1567201815666181011121750] [PMID: 30317997]
[51]
Tzeyung AS, Md S, Bhattamisra SK, et al. Fabrication, optimization, and evaluation of rotigotine-loaded chitosan nanoparticles for nose-to-brain delivery. Pharmaceutics 2019; 11(1): E26.
[http://dx.doi.org/10.3390/pharmaceutics11010026] [PMID: 30634665]
[52]
Lochner M, Thompson AJ. The muscarinic antagonists scopolamine and atropine are competitive antagonists at 5-HT3 receptors. Neuropharmacology 2016; 108: 220-8.
[http://dx.doi.org/10.1016/j.neuropharm.2016.04.027] [PMID: 27108935]
[53]
Bajo R, Pusil S, López ME, et al. Scopolamine effects on functional brain connectivity: A pharmacological model of Alzheimer’s disease. Sci Rep 2015; 5: 9748.
[http://dx.doi.org/10.1038/srep09748] [PMID: 26130273]
[54]
Lee JC, Kim IH, Cho JH, et al. Vanillin improves scopolamine induced memory impairment through restoration of ID1 expression in the mouse hippocampus. Mol Med Rep 2018; 17(3): 4399-405.
[http://dx.doi.org/10.3892/mmr.2018.8401] [PMID: 29328430]
[55]
Zhu F, Qian C. Berberine chloride can ameliorate the spatial memory impairment and increase the expression of interleukin-1beta and inducible nitric oxide synthase in the rat model of Alzheimer’s disease. BMC Neurosci 2006; 7: 78.
[http://dx.doi.org/10.1186/1471-2202-7-78] [PMID: 17137520]
[56]
Balbaa M, Abdulmalek SA, Khalil S. Oxidative stress and expression of insulin signaling proteins in the brain of diabetic rats: Role of Nigella sativa oil and antidiabetic drugs. PLoS One 2017; 12(5): e0172429.
[http://dx.doi.org/10.1371/journal.pone.0172429] [PMID: 28505155]
[57]
Venkatesan R, Subedi L, Yeo EJ, Kim SY. Lactucopicrin ameliorates oxidative stress mediated by scopolamine-induced neurotoxicity through activation of the NRF2 pathway. Neurochem Int 2016; 99: 133-46.
[http://dx.doi.org/10.1016/j.neuint.2016.06.010] [PMID: 27346436]
[58]
Lee G-Y, Lee C, Park GH, Jang J-H. Amelioration of scopolamine-induced learning and memory impairment by α-Pinene in C57BL/6 mice. Evid Based Complementary Altern Med 2017; 2017: 4926815.
[59]
Budzynska B, Boguszewska-Czubara A, Kruk-Slomka M, et al. Effects of imperatorin on scopolamine-induced cognitive impairment and oxidative stress in mice. Psychopharmacology (Berl) 2015; 232(5): 931-42.
[http://dx.doi.org/10.1007/s00213-014-3728-6] [PMID: 25189792]
[60]
El-Sherbiny DA, Khalifa AE, Attia AS. Eldenshary Eel-D. Hypericum perforatum extract demonstrates antioxidant properties against elevated rat brain oxidative status induced by amnestic dose of scopolamine. Pharmacol Biochem Behav 2003; 76(3-4): 525-33.
[http://dx.doi.org/10.1016/j.pbb.2003.09.014] [PMID: 14643852]
[61]
Jung HA, Min BS, Yokozawa T, Lee JH, Kim YS, Choi JS. Anti-Alzheimer and antioxidant activities of Coptidis rhizoma alkaloids. Biol Pharm Bull 2009; 32(8): 1433-8.
[http://dx.doi.org/10.1248/bpb.32.1433] [PMID: 19652386]
[62]
Jiang B, Song L, Huang C, Zhang W. P7C3 Attenuates the scopolamine-induced memory impairments in C57BL/6J mice. Neurochem Res 2016; 41(5): 1010-9.
[http://dx.doi.org/10.1007/s11064-015-1783-y] [PMID: 26646000]
[63]
El Khoury NB, Gratuze M, Papon MA, Bretteville A, Planel E. Insulin dysfunction and Tau pathology. Front Cell Neurosci 2014; 8: 22.
[http://dx.doi.org/10.3389/fncel.2014.00022] [PMID: 24574966]
[64]
Tang W-J. Targeting insulin-degrading enzyme to treat type 2 diabetes. Trends in Trends Endrocrinol Metab 2016; 27(1): 24-34.
[PMID: 26651592]
[65]
Russell RL, Siedlak SL, Raina AK, Bautista JM, Smith MA, Perry G. Increased neuronal glucose-6-phosphate dehydrogenase and sulfhydryl levels indicate reductive compensation to oxidative stress in Alzheimer disease. Arch Biochem Biophys 1999; 370(2): 236-9.
[http://dx.doi.org/10.1006/abbi.1999.1404] [PMID: 10510282]
[66]
Brorson JR, Schumacker PT, Zhang H. The Committees on Neurobiology and Cell Physiology. Nitric oxide acutely inhibits neuronal energy production. J Neurosci 1999; 19(1): 147-58.
[http://dx.doi.org/10.1523/JNEUROSCI.19-01-00147.1999] [PMID: 9870946]
[67]
Mietelska-Porowska A, Wasik U, Goras M, Filipek A, Niewiadomska G. Tau protein modifications and interactions: Their role in function and dysfunction. Int J Mol Sci 2014; 15(3): 4671-713.
[http://dx.doi.org/10.3390/ijms15034671] [PMID: 24646911]
[68]
Kulkarni KS, Kasture SB, Mengi SA. Efficacy study of Prunus amygdalus (almond) nuts in scopolamine-induced amnesia in rats. Indian J Pharmacol 2010; 42(3): 168-73.
[http://dx.doi.org/10.4103/0253-7613.66841] [PMID: 20871769]
[69]
Kolář D, Wimmerová L, Kádek R. Acetylcholinesterase and Butyrylcholinesterase inhibitory activities of Berberis vulgaris. Phytopharmacology 2010; 1(1): 7-11.
[70]
Ji HF, Shen L. Molecular basis of inhibitory activities of berberine against pathogenic enzymes in Alzheimer’s disease. Scientific World J 2012; 2012: 823201.
[http://dx.doi.org/10.1100/2012/823201] [PMID: 22262957]
[71]
Abd El-Wahab AE, Ghareeb DA, Sarhan EE, Abu-Serie MM, El Demellawy MA. In vitro biological assessment of Berberis vulgaris and its active constituent, berberine: Antioxidants, anti-acetylcholinesterase, anti-diabetic and anticancer effects. BMC Complement Altern Med 2013; 13: 218.
[http://dx.doi.org/10.1186/1472-6882-13-218] [PMID: 24007270]
[72]
Zimmermann M, Gardoni F, Marcello E, et al. Acetylcholinesterase inhibitors increase ADAM10 activity by promoting its trafficking in neuroblastoma cell lines. J Neurochem 2004; 90(6): 1489-99.
[http://dx.doi.org/10.1111/j.1471-4159.2004.02680.x] [PMID: 15341532]
[73]
Endres K, Deller T. Regulation of alpha-secretase ADAM10 in vitro and in vivo: Genetic, epigenetic, and protein-based mechanisms. Front Mol Neurosci 2017; 10: 56.
[http://dx.doi.org/10.3389/fnmol.2017.00056] [PMID: 28367112]
[74]
Zhu F, Wu F, Ma Y, et al. Decrease in the production of β-amyloid by berberine inhibition of the expression of β-secretase in HEK293 cells. BMC Neurosci 2011; 12: 125.
[http://dx.doi.org/10.1186/1471-2202-12-125] [PMID: 22152059]
[75]
Salehi S, Filtz TM. Berberine possesses muscarinic agonist-like properties in cultured rodent cardiomyocytes. Pharmacol Res 2011; 63(4): 335-40.
[http://dx.doi.org/10.1016/j.phrs.2010.12.004]
[76]
Wolozin B. Cholesterol and the biology of Alzheimer’s disease. Neuron 2004; 41(1): 7-10.
[http://dx.doi.org/10.1016/S0896-6273(03)00840-7] [PMID: 14715130]
[77]
Xiu J, Nordberg A, Qi X, Guan ZZ. Influence of cholesterol and lovastatin on alpha-form of secreted amyloid precursor protein and expression of alpha7 nicotinic receptor on astrocytes. Neurochem Int 2006; 49(5): 459-65.
[http://dx.doi.org/10.1016/j.neuint.2006.03.007] [PMID: 16675062]
[78]
Yan N, Yan Y, Cai Z. Statins are protective against beta-amyloid pathology in Alzheimer's disease? 2013. Corpus ID: 17210782
[79]
Kong W, Wei J, Abidi P, et al. Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med 2004; 10(12): 1344-51.
[http://dx.doi.org/10.1038/nm1135] [PMID: 15531889]
[80]
Yu G, Li Y, Tian Q, et al. Berberine attenuates calyculin A-induced cytotoxicity and Tau hyperphosphorylation in HEK293 cells. J Alzheimers Dis 2011; 24(3): 525-35.
[http://dx.doi.org/10.3233/JAD-2011-101779] [PMID: 21297267]
[81]
Tyler WJ, Alonso M, Bramham CR, Pozzo-Miller LD. From acquisition to consolidation: on the role of brain-derived neurotrophic factor signaling in hippocampal-dependent learning. Learn Mem 2002; 9(5): 224-37.
[http://dx.doi.org/10.1101/lm.51202] [PMID: 12359832]
[82]
Xu J, Rong S, Xie B, et al. Memory impairment in cognitively impaired aged rats associated with decreased hippocampal CREB phosphorylation: Reversal by procyanidins extracted from the lotus seedpod. J Gerontol A Biol Sci Med Sci 2010; 65(9): 933-40.
[http://dx.doi.org/10.1093/gerona/glq094] [PMID: 20530246]
[83]
Prior M, Dargusch R, Ehren JL, Chiruta C, Schubert D. The neurotrophic compound J147 reverses cognitive impairment in aged Alzheimer’s disease mice. Alzheimers Res Ther 2013; 5(3): 25.
[http://dx.doi.org/10.1186/alzrt179] [PMID: 23673233]

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