Generic placeholder image

Current Vascular Pharmacology

Editor-in-Chief

ISSN (Print): 1570-1611
ISSN (Online): 1875-6212

Review Article

Rhizoma Coptidis for Alzheimer’s Disease and Vascular Dementia: A Literature Review

Author(s): Zhiyong Wang, Yang Yang, Meixia Liu, Yun Wei, Jiangang Liu, Hui Pei and Hao Li*

Volume 18, Issue 4, 2020

Page: [358 - 368] Pages: 11

DOI: 10.2174/1570161117666190710151545

Price: $65

Abstract

Background: Alzheimer’s disease (AD) and vascular dementia (VaD) are major types of dementia, both of which cause heavy economic burdens for families and society. However, no currently available medicines can control dementia progression. Rhizoma coptidis, a Chinese herbal medicine, has been used for >2000 years and is now gaining attention as a potential treatment for AD and VaD.

Methods: We reviewed the mechanisms of the active ingredients of Rhizoma coptidis and Rhizoma coptidis-containing Chinese herbal compounds in the treatment of AD and VaD. We focused on studies on ameliorating the risk factors and the pathological changes of these diseases.

Results: The Rhizoma coptidis active ingredients include berberine, palmatine, coptisine, epiberberine, jatrorrhizine and protopine. The most widely studied ingredient is berberine, which has extensive therapeutic effects on the risk factors and pathogenesis of dementia. It can control blood glucose and lipid levels, regulate blood pressure, ameliorate atherosclerosis, inhibit cholinesterase activity, Aβ generation, and tau hyperphosphorylation, decrease neuroinflammation and oxidative stress and alleviate cognitive impairment. Other ingredients (such as jatrorrhizine, coptisine, epiberberine and palmatine) also regulate blood lipids and blood pressure; however, there are relatively few studies on them. Rhizoma coptidis-containing Chinese herbal compounds like Huanglian-Jie-Du-Tang, Huanglian Wendan Decoction, Banxia Xiexin Decoction and Huannao Yicong Formula have anti-inflammatory and antioxidant stress activities, regulate insulin signaling, inhibit γ-secretase activity, neuronal apoptosis, tau hyperphosphorylation, and Aβ deposition, and promote neural stem cell differentiation, thereby improving cognitive function.

Conclusion: The “One-Molecule, One-Target” paradigm has suffered heavy setbacks, but a “multitarget- directed ligands” strategy may be viable. Rhizoma coptidis active ingredients and Rhizoma coptidiscontaining Chinese herbal compounds have multi-aspect therapeutic effects on AD and VaD.

Keywords: Rhizoma coptidis, Chinese herbal medicine, berberine, Alzheimer's disease, vascular dementia, mental disease.

Graphical Abstract
[1]
Bacigalupo I, Mayer F, Lacorte E, et al. A systematic review and meta-analysis on the prevalence of dementia in Europe: estimates from the highest-quality studies adopting the DSM IV diagnostic criteria. J Alzheimers Dis 2018; 66(4): 1471-81.
[http://dx.doi.org/10.3233/JAD-180416] [PMID: 30412486]
[2]
Rizzi L, Rosset I, Roriz-Cruz M. Global epidemiology of dementia: Alzheimer’s and vascular types. BioMed Res Int 2014; 2014 908915
[http://dx.doi.org/10.1155/2014/908915] [PMID: 25089278]
[3]
Jia J, Wang F, Wei C, et al. The prevalence of dementia in urban and rural areas of China. Alzheimers Dement 2014; 10(1): 1-9.
[http://dx.doi.org/10.1016/j.jalz.2013.01.012] [PMID: 23871765]
[4]
Alzheimer Association. 2018 Alzheimer’s disease facts and figures. Alzheimers Dement 2018; 14: 367-429.
[http://dx.doi.org/10.1016/j.jalz.2018.02.001]
[5]
Qi L, Ma Y, Zhong F, Shen C. Comprehensive quality assessment for Rhizoma Coptidis based on quantitative and qualitative metabolic profiles using high performance liquid chromatography, Fourier transform near-infrared and Fourier transform mid-infrared combined with multivariate statistical analysis. J Pharm Biomed Anal 2018; 161: 436-43.
[http://dx.doi.org/10.1016/j.jpba.2018.09.012] [PMID: 30216792]
[6]
Ma BL, Ma YM. Pharmacokinetic properties, potential herb-drug interactions and acute toxicity of oral Rhizoma coptidis alkaloids. Expert Opin Drug Metab Toxicol 2013; 9(1): 51-61.
[http://dx.doi.org/10.1517/17425255.2012.722995] [PMID: 22998215]
[7]
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]
[8]
Meng FC, Wu ZF, Yin ZQ, Lin LG, Wang R, Zhang QW. Coptidis rhizoma and its main bioactive components: recent advances in chemical investigation, quality evaluation and pharmacological activity. Chin Med 2018; 13: 13.
[http://dx.doi.org/10.1186/s13020-018-0171-3] [PMID: 29541156]
[9]
Sun J, Ma JS, Jin J, et al. [Qualitative and quantitative determination of the main components of huanglianjiedu decoction by HPLC-UV/MS]. Yao Xue Xue Bao 2006; 41(4): 380-4.
[PMID: 16856488]
[10]
Fan J, Zhang K, Jin Y, et al. Pharmacological effects of berberine on mood disorders. J Cell Mol Med 2019; 23(1): 21-8.
[http://dx.doi.org/10.1111/jcmm.13930] [PMID: 30450823]
[11]
Pang B, Yu XT, Zhou Q, et al. Effect of Rhizoma coptidis (huang lian) on treating diabetes mellitus. Evid Based Complement Alternat Med 2015; 2015 921416
[http://dx.doi.org/10.1155/2015/921416] [PMID: 26508987]
[12]
Xing Y, Liu X, Lin Y, et al. Progress in pharmacological effects and clinical applications of berberine. Zhongguo Yaolixue Yu Dulixue Zazhi 2017; 31: 491-502.
[13]
Yao J, Kong W, Jiang J. Learning from berberine: Treating chronic diseases through multiple targets. Sci China Life Sci 2015; 58(9): 854-9.
[http://dx.doi.org/10.1007/s11427-013-4568-z] [PMID: 24174332]
[14]
Qian XC, Zhang L, Tao Y, et al. Simultaneous determination of ten alkaloids of crude and wine-processed Rhizoma Coptidis aqueous extracts in rat plasma by UHPLC-ESI-MS/MS and its application to a comparative pharmacokinetic study. J Pharm Biomed Anal 2015; 105: 64-73.
[http://dx.doi.org/10.1016/j.jpba.2014.11.049] [PMID: 25543284]
[15]
Zhang QS, Wang GW, Han ZQ, et al. Metabolic profile of Rhizoma coptidis in human plasma determined using ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry. Rapid Commun Mass Spectrom 2018; 32(1): 63-73.
[http://dx.doi.org/10.1002/rcm.7990] [PMID: 28926137]
[16]
Zhang Q, Wang G, Chen X, et al. Metabolism of Rhizoma coptidis in human urine by ultra-high-performance liquid chromatography coupled with high-resolution mass spectrometry. Eur J Drug Metab Pharmacokinet 2018; 43(4): 441-52.
[http://dx.doi.org/10.1007/s13318-018-0463-0] [PMID: 29450708]
[17]
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]
[18]
Su J, Miao Q, Miao P, et al. Pharmacokinetics and brain distribution and metabolite identification of coptisine, a protoberberine alkaloid with therapeutic potential for cns disorders, in rats. Biol Pharm Bull 2015; 38(10): 1518-28.
[http://dx.doi.org/10.1248/bpb.b15-00293] [PMID: 26228628]
[19]
Kalaria R. The pathology and pathophysiology of vascular dementia. Neuropharmacology 2018; 134(Pt B): 226-39.
[http://dx.doi.org/10.1016/j.neuropharm.2017.12.030]
[20]
Zhang J, Chen C, Hua S, et al. An updated meta-analysis of cohort studies: Diabetes and risk of Alzheimer’s disease. Diabetes Res Clin Pract 2017; 124: 41-7.
[http://dx.doi.org/10.1016/j.diabres.2016.10.024] [PMID: 28088029]
[21]
Gudala K, Bansal D, Schifano F, Bhansali A. Diabetes mellitus and risk of dementia: A meta-analysis of prospective observational studies. J Diabetes Investig 2013; 4(6): 640-50.
[http://dx.doi.org/10.1111/jdi.12087] [PMID: 24843720]
[22]
Katon W, Pedersen HS, Ribe AR, et al. Effect of depression and diabetes mellitus on the risk for dementia: a national population-based cohort study. JAMA Psychiatry 2015; 72(6): 612-9.
[http://dx.doi.org/10.1001/jamapsychiatry.2015.0082] [PMID: 25875310]
[23]
Biessels GJ, Reijmer YD. Brain changes underlying cognitive dysfunction in diabetes: what can we learn from MRI? Diabetes 2014; 63(7): 2244-52.
[http://dx.doi.org/10.2337/db14-0348] [PMID: 24931032]
[24]
Verdile G, Fuller SJ, Martins RN. The role of type 2 diabetes in neurodegeneration. Neurobiol Dis 2015; 84: 22-38.
[http://dx.doi.org/10.1016/j.nbd.2015.04.008] [PMID: 25926349]
[25]
Pasquier F, Boulogne A, Leys D, Fontaine P. Diabetes mellitus and dementia. Diabetes Metab 2006; 32(5 Pt 1): 403-14.
[http://dx.doi.org/10.1016/S1262-3636(07)70298-7] [PMID: 17110895]
[26]
Geng FH, Li GH, Zhang X, et al. Berberine improves mesenteric artery insulin sensitivity through up-regulating insulin receptor-mediated signalling in diabetic rats. Br J Pharmacol 2016; 173(10): 1569-79.
[http://dx.doi.org/10.1111/bph.13466] [PMID: 26914282]
[27]
Pérez-Rubio KG, González-Ortiz M, Martínez-Abundis E, Robles-Cervantes JA, Espinel-Bermúdez MC. Effect of berberine administration on metabolic syndrome, insulin sensitivity, and insulin secretion. Metab Syndr Relat Disord 2013; 11(5): 366-9.
[http://dx.doi.org/10.1089/met.2012.0183] [PMID: 23808999]
[28]
Yu Y, Hao G, Zhang Q, et al. Berberine induces GLP-1 secretion through activation of bitter taste receptor pathways. Biochem Pharmacol 2015; 97(2): 173-7.
[http://dx.doi.org/10.1016/j.bcp.2015.07.012] [PMID: 26206195]
[29]
Yue SJ, Liu J, Wang AT, et al. Berberine alleviates insulin resistance by reducing peripheral branched-chain amino acids. Am J Physiol Endocrinol Metab 2019; 316(1): E73-85.
[http://dx.doi.org/10.1152/ajpendo.00256.2018] [PMID: 30422704]
[30]
Wei S, Zhang M, Yu Y, et al. Berberine attenuates development of the hepatic gluconeogenesis and lipid metabolism disorder in type 2 diabetic mice and in palmitate-incubated HepG2 cells through suppression of the HNF-4α miR122 pathway. PLoS One 2016; 11(3) e0152097
[http://dx.doi.org/10.1371/journal.pone.0152097] [PMID: 27011261]
[31]
Chen Q, Mo R, Wu N, et al. Berberine ameliorates diabetes-associated cognitive decline through modulation of aberrant inflammation response and insulin signaling pathway in DM rats. Front Pharmacol 2017; 8: 334.
[http://dx.doi.org/10.3389/fphar.2017.00334] [PMID: 28634451]
[32]
Wang S, He B, Hang W, et al. Berberine alleviates tau hyperphosphorylation and axonopathy-associated with diabetic encephalopathy via restoring PI3K/Akt/GSK3β pathway. J Alzheimers Dis 2018; 65(4): 1385-400.
[http://dx.doi.org/10.3233/JAD-180497] [PMID: 30175975]
[33]
Yin S, Bai W, Li P, et al. Berberine suppresses the ectopic expression of miR-133a in endothelial cells to improve vascular dementia in diabetic rats. Clin Exp Hypertens 2018; 26: 1-9.
[PMID: 30472896]
[34]
Pegueroles J, Jiménez A, Vilaplana E, et al. Alzheimer’s Disease Neuroimaging Initiative. Obesity and Alzheimer’s disease, does the obesity paradox really exist? A magnetic resonance imaging study. Oncotarget 2018; 9(78): 34691-8.
[http://dx.doi.org/10.18632/oncotarget.26162] [PMID: 30410669]
[35]
Appleton JP, Scutt P, Sprigg N, Bath PM. Hypercholesterolaemia and vascular dementia. Clin Sci (Lond) 2017; 131(14): 1561-78.
[http://dx.doi.org/10.1042/CS20160382] [PMID: 28667059]
[36]
Pan ML, Hsu CC, Chen YM, Yu HK, Hu GC. Statin use and the risk of dementia in patients with stroke: a nationwide population-based cohort study. J Stroke Cerebrovasc Dis 2018; 27(11): 3001-7.
[http://dx.doi.org/10.1016/j.jstrokecerebrovasdis.2018.06.036] [PMID: 30087076]
[37]
Benseny-Cases N, Klementieva O, Cotte M, Ferrer I, Cladera J. Microspectroscopy (μFTIR) reveals co-localization of lipid oxidation and amyloid plaques in human Alzheimer disease brains. Anal Chem 2014; 86(24): 12047-54.
[http://dx.doi.org/10.1021/ac502667b] [PMID: 25415602]
[38]
Sripetchwandee J, Chattipakorn N, Chattipakorn SC. Links between obesity-induced brain insulin resistance, brain mitochondrial dysfunction, and dementia. Front Endocrinol (Lausanne) 2018; 9: 496.
[http://dx.doi.org/10.3389/fendo.2018.00496] [PMID: 30233495]
[39]
Frisardi V, Solfrizzi V, Seripa D, et al. Metabolic-cognitive syndrome: a cross-talk between metabolic syndrome and Alzheimer’s disease. Ageing Res Rev 2010; 9(4): 399-417.
[http://dx.doi.org/10.1016/j.arr.2010.04.007] [PMID: 20444434]
[40]
He K, Kou S, Zou Z, et al. Hypolipidemic effects of alkaloids from Rhizoma coptidis in diet-induced hyperlipidemic hamsters. Planta Med 2016; 82(8): 690-7.
[http://dx.doi.org/10.1055/s-0035-1568261] [PMID: 26848702]
[41]
Wang L, Peng LY, Wei GH, Ge H. Therapeutic effects of berberine capsule on patients with mild hyperlipidemia. Zhongguo Zhong Xi Yi Jie He Za Zhi 2016; 36(6): 681-4.
[PMID: 27491226]
[42]
Ju J, Li J, Lin Q, Xu H. Efficacy and safety of berberine for dyslipidaemias: A systematic review and meta-analysis of randomized clinical trials. Phytomedicine 2018; 50: 25-34.
[http://dx.doi.org/10.1016/j.phymed.2018.09.212] [PMID: 30466986]
[43]
Kou S, Han B, Wang Y, et al. Synergetic cholesterol-lowering effects of main alkaloids from Rhizoma Coptidis in HepG2 cells and hypercholesterolemia hamsters. Life Sci 2016; 151: 50-60.
[http://dx.doi.org/10.1016/j.lfs.2016.02.046] [PMID: 26876917]
[44]
Yang W, She L, Yu K, et al. Jatrorrhizine hydrochloride attenuates hyperlipidemia in a high-fat diet-induced obesity mouse model. Mol Med Rep 2016; 14(4): 3277-84.
[http://dx.doi.org/10.3892/mmr.2016.5634] [PMID: 27573054]
[45]
Zou ZY, Hu YR, Ma H, et al. Coptisine attenuates obesity-related inflammation through LPS/TLR-4-mediated signaling pathway in Syrian golden hamsters. Fitoterapia 2015; 105: 139-46.
[http://dx.doi.org/10.1016/j.fitote.2015.06.005] [PMID: 26073947]
[46]
Tadic M, Cuspidi C, Bombelli M, Facchetti R, Mancia G, Grassi G. Relationships between residual blood pressure variability and cognitive function in the general population of the PAMELA study. J Clin Hypertens (Greenwich) 2019; 21(1): 39-45.
[http://dx.doi.org/10.1111/jch.13428] [PMID: 30427125]
[47]
Jeon SY, Byun MS, Yi D, et al. Influence of hypertension on brain amyloid deposition and Alzheimer’s disease signature neurodegeneration. Neurobiol Aging 2019; 75: 62-70.
[http://dx.doi.org/10.1016/j.neurobiolaging.2018.11.001] [PMID: 30553154]
[48]
Peng M, Chen G, Tang KL, et al. Blood pressure at age 60-65 versus age 70-75 and vascular dementia: a population based observational study. BMC Geriatr 2017; 17(1): 252.
[http://dx.doi.org/10.1186/s12877-017-0649-3] [PMID: 29078750]
[49]
de Montgolfier O, Pinçon A, Pouliot P, et al. High systolic blood pressure induces cerebral microvascular endothelial dysfunction, neurovascular unit damage, and cognitive decline in mice. Hypertension 2019; 73(1): 217-28.
[http://dx.doi.org/10.1161/HYPERTENSIONAHA.118.12048] [PMID: 30571552]
[50]
Zhang H, Cui Y, Zhao Y, et al. Effects of sartans and low-dose statins on cerebral white matter hyperintensities and cognitive function in older patients with hypertension: a randomized, double-blind and placebo-controlled clinical trial. Hypertens Res 2019; 42(5): 717-29.
[http://dx.doi.org/10.1038/s41440-018-0165-7] [PMID: 30552406]
[51]
Ma YG, Liang L, Zhang YB, et al. Berberine reduced blood pressure and improved vasodilation in diabetic rats. J Mol Endocrinol 2017; 59(3): 191-204.
[http://dx.doi.org/10.1530/JME-17-0014] [PMID: 28515053]
[52]
Wang Y, Ding Y. Berberine protects vascular endothelial cells in hypertensive rats. Int J Clin Exp Med 2015; 8(9): 14896-905.
[PMID: 26628971]
[53]
Guo Z, Sun H, Zhang H, Zhang Y. Anti-hypertensive and renoprotective effects of berberine in spontaneously hypertensive rats. Clin Exp Hypertens 2015; 37(4): 332-9.
[http://dx.doi.org/10.3109/10641963.2014.972560] [PMID: 25867076]
[54]
Zhang P, Wang J, Zhao Y, et al. Discovery of novel antagonists on β2-adrenoceptor from natural products using a label-free cell phenotypic assay. Naunyn Schmiedebergs Arch Pharmacol 2018; 391(12): 1411-20.
[http://dx.doi.org/10.1007/s00210-018-1555-8] [PMID: 30155694]
[55]
Shabir O, Berwick J, Francis SE. Neurovascular dysfunction in vascular dementia, Alzheimer’s and atherosclerosis. BMC Neurosci 2018; 19(1): 62.
[http://dx.doi.org/10.1186/s12868-018-0465-5] [PMID: 30333009]
[56]
Kovacic JC, Fuster V. Atherosclerotic risk factors, vascular cognitive impairment, and Alzheimer disease. Mt Sinai J Med 2012; 79(6): 664-73.
[http://dx.doi.org/10.1002/msj.21347] [PMID: 23239205]
[57]
Shi Y, Hu J, Geng J, et al. Berberine treatment reduces atherosclerosis by mediating gut microbiota in apoE-/- mice. Biomed Pharmacother 2018; 107: 1556-63.
[http://dx.doi.org/10.1016/j.biopha.2018.08.148] [PMID: 30257374]
[58]
Zhu L, Zhang D, Zhu H, et al. Berberine treatment increases Akkermansia in the gut and improves high-fat diet-induced atherosclerosis in Apoe-/- mice. Atherosclerosis 2018; 268: 117-26.
[http://dx.doi.org/10.1016/j.atherosclerosis.2017.11.023] [PMID: 29202334]
[59]
Wan Q, Liu Z, Yang Y, Cui X. Suppressive effects of berberine on atherosclerosis via downregulating visfatin expression and attenuating visfatin-induced endothelial dysfunction. Int J Mol Med 2018; 41(4): 1939-48.
[http://dx.doi.org/10.3892/ijmm.2018.3440] [PMID: 29393413]
[60]
Jiang Y, Huang K, Lin X, et al. Berberine attenuates NLRP3 inflammasome activation in macrophages to reduce the secretion of interleukin-1β. Ann Clin Lab Sci 2017; 47(6): 720-8.
[PMID: 29263046]
[61]
Guo J, Wang L, Wang L, et al. Berberine protects human umbilical vein endothelial cells against LPS-induced apoptosis by blocking JNK-mediated signaling. Evid Based Complement Alternat Med 2016; 2016 6983956
[http://dx.doi.org/10.1155/2016/6983956] [PMID: 27478481]
[62]
Feng M, Kong SZ, Wang ZX, et al. The protective effect of coptisine on experimental atherosclerosis ApoE-/- mice is mediated by MAPK/NF-κB-dependent pathway. Biomed Pharmacother 2017; 93: 721-9.
[http://dx.doi.org/10.1016/j.biopha.2017.07.002] [PMID: 28700976]
[63]
Du X, Wang X, Geng M. Alzheimer’s disease hypothesis and related therapies. Transl Neurodegener 2018; 7: 2.
[http://dx.doi.org/10.1186/s40035-018-0107-y] [PMID: 29423193]
[64]
Kumar A, Singh A, Ekavali . A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep 2015; 67(2): 195-203.
[http://dx.doi.org/10.1016/j.pharep.2014.09.004] [PMID: 25712639]
[65]
de Oliveira JS, Abdalla FH, Dornelles GL, et al. Berberine protects against memory impairment and anxiogenic-like behavior in rats submitted to sporadic Alzheimer’s-like dementia: Involvement of acetylcholinesterase and cell death. Neurotoxicology 2016; 57: 241-50.
[http://dx.doi.org/10.1016/j.neuro.2016.10.008] [PMID: 27746125]
[66]
Patil S, Tawari S, Mundhada D, Nadeem S. Protective effect of berberine, an isoquinoline alkaloid ameliorates ethanol-induced oxidative stress and memory dysfunction in rats. Pharmacol Biochem Behav 2015; 136: 13-20.
[http://dx.doi.org/10.1016/j.pbb.2015.07.001] [PMID: 26159088]
[67]
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]
[68]
Braak H, Thal DR, Ghebremedhin E, Del Tredici K. Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol 2011; 70(11): 960-9.
[http://dx.doi.org/10.1097/NEN.0b013e318232a379] [PMID: 22002422]
[69]
Kametani F, Hasegawa M. Reconsideration of amyloid hypothesis and tau hypothesis in Alzheimer’s disease. Front Neurosci 2018; 12: 25.
[http://dx.doi.org/10.3389/fnins.2018.00025] [PMID: 29440986]
[70]
Zhang H, Zhao C, Cao G, et al. Berberine modulates amyloid-β peptide generation by activating AMP-activated protein kinase. Neuropharmacology 2017; 125: 408-17.
[http://dx.doi.org/10.1016/j.neuropharm.2017.08.013] [PMID: 28822725]
[71]
Cai Z, Wang C, He W, Chen Y. Berberine alleviates amyloid-beta pathology in the brain of APP/PS1 transgenic mice via inhibiting β/γ-secretases activity and enhancing α-secretases. Curr Alzheimer Res 2018; 15(11): 1045-52.
[http://dx.doi.org/10.2174/1567205015666180702105740] [PMID: 29962345]
[72]
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]
[73]
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]
[74]
Bradburn S, Murgatroyd C, Ray N. Neuroinflammation in mild cognitive impairment and Alzheimer’s disease: A meta-analysis. Ageing Res Rev 2019; 50: 1-8.
[http://dx.doi.org/10.1016/j.arr.2019.01.002] [PMID: 30610927]
[75]
Herrup K. Reimagining Alzheimer’s disease--an age-based hypothesis. J Neurosci 2010; 30(50): 16755-62.
[http://dx.doi.org/10.1523/JNEUROSCI.4521-10.2010] [PMID: 21159946]
[76]
Hopperton KE, Mohammad D, Trépanier MO, Giuliano V, Bazinet RP. Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: a systematic review. Mol Psychiatry 2018; 23(2): 177-98.
[http://dx.doi.org/10.1038/mp.2017.246] [PMID: 29230021]
[77]
Schmidt R, Schmidt H, Curb JD, Masaki K, White LR, Launer LJ. Early inflammation and dementia: a 25-year follow-up of the Honolulu-Asia Aging Study. Ann Neurol 2002; 52(2): 168-74.
[http://dx.doi.org/10.1002/ana.10265] [PMID: 12210786]
[78]
Darweesh SKL, Wolters FJ, Ikram MA, de Wolf F, Bos D, Hofman A. Inflammatory markers and the risk of dementia and Alzheimer’s disease: A meta-analysis. Alzheimers Dement 2018; 14(11): 1450-9.
[http://dx.doi.org/10.1016/j.jalz.2018.02.014] [PMID: 29605221]
[79]
Cheignon C, Tomas M, Bonnefont-Rousselot D, Faller P, Hureau C, Collin F. Oxidative stress and the amyloid beta peptide in Alzheimer’s disease. Redox Biol 2018; 14: 450-64.
[http://dx.doi.org/10.1016/j.redox.2017.10.014] [PMID: 29080524]
[80]
Luca M, Luca A, Calandra C. The role of oxidative damage in the pathogenesis and progression of Alzheimer’s disease and vascular dementia. Oxid Med Cell Longev 2015; 2015 504678
[http://dx.doi.org/10.1155/2015/504678] [PMID: 26301043]
[81]
Liu H, Zhang J. Cerebral hypoperfusion and cognitive impairment: the pathogenic role of vascular oxidative stress. Int J Neurosci 2012; 122(9): 494-9.
[http://dx.doi.org/10.3109/00207454.2012.686543] [PMID: 22519891]
[82]
Sadraie S, Kiasalari Z, Razavian M, et al. Berberine ameliorates lipopolysaccharide-induced learning and memory deficit in the rat: insights into underlying molecular mechanisms. Metab Brain Dis 2019; 34(1): 245-55.
[http://dx.doi.org/10.1007/s11011-018-0349-5] [PMID: 30456649]
[83]
Jia L, Liu J, Song Z, et al. Berberine suppresses amyloid-beta-induced inflammatory response in microglia by inhibiting nuclear factor-kappaB and mitogen-activated protein kinase signalling pathways. J Pharm Pharmacol 2012; 64(10): 1510-21.
[http://dx.doi.org/10.1111/j.2042-7158.2012.01529.x] [PMID: 22943182]
[84]
He W, Wang C, Chen Y, He Y, Cai Z. Berberine attenuates cognitive impairment and ameliorates tau hyperphosphorylation by limiting the self-perpetuating pathogenic cycle between NF-κB signaling, oxidative stress and neuroinflammation. Pharmacol Rep 2017; 69(6): 1341-8.
[http://dx.doi.org/10.1016/j.pharep.2017.06.006] [PMID: 29132092]
[85]
Sadeghnia HR, Kolangikhah M, Asadpour E, Forouzanfar F, Hosseinzadeh H. Berberine protects against glutamate-induced oxidative stress and apoptosis in PC12 and N2a cells. Iran J Basic Med Sci 2017; 20(5): 594-603.
[PMID: 28656094]
[86]
Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 2011; 147(4): 728-41.
[http://dx.doi.org/10.1016/j.cell.2011.10.026] [PMID: 22078875]
[87]
Uddin MS, Mamun AA, Labu ZK, Hidalgo-Lanussa O, Barreto GE, Ashraf GM. Autophagic dysfunction in Alzheimer’s disease: Cellular and molecular mechanistic approaches to halt Alzheimer’s pathogenesis. J Cell Physiol 2019; 234(6): 8094-112.
[http://dx.doi.org/10.1002/jcp.27588] [PMID: 30362531]
[88]
Gibas KJ. The starving brain: Overfed meets undernourished in the pathology of mild cognitive impairment (MCI) and Alzheimer’s disease (AD). Neurochem Int 2017; 110: 57-68.
[http://dx.doi.org/10.1016/j.neuint.2017.09.004] [PMID: 28899812]
[89]
Aski ML, Rezvani ME, Khaksari M, et al. Neuroprotective effect of berberine chloride on cognitive impairment and hippocampal damage in experimental model of vascular dementia. Iran J Basic Med Sci 2018; 21(1): 53-8.
[PMID: 29372037]
[90]
Viegas ATB, Guedes JR, Oliveira AR, Cardoso AMS, Cardoso ALC. MiRNAs: new biomarkers and therapeutic targets in dementia. Curr Pharm Des 2017; 23(5): 669-92.
[http://dx.doi.org/10.2174/1381612823666170111094702] [PMID: 28078993]
[91]
Zhan PY, Peng CX, Zhang LH. Berberine rescues D-galactose-induced synaptic/memory impairment by regulating the levels of Arc. Pharmacol Biochem Behav 2014; 117: 47-51.
[http://dx.doi.org/10.1016/j.pbb.2013.12.006] [PMID: 24342459]
[92]
Durairajan SSK, Iyaswamy A, Shetty SG, et al. A modified formulation of Huanglian-Jie-Du-Tang reduces memory impairments and β-amyloid plaques in a triple transgenic mouse model of Alzheimer’s disease. Sci Rep 2017; 7(1): 6238.
[http://dx.doi.org/10.1038/s41598-017-06217-9] [PMID: 28740171]
[93]
Durairajan SS, Huang YY, Yuen PY, et al. Effects of Huanglian-Jie-Du-Tang and its modified formula on the modulation of amyloid-β precursor protein processing in Alzheimer’s disease models. PLoS One 2014; 9(3) e92954
[http://dx.doi.org/10.1371/journal.pone.0092954] [PMID: 24671102]
[94]
Dong X, Du H, Han Z, et al. Effects of Huanglian Jiedu decoction on the activities of SOD, contents of MDA, expressions of I-κB and NF-κB in Alzheimer’s disease model rat’s brain. Zhonghua Zhongyiyao Xuekan 2012; 30: 1730-2.
[95]
Li YB, Zhang WH, Liu HD, Liu Z, Ma SP. Protective effects of Huanglian Wendan Decoction aganist cognitive deficits and neuronal damages in rats with diabetic encephalopathy by inhibiting the release of inflammatory cytokines and repairing insulin signaling pathway in hippocampus. Chin J Nat Med 2016; 14(11): 813-22.
[http://dx.doi.org/10.1016/S1875-5364(16)30098-X] [PMID: 27914525]
[96]
Chen F, He Y, Wang P, et al. Banxia Xiexin decoction ameliorated cognition via the regulation of insulin pathways and glucose transporters in the hippocampus of APPswe/PS1dE9 mice. Int J Immunopathol Pharmacol 2018; 32 2058738418780066
[http://dx.doi.org/10.1177/2058738418780066] [PMID: 29873261]
[97]
Yang Y, Liu J, Fang J, et al. Effect and safety of Huannao Yicong formula on patients with mild-to-moderate Alzheimer’s disease: a randomized, double-blinded, donepezil-controlled trial. Chin J Integr Med 2019; 25(8): 574-81.
[PMID: 30109588]
[98]
Cao Y, Jia X, Wei Y, Liu M, Liu J, Li H. Traditional Chinese medicine Huannao Yicong decoction extract decreases Tau hyperphosphorylation in the brain of Alzheimer’s disease model rats induced by Aβ1-42. Evid Based Complement Alternat Med 2016; 2016 6840432
[http://dx.doi.org/10.1155/2016/6840432]
[99]
Liu M, Wei Y, Yang Y, et al. Effects and mechanism of Huannao Yicong decoction extract on the ethology of transgenic APP/PS1 mice. Evid Based Complement Alternat Med 2017; 2017 9502067
[http://dx.doi.org/10.1155/2017/9502067] [PMID: 29422937]
[100]
Wang Q, Li H, Wang FX, et al. Huannao Yicong Decoction extract reduces inflammation and cell apoptosis in Aβ1-42-induced Alzheimer’s disease model of rats. Chin J Integr Med 2017; 23(9): 672-80.
[http://dx.doi.org/10.1007/s11655-016-2255-1] [PMID: 27022730]
[101]
Liu F, Niu K, Wu Z, et al. [Effects of Jiji decoction on the cognitive function and oxidative stress in mice with vascular dementia induced by cerebral ischemia/reperfusion]. Zhongguo Ying Yong Sheng Li Xue Za Zhi 2015; 31: 170-3.
[102]
Wu Y, Jing Z, Qin X, et al. Qingnaoyizhi decoction suppresses the formation of glial fibrillary acidic protein-positive cells in cultured neural stem cells by inhibiting the Janus kinase 2/signal transducer and activator of transcription 3 signaling pathway. J Tradit Chin Med 2015; 35(1): 69-76.
[http://dx.doi.org/10.1016/S0254-6272(15)30011-X] [PMID: 25842731]
[103]
Dias KS, Viegas C Jr. Multi-target directed drugs: a modern approach for design of new drugs for the treatment of Alzheimer’s disease. Curr Neuropharmacol 2014; 12(3): 239-55.
[http://dx.doi.org/10.2174/1570159X1203140511153200] [PMID: 24851088]
[104]
Oset-Gasque MJ, Marco-Contelles J. Alzheimer’s disease, the one-molecule, one-target paradigm, and the multitarget directed ligand approach. ACS Chem Neurosci 2018; 9(3): 401-3.
[http://dx.doi.org/10.1021/acschemneuro.8b00069] [PMID: 29465220]

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