Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Exploring the Multifunctional Neuroprotective Promise of Rasagiline Derivatives for Multi-Dysfunctional Alzheimer’s Disease

Author(s): Md. Sahab Uddin*, Md. Tanvir Kabir, Md. Habibur Rahman, Md. Abdul Alim, Md. Motiar Rahman, Anurag Khatkar, Abdullah Al Mamun, Abdur Rauf, Bijo Mathew and Ghulam Md. Ashraf

Volume 26, Issue 37, 2020

Page: [4690 - 4698] Pages: 9

DOI: 10.2174/1381612826666200406075044

Price: $65

Abstract

Alzheimer's disease (AD) is a chronic, age-related, and irreversible brain disorder that typically develops slowly and gets worse over time. The potent auspicious drug candidate for the treatment of AD is supposed to perform the simultaneous modulation of several targets linked to AD. The new therapeutic approach involves drug candidates that are designed to act on multiple targets and have various pharmacological properties. This trend has triggered the development of various multimodal drugs including TV-3326 (i.e. ladostigil) and M-30 (i.e. a new multitarget iron chelator). TV-3326 combines the neurorestorative/neuroprotective effects of the cholinesterase (ChE) inhibitory activity of rivastigmine with rasagiline (a selective monoamine oxidase-B inhibitor and novel antiparkinsonian agent) in a single molecule. M-30, the second derivative of rasagiline, was developed by combining the propargyl moiety of rasagiline into the skeleton of VK-28 (i.e. a novel brain permeable neuroprotective iron chelator). It has been revealed that both the compounds possess anti-AD effects and therefore, the clinical development is directed to the treatment of this type of neurodegenerative diseases (NDs). In this article, we have reviewed the neuroprotective molecular mechanisms and multimodal effects of TV-3326 and M-30.

Keywords: TV-3326, ladostigil, M-30, cholinesterase inhibitor, monoamine oxidase inhibitor, Alzheimer's disease.

[1]
Uddin MS, Mamun AA, Jakaria M, et al. Emerging promise of sulforaphane-mediated Nrf2 signaling cascade against neurological disorders. Sci Total Environ 2020; 707: 135624.
[http://dx.doi.org/10.1016/j.scitotenv.2019.135624 ] [PMID: 31784171]
[2]
Al Mamun A, Uddin MS, Kabir MT, et al. Exploring the Promise of Targeting Ubiquitin-Proteasome System to Combat Alzheimer’s Disease. Neurotox Res 2020; 1-10.
[http://dx.doi.org/10.1007/s12640-020-00185-1 ] [PMID: 32157628]
[3]
Bullock R. Future directions in the treatment of Alzheimer’s disease. Expert Opin Investig Drugs 2004; 13(4): 303-14.
[http://dx.doi.org/10.1517/13543784.13.4.303 ] [PMID: 15102582]
[4]
Uddin MS, Al Mamun A, Kabir MT, et al. Nootropic and Anti-Alzheimer’s Actions of Medicinal Plants: Molecular Insight into Therapeutic Potential to Alleviate Alzheimer’s Neuropathology. Mol Neurobiol 2019; 56(7): 4925-44.
[http://dx.doi.org/10.1007/s12035-018-1420-2 ] [PMID: 30414087]
[5]
Eckert A, Keil U, Marques CA, et al. Mitochondrial dysfunction, apoptotic cell death, and Alzheimer’s disease. Biochem Pharmacol 2003; 66(8): 1627-34.
[http://dx.doi.org/10.1016/S0006-2952(03)00534-3 ] [PMID: 14555243]
[6]
Maccioni RB, Muñoz JP, Barbeito L. The molecular bases of Alzheimer’s disease and other neurodegenerative disorders. Arch Med Res 2001; 32(5): 367-81.
[http://dx.doi.org/10.1016/S0188-4409(01)00316-2 ] [PMID: 11578751]
[7]
Uddin MS, Kabir MT, Al Mamun A, Abdel-Daim MM, Barreto GE, Ashraf GM. APOE and Alzheimer’s Disease: Evidence Mounts That Targeting APOE4 May Combat Alzheimer’s Pathogenesis. Mol Neurobiol 2019; 56: 2450-65.
[PMID: 30032423]
[8]
Tsolaki M, Kokarida K, Iakovidou V, Stilopoulos E, Meimaris J, Kazis A. Extrapyramidal symptoms and signs in Alzheimer’s disease: prevalence and correlation with the first symptom. Am J Alzheimers Dis Other Demen 2001; 16(5): 268-78.
[http://dx.doi.org/10.1177/153331750101600512 ] [PMID: 11603162]
[9]
Kabir MT, Uddin MS, Begum MM, et al. Cholinesterase Inhibitors for Alzheimer’s Disease: Multitargeting Strategy Based on Anti-Alzheimer’s Drugs Repositioning. Curr Pharm Des 2019; 25(33): 3519-35.
[http://dx.doi.org/10.2174/1381612825666191008103141 ] [PMID: 31593530]
[10]
Uddin MS, Kabir MT, Niaz K, et al. Molecular Insight into the Therapeutic Promise of Flavonoids against Alzheimer’s Disease. Mol 2020; 25: 25-1267.
[11]
Selkoe DJ, Schenk D. Alzheimer’s disease: molecular understanding predicts amyloid-based therapeutics. Annu Rev Pharmacol Toxicol 2003; 43: 545-84.
[http://dx.doi.org/10.1146/annurev.pharmtox.43.100901.140248 ] [PMID: 12415125]
[12]
Schliebs R. Basal forebrain cholinergic dysfunction in Alzheimer’s disease‐interrelationship with β-amyloid, inflammation and neurotrophin signaling. Neurochem Res 2005; 30(6-7): 895-908.
[http://dx.doi.org/10.1007/s11064-005-6962-9 ] [PMID: 16187224]
[13]
Mamun AA, Uddin MS, Mathew B, Ashraf GM. Toxic tau: structural origins of tau aggregation in Alzheimer’s disease. Neural Regen Res 2020; 15(8): 1417-20.
[http://dx.doi.org/10.4103/1673-5374.274329 ] [PMID: 31997800]
[14]
Al Mamun A, Uddin MS. KDS2010: A Potent Highly Selective and Reversible MAO-B Inhibitor to Abate Alzheimer’s Disease. Comb Chem High Throughput Screen 2020; 23.
[http://dx.doi.org/10.2174/1386207323666200117103144 ] [PMID: 31957612]
[15]
Palmer AM, Stratmann GC, Procter AW, Bowen DM. Possible neurotransmitter basis of behavioral changes in Alzheimer’s disease. Ann Neurol 1988; 23(6): 616-20.
[http://dx.doi.org/10.1002/ana.410230616 ] [PMID: 2457353]
[16]
Newman SC. The prevalence of depression in Alzheimer’s disease and vascular dementia in a population sample. J Affect Disord 1999; 52(1-3): 169-76.
[http://dx.doi.org/10.1016/S0165-0327(98)00070-6 ] [PMID: 10357030]
[17]
Uddin MS, Mamun AA, Kabir MT, et al. Neurochemistry of Neurochemicals: Messengers of Brain Functions. J Intellect Disabil Diagnosis Treat 2018; 5: 137-51.
[18]
Kabir MT, Sufian MA, Uddin MS, et al. NMDA Receptor Antagonists: Repositioning of Memantine as a Multitargeting Agent for Alzheimer’s Therapy. Curr Pharm Des 2019; 25(33): 3506-18.
[http://dx.doi.org/10.2174/1381612825666191011102444 ] [PMID: 31604413]
[19]
Uddin MS, Kabir MT, Tewari D, Mathew B, Aleya L. Emerging signal regulating potential of small molecule biflavonoids to combat neuropathological insults of Alzheimer’s disease. Sci Total Environ 2020; 700: 134836.
[http://dx.doi.org/10.1016/j.scitotenv.2019.134836 ] [PMID: 31704512]
[20]
Mathew B, Parambi DGT, Mathew GE, et al. Emerging Therapeutic Potentials of Dual‐acting MAO and AChE Inhibitors in Alzheimer’s and Parkinson’s Diseases Arch Pharm (Weinheim) 2019.
[http://dx.doi.org/10.1002/ardp.201900177]
[21]
Uddin MS, Rashid M. Advances in Neuropharmacology : Drugs and Therapeutics. Canada: Apple Academic Press 2020.
[22]
Clark CM, Karlawish JHT. Alzheimer disease: current concepts and emerging diagnostic and therapeutic strategies. Ann Intern Med 2003; 138(5): 400-10.
[http://dx.doi.org/10.7326/0003-4819-138-5-200303040-00010 ] [PMID: 12614093]
[23]
Klafki HW, Staufenbiel M, Kornhuber J, Wiltfang J. Therapeutic approaches to Alzheimer’s disease. Brain 2006; 129(Pt 11): 2840-55.
[http://dx.doi.org/10.1093/brain/awl280 ] [PMID: 17018549]
[24]
Hossain MF, Uddin MS, Uddin GMS, et al. Melatonin in Alzheimer’s Disease: A Latent Endogenous Regulator of Neurogenesis to Mitigate Alzheimer’s Neuropathology. Mol Neurobiol 2019; 56(12): 8255-76.
[http://dx.doi.org/10.1007/s12035-019-01660-3 ] [PMID: 31209782]
[25]
Ramsay RR, Popovic-Nikolic MR, Nikolic K, Uliassi E, Bolognesi ML. A perspective on multi-target drug discovery and design for complex diseases. Clin Transl Med 2018; 7(1): 3.
[http://dx.doi.org/10.1186/s40169-017-0181-2 ] [PMID: 29340951]
[26]
Folch J, Petrov D, Ettcheto M. Current Research Therapeutic Strategies for Alzheimer’s Disease Treatment Neural Plast 2016.
[27]
Youdim MBH. Multi target neuroprotective and neurorestorative anti-Parkinson and anti-Alzheimer drugs ladostigil and m30 derived from rasagiline. Exp Neurobiol 2013; 22(1): 1-10.
[http://dx.doi.org/10.5607/en.2013.22.1.1 ] [PMID: 23585716]
[28]
Weinstock M, Bejar C, Wang R-H, et al. TV3326, a Novel Neuroprotective Drug with Cholinesterase and Monoamine Oxidase Inhibitory Activities for the Treatment of Alzheimer’s DiseaseAdvances in Research on Neurodegeneration. Vienna: Springer Vienna 2000; pp. 157-69.
[http://dx.doi.org/10.1007/978-3-7091-6301-6_10]
[29]
Weinstock M. Selectivity of Cholinesterase Inhibition: Clinical Implications for the Treatment of Alzheimer’s Disease. CNS Drugs 1999; 12: 307-23.
[http://dx.doi.org/10.2165/00023210-199912040-00005]
[30]
Yogev-Falach M, Amit T, Bar-Am O, Weinstock M, Youdim MBH. Involvement of MAP kinase in the regulation of amyloid precursor protein processing by novel cholinesterase inhibitors derived from rasagiline. FASEB J 2002; 16(12): 1674-6.
[http://dx.doi.org/10.1096/fj.02-0198fje ] [PMID: 12206996]
[31]
Youdim MBH, Amit T, Bar-Am O, Weinstock M, Yogev-Falach M. M. Amyloid Processing and Signal Transduction Properties of Antiparkinson Antialzheimer Neuroprotective Drugs Rasagiline and TV3326 Proceedings of the Annals of. Vol. 993: 378-86.
[http://dx.doi.org/10.1111/j.1749-6632.2003.tb07548.x]
[32]
Yogev-Falach M, Bar-Am O, Amit T, Weinreb O, Youdim MBHA. A multifunctional, neuroprotective drug, ladostigil (TV3326), regulates holo-APP translation and processing. FASEB J 2006; 20(12): 2177-9.
[http://dx.doi.org/10.1096/fj.05-4910fje ] [PMID: 16935943]
[33]
Uddin MS, Kabir MT, Rahman MM, Mathew B, Shah MA, Ashraf GM. TV 3326 for Alzheimer’s Dementia: A Novel Multimodal ChE and MAO Inhibitors to Mitigate Alzheimer’s-like Neuropathology. J Pharm Pharmacol 2020; 13244. jphp.
[http://dx.doi.org/10.1111/jphp.13244]
[34]
Parkinson Study Group. A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005; 62(2): 241-8.
[http://dx.doi.org/10.1001/archneur.62.2.241 ] [PMID: 15710852]
[35]
Biglan KM, Schwid S, Eberly S, et al. Parkinson Study Group. Rasagiline improves quality of life in patients with early Parkinson’s disease. Mov Disord 2006; 21(5): 616-23.
[http://dx.doi.org/10.1002/mds.20764 ] [PMID: 16450340]
[36]
Chen JJ, Swope DM. Clinical pharmacology of rasagiline: a novel, second-generation propargylamine for the treatment of Parkinson disease. J Clin Pharmacol 2005; 45(8): 878-94.
[http://dx.doi.org/10.1177/0091270005277935 ] [PMID: 16027398]
[37]
Weinstock M, Razin M, Chorev M, Enz A. Pharmacological evaluation of phenyl-carbamates as CNS-selective acetylcholinesterase inhibitors. J Neural Transm Suppl 1994; 43: 219-25.
[PMID: 7884403]
[38]
Machado JC. Review: rivastigmine reduces rate of cognitive decline and improves performance in mild to moderate Alzheimer’s. Evid Based Ment Health 2009; 12(4): 113.
[http://dx.doi.org/10.1136/ebmh.12.4.113 ] [PMID: 19854775]
[39]
Weinreb O, Mandel S, Bar-Am O, et al. Multifunctional neuroprotective derivatives of rasagiline as anti-Alzheimer’s disease drugs. Neurotherapeutics 2009; 6(1): 163-74.
[http://dx.doi.org/10.1016/j.nurt.2008.10.030 ] [PMID: 19110207]
[40]
Johnson S, Tazik S, Lu D, et al. The New Inhibitor of Monoamine Oxidase, M30, has a Neuroprotective Effect Against Dexamethasone-Induced Brain Cell Apoptosis. Front Neurosci 2010; 4: 180-0.
[http://dx.doi.org/10.3389/fnins.2010.00180 ] [PMID: 21103012]
[41]
Zheng H, Gal S, Weiner LM, et al. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases: in vitro studies on antioxidant activity, prevention of lipid peroxide formation and monoamine oxidase inhibition. J Neurochem 2005; 95(1): 68-78.
[http://dx.doi.org/10.1111/j.1471-4159.2005.03340.x ] [PMID: 16181413]
[42]
Weinstock M, Gorodetsky E, Poltyrev T, Gross A, Sagi Y, Youdim M. A novel cholinesterase and brain-selective monoamine oxidase inhibitor for the treatment of dementia comorbid with depression and Parkinson’s disease. Prog Neuropsychopharmacol Biol Psychiatry 2003; 27(4): 555-61.
[http://dx.doi.org/10.1016/S0278-5846(03)00053-8 ] [PMID: 12787840]
[43]
Weinstock M, Bejar C, Wang RH, et al. TV3326, a novel neuroprotective drug with cholinesterase and monoamine oxidase inhibitory activities for the treatment of Alzheimer’s disease. J Neural Transm Suppl 2000; 60(60): 157-69.
[http://dx.doi.org/10.1007/978-3-7091-6301-6_10 ] [PMID: 11205137]
[44]
Sagi Y, Driguès N, Youdim MBH. The neurochemical and behavioral effects of the novel cholinesterase-monoamine oxidase inhibitor, ladostigil, in response to L-dopa and L-tryptophan, in rats. Br J Pharmacol 2005; 146(4): 553-60.
[http://dx.doi.org/10.1038/sj.bjp.0706355 ] [PMID: 16086033]
[45]
Davis KL, Rosenberg GS. Induction of behavioral supersensitivity to apomorphine by DFP treatment. Life Sci 1981; 28(17): 1953-9.
[http://dx.doi.org/10.1016/0024-3205(81)90304-0 ] [PMID: 7253803]
[46]
Rastogi SK, Rastogi RB, Singhal RL, Lapierre YD. Effects of cholinergic and GABAergic drugs on apomorphine-induced gnawing behavior in rats. Biol Psychiatry 1982; 17(8): 937-46.
[PMID: 6288127]
[47]
Scali C, Casamenti F, Bellucci A, Costagli C, Schmidt B, Pepeu G. Effect of subchronic administration of metrifonate, rivastigmine and donepezil on brain acetylcholine in aged F344 rats. J Neural Transm (Vienna) 2002; 109(7-8): 1067-80.
[http://dx.doi.org/10.1007/s007020200090 ] [PMID: 12111444]
[48]
Sterling J, Herzig Y, Youdim MBH, Weinstock M. The Design and Synthesis of Dual Action Compounds for the Treatment of Alzheimer’s Disease 2000.
[49]
Weinstock M, Kirschbaum-Slager N, Lazarovici P, Bejar C, Shoami E, Youdim MB. Cell Culture and In Vivo Neuroprotective Effects of Novel Cholinesterase-MAO Inhibitors. Ann N Y Acad Sci 2000; 939: 148-61.
[http://dx.doi.org/10.1111/j.1749-6632.2001.tb03622.x ] [PMID: 11462767]
[50]
Sagi Y, Weinstock M, Youdim MBH. Attenuation of MPTPinduced dopaminergic neurotoxicity by TV3326, a cholinesterase monoamine oxidase inhibitor. J Neurochem 2003; 86(2): 290-7.
[http://dx.doi.org/10.1046/j.1471-4159.2003.01801.x ] [PMID: 12871570]
[51]
Hubálek F, Binda C, Li M, et al. Inactivation of purified human recombinant monoamine oxidases A and B by rasagiline and its analogues. J Med Chem 2004; 47(7): 1760-6.
[http://dx.doi.org/10.1021/jm0310885 ] [PMID: 15027867]
[52]
Sterling J, Herzig Y, Goren T, et al. Novel dual inhibitors of AChE and MAO derived from hydroxy aminoindan and phenethylamine as potential treatment for Alzheimer’s disease. J Med Chem 2002; 45(24): 5260-79.
[http://dx.doi.org/10.1021/jm020120c ] [PMID: 12431053]
[53]
Weinstock M, Poltyrev T, Bejar C, Youdim MB. Effect of TV3326, a novel monoamine-oxidase cholinesterase inhibitor, in rat models of anxiety and depression. Psychopharmacology (Berl) 2002; 160(3): 318-24.
[http://dx.doi.org/10.1007/s00213-001-0978-x ] [PMID: 11889501]
[54]
Buccafusco JJ, Terry AV Jr, Goren T, Blaugrun E. Potential cognitive actions of (n-propargly-(3r)-aminoindan-5-yl)-ethyl, methyl carbamate (tv3326), a novel neuroprotective agent, as assessed in old rhesus monkeys in their performance of versions of a delayed matching task. Neuroscience 2003; 119(3): 669-78.
[http://dx.doi.org/10.1016/S0306-4522(02)00937-5 ] [PMID: 12809688]
[55]
Weinstock M, Kirschbaum-Slager N, Lazarovici P, Bejar C, Youdim MBH, Shoham S. Neuroprotective Effects of Novel Cholinesterase Inhibitors Derived from Rasagiline as Potential Anti-Alzheimer Drugs. Ann N Y Acad Sci 2006; 939: 148-61.
[http://dx.doi.org/10.1111/j.1749-6632.2001.tb03622.x]
[56]
Shoham S, Bejar C, Kovalev E, Schorer-Apelbaum D, Weinstock M. Ladostigil prevents gliosis, oxidative-nitrative stress and memory deficits induced by intracerebroventricular injection of streptozotocin in rats. Neuropharmacology 2007; 52(3): 836-43.
[http://dx.doi.org/10.1016/j.neuropharm.2006.10.005 ] [PMID: 17123555]
[57]
Luques L, Shoham S, Weinstock M. Chronic brain cytochrome oxidase inhibition selectively alters hippocampal cholinergic innervation and impairs memory: prevention by ladostigil. Exp Neurol 2007; 206(2): 209-19.
[http://dx.doi.org/10.1016/j.expneurol.2007.04.007 ] [PMID: 17580085]
[58]
Weinreb O, Drigues N, Sagi Y, Reznick AZ, Amit T, Youdim MBH. The application of proteomics and genomics to the study of age-related neurodegeneration and neuroprotection. Antioxid Redox Signal 2007; 9(2): 169-79.
[http://dx.doi.org/10.1089/ars.2007.9.169 ] [PMID: 17115941]
[59]
Kern A, Roempp B, Prager K, Walter J, Behl C. Down-regulation of endogenous amyloid precursor protein processing due to cellular aging. J Biol Chem 2006; 281(5): 2405-13.
[http://dx.doi.org/10.1074/jbc.M505625200 ] [PMID: 16303768]
[60]
Vetrivel KS, Thinakaran G. Amyloidogenic processing of β-amyloid precursor protein in intracellular compartments. Neurology 2006; 66(2)(Suppl. 1): S69-73.
[http://dx.doi.org/10.1212/01.wnl.0000192107.17175.39 ] [PMID: 16432149]
[61]
Maruyama W, Weinstock M, Youdim MBH, Nagai M, Naoi M. Anti-apoptotic action of anti-Alzheimer drug, TV3326 [(Npropargyl)-(3R)-aminoindan-5-yl]-ethyl methyl carbamate, a novel cholinesterase-monoamine oxidase inhibitor. Neurosci Lett 2003; 341(3): 233-6.
[http://dx.doi.org/10.1016/S0304-3940(03)00211-8 ] [PMID: 12697291]
[62]
Weinreb O, Bar-Am O, Amit T, Drigues N, Sagi Y, Youdim MBH. The neuroprotective effect of ladostigil against hydrogen peroxidemediated cytotoxicity. Chem Biol Interact 2008; 175(1-3): 318-26.
[http://dx.doi.org/10.1016/j.cbi.2008.05.038 ] [PMID: 18598687]
[63]
Bar-Am O, Weinreb O, Amit T, Youdim MBH. The novel cholinesterase-monoamine oxidase inhibitor and antioxidant, ladostigil, confers neuroprotection in neuroblastoma cells and aged rats. J Mol Neurosci 2009; 37(2): 135-45.
[http://dx.doi.org/10.1007/s12031-008-9139-6 ] [PMID: 18751929]
[64]
Weinreb O, Amit T, Bar-Am O, Youdim MBH. Induction of Neurotrophic Factors GDNF and BDNF Associated with the Mechanism of Neurorescue Action of Rasagiline and Ladostigil: New Insights and Implications for Therapy Proceedings of the Annals of. Vol. 1122: 155-68.
[http://dx.doi.org/10.1196/annals.1403.011]
[65]
Pakaski M, Kasa P. Role of acetylcholinesterase inhibitors in the metabolism of amyloid precursor protein. Curr Drug Targets CNS Neurol Disord 2003; 2(3): 163-71.
[http://dx.doi.org/10.2174/1568007033482869 ] [PMID: 12769797]
[66]
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]
[67]
Uddin MS, Mamun AA, Takeda S, Sarwar MS, Begum MM. Analyzing the chance of developing dementia among geriatric people: a cross-sectional pilot study in Bangladesh. Psychogeriatrics 2019; 19(2): 87-94.
[http://dx.doi.org/10.1111/psyg.12368 ] [PMID: 30221441]
[68]
Murphy MP, LeVine H III. Alzheimer’s disease and the amyloid-β peptide. J Alzheimers Dis 2010; 19(1): 311-23.
[http://dx.doi.org/10.3233/JAD-2010-1221 ] [PMID: 20061647]
[69]
Zhang X, Fu Z, Meng L, He M, Zhang Z. The Early Events That Initiate β-Amyloid Aggregation in Alzheimer’s Disease. Front Aging Neurosci 2018; 10: 359.
[http://dx.doi.org/10.3389/fnagi.2018.00359 ] [PMID: 30542277]
[70]
Lee JC, Kim SJ, Hong S, Kim Y. Diagnosis of Alzheimer’s disease utilizing amyloid and tau as fluid biomarkers. Exp Mol Med 2019; 51(5): 1-10.
[http://dx.doi.org/10.1038/s12276-019-0250-2 ] [PMID: 31073121]
[71]
Bar-Am O, Amit T, Weinreb O, Youdim MBH, Mandel S. Propargylamine containing compounds as modulators of proteolytic cleavage of amyloid-beta protein precursor: involvement of MAPK and PKC activation. J Alzheimers Dis 2010; 21(2): 361-71.
[http://dx.doi.org/10.3233/JAD-2010-100150 ] [PMID: 20555137]
[72]
Uddin MS, Kabir MT. Emerging Signal Regulating Potential of Genistein Against Alzheimer’s Disease: A Promising Molecule of Interest. Front Cell Dev Biol 2019; 7: 197.
[http://dx.doi.org/10.3389/fcell.2019.00197 ] [PMID: 31620438]
[73]
Harilal S, Jose J, Parambi DGT, et al. Advancements in nanotherapeutics for Alzheimer’s disease: current perspectives. J Pharm Pharmacol 2019; 71(9): 1370-83.
[http://dx.doi.org/10.1111/jphp.13132 ] [PMID: 31304982]
[74]
Mills J, Reiner PB. Regulation of amyloid precursor protein cleavage. J Neurochem 1999; 72(2): 443-60.
[http://dx.doi.org/10.1046/j.1471-4159.1999.0720443.x ] [PMID: 9930716]
[75]
Mills J, Reiner PB. Mitogen-activated protein kinase is involved in N-methyl-D-aspartate receptor regulation of amyloid precursor protein cleavage. Neuroscience 1999; 94(4): 1333-8.
[http://dx.doi.org/10.1016/S0306-4522(99)00381-4 ] [PMID: 10625071]
[76]
Zhang YW, Xu H. Molecular and cellular mechanisms for Alzheimer’s disease: understanding APP metabolism. Curr Mol Med 2007; 7(7): 687-96.
[http://dx.doi.org/10.2174/156652407782564462 ] [PMID: 18045146]
[77]
Hung AY, Haass C, Nitsch RM, et al. Activation of protein kinase C inhibits cellular production of the amyloid β-protein. J Biol Chem 1993; 268(31): 22959-62.
[PMID: 8226807]
[78]
Bar-Am O, Yogev-Falach M, Amit T, Sagi Y, Youdim MB. Regulation of protein kinase C by the anti-Parkinson drug, MAO-B inhibitor, rasagiline and its derivatives, in vivo. J Neurochem 2004; 89(5): 1119-25.
[http://dx.doi.org/10.1111/j.1471-4159.2004.02425.x ] [PMID: 15147504]
[79]
Yogev-Falach M, Amit T, Bar-Am O, Youdim MB. The importance of propargylamine moiety in the anti-Parkinson drug rasagiline and its derivatives in MAPK-dependent amyloid precursor protein processing. FASEB J 2003; 17(15): 2325-7.
[http://dx.doi.org/10.1096/fj.03-0078fje ] [PMID: 14525944]
[80]
Zhu X, Raina AK, Lee HG, Casadesus G, Smith MA, Perry G. Oxidative stress signalling in Alzheimer’s disease. Brain Res 2004; 1000(1-2): 32-9.
[http://dx.doi.org/10.1016/j.brainres.2004.01.012 ] [PMID: 15053949]
[81]
Uddin MS, Upaganlawar AB. Oxidative Stress and Antioxidant Defense: Biomedical Value in Health and Diseases. USA: Nova Science Publishers 2019.
[82]
Uddin MS, Kabir MT. Oxidative Stress in Alzheimer' Disease: Molecular Hallmarks of Underlying Vulnerability. In: Biological, Diagnostic and Therapeutic Advances in Alzheimer' Disease; Springer Singapore, 2019; pp. 91-115.
[83]
Rhee SG, Kim KH, Chae HZ, et al. Antioxidant Defense Mechanisms: A New Thiol-Specific Antioxidant Enzyme. Ann N Y Acad Sci 1994; 738: 86-92.
[84]
Yaffe K, Laffan AM, Harrison SL, et al. Sleep-disordered breathing, hypoxia, and risk of mild cognitive impairment and dementia in older women. JAMA 2011; 306(6): 613-9.
[http://dx.doi.org/10.1001/jama.2011.1115 ] [PMID: 21828324]
[85]
Halliwell B. Reactive oxygen species and the central nervous system. J Neurochem 1992; 59(5): 1609-23.
[http://dx.doi.org/10.1111/j.1471-4159.1992.tb10990.x ] [PMID: 1402908]
[86]
Zhang L, Yu H, Sun Y, et al. Protective effects of salidroside on hydrogen peroxide-induced apoptosis in SH-SY5Y human neuroblastoma cells. Eur J Pharmacol 2007; 564(1-3): 18-25.
[http://dx.doi.org/10.1016/j.ejphar.2007.01.089 ] [PMID: 17349619]
[87]
Halliwell B. Role of free radicals in the neurodegenerative diseases: therapeutic implications for antioxidant treatment. Drugs Aging 2001; 18(9): 685-716.
[http://dx.doi.org/10.2165/00002512-200118090-00004 ] [PMID: 11599635]
[88]
Youdim MBH, Edmondson D, Tipton KF. The therapeutic potential of monoamine oxidase inhibitors. Nat Rev Neurosci 2006; 7(4): 295-309.
[http://dx.doi.org/10.1038/nrn1883 ] [PMID: 16552415]
[89]
Weinreb O, Amit T, Bar-Am O, Youdim MBH. Rasagiline: a novel anti-Parkinsonian monoamine oxidase-B inhibitor with neuroprotective activity. Prog Neurobiol 2010; 92(3): 330-44.
[http://dx.doi.org/10.1016/j.pneurobio.2010.06.008 ] [PMID: 20600573]
[90]
Weinreb O, Amit T, Bar-Am O, Youdim MBH. A Novel Anti-Alzheimer’s Disease Drug, Ladostigil Neuroprotective, Multimodal Brain-Selective Monoamine Oxidase and Cholinesterase InhibitorInternational Review of Neurobiology. Academic Press Inc. 2011; Vol. 100: pp. 191-215.
[91]
Gal S, Zheng H, Fridkin M, Youdim MBH. Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. In vivo selective brain monoamine oxidase inhibition and prevention of MPTP-induced striatal dopamine depletion. J Neurochem 2005; 95(1): 79-88.
[http://dx.doi.org/10.1111/j.1471-4159.2005.03341.x ] [PMID: 16181414]
[92]
Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MBH. Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology 2004; 46(2): 254-63.
[http://dx.doi.org/10.1016/j.neuropharm.2003.09.005 ] [PMID: 14680763]
[93]
Zheng H, Weiner LM, Bar-Am O, et al. Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer’s, Parkinson’s, and other neurodegenerative diseases. Bioorg Med Chem 2005; 13(3): 773-83.
[http://dx.doi.org/10.1016/j.bmc.2004.10.037 ] [PMID: 15653345]
[94]
Borisenko GG, Kagan VE, Hsia CJC, Schor NF. Interaction between 6-hydroxydopamine and transferrin: “Let my iron go". Biochemistry 2000; 39(12): 3392-400.
[http://dx.doi.org/10.1021/bi992296v ] [PMID: 10727233]
[95]
Van der Schyf CJ, Gal S, Geldenhuys WJ, Youdim MBH. Multifunctional neuroprotective drugs targeting monoamine oxidase inhibition, iron chelation, adenosine receptors, and cholinergic and glutamatergic action for neurodegenerative diseases. Expert Opin Investig Drugs 2006; 15(8): 873-86.
[http://dx.doi.org/10.1517/13543784.15.8.873 ] [PMID: 16859391]
[96]
Youdim MBH, Buccafusco JJ. Multi-functional drugs for various CNS targets in the treatment of neurodegenerative disorders. Trends Pharmacol Sci 2005; 26(1): 27-35.
[http://dx.doi.org/10.1016/j.tips.2004.11.007 ] [PMID: 15629202]
[97]
Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H, Fridkin M, Youdim MBH. Therapeutic targets and potential of the novel brain permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, for the treatment of Alzheimer’s disease. J Neurochem 2007; 100(2): 490-502.
[http://dx.doi.org/10.1111/j.1471-4159.2006.04258.x ] [PMID: 17144902]
[98]
Avramovich-Tirosh Y, Reznichenko L, Mit T, et al. Neurorescue activity, APP regulation and amyloid-beta peptide reduction by novel multi-functional brain permeable iron- chelating- antioxidants, M-30 and green tea polyphenol, EGCG. Curr Alzheimer Res 2007; 4(4): 403-11.
[http://dx.doi.org/10.2174/156720507781788927 ] [PMID: 17908043]
[99]
Ward MW, Kögel D, Prehn JHM. Neuronal apoptosis: BH3-only proteins the real killers? J Bioenerg Biomembr 2004; 36(4): 295-8.
[http://dx.doi.org/10.1023/B:JOBB.0000041756.23918.11 ] [PMID: 15377860]
[100]
Copani A, Condorelli F, Caruso A, et al. Mitotic signaling by betaamyloid causes neuronal death. FASEB J 1999; 13(15): 2225-34.
[http://dx.doi.org/10.1096/fasebj.13.15.2225 ] [PMID: 10593870]
[101]
Wu Q, Combs C, Cannady SB, Geldmacher DS, Herrup K. Betaamyloid activated microglia induce cell cycling and cell death in cultured cortical neurons. Neurobiol Aging 2000; 21(6): 797-806.
[http://dx.doi.org/10.1016/S0197-4580(00)00219-0 ] [PMID: 11124423]

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