Generic placeholder image

Current Drug Therapy

Editor-in-Chief

ISSN (Print): 1574-8855
ISSN (Online): 2212-3903

Research Article

Neuroprotective Effect of Levetiracetam in Combination with Berberine on Scopolamine Induced-Cognitive Impairment in Mice: A Behavioral and Biochemical Approach

Author(s): Anuradha Singh, Suneela Sunil Dhaneshwar*, Avijit Mazumder, Swatantra Kumar and Shailendra Kumar Saxena

Volume 18, Issue 5, 2023

Published on: 02 May, 2023

Page: [415 - 432] Pages: 18

DOI: 10.2174/1574885518666230213151508

Price: $65

conference banner
Abstract

Background: Presently, only four drugs have been approved by FDA for Alzheimer’s disease (AD). A drug repurposing approach can be fruitful in searching for promising candidates for AD.

Objective: The objective of the work was to evaluate the neuroprotective effect of levetiracetam (LEV) in combination with berberine (BER) in scopolamine-induced cognitive impairment in mice by applying a drug repositioning approach owing to their antioxidant potential.

Methods: Cognitive impairment was induced in mice by scopolamine. Morris water maze, elevated plus maze, and Y-maze were used to evaluate behavioral parameters. Assays for acetylcholinesterase (AChE), reduced glutathione (GSH), malondialdehyde (MDA), catalase, nitrite, TNF-α, and brain histopathology were performed.

Results: The transfer latency time and percentage of spontaneous alternation were significantly reduced and significant alterations in AChE and MDA levels, GSH concentration, and improvement in nitrite and catalase levels were also evidenced after the treatment of mice with a combination of LEV and BER in comparison to independent drugs, standard and disease control groups. The antioxidant defense was also improved and TNF-α levels were significantly reduced by a combination of LEV and BER. Improvement in neuronal damage by restoration of the cytoarchitecture of the brain was also seen in the histopathological study of the brain of treatment groups.

Conclusion: The present study has demonstrated that the combination of LEV and BER has significantly improved cognition in mice by lipid peroxidation inhibition, augmentation of endogenous antioxidant enzymes, the decline in TNF- α levels, and AChE activity in the brain when compared to individual drugs, standard and disease control owing to their strong antioxidant and anti-inflammatory potentials.

Graphical Abstract

[1]
Ballatore C, Lee VMY, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci 2007; 8(9): 663-72.
[http://dx.doi.org/10.1038/nrn2194] [PMID: 17684513]
[2]
Alzheimer’s Association. . Includes a special report on the next frontier of Alzheimer’s research. Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement 2017; 13: 325-73.
[3]
Ghumatkar PJ, Patil SP, Jain PD, Tambe RM, Sathaye S. Nootropic, neuroprotective and neurotrophic effects of phloretin in scopolamine induced amnesia in mice. Pharmacol Biochem Behav 2015; 135: 182-91.
[http://dx.doi.org/10.1016/j.pbb.2015.06.005] [PMID: 26071678]
[4]
Yatin SM, Varadarajan S, Link CD, Butterfield DA. In vitro and in vivo oxidative stress associated with Alzheimer’s amyloid beta-peptide (1-42). Neurobiol Aging 1999; 20(3): 325-30.
[PMID: 10588580]
[5]
Butterfield DA, Lauderback CM. Lipid peroxidation and protein oxidation in Alzheimer’s disease brain: potential causes and consequences involving amyloid β-peptide-associated free radical oxidative stress 1,2 1Guest Editors: Mark A. Smith and George Perry 2This article is part of a series of reviews on “Causes and Consequences of Oxidative Stress in Alzheimer’s Disease.” The full list of papers may be found on the homepage of the journal. Free Radic Biol Med 2002; 32: pp. (11)1050-60.
[http://dx.doi.org/10.1016/S0891-5849(02)00794-3]] [PMID: 12031889]
[6]
Ding Q, Dimayuga E, Keller J. Oxidative damage, protein synthesis, and protein degradation in Alzheimer’s disease. Curr Alzheimer Res 2007; 4(1): 73-9.
[http://dx.doi.org/10.2174/156720507779939788] [PMID: 17316168]
[7]
Joshi L, Ponnana M, Sivangala R, et al. Evaluation of TNF-α IL-10 and IL-6 cytokine production and their correlation with genotype variants amongst tuberculosis patients and their household contacts. PLoS One 2015; 10(9): e0137727.
[http://dx.doi.org/10.1371/journal.pone.0137727] [PMID: 26359865]
[8]
Huang WJ, Zhang X, Chen WW. Role of oxidative stress in Alzheimer’s disease. Biomed Rep 2016; 4(5): 519-22.
[http://dx.doi.org/10.3892/br.2016.630] [PMID: 27123241]
[9]
Smith MA, Rottkamp CA, Nunomura A, Raina AK, Perry G. Oxidative stress in Alzheimer’s disease. Biochim Biophys Acta Mol Basis Dis 2000; 1502(1): 139-44.
[http://dx.doi.org/10.1016/S0925-4439(00)00040-5] [PMID: 10899439]
[10]
Zhao B, Zhao B. Natural antioxidants in prevention and management of Alzheimer s disease. Front Biosci (Elite Ed) 2012; E4(3): 794-808.
[http://dx.doi.org/10.2741/e419] [PMID: 22201914]
[11]
Rudrapal M, J. Khairnar S,G, Jadhav A. Drug Repurposing (DR): An emerging approach in drug discovery. Drug repurposing - hypothesis, molecular aspects and therapeutic applications [Internet]. 2020 Dec 2; Available from:
[http://dx.doi.org/10.5772/intechopen.93193]
[12]
Siavelis JC, Bourdakou MM, Athanasiadis EI, Spyrou GM, Nikita KS. Bioinformatics methods in drug repurposing for Alzheimer’s disease. Brief Bioinform 2016; 17(2): 322-35.
[http://dx.doi.org/10.1093/bib/bbv048] [PMID: 26197808]
[13]
Durães F, Pinto M, Sousa E. Old drugs as new treatments for neurodegenerative diseases. Pharmaceuticals 2018; 11(2): 44.
[http://dx.doi.org/10.3390/ph11020044] [PMID: 29751602]
[14]
Williams G, Gatt A, Clarke E, et al. Drug repurposing for Alzheimer’s disease based on transcriptional profiling of human iPSC-derived cortical neurons. Transl Psychiatry 2019; 9(1): 220.
[http://dx.doi.org/10.1038/s41398-019-0555-x] [PMID: 31492831]
[15]
Kim MS, Jeon WK, Lee KW, Park YH, Han JS. Ameliorating effects of ethanol extract of fructus mume on scopolamine-induced memory impairment in mice. Evid.-based Complement. Altern Med 2015; 2015: 1-8.
[16]
Spagnuolo C, Napolitano M, Tedesco I, Moccia S, Milito A, Luigi Russo G. Neuroprotective role of natural polyphenols. Curr Top Med Chem 2016; 16(17): 1943-50.
[http://dx.doi.org/10.2174/1568026616666160204122449] [PMID: 26845551]
[17]
Venigalla M, Sonego S, Gyengesi E, Sharman MJ, Münch G. Novel promising therapeutics against chronic neuroinflammation and neurodegeneration in Alzheimer’s disease. Neurochem Int 2016; 95: 63-74.
[http://dx.doi.org/10.1016/j.neuint.2015.10.011] [PMID: 26529297]
[18]
Uddin MS, Al Mamun A, Kabir MT, et al. Neuroprotective role of polyphenols against oxidative stress-mediated neurodegeneration. Eur J Pharmacol 2020; 886: 173412.
[http://dx.doi.org/10.1016/j.ejphar.2020.173412] [PMID: 32771668]
[19]
Liu J, Wang LN, Wu LY, Wang YP. Treatment of epilepsy for people with Alzheimer’s disease. Cochrane Libr 2016; 11(11): CD011922.
[http://dx.doi.org/10.1002/14651858.CD011922.pub2] [PMID: 27805721]
[20]
Piccialli I, Tedeschi V, Caputo L, et al. Exploring the therapeutic potential of phytochemicals in Alzheimer’s disease: Focus on polyphenols and monoterpenes. Front Pharmacol 2022; 13: 876614.
[http://dx.doi.org/10.3389/fphar.2022.876614] [PMID: 35600880]
[21]
Gibbs JE, Walker MC, Cock HR. Levetiracetam: Antiepileptic properties and protective effects on mitochondrial dysfunction in experimental status epilepticus. Epilepsia 2006; 47(3): 469-78.
[http://dx.doi.org/10.1111/j.1528-1167.2006.00454.x] [PMID: 16529608]
[22]
Bakker A, Krauss GL, Albert MS, et al. Reduction of hippocampal hyperactivity improves cognition in amnestic mild cognitive impairment. Neuron 2012; 74(3): 467-74.
[http://dx.doi.org/10.1016/j.neuron.2012.03.023] [PMID: 22578498]
[23]
Lyseng-Williamson KA. Levetiracetam: A review of its use in epilepsy. Drugs 2011; 71(4): 489-514.
[PMID: 21395360]
[24]
Tang J, Feng Y, Tsao S, Wang N, Curtain R, Wang Y. Berberine and Coptidis Rhizoma as novel antineoplastic agents: A review of traditional use and biomedical investigations. J Ethnopharmacol 2009; 126(1): 5-17.
[http://dx.doi.org/10.1016/j.jep.2009.08.009] [PMID: 19686830]
[25]
Kulkarni SK, Dhir A. Berberine: a plant alkaloid with therapeutic potential for central nervous system disorders. Phytother Res 2010; 24(3): 317-24.
[http://dx.doi.org/10.1002/ptr.2968] [PMID: 19998323]
[26]
Li B, Stribley JA, Ticu A, et al. Abundant tissue butyrylcholinesterase and its possible function in the acetylcholinesterase knockout mouse. J Neurochem 2000; 75(3): 1320-31.
[http://dx.doi.org/10.1046/j.1471-4159.2000.751320.x] [PMID: 10936216]
[27]
Cooper JR. Unsolved problems in the cholinergic nervous system. J Neurochem 1994; 63(2): 395-9.
[http://dx.doi.org/10.1046/j.1471-4159.1994.63020395.x] [PMID: 8035169]
[28]
Sunderland T, Tariot PN, Weingartner H, et al. Pharmacologic modelling of Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry 1986; 10(3-5): 599-610.
[http://dx.doi.org/10.1016/0278-5846(86)90030-8] [PMID: 3541054]
[29]
Ebert U, Kirch W. Scopolamine model of dementia: electroencephalogram findings and cognitive performance. Eur J Clin Invest 1998; 28(11): 944-9.
[http://dx.doi.org/10.1046/j.1365-2362.1998.00393.x] [PMID: 9824440]
[30]
Kulshreshtha A, Piplani P. Ameliorative effects of amide derivatives of 1,3,4-thiadiazoles on scopolamine induced cognitive dysfunction. Eur J Med Chem 2016; 122: 557-73.
[http://dx.doi.org/10.1016/j.ejmech.2016.06.046] [PMID: 27448914]
[31]
Mahmoud YI, Sayed SS. Effects of L-cysteine on lead acetate induced neurotoxicity in albino mice. Biotech Histochem 2016; 91(5): 327-32.
[http://dx.doi.org/10.3109/10520295.2016.1164897] [PMID: 27045382]
[32]
Goverdhan P, Akina S, Thati M, Puchchakayala G. Neuroprotective effect of ceftriaxone and selegiline on scopolamine induced cognitive impairment in mice. Adv Biol Res 2013; 7: 266-75.
[33]
Sanchez PE, Zhu L, Verret L, et al. Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model. Proc Natl Acad Sci 2012; 109(42): E2895-903.
[http://dx.doi.org/10.1073/pnas.1121081109] [PMID: 22869752]
[34]
Abdel-Latif MS, Abady MMA, Saleh SR, Abdel-Monaem N, Gareeb DA. Effect of berberine and ipriflavone mixture against scopolamine-induced Alzheimer-like disease. Int J Pharm Phytopharmacological Res 2009; 9: 48-63.
[35]
Kaur R, Mehan S, Khanna D, Kalra S. Ameliorative treatment with ellagic acid in scopolamine-induced Alzheimer’s type memory and cognitive dysfunctions in rats. Austin J Clin Neurol 2015; 2: 1-11.
[36]
Skirzewski M, Hernandez L, Schechter LE, Rada P. Acute lecozotan administration increases learning and memory in rats without affecting anxiety or behavioral depression. Pharmacol Biochem Behav 2010; 95(3): 325-30.
[http://dx.doi.org/10.1016/j.pbb.2010.02.008] [PMID: 20170670]
[37]
Lee GY, Lee C, Park GH, Jang JH. Amelioration of scopolamine-induced learning and memory impairment by -α pinene in C57BL/6 mice. Evid Based Complement Alternat Med 2017; 2017: 1-9.
[http://dx.doi.org/10.1155/2017/4926815] [PMID: 29234406]
[38]
Foyet HS, Armand B, Abaissou Herve N, et al. Neuroprotective and memory improvement effects of a standardized extract of Emilia coccinea (SIMS) G. on animal models of anxiety and depression. J Pharmacogn Phytochem 2014; 3: 146-54.
[39]
Kumar A, Dogra S, Prakash A. Neuroprotective effects of Centella asiatica against intracerebroventricular colchicine-induced cognitive impairment and oxidative stress. Int J Alzheimers Dis 2009; 2009: 1-8.
[http://dx.doi.org/10.4061/2009/972178] [PMID: 20798885]
[40]
Goverdhan P, Sravanthi A, Mamatha T. Neuroprotective effects of meloxicam and selegiline in scopolamine-induced cognitive impairment and oxidative stress. Int J Alzheimers Dis 2012; 2012: 1-8.
[http://dx.doi.org/10.1155/2012/974013] [PMID: 22536538]
[41]
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]
[42]
Ohkawa H, Ohishi N, Yagi K. Assay for lipid peroxides in animal tissues by thiobarbituric acid reaction. Anal Biochem 1979; 95(2): 351-8.
[http://dx.doi.org/10.1016/0003-2697(79)90738-3] [PMID: 36810]
[43]
Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR. Analysis of nitrate, nitrite, and [15N]nitrate in biological fluids. Anal Biochem 1982; 126(1): 131-8.
[http://dx.doi.org/10.1016/0003-2697(82)90118-X] [PMID: 7181105]
[44]
Saikia B, Barua C, Sarma J, et al. Zanthoxylum alatum ameliorates scopolamine-induced amnesia in rats: Behavioral, biochemical, and molecular evidence. Indian J Pharmacol 2018; 50(1): 30-8.
[http://dx.doi.org/10.4103/ijp.IJP_417_17] [PMID: 29861525]
[45]
Fisher A. Cholinergic treatments with emphasis on M1 muscarinic agonists as potential disease-modifying agents for Alzheimer’s disease. Neurotherapeutics 2008; 5(3): 433-42.
[http://dx.doi.org/10.1016/j.nurt.2008.05.002] [PMID: 18625455]
[46]
Bartus RT, Dean RL III, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982; 217(4558): 408-14.
[http://dx.doi.org/10.1126/science.7046051] [PMID: 7046051]
[47]
Mahadevan S, Park Y. Multifaceted therapeutic benefits of Ginkgo biloba L.: Chemistry, efficacy, safety, and uses. J Food Sci 2008; 73(1): R14-9.
[http://dx.doi.org/10.1111/j.1750-3841.2007.00597.x] [PMID: 18211362]
[48]
Ramassamy C. Emerging role of polyphenolic compounds in the treatment of neurodegenerative diseases: A review of their intracellular targets. Eur J Pharmacol 2006; 545(1): 51-64.
[http://dx.doi.org/10.1016/j.ejphar.2006.06.025] [PMID: 16904103]
[49]
Masella R, Di Benedetto R, Varì R, Filesi C, Giovannini C. Novel mechanisms of natural antioxidant compounds in biological systems: involvement of glutathione and glutathione-related enzymes. J Nutr Biochem 2005; 16(10): 577-86.
[http://dx.doi.org/10.1016/j.jnutbio.2005.05.013] [PMID: 16111877]
[50]
Stevenson DE, Hurst RD. Polyphenolic phytochemicals-just antioxidants or much more? Cell Mol Life Sci 2007; 64(22): 2900-16.
[http://dx.doi.org/10.1007/s00018-007-7237-1] [PMID: 17726576]
[51]
Nabavi SF, Nabavi SM, Mirzaei M, Moghaddam AH. Protective effect of quercetin against sodium fluoride induced oxidative stress in rat’s heart. Food Funct 2012; 3(4): 437-41.
[http://dx.doi.org/10.1039/c2fo10264a] [PMID: 22314573]
[52]
Budzynska B, Boguszewska-Czubara A, Kruk-Slomka M, et al. Effects of imperatorin on scopolamine-induced cognitive impairment and oxidative stress in mice. Psychopharmacology 2015; 232(5): 931-42.
[http://dx.doi.org/10.1007/s00213-014-3728-6] [PMID: 25189792]
[53]
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]
[54]
Ueda Y, Doi T, Takaki M, Nagatomo K, Nakajima A, Willmore LJ. Levetiracetam enhances endogenous antioxidant in the hippocampus of rats: In vivo evaluation by brain microdialysis combined with ESR spectroscopy. Brain Res 2009; 1266: 1-7.
[http://dx.doi.org/10.1016/j.brainres.2009.02.040] [PMID: 19268434]
[55]
Jang MH, Kim HY, Kang KS, Yokozawa T, Park JH. Hydroxyl radical scavenging activities of isoquinoline alkaloids isolated from Coptis chinensis. Arch Pharm Res 2009; 32(3): 341-5.
[http://dx.doi.org/10.1007/s12272-009-1305-z] [PMID: 19387576]
[56]
Li Z, Geng YN, Jiang JD, Kong WJ. Antioxidant and anti-inflammatory activities of berberine in the treatment of diabetes mellitus. Evid Based Complement Alternat Med 2014; 2014: 1-12.
[http://dx.doi.org/10.1155/2014/289264] [PMID: 24669227]
[57]
Hasanein P, Ghafari-Vahed M, Khodadadi I. Effects of isoquinoline alkaloid berberine on lipid peroxidation, antioxidant defense system, and liver damage induced by lead acetate in rats. Redox Rep 2017; 22(1): 42-50.
[http://dx.doi.org/10.1080/13510002.2016.1140406] [PMID: 26871196]
[58]
Chandirasegaran G, Elanchezhiyan C, Ghosh K. Modulatory effects of berberine chloride on lipid profile, oxidant status and insulin signaling molecules in streptozotocin-induced diabetic rats. Indian J Clin Biochem 2019; 34(3): 254-62.
[http://dx.doi.org/10.1007/s12291-018-0754-x] [PMID: 31391714]
[59]
Poorheidari G, Pratt JA, Dehghani N. Effects of low-dose scopolamine on locomotor activity: No dissociation between cognitive and non-effects. Neurosci Res Commun 2002; 31(3): 165-74.
[http://dx.doi.org/10.1002/nrc.10049]
[60]
Chintoh A, Fulton J, Koziel N, Aziz M, Sud M, Yeomans JS. Role of cholinergic receptors in locomotion induced by scopolamine and oxotremorine-M. Pharmacol Biochem Behav 2003; 76(1): 53-61.
[http://dx.doi.org/10.1016/S0091-3057(03)00196-5] [PMID: 13679217]
[61]
Chen J, Long Y, Han M, Wang T, Chen Q, Wang R. Water-soluble derivative of propolis mitigates scopolamine-induced learning and memory impairment in mice. Pharmacol Biochem Behav 2008; 90(3): 441-6.
[http://dx.doi.org/10.1016/j.pbb.2008.03.029] [PMID: 18485465]
[62]
Rahimzadegan M, Soodi M. Comparison of memory impairment and oxidative stress following single or repeated doses administration of scopolamine in rat hippocampus. Basic Clin Neurosci 2018; 9(1): 5-14.
[http://dx.doi.org/10.29252/nirp.bcn.9.1.5] [PMID: 29942435]
[63]
Pattanashetti LA, Taranalli AD, Parvatrao V, Malabade RH, Kumar D. Evaluation of neuroprotective effect of quercetin with donepezil in scopolamine-induced amnesia in rats. Indian J Pharmacol 2017; 49(1): 60-4.
[PMID: 28458424]
[64]
Yang JH, Han SJ, Ryu JH, Jang IS, Kim DH. Ginsenoside Rh2 ameliorates scopolamine-induced learning deficit in mice. Biol Pharm Bull 2009; 32(10): 1710-5.
[http://dx.doi.org/10.1248/bpb.32.1710] [PMID: 19801832]
[65]
Olton DS, Paras BC. Spatial memory and hippocampal function. Neuropsychologia 1979; 17(6): 669-82.
[http://dx.doi.org/10.1016/0028-3932(79)90042-3] [PMID: 522981]
[66]
dela Peña IJI, Kim HJ, Botanas CJ, et al. The psychopharmacological activities of Vietnamese ginseng in mice: characterization of its psychomotor, sedative–hypnotic, antistress, anxiolytic, and cognitive effects. J Ginseng Res 2017; 41(2): 201-8.
[http://dx.doi.org/10.1016/j.jgr.2016.03.005] [PMID: 28413325]
[67]
Shin CY, Kim HS, Cha KH, et al. The effects of donepezil, an acetylcholinesterase inhibitor, on impaired learning and memory in rodents. Biomol Ther 2018; 26(3): 274-81.
[http://dx.doi.org/10.4062/biomolther.2017.189] [PMID: 29463072]
[68]
Francis PT, Palmer AM, Snape M, Wilcock GK. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J Neurol Neurosurg Psychiatry 1999; 66(2): 137-47.
[http://dx.doi.org/10.1136/jnnp.66.2.137] [PMID: 10071091]
[69]
Maurer SV, Williams CL. The cholinergic system modulates memory and hippocampal plasticity via its interactions with non-neuronal cells. Front Immunol 2017; 8: 1489.
[http://dx.doi.org/10.3389/fimmu.2017.01489] [PMID: 29167670]
[70]
Flood JF, Cherkin A. Scopolamine effects on memory retention in mice: A model of dementia? Behav Neural Biol 1986; 45(2): 169-84.
[http://dx.doi.org/10.1016/S0163-1047(86)90750-8] [PMID: 3964171]
[71]
Kanwal A, Mehla J, Kuncha M, Naidu VGM, Gupta YK, Sistla R. Anti-amnesic activity of Vitex negundo in scopolamine induced amnesia in rats. Pharmacol Pharm 2010; 1(1): 1-8.
[http://dx.doi.org/10.4236/pp.2010.11001]
[72]
Bihaqi S, Tiwari M, Singh AP. In vivo investigation of the neuroprotective property of Convolvulus pluricaulis in scopolamine-induced cognitive impairments in Wistar rats. Indian J Pharmacol 2011; 43(5): 520-5.
[http://dx.doi.org/10.4103/0253-7613.84958] [PMID: 22021993]
[73]
Pushpalatha B, Venumadhav N, Swathi M, Raju B. Neuroprotective effect of resveratrol against scopolamine-induced cognitive impairment and oxidative stress in rats. Arch Biol Sci 2013; 65(4): 1381-6.
[http://dx.doi.org/10.2298/ABS1304381P]
[74]
Lee S, Kim J, Seo SG, et al. Sulforaphane alleviates scopolamine-induced memory impairment in mice. Pharmacol Res 2014; 85: 23-32.
[http://dx.doi.org/10.1016/j.phrs.2014.05.003] [PMID: 24836869]
[75]
Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic Biol Med 1997; 23(1): 134-47.
[http://dx.doi.org/10.1016/S0891-5849(96)00629-6] [PMID: 9165306]
[76]
Baek SY, Li FY, Kim DH, Kim SJ, Kim MR. Enteromorpha prolifera Extract Improves Memory in Scopolamine-Treated Mice via Downregulating Amyloid-β Expression and Upregulating BDNF/TrkB Pathway. Antioxidants 2020; 9(7): 620.
[http://dx.doi.org/10.3390/antiox9070620] [PMID: 32679768]
[77]
Yadang FSA, Nguezeye Y, Kom CW, et al. Scopolamine-induced memory impairment in mice: Neuroprotective effects of Carissa edulis (Forssk.) Valh (Apocynaceae) aqueous extract. Int J Alzheimers Dis 2020; 2020: 1-10.
[http://dx.doi.org/10.1155/2020/6372059] [PMID: 32934845]
[78]
Schuessel K, Leutner S, Cairns NJ, Müller WE, Eckert A. Impact of gender on upregulation of antioxidant defence mechanisms in Alzheimer’s disease brain. J Neural Transm 2004; 111(9): 1167-82.
[http://dx.doi.org/10.1007/s00702-004-0156-5] [PMID: 15338332]
[79]
Bains JS, Shaw CA. Neurodegenerative disorders in humans: the role of glutathione in oxidative stress-mediated neuronal death. Brain Res Brain Res Rev 1997; 25(3): 335-58.
[http://dx.doi.org/10.1016/S0165-0173(97)00045-3] [PMID: 9495562]
[80]
Malinski T. Nitric oxide and nitroxidative stress in Alzheimer’s disease. J Alzheimers Dis 2007; 11(2): 207-18.
[http://dx.doi.org/10.3233/JAD-2007-11208] [PMID: 17522445]
[81]
Kim MS, Lee DY, Lee J, et al. Terminalia chebula extract prevents scopolamine-induced amnesia via cholinergic modulation and anti-oxidative effects in mice. BMC Complement Altern Med 2018; 18(1): 136.
[http://dx.doi.org/10.1186/s12906-018-2212-y] [PMID: 29716575]
[82]
Marcus DL, Thomas C, Rodriguez C, et al. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer’s disease. Exp Neurol 1998; 150(1): 40-4.
[http://dx.doi.org/10.1006/exnr.1997.6750] [PMID: 9514828]
[83]
Nandi A, Yan LJ, Jana CK, Das N. Role of catalase in oxidative stress-and age-associated degenerative diseases. Oxid Med Cell Longev 2019; 2019: 1-19.
[http://dx.doi.org/10.1155/2019/9613090] [PMID: 31827713]
[84]
Sayanti S, Mukund H. Evaluation of memory enhancing activity of leaf extract of Dalbergia sissoo in mice. Int J Pharm Sci Drug Res 2015; 7: 263-9.
[85]
Goschorska M, Gutowska I, Baranowska-Bosiacka I, et al. Influence of acetylcholinesterase inhibitors used in Alzheimer’s disease treatment on the activity of antioxidant enzymes and the concentration of glutathione in THP-1 macrophages under fluorideinduced oxidative stress. Int J Environ Res Public Health 2018; 16(1): 10.
[http://dx.doi.org/10.3390/ijerph16010010] [PMID: 30577562]
[86]
Heneka MT, Carson MJ, Khoury JE, et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol 2015; 14(4): 388-405.
[http://dx.doi.org/10.1016/S1474-4422(15)70016-5] [PMID: 25792098]
[87]
Nemy M, Cedres N, Grothe MJ, et al. Cholinergic white matter pathways make a stronger contribution to attention and memory in normal aging than cerebrovascular health and nucleus basalis of Meynert. Neuroimage 2020; 211: 116607.
[http://dx.doi.org/10.1016/j.neuroimage.2020.116607] [PMID: 32035186]
[88]
Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer’s disease. Neurobiol Aging 2000; 21(3): 383-421.
[http://dx.doi.org/10.1016/S0197-4580(00)00124-X] [PMID: 10858586]
[89]
Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry 2003; 74(9): 1200-5.
[http://dx.doi.org/10.1136/jnnp.74.9.1200] [PMID: 12933918]
[90]
Cheng X, Shen Y, Li R. Targeting TNF: a therapeutic strategy for Alzheimer’s disease. Drug Discov Today 2014; 19(11): 1822-7.
[http://dx.doi.org/10.1016/j.drudis.2014.06.029] [PMID: 24998784]
[91]
McCaulley ME, Grush KA. Alzheimer’s disease: Exploring the role of inflammation and implications for treatment. Int J Alzheimers Dis 2015; 2015: 1-10.
[http://dx.doi.org/10.1155/2015/515248] [PMID: 26664821]
[92]
Bruunsgaard H, Andersen-Ranberg K, Jeune B, Pedersen AN, Skinhøj P, Pedersen BK. A high plasma concentration of TNF-α is associated with dementia in centenarians. J Gerontol A Biol Sci Med Sci 1999; 54(7): M357-64.
[http://dx.doi.org/10.1093/gerona/54.7.M357] [PMID: 10462168]
[93]
Cheon SY, Koo BN, Kim SY, Kam EH, Nam J, Kim EJ. Scopolamine promotes neuroinflammation and delirium-like neuropsychiatric disorder in mice. Sci Rep 2021; 11(1): 8376.
[http://dx.doi.org/10.1038/s41598-021-87790-y] [PMID: 33863952]
[94]
Arikawa M, Kakinuma Y, Noguchi T, Todaka H, Sato T. Donepezil, an acetylcholinesterase inhibitor, attenuates LPS-induced inflammatory response in murine macrophage cell line RAW 264.7 through inhibition of nuclear factor kappa B translocation. Eur J Pharmacol 2016; 789: 17-26.
[http://dx.doi.org/10.1016/j.ejphar.2016.06.053] [PMID: 27373848]

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