[1]
Mount, C.; Downton, C. Alzheimer disease: Progress or profit? Nat. Med., 2006, 12(7), 780-784. [http://dx.doi.org/ 10.1038/nm0706-780]. [PMID: 16829947].
[2]
Hebert, L.E.; Scherr, P.A.; Bienias, J.L.; Bennett, D.A.; Evans, D.A. State-specific projections through 2025 of Alzheimer disease prevalence. Neurology, 2004, 62(9), 1645. [http://dx.doi.org/ 10.1212/01.WNL.0000123018.01306.10]. [PMID: 15136705].
[3]
Ashford, J.W. APOE genotype effects on Alzheimer’s disease onset and epidemiology. J. Mol. Neurosci., 2004, 23(3), 157-165. [http://dx.doi.org/10.1385/JMN:23:3:157]. [PMID: 15181244].
[4]
Katzman, R. Alzheimer’s disease. N. Engl. J. Med., 1986, 314(15), 964-973. [http://dx.doi.org/10.1056/NEJM198604103141506]. [PMID: 2870433].
[5]
Blass, J.P. Li-wen Ko, B.V.M.; Wisniewski, H.M. Pathology of Alzheimer’s disease. Psychiatr. Clin. (Basel), 1991, 14(2), 397-420.
[6]
Okura, T.; Plassman, B.L.; Steffens, D.C.; Llewellyn, D.J.; Potter, G.G.; Langa, K.M. Neuropsychiatric symptoms and the risk of institutionalization and death: the aging, demographics, and memory study. J. Am. Geriatr. Soc., 2011, 59(3), 473-481. [http://dx.doi.org/10.1111/j.1532-5415.2011.03314.x]. [PMID: 21391937].
[7]
Gauthier, S.; Cummings, J.; Ballard, C.; Brodaty, H.; Grossberg, G.; Robert, P.; Lyketsos, C. Management of behavioral problems in Alzheimer’s disease. Int. Psychogeriatr., 2010, 22(3), 346-372. [http://dx.doi.org/10.1017/S1041610209991505]. [PMID: 20096151].
[8]
Borisovskaya, A.; Pascualy, M.; Borson, S. Cognitive and neuropsychiatric impairments in Alzheimer’s disease: Current treatment strategies. Curr. Psychiatry Rep., 2014, 16(9), 470. [http://dx.doi.org/10.1007/s11920-014-0470-z]. [PMID: 25023513].
[9]
Goedert, M.; Spillantini, M.G. A century of Alzheimer’s disease. Science, 2006, 314(5800), 777-781. [http://dx.doi.org/ 10.1126/science.1132814]. [PMID: 17082447].
[10]
Kidd, M. Paired helical filaments in electron microscopy of Alzheimer’s disease. Nature, 1963, 197, 192-193. [http://dx.doi.org/10.1038/197192b0]. [PMID: 14032480].
[11]
Šimić, G.; Babić Leko, M.; Wray, S.; Harrington, C.R.; Delalle, I.; Jovanov-Milošević, N.; Bažadona, D.; Buée, L.; de Silva, R.; Di Giovanni, G.; Wischik, C.M.; Hof, P.R. Monoaminergic neuropathology in Alzheimer’s disease. Prog. Neurobiol., 2017, 151, 101-138. [http://dx.doi.org/10.1016/j.pneurobio.2016.04.001]. [PMID: 27084356].
[12]
Zetterberg, H.; Blennow, K.; Hanse, E. Amyloid beta and APP as biomarkers for Alzheimer’s disease. Exp. Gerontol., 2010, 45(1), 23-29. [http://dx.doi.org/10.1016/j.exger.2009.08.002]. [PMID: 19698775].
[13]
Brandt, R.; Hundelt, M.; Shahani, N. Tau alteration and neuronal degeneration in tauopathies: Mechanisms and models. Biochim. Biophys. Acta, 2005, 1739(2-3), 331-354. [http://dx.doi.org/ 10.1016/j.bbadis.2004.06.018]. [PMID: 15615650].
[14]
Zhang, Y.; Tian, Q.; Zhang, Q.; Zhou, X.; Liu, S.; Wang, J.Z. Hyperphosphorylation of microtubule-associated tau protein plays dual role in neurodegeneration and neuroprotection. Pathophysiology, 2009, 16(4), 311-316. [http://dx.doi.org/10.1016/ j.pathophys.2009.02.003]. [PMID: 19410438].
[15]
Craig, L.A.; Hong, N.S.; McDonald, R.J. Revisiting the cholinergic hypothesis in the development of Alzheimer’s disease. Neurosci. Biobehav. Rev., 2011, 35(6), 1397-1409. [http://dx.doi.org/10.1016/ j.neubiorev.2011.03.001]. [PMID: 21392524].
[16]
Trepanier, C.H.; Milgram, N.W. Neuroinflammation in Alzheimer’s disease: are NSAIDs and selective COX-2 inhibitors the next line of therapy? J. Alzheimers Dis., 2010, 21(4), 1089-1099. [http://dx.doi.org/10.3233/JAD-2010-090667]. [PMID: 21504126].
[17]
Tan, M.S.; Yu, J.T.; Jiang, T.; Zhu, X.C.; Tan, L. The NLRP3 inflammasome in Alzheimer’s disease. Mol. Neurobiol., 2013, 48(3), 875-882. [http://dx.doi.org/10.1007/s12035-013-8475-x]. [PMID: 23686772].
[18]
Sultana, R.; Butterfield, D.A. Role of oxidative stress in the progression of Alzheimer’s disease. J. Alzheimers Dis., 2010, 19(1), 341-353. [http://dx.doi.org/10.3233/JAD-2010-1222]. [PMID: 20061649].
[19]
Wyllie, A.H. Apoptosis: Cell death in tissue regulation. J. Pathol., 1987, 153(4), 313-316. [http://dx.doi.org/10.1002/ path.1711530404]. [PMID: 3323435].
[20]
Axelsen, P.H.; Komatsu, H.; Murray, I.V. Oxidative stress and cell membranes in the pathogenesis of Alzheimer’s disease. Physiology (Bethesda), 2011, 26(1), 54-69. [http://dx.doi.org/10.1152/ physiol.00024.2010]. [PMID: 21357903].
[21]
Shimohama, S. Apoptosis in Alzheimer’s disease--An update. Apoptosis, 2000, 5(1), 9-16. [http://dx.doi.org/10.1023/ A:1009625323388]. [PMID: 11227497].
[22]
Bezprozvanny, I.; Mattson, M.P. Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease. Trends Neurosci., 2008, 31(9), 454-463. [http://dx.doi.org/10.1016/ j.tins.2008.06.005]. [PMID: 18675468].
[23]
Kocahan, S.; Doğan, Z. Mechanisms of Alzheimer’s disease pathogenesis and prevention: The brain, neural pathology, N-methyl-D-aspartate receptors, tau protein and other risk factors. Clin. Psychopharmacol. Neurosci., 2017, 15(1), 1-8. [http://dx.doi.org/ 10.9758/cpn.2017.15.1.1]. [PMID: 28138104].
[24]
Weldon, D.T.; Rogers, S.D.; Ghilardi, J.R.; Finke, M.P.; Cleary, J.P.; O’Hare, E.; Esler, W.P.; Maggio, J.E.; Mantyh, P.W. Fibrillar beta-amyloid induces microglial phagocytosis, expression of inducible nitric oxide synthase, and loss of a select population of neurons in the rat CNS in vivo. J. Neurosci., 1998, 18(6), 2161-2173. [http://dx.doi.org/10.1523/JNEUROSCI.18-06-02161.1998]. [PMID: 9482801].
[25]
Zhu, X.; Castellani, R.J.; Takeda, A.; Nunomura, A.; Atwood, C.S.; Perry, G.; Smith, M.A. Differential activation of neuronal ERK, JNK/SAPK and p38 in Alzheimer disease: the ‘two hit’ hypothesis. Mech. Ageing Dev., 2001, 123(1), 39-46. [http://dx.doi.org/ 10.1016/S0047-6374(01)00342-6]. [PMID: 11640950].
[26]
Smith, M.A.; Nunomura, A.; Zhu, X.; Takeda, A.; Perry, G. Metabolic, metallic, and mitotic sources of oxidative stress in Alzheimer disease. Antioxid. Redox Signal., 2000, 2(3), 413-420. [http://dx.doi.org/10.1089/15230860050192198]. [PMID: 11229355].
[27]
Lim, D.; Iyer, A.; Ronco, V.; Grolla, A.A.; Canonico, P.L.; Aronica, E.; Genazzani, A.A. Amyloid beta deregulates astroglial mGluR5-mediated calcium signaling via calcineurin and Nf-kB. Glia, 2013, 61(7), 1134-1145. [http://dx.doi.org/10.1002/ glia.22502]. [PMID: 23616440].
[28]
Garcez, M.L.; Mina, F.; Bellettini-Santos, T.; Carneiro, F.G.; Luz, A.P.; Schiavo, G.L.; Andrighetti, M.S.; Scheid, M.G.; Bolfe, R.P.; Budni, J. Minocycline reduces inflammatory parameters in the brain structures and serum and reverses memory impairment caused by the administration of amyloid β (1-42) in mice. Prog. Neuropsychopharmacol. Biol. Psychiatry, 2017, 77, 23-31. [http://dx.doi.org/10.1016/j.pnpbp.2017.03.010]. [PMID: 28336494].
[29]
Muñoz-Torrero, D. Acetylcholinesterase inhibitors as disease-modifying therapies for Alzheimer’s disease. Curr. Med. Chem., 2008, 15(24), 2433-2455. [http://dx.doi.org/10.2174/ 092986708785909067]. [PMID: 18855672].
[30]
Haas, C. Strategies, development, and pitfalls of therapeutic options for Alzheimer’s disease. J. Alzheimers Dis., 2012, 28(2), 241-281. [http://dx.doi.org/10.3233/JAD-2011-110986]. [PMID: 21987594].
[31]
Ansari, N.; Khodagholi, F. Natural products as promising drug candidates for the treatment of Alzheimer’s disease: Molecular mechanism aspect. Curr. Neuropharmacol., 2013, 11(4), 414-429. [http://dx.doi.org/10.2174/1570159X11311040005]. [PMID: 24381531].
[32]
McGleenon, B.M.; Dynan, K.B.; Passmore, A.P. Acetylcholinesterase inhibitors in Alzheimer’s disease. Br. J. Clin. Pharmacol., 1999, 48(4), 471-480. [http://dx.doi.org/10.1046/j.1365-2125.1999.00026.x]. [PMID: 10583015].
[33]
Watkins, P.B.; Zimmerman, H.J.; Knapp, M.J.; Gracon, S.I.; Lewis, K.W. Hepatotoxic effects of tacrine administration in patients with Alzheimer’s disease. JAMA, 1994, 271(13), 992-998. [http://dx.doi.org/10.1001/jama.1994.03510370044030]. [PMID: 8139084].
[34]
Cummings, J.L. Treatment of Alzheimer’s disease: current and future therapeutic approaches. Rev. Neurol. Dis., 2004, 1(2), 60-69. [PMID: 16400259].
[35]
Standridge, J.B. Pharmacotherapeutic approaches to the prevention of Alzheimer’s disease. Am. J. Geriatr. Pharmacother., 2004, 2(2), 119-132. [http://dx.doi.org/10.1016/S1543-5946(04)90017-7]. [PMID: 15555488].
[36]
Shal, B.; Ding, W.; Ali, H.; Kim, Y.S.; Khan, S. Anti-Neuroinflammatory potential of natural products in attenuation of Alzheimer’s disease. Front. Pharmacol., 2018, 9, 548. [http://dx.doi.org/10.3389/fphar.2018.00548]. [PMID: 29896105].
[37]
Deng, Y.H.; Wang, N.N.; Zou, Z.X.; Zhang, L.; Xu, K.P.; Chen, A.F.; Cao, D.S.; Tan, G.S. Multi-Target screening and experimental validation of natural products from Selaginella plants against Alzheimer’s disease. Front. Pharmacol., 2017, 8, 539. [http://dx.doi.org/10.3389/fphar.2017.00539]. [PMID: 28890698].
[38]
Farver, D. The use of “natural products” in clinical medicine. S. D. J. Med., 1996, 49(4), 129-130. [PMID: 8936438].
[39]
Anekonda, T.S.; Reddy, P.H. Can herbs provide a new generation of drugs for treating Alzheimer’s disease? Brain Res. Brain Res. Rev., 2005, 50(2), 361-376. [http://dx.doi.org/10.1016/ j.brainresrev.2005.09.001]. [PMID: 16263176].
[40]
Essa, M.M.; Vijayan, R.K.; Castellano-Gonzalez, G.; Memon, M.A.; Braidy, N.; Guillemin, G.J. Neuroprotective effect of natural products against Alzheimer’s disease. Neurochem. Res., 2012, 37(9), 1829-1842. [http://dx.doi.org/10.1007/s11064-012-0799-9]. [PMID: 22614926].
[41]
Morales, I.; Guzmán-Martínez, L.; Cerda-Troncoso, C.; Farías, G.A.; Maccioni, R.B. Neuroinflammation in the pathogenesis of alzheimer’s disease. A rational framework for the search of novel therapeutic approaches. Front. Cell. Neurosci., 2014, 8, 112. [http://dx.doi.org/10.3389/fncel.2014.00112]. [PMID: 24795567].
[42]
Rees, T.M.; Brimijoin, S. The role of acetylcholinesterase in the pathogenesis of Alzheimer’s disease. Drugs Today (Barc), 2003, 39(1), 75-83. [http://dx.doi.org/10.1358/dot.2003.39.1.740206]. [PMID: 12669110].
[43]
Babic, T. The cholinergic hypothesis of Alzheimer’s disease: A review of progress. J. Neurol. Neurosurg. Psychiatry, 1999, 67(4), 558. [http://dx.doi.org/10.1136/jnnp.67.4.558]. [PMID: 10610396].
[44]
Pappas, B.A.; Bayley, P.J.; Bui, B.K.; Hansen, L.A.; Thal, L.J. Choline acetyltransferase activity and cognitive domain scores of Alzheimer’s patients. Neurobiol. Aging, 2000, 21(1), 11-17. [http://dx.doi.org/10.1016/S0197-4580(00)00090-7]. [PMID: 10794843].
[45]
Pepping, J.; Huperzine, A. Huperzine A. Am. J. Health Syst. Pharm., 2000, 57(6), 530-533-534. [http://dx.doi.org/10.1093/ ajhp/57.6.530] [PMID: 10754762]
[46]
Ha, G.T.; Wong, R.K.; Zhang, Y. Huperzine a as potential treatment of Alzheimer’s disease: An assessment on chemistry, pharmacology, and clinical studies. Chem. Biodivers., 2011, 8(7), 1189-1204. [http://dx.doi.org/10.1002/cbdv.201000269]. [PMID: 21766442].
[47]
Wang, Y.; Tang, X.C.; Zhang, H.Y. Huperzine A alleviates synaptic deficits and modulates amyloidogenic and nonamyloidogenic pathways in APPswe/PS1dE9 transgenic mice. J. Neurosci. Res., 2012, 90(2), 508-517. [http://dx.doi.org/10.1002/jnr.22775]. [PMID: 22002568].
[48]
Wang, C.Y.; Zheng, W.; Wang, T.; Xie, J.W.; Wang, S.L.; Zhao, B.L.; Teng, W.P.; Wang, Z.Y. Huperzine A activates Wnt/β-catenin signaling and enhances the nonamyloidogenic pathway in an Alzheimer transgenic mouse model. Neuropsychopharmacology, 2011, 36(5), 1073-1089. [http://dx.doi.org/10.1038/ npp.2010.245]. [PMID: 21289607].
[49]
Gordon, R.K.; Nigam, S.V.; Weitz, J.A.; Dave, J.R.; Doctor, B.P.; Ved, H.S. The NMDA receptor ion channel: A site for binding of Huperzine A. J. Appl. Toxicol., 2001, 21(Suppl. 1), S47-S51. [http://dx.doi.org/10.1002/jat.805]. [PMID: 11920920].
[50]
Yang, L.; Ye, C.Y.; Huang, X.T.; Tang, X.C.; Zhang, H.Y. Decreased accumulation of subcellular amyloid-β with improved mitochondrial function mediates the neuroprotective effect of huperzine A. J. Alzheimers Dis., 2012, 31(1), 131-142. [http://dx.doi.org/10.3233/JAD-2012-120274]. [PMID: 22531425].
[51]
Camps, P.; Morral, J.; Muñoz-Torrero, D.; Badia, A.; Baños, J.E.; Vivas, N.M.; Barril, X.; Orozco, M.; Luque, F.J.; Luque, F.J. New tacrine-huperzine A hybrids (huprines): highly potent tight-binding acetylcholinesterase inhibitors of interest for the treatment of Alzheimer’s disease. J. Med. Chem., 2000, 43(24), 4657-4666. [http://dx.doi.org/10.1021/jm000980y]. [PMID: 11101357].
[52]
Camps, P.; Muñoz-Torrero, D. Tacrine-huperzine A hybrids (huprines):A new class of highly potent and selective acetylcholinesterase inhibitors of interest for the treatment of Alzheimer’s disease. Mini Rev. Med. Chem., 2001, 1(2), 163-174. [http://dx.doi.org/10.2174/1389557013406972]. [PMID: 12369981].
[53]
Rafii, M.S.; Walsh, S.; Little, J.T.; Behan, K.; Reynolds, B.; Ward, C.; Jin, S.; Thomas, R.; Aisen, P.S. A phase II trial of huperzine A in mild to moderate Alzheimer disease. Neurology, 2011, 76(16), 1389-1394. [http://dx.doi.org/10.1212/WNL.0b013e318216eb7b]. [PMID: 21502597].
[54]
Kulkarni, S.K.; Dhir, A. Berberine: A plant alkaloid with therapeutic potential for central nervous system disorders. Phytother. Res., 2010, 24(3), 317-324. [http://dx.doi.org/10.1002/ptr.2968]. [PMID: 19998323].
[55]
Vuddanda, P.R.; Chakraborty, S.; Singh, S. Berberine: A potential phytochemical with multispectrum therapeutic activities. Expert Opin. Investig. Drugs, 2010, 19(10), 1297-1307. [http://dx.doi.org/ 10.1517/13543784.2010.517745]. [PMID: 20836620].
[56]
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].
[57]
Zhu, F.; Wu, F.; Ma, Y.; Liu, G.; Li, Z.; Sun, Y.; Pei, Z. 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].
[58]
Yu, G.; Li, Y.; Tian, Q.; Liu, R.; Wang, Q.; Wang, J.Z.; Wang, X. Berberine attenuates calyculin A-induced cytotoxicity and Tau hyperphosphorylation in HEK293 cells. J. Alzheimers Dis., 2011, 24(3), 525-535. [http://dx.doi.org/10.3233/JAD-2011-101779]. [PMID: 21297267].
[59]
Jia, L.; Liu, J.; Song, Z.; Pan, X.; Chen, L.; Cui, X.; Wang, M. 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), 1510-1521. [http://dx.doi.org/10.1111/j.2042-7158.2012.01529.x]. [PMID: 22943182].
[60]
Durairajan, S.S.; Liu, L.F.; Lu, J.H.; Chen, L.L.; Yuan, Q.; Chung, S.K.; Huang, L.; Li, X.S.; Huang, J.D.; Li, M. Berberine ameliorates β-amyloid pathology, gliosis, and cognitive impairment in an Alzheimer’s disease transgenic mouse model. Neurobiol. Aging, 2012, 33(12), 2903-2919. [http://dx.doi.org/10.1016/ j.neurobiolaging.2012.02.016]. [PMID: 22459600].
[61]
Maczurek, A.; Hager, K.; Kenklies, M.; Sharman, M.; Martins, R.; Engel, J.; Carlson, D.A.; Münch, G. Lipoic acid as an anti-inflammatory and neuroprotective treatment for Alzheimer’s disease. Adv. Drug Deliv. Rev., 2008, 60(13-14), 1463-1470. [http://dx.doi.org/10.1016/j.addr.2008.04.015]. [PMID: 18655815].
[62]
Meyerhoff, J.L.; Yoorick, D.L.; Koenig, M.L.; Yourick, D.L. Treatment of central nervous system injuries or diseases comprises administering at least one lipoic acid, 2001, WO200180851A1.
[63]
Oboh, G.; Ademiluyi, A.O.; Akinyemi, A.J. Inhibition of acetylcholinesterase activities and some pro-oxidant induced lipid peroxidation in rat brain by two varieties of ginger (Zingiber officinale). Exp. Toxicol. Pathol., 2012, 64(4), 315-319. [http://dx.doi.org/10.1016/j.etp.2010.09.004]. [PMID: 20952170].
[64]
Rishton, G.M.; Arai, H.; Kai, Z.; Fullenwider, C.L.; Beierle, K Method of inhibiting, treating, or abatement of cognitive decline and Alzheimer's disease in a mammal, comprises administering a derivative of ginger oil to a mammal., 2011.
[65]
Martinez, G.A.; Alonso, G.D.; Dorronsoro, D.I.; Garcia, P.E. De-, Austria; De-, L.C.; Usan, E.P New spiro heterocyclic compounds are calcium channel blockers useful in the manufacture of a medicament for the treatment of brain ischemia, stroke, cognitive disorders, cerebrovascular dementia or neurodegenerative dementing disease. EP1609783A1; WO2005123073A1; EP1761262A1, 2005.
[66]
Bui, T.T.; Nguyen, T.H. Natural product for the treatment of Alzheimer’s disease. J. Basic Clin. Physiol. Pharmacol., 2017, 28(5), 413-423. [http://dx.doi.org/10.1515/jbcpp-2016-0147]. [PMID: 28708573].
[67]
Hardy, J.; Allsop, D. Amyloid deposition as the central event in the aetiology of Alzheimer’s disease. Trends Pharmacol. Sci., 1991, 12(10), 383-388. [http://dx.doi.org/10.1016/0165-6147(91)90609-V]. [PMID: 1763432].
[68]
Priller, C.; Bauer, T.; Mitteregger, G.; Krebs, B.; Kretzschmar, H.A.; Herms, J. Synapse formation and function is modulated by the amyloid precursor protein. J. Neurosci., 2006, 26(27), 7212-7221. [http://dx.doi.org/10.1523/JNEUROSCI.1450-06.2006]. [PMID: 16822978].
[69]
Turner, P.R.; O’Connor, K.; Tate, W.P.; Abraham, W.C. Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory. Prog. Neurobiol., 2003, 70(1), 1-32. [http://dx.doi.org/10.1016/S0301-0082(03)00089-3]. [PMID: 12927332].
[70]
Duce, J.A.; Tsatsanis, A.; Cater, M.A.; James, S.A.; Robb, E.; Wikhe, K.; Leong, S.L.; Perez, K.; Johanssen, T.; Greenough, M.A.; Cho, H.H.; Galatis, D.; Moir, R.D.; Masters, C.L.; McLean, C.; Tanzi, R.E.; Cappai, R.; Barnham, K.J.; Ciccotosto, G.D.; Rogers, J.T.; Bush, A.I. Iron-export ferroxidase activity of β-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell, 2010, 142(6), 857-867. [http://dx.doi.org/10.1016/ j.cell.2010.08.014]. [PMID: 20817278].
[71]
Haass, C.; Selkoe, D.J. Soluble protein oligomers in neurodegeneration: Lessons from the Alzheimer’s amyloid beta-peptide. Nat. Rev. Mol. Cell Biol., 2007, 8(2), 101-112. [http://dx.doi.org/ 10.1038/nrm2101]. [PMID: 17245412].
[72]
Nussbaum, J.M.; Seward, M.E.; Bloom, G.S. Alzheimer disease: A tale of two prions. Prion, 2013, 7(1), 14-19. [http://dx.doi.org/ 10.4161/pri.22118]. [PMID: 22965142].
[73]
Pulawski, W.; Ghoshdastider, U.; Andrisano, V.; Filipek, S. Ubiquitous amyloids. Appl. Biochem. Biotechnol., 2012, 166(7), 1626-1643. [http://dx.doi.org/10.1007/s12010-012-9549-3]. [PMID: 22350870].
[74]
Mudher, A.; Lovestone, S. Alzheimer’s disease-do tauists and baptists finally shake hands? Trends Neurosci., 2002, 25(1), 22-26. [http://dx.doi.org/10.1016/S0166-2236(00)02031-2]. [PMID: 11801334].
[75]
Esatbeyoglu, T.; Huebbe, P.; Ernst, I.M.; Chin, D.; Wagner, A.E.; Rimbach, G. Curcumin--From molecule to biological function. Angew. Chem. Int. Ed. Engl., 2012, 51(22), 5308-5332. [http://dx.doi.org/10.1002/anie.201107724]. [PMID: 22566109].
[76]
Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. Adv. Exp. Med. Biol., 2007, 595, 1-75. [http://dx.doi.org/10.1007/978-0-387-46401-5_1]. [PMID: 17569205].
[77]
Jiang, S.; Han, J.; Li, T.; Xin, Z.; Ma, Z.; Di, W.; Hu, W.; Gong, B.; Di, S.; Wang, D.; Yang, Y. Curcumin as a potential protective compound against cardiac diseases. Pharmacol. Res., 2017, 119, 373-383. [http://dx.doi.org/10.1016/j.phrs.2017.03.001]. [PMID: 28274852].
[78]
Hamaguchi, T.; Ono, K.; Yamada, M. REVIEW: Curcumin and Alzheimer’s disease. CNS Neurosci. Ther., 2010, 16(5), 285-297. [http://dx.doi.org/10.1111/j.1755-5949.2010.00147.x]. [PMID: 20406252].
[79]
Yang, F.; Lim, G.P.; Begum, A.N.; Ubeda, O.J.; Simmons, M.R.; Ambegaokar, S.S.; Chen, P.P.; Kayed, R.; Glabe, C.G.; Frautschy, S.A.; Cole, G.M. Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J. Biol. Chem., 2005, 280(7), 5892-5901. [http://dx.doi.org/ 10.1074/jbc.M404751200]. [PMID: 15590663].
[80]
Veldman, E.R.; Jia, Z.; Halldin, C.; Svedberg, M.M. Amyloid binding properties of curcumin analogues in Alzheimer’s disease postmortem brain tissue. Neurosci. Lett., 2016, 630, 183-188. [http://dx.doi.org/10.1016/j.neulet.2016.07.045]. [PMID: 27461789].
[81]
Sahu, P.K. Design, structure activity relationship, cytotoxicity and evaluation of antioxidant activity of curcumin derivatives/analogues. Eur. J. Med. Chem., 2016, 121, 510-516. [http://dx.doi.org/10.1016/j.ejmech.2016.05.037]. [PMID: 27318975].
[82]
Butterfield, D.; Castegna, A.; Pocernich, C.; Drake, J.; Scapagnini, G.; Calabrese, V. Nutritional approaches to combat oxidative stress in Alzheimer’s disease. J. Nutr. Biochem., 2002, 13(8), 444. [http://dx.doi.org/10.1016/S0955-2863(02)00205-X]. [PMID: 12165357].
[83]
DiSilvestro, R.A.; Joseph, E.; Zhao, S.; Bomser, J. Diverse effects of a low dose supplement of lipidated curcumin in healthy middle aged people. Nutr. J., 2012, 11, 79. [http://dx.doi.org/10.1186/ 1475-2891-11-79]. [PMID: 23013352].
[84]
Pocernich, C.B.; Lange, M.L.; Sultana, R.; Butterfield, D.A. Nutritional approaches to modulate oxidative stress in Alzheimer’s disease. Curr. Alzheimer Res., 2011, 8(5), 452-469. [http://dx.doi.org/10.2174/156720511796391908]. [PMID: 21605052].
[85]
López-Lázaro, M. Distribution and biological activities of the flavonoid luteolin. Mini Rev. Med. Chem., 2009, 9(1), 31-59. [http://dx.doi.org/10.2174/138955709787001712]. [PMID: 19149659].
[86]
Seelinger, G.; Merfort, I.; Schempp, C.M. Anti-oxidant, anti-inflammatory and anti-allergic activities of luteolin. Planta Med., 2008, 74(14), 1667-1677. [http://dx.doi.org/10.1055/s-0028-1088314]. [PMID: 18937165].
[87]
Zhou, F.; Chen, S.; Xiong, J.; Li, Y.; Qu, L. Luteolin reduces zinc-induced tau phosphorylation at Ser262/356 in an ROS-dependent manner in SH-SY5Y cells. Biol. Trace Elem. Res., 2012, 149(2), 273-279. [http://dx.doi.org/10.1007/s12011-012-9411-z]. [PMID: 22528780].
[88]
Liu, R.; Meng, F.; Zhang, L.; Liu, A.; Qin, H.; Lan, X.; Li, L.; Du, G. Luteolin isolated from the medicinal plant Elsholtzia rugulosa (Labiatae) prevents copper-mediated toxicity in β-amyloid precursor protein Swedish mutation overexpressing SH-SY5Y cells. Molecules, 2011, 16(3), 2084-2096. [http://dx.doi.org/10.3390/ molecules16032084]. [PMID: 21368720].
[89]
Zhao, G.; Yao-Yue, C.; Qin, G.W.; Guo, L.H. Luteolin from Purple Perilla mitigates ROS insult particularly in primary neurons. Neurobiol. Aging, 2012, 33(1), 176-186. [http://dx.doi.org/10.1016/ j.neurobiolaging.2010.02.013]. [PMID: 20382451].
[90]
Wruck, C.J.; Claussen, M.; Fuhrmann, G.; Römer, L.; Schulz, A.; Pufe, T.; Waetzig, V.; Peipp, M.; Herdegen, T.; Götz, M.E. Luteolin protects rat PC12 and C6 cells against MPP+ induced toxicity via an ERK dependent Keap1-Nrf2-ARE pathway. J. Neural Transm. Suppl., 2007, (72), 57-67. [PMID: 17982879].
[91]
Xu, B.; Li, X.X.; He, G.R.; Hu, J.J.; Mu, X.; Tian, S.; Du, G.H. Luteolin promotes long-term potentiation and improves cognitive functions in chronic cerebral hypoperfused rats. Eur. J. Pharmacol., 2010, 627(1-3), 99-105. [http://dx.doi.org/10.1016/ j.ejphar.2009.10.038]. [PMID: 19857483].
[92]
Moreno, L.C.G.E.I.; Puerta, E.; Suárez-Santiago, J.E.; Santos-Magalhães, N.S.; Ramirez, M.J.; Irache, J.M. Effect of the oral administration of nanoencapsulated quercetin on a mouse model of Alzheimer’s disease. Int. J. Pharm., 2017, 517(1-2), 50-57. [http://dx.doi.org/10.1016/j.ijpharm.2016.11.061]. [PMID: 27915007].
[93]
Lu, J.; Wu, D.M.; Zheng, Y.L.; Hu, B.; Zhang, Z.F.; Shan, Q.; Zheng, Z.H.; Liu, C.M.; Wang, Y.J. Quercetin activates AMP-activated protein kinase by reducing PP2C expression protecting old mouse brain against high cholesterol-induced neurotoxicity. J. Pathol., 2010, 222(2), 199-212. [http://dx.doi.org/10.1002/ path.2754]. [PMID: 20690163].
[94]
Jiménez-Aliaga, K.; Bermejo-Bescós, P.; Benedí, J.; Martín-Aragón, S. Quercetin and rutin exhibit antiamyloidogenic and fibril-disaggregating effects in vitro and potent antioxidant activity in APPswe cells. Life Sci., 2011, 89(25-26), 939-945. [http://dx.doi.org/10.1016/j.lfs.2011.09.023]. [PMID: 22008478].
[95]
Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Flavonols and flavones as BACE-1 inhibitors: Structure-activity relationship in cell-free, cell-based and in silico studies reveal novel pharmacophore features. Biochim. Biophys. Acta, 2008, 1780(5), 819-825. [http://dx.doi.org/10.1016/j.bbagen.2008.01.017]. [PMID: 18295609].
[96]
Bartolini, M.; Naldi, M.; Fiori, J.; Valle, F.; Biscarini, F.; Nicolau, D.V.; Andrisano, V. Kinetic characterization of amyloid-beta 1-42 aggregation with a multimethodological approach. Anal. Biochem., 2011, 414(2), 215-225. [http://dx.doi.org/10.1016/ j.ab.2011.03.020]. [PMID: 21435333].
[97]
Shimmyo, Y.; Kihara, T.; Akaike, A.; Niidome, T.; Sugimoto, H. Multifunction of myricetin on A beta: neuroprotection via a conformational change of A beta and reduction of A beta via the interference of secretases. J. Neurosci. Res., 2008, 86(2), 368-377. [http://dx.doi.org/10.1002/jnr.21476]. [PMID: 17722071].
[98]
Yang, S.; Liu, W.; Lu, S.; Tian, Y.Z.; Wang, W.Y.; Ling, T.J.; Liu, R.T. A novel multifunctional compound Camellikaempferoside B decreases aβ production, interferes with Aβ Aggregation, and Prohibits Aβ-Mediated Neurotoxicity and Neuroinflammation. ACS Chem. Neurosci., 2016, 7(4), 505-518. [http://dx.doi.org/10.1021/ acschemneuro.6b00091]. [PMID: 27015590].
[99]
Kosmeder, J.W., II; Pezzuto, J.M.; Pezzuto, J.M. Biological effects of resveratrol. Antioxid. Redox Signal., 2001, 3(6), 1041-1064. [http://dx.doi.org/10.1089/152308601317203567]. [PMID: 11813979].
[100]
Li, F.; Gong, Q.; Dong, H.; Shi, J. Resveratrol, A neuroprotective supplement for Alzheimer’s disease. Curr. Pharm. Des., 2012, 18(1), 27-33. [http://dx.doi.org/10.2174/138161212798919075]. [PMID: 22211686].
[101]
Ge, J.F.; Qiao, J.P.; Qi, C.C.; Wang, C.W.; Zhou, J.N. The binding of resveratrol to monomer and fibril amyloid beta. Neurochem. Int., 2012, 61(7), 1192-1201. [http://dx.doi.org/10.1016/ j.neuint.2012.08.012]. [PMID: 22981725].
[102]
Feng, Y.; Wang, X.P.; Yang, S.G.; Wang, Y.J.; Zhang, X.; Du, X.T.; Sun, X.X.; Zhao, M.; Huang, L.; Liu, R.T. Resveratrol inhibits beta-amyloid oligomeric cytotoxicity but does not prevent oligomer formation. Neurotoxicology, 2009, 30(6), 986-995. [http://dx.doi.org/10.1016/j.neuro.2009.08.013]. [PMID: 19744518].
[103]
Turner, R.S.; Thomas, R.G.; Craft, S.; van Dyck, C.H.; Mintzer, J.; Reynolds, B.A.; Brewer, J.B.; Rissman, R.A.; Raman, R.; Aisen, P.S. A randomized, double-blind, placebo-controlled trial of resveratrol for Alzheimer disease. Neurology, 2015, 85(16), 1383-1391. [http://dx.doi.org/10.1212/WNL.0000000000002035]. [PMID: 26362286].
[104]
Luo, L.; Huang, Y.M. Effect of resveratrol on the cognitive ability of Alzheimeros mice. Zhong Nan Da Xue Xue Bao Yi Xue Ban, 2006, 31(4), 566-569. [Effect of resveratrol on the cognitive ability of Alzheimeros mice]. [PMID: 16951520].
[105]
Kumar, A.; Naidu, P.S.; Seghal, N.; Padi, S.S. Neuroprotective effects of resveratrol against intracerebroventricular colchicine-induced cognitive impairment and oxidative stress in rats. Pharmacology, 2007, 79(1), 17-26. [http://dx.doi.org/10.1159/000097511]. [PMID: 17135773].
[106]
Wight, R.D.; Tull, C.A.; Deel, M.W.; Stroope, B.L.; Eubanks, A.G.; Chavis, J.A.; Drew, P.D.; Hensley, L.L. Resveratrol effects on astrocyte function: relevance to neurodegenerative diseases. Biochem. Biophys. Res. Commun., 2012, 426(1), 112-115. [http://dx.doi.org/10.1016/j.bbrc.2012.08.045]. [PMID: 22917537].
[107]
Karthick, C.; Periyasamy, S.; Jayachandran, K.S.; Anusuyadevi, M. Intrahippocampal administration of ibotenic acid induced cholinergic dysfunction via NR2A/NR2B expression: Implications of resveratrol against Alzheimer disease pathophysiology. Front. Mol. Neurosci., 2016, 9, 28. [http://dx.doi.org/10.3389/ fnmol.2016.00028]. [PMID: 27199654].
[108]
Moussa, C.; Hebron, M.; Huang, X.; Ahn, J.; Rissman, R.A.; Aisen, P.S.; Turner, R.S. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflammation, 2017, 14(1), 1. [http://dx.doi.org/10.1186/s12974-016-0779-0]. [PMID: 28086917].
[109]
Satake, M.; Murata, M.; Yasumoto, T. Gambierol: A new toxic polyether compound isolated from the marine dinoflagellate Gambierdiscus toxicus. J. Am. Chem. Soc., 1993, 115(1), 361-362. [http://dx.doi.org/10.1021/ja00054a061].
[110]
Alonso, E.; Fuwa, H.; Vale, C.; Suga, Y.; Goto, T.; Konno, Y.; Sasaki, M.; LaFerla, F.M.; Vieytes, M.R.; Giménez-Llort, L.; Botana, L.M. Design and synthesis of skeletal analogues of gambierol: attenuation of amyloid-β and tau pathology with voltage-gated potassium channel and N-methyl-D-aspartate receptor implications. J. Am. Chem. Soc., 2012, 134(17), 7467-7479. [http://dx.doi.org/ 10.1021/ja300565t]. [PMID: 22475455].
[111]
Birinyi-Strachan, L.C.; Gunning, S.J.; Lewis, R.J.; Nicholson, G.M. Block of voltage-gated potassium channels by Pacific ciguatoxin-1 contributes to increased neuronal excitability in rat sensory neurons. Toxicol. Appl. Pharmacol., 2005, 204(2), 175-186. [http://dx.doi.org/10.1016/j.taap.2004.08.020]. [PMID: 15808523].
[112]
Hidalgo, J.; Liberona, J.L.; Molgó, J.; Jaimovich, E. Pacific ciguatoxin-1b effect over Na+ and K+ currents, inositol 1,4,5-triphosphate content and intracellular Ca2+ signals in cultured rat myotubes. Br. J. Pharmacol., 2002, 137(7), 1055-1062. [http://dx.doi.org/10.1038/sj.bjp.0704980]. [PMID: 12429578].
[113]
Schlumberger, S.; Mattei, C.; Molgó, J.; Benoit, E. Dual action of a dinoflagellate-derived precursor of Pacific ciguatoxins (P-CTX-4B) on voltage-dependent K(+) and Na(+) channels of single myelinated axons. Toxicon, 2010, 56(5), 768-775. [http://dx.doi.org/ 10.1016/j.toxicon.2009.06.035]. [PMID: 19589350].
[114]
Oddo, S.; Caccamo, A.; Shepherd, J.D.; Murphy, M.P.; Golde, T.E.; Kayed, R.; Metherate, R.; Mattson, M.P.; Akbari, Y.; LaFerla, F.M. Triple-transgenic model of Alzheimer’s disease with plaques and tangles: intracellular Abeta and synaptic dysfunction. Neuron, 2003, 39(3), 409-421. [http://dx.doi.org/10.1016/S0896-6273(03)00434-3]. [PMID: 12895417].
[115]
Dräger, U.C. Retinoic acid signaling in the functioning brain. Sci. STKE, 2006, 2006(324), pe10. [PMID: 16507818].
[116]
Duester, G. Retinoic acid synthesis and signaling during early organogenesis. Cell, 2008, 134(6), 921-931. [http://dx.doi.org/ 10.1016/j.cell.2008.09.002]. [PMID: 18805086].
[117]
Niederreither, K.; Dollé, P. Retinoic acid in development: Towards an integrated view. Nat. Rev. Genet., 2008, 9(7), 541-553. [http://dx.doi.org/10.1038/nrg2340]. [PMID: 18542081].
[118]
Theodosiou, M.; Laudet, V.; Schubert, M. From carrot to clinic: An overview of the retinoic acid signaling pathway. Cell. Mol. Life Sci., 2010, 67(9), 1423-1445. [http://dx.doi.org/10.1007/s00018-010-0268-z]. [PMID: 20140749].
[119]
Beckman, M.; Iverfeldt, K. Increased gene expression of beta-amyloid precursor protein and its homologues APLP1 and APLP2 in human neuroblastoma cells in response to retinoic acid. Neurosci. Lett., 1997, 221(2-3), 73-76. [http://dx.doi.org/10.1016/S0304-3940(96)13292-4]. [PMID: 9121703].
[120]
Misner, D.L.; Jacobs, S.; Shimizu, Y.; de Urquiza, A.M.; Solomin, L.; Perlmann, T.; De Luca, L.M.; Stevens, C.F.; Evans, R.M. Vitamin A deprivation results in reversible loss of hippocampal long-term synaptic plasticity. Proc. Natl. Acad. Sci. USA, 2001, 98(20), 11714-11719. [http://dx.doi.org/10.1073/pnas.191369798]. [PMID: 11553775].
[121]
Ding, Y.; Qiao, A.; Wang, Z.; Goodwin, J.S.; Lee, E.S.; Block, M.L.; Allsbrook, M.; McDonald, M.P.; Fan, G.H. Retinoic acid attenuates beta-amyloid deposition and rescues memory deficits in an Alzheimer’s disease transgenic mouse model. J. Neurosci., 2008, 28(45), 11622-11634. [http://dx.doi.org/10.1523/ JNEUROSCI.3153-08.2008]. [PMID: 18987198].
[122]
Jarvis, C.I.; Goncalves, M.B.; Clarke, E.; Dogruel, M.; Kalindjian, S.B.; Thomas, S.A.; Maden, M.; Corcoran, J.P. Retinoic acid receptor-α signalling antagonizes both intracellular and extracellular amyloid-β production and prevents neuronal cell death caused by amyloid-β. Eur. J. Neurosci., 2010, 32(8), 1246-1255. [http://dx.doi.org/10.1111/j.1460-9568.2010.07426.x]. [PMID: 20950278].
[123]
Corcoran, J.P.; So, P.L.; Maden, M. Disruption of the retinoid signalling pathway causes a deposition of amyloid beta in the adult rat brain. Eur. J. Neurosci., 2004, 20(4), 896-902. [http://dx.doi.org/10.1111/j.1460-9568.2004.03563.x]. [PMID: 15305858].
[124]
Kapoor, A.; Wang, B.J.; Hsu, W.M.; Chang, M.Y.; Liang, S.M.; Liao, Y.F. Retinoic acid-elicited RARα/RXRα signaling attenuates Aβ production by directly inhibiting γ-secretase-mediated cleavage of amyloid precursor protein. ACS Chem. Neurosci., 2013, 4(7), 1093-1100. [http://dx.doi.org/10.1021/cn400039s]. [PMID: 23530929].
[125]
Ali, M.Y.; Jannat, S.; Jung, H.A.; Choi, R.J.; Roy, A.; Choi, J.S. Anti-Alzheimer’s disease potential of coumarins from Angelica decursiva and Artemisia capillaris and structure-activity analysis. Asian Pac. J. Trop. Med., 2016, 9(2), 103-111. [http://dx.doi.org/ 10.1016/j.apjtm.2016.01.014]. [PMID: 26919937].
[126]
Ali, M.Y.; Seong, S.H.; Reddy, M.R.; Seo, S.Y.; Choi, J.S.; Jung, H.A. Kinetics and Molecular Docking Studies of 6-Formyl Umbelliferone Isolated from Angelica decursiva as an Inhibitor of Cholinesterase and BACE1. Molecules, 2017, 22(10), E1604. [http://dx.doi.org/10.3390/molecules22101604]. [PMID: 28946641].
[127]
Weggen, S.; Eriksen, J.L.; Das, P.; Sagi, S.A.; Wang, R.; Pietrzik, C.U.; Findlay, K.A.; Smith, T.E.; Murphy, M.P.; Bulter, T.; Kang, D.E.; Marquez-Sterling, N.; Golde, T.E.; Koo, E.H. A subset of NSAIDs lower amyloidogenic Abeta42 independently of cyclooxygenase activity. Nature, 2001, 414(6860), 212-216. [http://dx.doi.org/10.1038/35102591]. [PMID: 11700559].
[128]
Haugabook, S.J.; Yager, D.M.; Eckman, E.A.; Golde, T.E.; Younkin, S.G.; Eckman, C.B. High throughput screens for the identification of compounds that alter the accumulation of the Alzheimer’s amyloid beta peptide (Abeta). J. Neurosci. Methods, 2001, 108(2), 171-179. [http://dx.doi.org/10.1016/S0165-0270(01)00388-0]. [PMID: 11478976].
[129]
Yager, D.; Watson, M.; Healy, B.; Eckman, E.A.; Eckman, C.B. Natural product extracts that reduce accumulation of the Alzheimer’s amyloid beta peptide: selective reduction in A beta42. J. Mol. Neurosci., 2002, 19(1-2), 129-133. [http://dx.doi.org/ 10.1007/s12031-002-0023-5]. [PMID: 12212770].
[130]
Findeis, M.A. The role of amyloid beta peptide 42 in Alzheimer’s disease. Pharmacol. Ther., 2007, 116(2), 266-286. [http://dx.doi.org/10.1016/j.pharmthera.2007.06.006]. [PMID: 17716740].
[131]
Findeis, M.A.; Schroeder, F.; McKee, T.D.; Yager, D.; Fraering, P.C.; Creaser, S.P.; Austin, W.F.; Clardy, J.; Wang, R.; Selkoe, D.; Eckman, C.B. Discovery of a novel pharmacological and structural class of gamma secretase modulators derived from the extract of Actaea racemosa. ACS Chem. Neurosci., 2012, 3(11), 941-951. [http://dx.doi.org/10.1021/cn3000857]. [PMID: 23205187].
[132]
Fuller, N.O.; Hubbs, J.L.; Austin, W.F.; Creaser, S.P.; McKee, T.D.; Loureiro, R.M.; Tate, B.; Xia, W.; Ives, J.L.; Findeis, M.A.; Bronk, B.S. Initial optimization of a new series of γ-secretase modulators derived from a triterpene glycoside. ACS Med. Chem. Lett., 2012, 3(11), 908-913. [http://dx.doi.org/10.1021/ml300256p]. [PMID: 24900406].
[133]
Hubbs, J.L.; Fuller, N.O.; Austin, W.F.; Shen, R.; Creaser, S.P.; McKee, T.D.; Loureiro, R.M.B.; Tate, B.; Xia, W.; Ives, J.; Bronk, B.S. Optimization of a natural product-based class of γ-secretase modulators. J. Med. Chem., 2012, 55(21), 9270-9282. [http://dx.doi.org/10.1021/jm300976b]. [PMID: 23030762].
[134]
Tate, B.; McKee, T.D.; Loureiro, R.M.; Dumin, J.A.; Xia, W.; Pojasek, K.; Austin, W.F.; Fuller, N.O.; Hubbs, J.L.; Shen, R.; Jonker, J.; Ives, J.; Bronk, B.S. Modulation of gamma-secretase for the treatment of Alzheimer’s disease. Int. J. Alzheimers Dis., 2012, 2012, 210756. [http://dx.doi.org/10.1155/2012/210756]. [PMID: 23320246].
[135]
Hubbs, J.L.; Fuller, N.O.; Austin, W.F.; Shen, R.; Ma, J.; Gong, Z.; Li, J.; McKee, T.D.; Loureiro, R.M.; Tate, B.; Dumin, J.A.; Ives, J.; Bronk, B.S. Minimization of drug-drug interaction risk and candidate selection in a natural product-based class of gamma-secretase modulators. Bioorg. Med. Chem. Lett., 2015, 25(7), 1621-1626. [http://dx.doi.org/10.1016/j.bmcl.2015.01.051]. [PMID: 25708617].
[136]
Austin, W.F.H.J.L.; Fuller, N.O.; Creaser, S.P.; McKee, T.D.; Loureiro, R.M.B.; Findeis, M.A.; Tate, B.; Ives, J.L.; Bronk, B.S. SAR investigations on a novel class of gamma-secretase modulators based on a unique scaffold. MedChemComm, 2013, 4, 569-574. [http://dx.doi.org/10.1039/c3md20357c].
[137]
Findeis, M.A.; Schroeder, F.C.; Creaser, S.P.; McKee, T.D.; Xia, W. Natural product and natural product-derived gamma secretase modulators from Actaea racemosa extracts. Medicines (Basel), 2015, 2(3), 127-140. [http://dx.doi.org/10.3390/medicines2030127]. [PMID: 28930205].
[138]
Malik, J.; Karan, M.; Vasisht, K. Nootropic, anxiolytic and CNS-depressant studies on different plant sources of shankhpushpi. Pharm. Biol., 2011, 49(12), 1234-1242. [http://dx.doi.org/10.3109/ 13880209.2011.584539]. [PMID: 21846173].
[139]
Sethiya, N.K.; Nahata, A.; Mishra, S.H.; Dixit, V.K. An update on Shankhpushpi, a cognition-boosting Ayurvedic medicine. J. Chin. Integr. Med., 2009, 7(11), 1001-1022. [http://dx.doi.org/ 10.3736/jcim20091101]. [PMID: 19912732].
[140]
Nahata, A.; Patil, U.K.; Dixit, V.K. Effect of Convulvulus pluricaulis Choisy. on learning behaviour and memory enhancement activity in rodents. Nat. Prod. Res., 2008, 22(16), 1472-1482. [http://dx.doi.org/10.1080/14786410802214199]. [PMID: 19023811].
[141]
Bihaqi, S.W.; Singh, A.P.; Tiwari, M. Supplementation of Convolvulus pluricaulis attenuates scopolamine-induced increased tau and amyloid precursor protein (AβPP) expression in rat brain. Indian J. Pharmacol., 2012, 44(5), 593-598. [http://dx.doi.org/ 10.4103/0253-7613.100383]. [PMID: 23112420].
[142]
Zhang, Q.; Zhao, J.J.; Xu, J.; Feng, F.; Qu, W. Medicinal uses, phytochemistry and pharmacology of the genus Uncaria. J. Ethnopharmacol., 2015, 173, 48-80. [http://dx.doi.org/ 10.1016/j.jep.2015.06.011]. [PMID: 26091967].
[143]
Lake, T.; Snow, A. Treatment of amyloidosis e.g. Alzheimer's in a mammal involves administration of a composition comprising two of the substances selected from uncaria tomentosa extract, Gingko biloba, green tea extract, grape seed extract and curcumin. 2009.
[144]
Cummings, J.; Lee, G.; Mortsdorf, T.; Ritter, A.; Zhong, K. Alzheimer’s disease drug development pipeline: 2017. Alzheimers Dement. (N. Y.), 2017, 3(3), 367-384. [http://dx.doi.org/ 10.1016/j.trci.2017.05.002]. [PMID: 29067343].
[145]
Ballatore, C.; Brunden, K.R.; Huryn, D.M.; Trojanowski, J.Q.; Lee, V.M.Y.; Smith, A.B. III Microtubule stabilizing agents as potential treatment for Alzheimer’s disease and related neurodegenerative tauopathies. J. Med. Chem., 2012, 55(21), 8979-8996. [http://dx.doi.org/10.1021/jm301079z]. [PMID: 23020671].
[146]
Beer, T.M.; Higano, C.S.; Saleh, M.; Dreicer, R.; Hudes, G.; Picus, J.; Rarick, M.; Fehrenbacher, L.; Hannah, A.L. Phase II study of KOS-862 in patients with metastatic androgen independent prostate cancer previously treated with docetaxel. Invest. New Drugs, 2007, 25(6), 565-570. [http://dx.doi.org/10.1007/s10637-007-9068-1]. [PMID: 17618407].
[147]
Brunden, K.R.; Gardner, N.M.; James, M.J.; Yao, Y.; Trojanowski, J.Q.; Lee, V.M.Y.; Paterson, I.; Ballatore, C.; Smith, A.B., III MT-stabilizer, dictyostatin, exhibits prolonged brain retention and activity: Potential therapeutic implications. ACS Med. Chem. Lett., 2013, 4(9), 886-889. [http://dx.doi.org/10.1021/ml400233e]. [PMID: 24900764].
[148]
Furukawa, H.; Singh, S.K.; Mancusso, R.; Gouaux, E. Subunit arrangement and function in NMDA receptors. Nature, 2005, 438(7065), 185-192. [http://dx.doi.org/10.1038/nature04089]. [PMID: 16281028].
[149]
Li, F.; Tsien, J.Z. Memory and the NMDA receptors. N. Engl. J. Med., 2009, 361(3), 302-303. [http://dx.doi.org/10.1056/ NEJMcibr0902052]. [PMID: 19605837].
[150]
Lesné, S.; Ali, C.; Gabriel, C.; Croci, N.; MacKenzie, E.T.; Glabe, C.G.; Plotkine, M.; Marchand-Verrecchia, C.; Vivien, D.; Buisson, A. NMDA receptor activation inhibits alpha-secretase and promotes neuronal amyloid-beta production. J. Neurosci., 2005, 25(41), 9367-9377. [http://dx.doi.org/10.1523/JNEUROSCI.0849-05.2005]. [PMID: 16221845].
[151]
Parameshwaran, K.; Dhanasekaran, M.; Suppiramaniam, V. Amyloid beta peptides and glutamatergic synaptic dysregulation. Exp. Neurol., 2008, 210(1), 7-13. [http://dx.doi.org/10.1016/ j.expneurol.2007.10.008]. [PMID: 18053990].
[152]
Kim, Y.S.; Woo, J.Y.; Han, C.K.; Chang, I.M. Safety analysis of Panax ginseng in randomized clinical trials: A Systematic Review. Medicines (Basel), 2015, 2(2), 106-126. [http://dx.doi.org/ 10.3390/medicines2020106]. [PMID: 28930204].
[153]
Chang, Y.; Huang, W.J.; Tien, L.T.; Wang, S.J. Ginsenosides Rg1 and Rb1 enhance glutamate release through activation of protein kinase A in rat cerebrocortical nerve terminals (synaptosomes). Eur. J. Pharmacol., 2008, 578(1), 28-36. [http://dx.doi.org/ 10.1016/j.ejphar.2007.09.023]. [PMID: 17949708].
[154]
Finger, A.; Kuhr, S.; Engelhardt, U.H. Chromatography of tea constituents. J. Chromatogr. A, 1992, 624(1-2), 293-315. [http://dx.doi.org/10.1016/0021-9673(92)85685-M]. [PMID: 1494009].
[155]
Kakuda, T.; Nozawa, A.; Sugimoto, A.; Niino, H. Inhibition by theanine of binding of [3H]AMPA, [3H]kainate, and [3H]MDL 105,519 to glutamate receptors. Biosci. Biotechnol. Biochem., 2002, 66(12), 2683-2686. [http://dx.doi.org/10.1271/bbb.66.2683]. [PMID: 12596867].
[156]
Di, X.; Yan, J.; Zhao, Y.; Zhang, J.; Shi, Z.; Chang, Y.; Zhao, B. L-theanine protects the APP (Swedish mutation) transgenic SH-SY5Y cell against glutamate-induced excitotoxicity via inhibition of the NMDA receptor pathway. Neuroscience, 2010, 168(3), 778-786. [http://dx.doi.org/10.1016/j.neuroscience.2010.04.019]. [PMID: 20416364].
[157]
Hynd, M.R.; Scott, H.L.; Dodd, P.R. Glutamate-mediated excitotoxicity and neurodegeneration in Alzheimer’s disease. Neurochem. Int., 2004, 45(5), 583-595. [http://dx.doi.org/10.1016/ j.neuint.2004.03.007]. [PMID: 15234100].
[158]
Kim, T.I.; Lee, Y.K.; Park, S.G.; Choi, I.S.; Ban, J.O.; Park, H.K.; Nam, S.Y.; Yun, Y.W.; Han, S.B.; Oh, K.W.; Hong, J.T. l-Theanine, an amino acid in green tea, attenuates beta-amyloid-induced cognitive dysfunction and neurotoxicity: reduction in oxidative damage and inactivation of ERK/p38 kinase and NF-kappaB pathways. Free Radic. Biol. Med., 2009, 47(11), 1601-1610. [http://dx.doi.org/10.1016/j.freeradbiomed.2009.09.008]. [PMID: 19766184].
[159]
Mattson, M.P. Cellular actions of beta-amyloid precursor protein and its soluble and fibrillogenic derivatives. Physiol. Rev., 1997, 77(4), 1081-1132. [http://dx.doi.org/10.1152/ physrev.1997.77.4.1081]. [PMID: 9354812].
[160]
Marcus, D.L.; Thomas, C.; Rodriguez, C.; Simberkoff, K.; Tsai, J.S.; Strafaci, J.A.; Freedman, M.L. Increased peroxidation and reduced antioxidant enzyme activity in Alzheimer’s disease. Exp. Neurol., 1998, 150(1), 40-44. [http://dx.doi.org/10.1006/ exnr.1997.6750]. [PMID: 9514828].
[161]
Ansari, M.A.; Scheff, S.W. Oxidative stress in the progression of Alzheimer disease in the frontal cortex. J. Neuropathol. Exp. Neurol., 2010, 69(2), 155-167. [http://dx.doi.org/10.1097/ NEN.0b013e3181cb5af4]. [PMID: 20084018].
[162]
Padurariu, M.; Ciobica, A.; Hritcu, L.; Stoica, B.; Bild, W.; Stefanescu, C. Changes of some oxidative stress markers in the serum of patients with mild cognitive impairment and Alzheimer’s disease. Neurosci. Lett., 2010, 469(1), 6-10. [http://dx.doi.org/ 10.1016/j.neulet.2009.11.033]. [PMID: 19914330].
[163]
Omar, R.A.; Chyan, Y.J.; Andorn, A.C.; Poeggeler, B.; Robakis, N.K.; Pappolla, M.A. Increased Expression but Reduced Activity of Antioxidant Enzymes in Alzheimer’s Disease. J. Alzheimers Dis., 1999, 1(3), 139-145. [http://dx.doi.org/10.3233/JAD-1999-1301]. [PMID: 12213999].
[164]
Matsuoka, Y.; Picciano, M.; La Francois, J.; Duff, K. Fibrillar beta-amyloid evokes oxidative damage in a transgenic mouse model of Alzheimer’s disease. Neuroscience, 2001, 104(3), 609-613. [http://dx.doi.org/10.1016/S0306-4522(01)00115-4]. [PMID: 11440793].
[165]
De Felice, F.G.; Velasco, P.T.; Lambert, M.P.; Viola, K.; Fernandez, S.J.; Ferreira, S.T.; Klein, W.L. Abeta oligomers induce neuronal oxidative stress through an N-methyl-D-aspartate receptor-dependent mechanism that is blocked by the Alzheimer drug memantine. J. Biol. Chem., 2007, 282(15), 11590-11601. [http://dx.doi.org/10.1074/jbc.M607483200]. [PMID: 17308309].
[166]
Li, F.; Calingasan, N.Y.; Yu, F.; Mauck, W.M.; Toidze, M.; Almeida, C.G.; Takahashi, R.H.; Carlson, G.A.; Flint Beal, M.; Lin, M.T.; Gouras, G.K. Increased plaque burden in brains of APP mutant MnSOD heterozygous knockout mice. J. Neurochem., 2004, 89(5), 1308-1312. [http://dx.doi.org/10.1111/j.1471-4159.2004.02455.x]. [PMID: 15147524].
[167]
Nishida, Y.; Yokota, T.; Takahashi, T.; Uchihara, T.; Jishage, K.; Mizusawa, H. Deletion of vitamin E enhances phenotype of Alzheimer disease model mouse. Biochem. Biophys. Res. Commun., 2006, 350(3), 530-536. [http://dx.doi.org/10.1016/ j.bbrc.2006.09.083]. [PMID: 17026966].
[168]
Dal Prà, I.; Chiarini, A.; Gui, L.; Chakravarthy, B.; Pacchiana, R.; Gardenal, E.; Whitfield, J.F.; Armato, U. Do astrocytes collaborate with neurons in spreading the “infectious” aβ and Tau drivers of Alzheimer’s disease? Neuroscientist, 2015, 21(1), 9-29. [http://dx.doi.org/10.1177/1073858414529828]. [PMID: 24740577].
[169]
Domanski, D.; Zegrocka-Stendel, O.; Perzanowska, A.; Dutkiewicz, M.; Kowalewska, M.; Grabowska, I.; Maciejko, D.; Fogtman, A.; Dadlez, M.; Koziak, K. Molecular Mechanism for Cellular Response to β-Escin and its therapeutic implications. PLoS One, 2016, 11(10), e0164365. [http://dx.doi.org/10.1371/ journal.pone.0164365]. [PMID: 27727329].
[170]
Zhao, J.; O’Connor, T.; Vassar, R. The contribution of activated astrocytes to Aβ production: implications for Alzheimer’s disease pathogenesis. J. Neuroinflammation, 2011, 8, 150. [http://dx.doi.org/10.1186/1742-2094-8-150]. [PMID: 22047170].
[171]
Koziak, K.; Kowalewska, M.; Maciejko, D.; Zegrocka-Stendel, O.; Domanski, D.; Perzanowska, A. Agent used to treat and/or prevent Alzheimer's disease, comprises horse chestnut extract, escins, or their salts and derivatives, 2015, EP3078381A1.
[172]
de Oliveira, M.R. The effects of ellagic acid upon brain cells: A mechanistic view and future directions. Neurochem. Res., 2016, 41(6), 1219-1228. [http://dx.doi.org/10.1007/s11064-016-1853-9]. [PMID: 26846140].
[173]
Tenchov, B.; Abarova, S.; Koynova, R.; Traikov, L.; Dragomanova, S.; Tancheva, L. A new approach for investigating neurodegenerative disorders in mice based on DSC. J. Therm. Anal. Calorim., 2017, 127(1), 483-486. [http://dx.doi.org/10.1007/s10973-016-5749-3].
[174]
Abarova, S.; Koynova, R.; Tancheva, L.; Tenchov, B. A novel DSC approach for evaluating protectant drugs efficacy against dementia. Biochim. Biophys. Acta Mol. Basis. Dis., 2017, 1863(11), 2934-2941. [http://dx.doi.org/10.1016/j.bbadis.2017.07.033]. [PMID: 28778589].
[175]
Ahmed, T.; Setzer, W.N.; Nabavi, S.F.; Orhan, I.E.; Braidy, N.; Sobarzo-Sanchez, E.; Nabavi, S.M. Insights into effects of ellagic acid on the nervous system: A mini review. Curr. Pharm. Des., 2016, 22(10), 1350-1360. [http://dx.doi.org/10.2174/ 1381612822666160125114503]. [PMID: 26806345].
[176]
Ahmad, N.; Feyes, D.K.; Nieminen, A.L.; Agarwal, R.; Mukhtar, H. Green tea constituent epigallocatechin-3-gallate and induction of apoptosis and cell cycle arrest in human carcinoma cells. J. Natl. Cancer Inst., 1997, 89(24), 1881-1886. [http://dx.doi.org/10.1093/ jnci/89.24.1881]. [PMID: 9414176].
[177]
Mandel, S.A.; Amit, T.; Kalfon, L.; Reznichenko, L.; Weinreb, O.; Youdim, M.B. Cell signaling pathways and iron chelation in the neurorestorative activity of green tea polyphenols: special reference to epigallocatechin gallate (EGCG). J. Alzheimers Dis., 2008, 15(2), 211-222. [http://dx.doi.org/10.3233/JAD-2008-15207]. [PMID: 18953110].
[178]
Biasibetti, R.; Tramontina, A.C.; Costa, A.P.; Dutra, M.F.; Quincozes-Santos, A.; Nardin, P.; Bernardi, C.L.; Wartchow, K.M.; Lunardi, P.S.; Gonçalves, C.A. Green tea (-)epigallocatechin-3-gallate reverses oxidative stress and reduces acetylcholinesterase activity in a streptozotocin-induced model of dementia. Behav. Brain Res., 2013, 236(1), 186-193. [http://dx.doi.org/10.1016/ j.bbr.2012.08.039]. [PMID: 22964138].
[179]
Rezai-Zadeh, K.; Shytle, D.; Sun, N.; Mori, T.; Hou, H.; Jeanniton, D.; Ehrhart, J.; Townsend, K.; Zeng, J.; Morgan, D.; Hardy, J.; Town, T.; Tan, J. Green tea epigallocatechin-3-gallate (EGCG) modulates amyloid precursor protein cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J. Neurosci., 2005, 25(38), 8807-8814. [http://dx.doi.org/10.1523/JNEUROSCI.1521-05.2005]. [PMID: 16177050].
[180]
Lee, J.W.; Lee, Y.K.; Ban, J.O.; Ha, T.Y.; Yun, Y.P.; Han, S.B.; Oh, K.W.; Hong, J.T. Green tea (-)-epigallocatechin-3-gallate inhibits beta-amyloid-induced cognitive dysfunction through modification of secretase activity via inhibition of ERK and NF-kappaB pathways in mice. J. Nutr., 2009, 139(10), 1987-1993. [http://dx.doi.org/10.3945/jn.109.109785]. [PMID: 19656855].
[181]
Lee, Y.J.; Choi, D.Y.; Yun, Y.P.; Han, S.B.; Oh, K.W.; Hong, J.T. Epigallocatechin-3-gallate prevents systemic inflammation-induced memory deficiency and amyloidogenesis via its anti-neuroinflammatory properties. J. Nutr. Biochem., 2013, 24(1), 298-310. [http://dx.doi.org/10.1016/j.jnutbio.2012.06.011]. [PMID: 22959056].
[182]
Li, R.; Shen, Y. Estrogen and brain: synthesis, function and diseases. Front. Biosci., 2005, 10, 257-267. [http://dx.doi.org/ 10.2741/1525]. [PMID: 15574366].
[183]
Behl, C.; Skutella, T.; Lezoualc’h, F.; Post, A.; Widmann, M.; Newton, C.J.; Holsboer, F. Neuroprotection against oxidative stress by estrogens: structure-activity relationship. Mol. Pharmacol., 1997, 51(4), 535-541. [http://dx.doi.org/10.1124/mol.51.4.535]. [PMID: 9106616].
[184]
Zec, R.F.; Trivedi, M.A. The effects of estrogen replacement therapy on neuropsychological functioning in postmenopausal women with and without dementia: a critical and theoretical review. Neuropsychol. Rev., 2002, 12(2), 65-109. [http://dx.doi.org/10.1023/ A:1016880127635]. [PMID: 12371603].
[185]
Guo, Q.; Rimbach, G.; Moini, H.; Weber, S.; Packer, L. ESR and cell culture studies on free radical-scavenging and antioxidant activities of isoflavonoids. Toxicology, 2002, 179(1-2), 171-180. [http://dx.doi.org/10.1016/S0300-483X(02)00241-X]. [PMID: 12204553].
[186]
Chan, W.H.; Yu, J.S. Inhibition of UV irradiation-induced oxidative stress and apoptotic biochemical changes in human epidermal carcinoma A431 cells by genistein. J. Cell. Biochem., 2000, 78(1), 73-84. [http://dx.doi.org/10.1002/(SICI)1097-4644(20000701)78:1 <73:AID-JCB7>3.0.CO;2-P]. [PMID: 10797567].
[187]
Kim, H.; Xia, H.; Li, L.; Gewin, J. Attenuation of neurodegeneration-relevant modifications of brain proteins by dietary soy. Biofactors, 2000, 12(1-4), 243-250. [http://dx.doi.org/ 10.1002/ biof.5520120137]. [PMID: 11216492].
[188]
Vallés, S.L.; Borrás, C.; Gambini, J.; Furriol, J.; Ortega, A.; Sastre, J.; Pallardó, F.V.; Viña, J. Oestradiol or genistein rescues neurons from amyloid beta-induced cell death by inhibiting activation of p38. Aging Cell, 2008, 7(1), 112-118. [http://dx.doi.org/ 10.1111/j.1474-9726.2007.00356.x]. [PMID: 18031570].
[189]
Gutierrez-Zepeda, A.; Santell, R.; Wu, Z.; Brown, M.; Wu, Y.; Khan, I.; Link, C.D.; Zhao, B.; Luo, Y. Soy isoflavone glycitein protects against beta amyloid-induced toxicity and oxidative stress in transgenic Caenorhabditis elegans. BMC Neurosci., 2005, 6, 54. [http://dx.doi.org/10.1186/1471-2202-6-54]. [PMID: 16122394].
[190]
Zhao, L.; Brinton, R.D. WHI and WHIMS follow-up and human studies of soy isoflavones on cognition. Expert Rev. Neurother., 2007, 7(11), 1549-1564. [http://dx.doi.org/10.1586/ 14737175.7.11.1549]. [PMID: 17997703].
[191]
Liu, Q.; Zhao, B. Nicotine attenuates beta-amyloid peptide-induced neurotoxicity, free radical and calcium accumulation in hippocampal neuronal cultures. Br. J. Pharmacol., 2004, 141(4), 746-754. [http://dx.doi.org/10.1038/sj.bjp.0705653]. [PMID: 14757701].
[192]
Xie, Y.X.; Bezard, E.; Zhao, B.L. Investigating the receptor-independent neuroprotective mechanisms of nicotine in mitochondria. J. Biol. Chem., 2005, 280(37), 32405-32412. [http://dx.doi.org/10.1074/jbc.M504664200]. [PMID: 15985439].
[193]
Lovell, M.A.; Robertson, J.D.; Teesdale, W.J.; Campbell, J.L.; Markesbery, W.R. Copper, iron and zinc in Alzheimer’s disease senile plaques. J. Neurol. Sci., 1998, 158(1), 47-52. [http://dx.doi.org/10.1016/S0022-510X(98)00092-6]. [PMID: 9667777].
[194]
Zhang, J.; Liu, Q.; Chen, Q.; Liu, N.Q.; Li, F.L.; Lu, Z.B.; Qin, C.; Zhu, H.; Huang, Y.Y.; He, W.; Zhao, B.L. Nicotine attenuates beta-amyloid-induced neurotoxicity by regulating metal homeostasis. FASEB J., 2006, 20(8), 1212-1214. [http://dx.doi.org/ 10.1096/fj.05-5214fje]. [PMID: 16627626].
[195]
Srivareerat, M.; Tran, T.T.; Salim, S.; Aleisa, A.M.; Alkadhi, K.A. Chronic nicotine restores normal Aβ levels and prevents short-term memory and E-LTP impairment in Aβ rat model of Alzheimer’s disease. Neurobiol. Aging, 2011, 32(5), 834-844. [http://dx.doi.org/ 10.1016/j.neurobiolaging.2009.04.015]. [PMID: 19464074].
[196]
Petersen, M.; Simmonds, M.S. Rosmarinic acid. Phytochemistry, 2003, 62(2), 121-125. [http://dx.doi.org/10.1016/S0031-9422(02)00513-7]. [PMID: 12482446].
[197]
Shaerzadeh, F.; Ahmadiani, A.; Esmaeili, M.A.; Ansari, N.; Asadi, S.; Tusi, S.K.; Sonboli, A.; Ghahremanzamaneh, M.; Khodagholi, F. Antioxidant and antiglycating activities of Salvia sahendica and its protective effect against oxidative stress in neuron-like PC12 cells. J. Nat. Med., 2011, 65(3-4), 455-465. [http://dx.doi.org/ 10.1007/s11418-011-0519-9]. [PMID: 21424254].
[198]
Bulgakov, V.P.; Inyushkina, Y.V.; Fedoreyev, S.A. Rosmarinic acid and its derivatives: biotechnology and applications. Crit. Rev. Biotechnol., 2012, 32(3), 203-217. [http://dx.doi.org/10.3109/ 07388551.2011.596804]. [PMID: 21838541].
[199]
Alkam, T.; Nitta, A.; Mizoguchi, H.; Itoh, A.; Nabeshima, T. A natural scavenger of peroxynitrites, rosmarinic acid, protects against impairment of memory induced by Abeta(25-35). Behav. Brain Res., 2007, 180(2), 139-145. [http://dx.doi.org/ 10.1016/j.bbr.2007.03.001]. [PMID: 17420060].
[200]
Iuvone, T.; De Filippis, D.; Esposito, G.; D’Amico, A.; Izzo, A.A. The spice sage and its active ingredient rosmarinic acid protect PC12 cells from amyloid-beta peptide-induced neurotoxicity. J. Pharmacol. Exp. Ther., 2006, 317(3), 1143-1149. [http://dx.doi.org/10.1124/jpet.105.099317]. [PMID: 16495207].
[201]
Kantar Gok, D.; Ozturk, N.; Er, H.; Aslan, M.; Demir, N.; Derin, N.; Agar, A.; Yargicoglu, P. Effects of rosmarinic acid on cognitive and biochemical alterations in ovariectomized rats treated with D-galactose. Folia Histochem. Cytobiol., 2015, 53(4), 283-293. [http://dx.doi.org/10.5603/fhc.a2015.0034]. [PMID: 26714446].
[202]
Horrocks, L.A.; Farooqui, A.A. Docosahexaenoic acid in the diet: its importance in maintenance and restoration of neural membrane function. Prostaglandins Leukot. Essent. Fatty Acids, 2004, 70(4), 361-372. [http://dx.doi.org/10.1016/j.plefa.2003.12.011]. [PMID: 15041028].
[203]
Morris, M.C.; Evans, D.A.; Bienias, J.L.; Tangney, C.C.; Bennett, D.A.; Wilson, R.S.; Aggarwal, N.; Schneider, J. Consumption of fish and n-3 fatty acids and risk of incident Alzheimer disease. Arch. Neurol., 2003, 60(7), 940-946. [http://dx.doi.org/ 10.1001/archneur.60.7.940]. [PMID: 12873849].
[204]
Schaefer, E.J.; Bongard, V.; Beiser, A.S.; Lamon-Fava, S.; Robins, S.J.; Au, R.; Tucker, K.L.; Kyle, D.J.; Wilson, P.W.; Wolf, P.A. Plasma phosphatidylcholine docosahexaenoic acid content and risk of dementia and Alzheimer disease: the Framingham Heart Study. Arch. Neurol., 2006, 63(11), 1545-1550. [http://dx.doi.org/10.1001/archneur.63.11.1545]. [PMID: 17101822].
[205]
Jicha, G.A.; Markesbery, W.R. Omega-3 fatty acids: potential role in the management of early Alzheimer’s disease. Clin. Interv. Aging, 2010, 5, 45-61. [http://dx.doi.org/10.2147/CIA.S5231]. [PMID: 20396634].
[206]
Lukiw, W.J.; Bazan, N.G. Docosahexaenoic acid and the aging brain. J. Nutr., 2008, 138(12), 2510-2514. [http://dx.doi.org/ 10.3945/jn.108.096016]. [PMID: 19022980].
[207]
Cole, G.M.; Frautschy, S.A. Docosahexaenoic acid protects from amyloid and dendritic pathology in an Alzheimer’s disease mouse model. Nutr. Health, 2006, 18(3), 249-259. [http://dx.doi.org/ 10.1177/026010600601800307]. [PMID: 17180870].
[208]
Hossain, M.S.; Hashimoto, M.; Gamoh, S.; Masumura, S. Antioxidative effects of docosahexaenoic acid in the cerebrum versus cerebellum and brainstem of aged hypercholesterolemic rats. J. Neurochem., 1999, 72(3), 1133-1138. [http://dx.doi.org/ 10.1046/j.1471-4159.1999.0721133.x]. [PMID: 10037485].
[209]
Komatsu, W.; Ishihara, K.; Murata, M.; Saito, H.; Shinohara, K. Docosahexaenoic acid suppresses nitric oxide production and inducible nitric oxide synthase expression in interferon-gamma plus lipopolysaccharide-stimulated murine macrophages by inhibiting the oxidative stress. Free Radic. Biol. Med., 2003, 34(8), 1006-1016. [http://dx.doi.org/10.1016/S0891-5849(03)00027-3]. [PMID: 12684085].
[210]
Winter, J.C. The effects of an extract of Ginkgo biloba, EGb 761, on cognitive behavior and longevity in the rat. Physiol. Behav., 1998, 63(3), 425-433. [http://dx.doi.org/10.1016/S0031-9384(97)00464-2]. [PMID: 9469738].
[211]
Ni, Y.; Zhao, B.; Hou, J.; Xin, W. Preventive effect of Ginkgo biloba extract on apoptosis in rat cerebellar neuronal cells induced by hydroxyl radicals. Neurosci. Lett., 1996, 214(2-3), 115-118. [http://dx.doi.org/10.1016/0304-3940(96)12897-4]. [PMID: 8878097].
[212]
Wei, T.; Ni, Y.; Hou, J.; Chen, C.; Zhao, B.; Xin, W. Hydrogen peroxide-induced oxidative damage and apoptosis in cerebellar granule cells: protection by Ginkgo biloba extract. Pharmacol. Res., 2000, 41(4), 427-433. [http://dx.doi.org/ 10.1006/phrs.1999.0604]. [PMID: 10704267].
[213]
Chen, C.; Wei, T.; Gao, Z.; Zhao, B.; Hou, J.; Xu, H.; Xin, W.; Packer, L. Different effects of the constituents of EGb761 on apoptosis in rat cerebellar granule cells induced by hydroxyl radicals. Biochem. Mol. Biol. Int., 1999, 47(3), 397-405. [PMID: 10204076].
[214]
Bastianetto, S.; Zheng, W.H.; Quirion, R. The Ginkgo biloba extract (EGb 761) protects and rescues hippocampal cells against nitric oxide-induced toxicity: involvement of its flavonoid constituents and protein kinase C. J. Neurochem., 2000, 74(6), 2268-2277. [http://dx.doi.org/10.1046/j.1471-4159.2000.0742268.x]. [PMID: 10820186].
[215]
Bastianetto, S.; Ramassamy, C.; Doré, S.; Christen, Y.; Poirier, J.; Quirion, R. The Ginkgo biloba extract (EGb 761) protects hippocampal neurons against cell death induced by beta-amyloid. Eur. J. Neurosci., 2000, 12(6), 1882-1890. [http://dx.doi.org/10.1046/ j.1460-9568.2000.00069.x]. [PMID: 10886329].
[216]
Wu, Y.; Wu, Z.; Butko, P.; Christen, Y.; Lambert, M.P.; Klein, W.L.; Link, C.D.; Luo, Y. Amyloid-beta-induced pathological behaviors are suppressed by Ginkgo biloba extract EGb 761 and ginkgolides in transgenic Caenorhabditis elegans. J. Neurosci., 2006, 26(50), 13102-13113. [http://dx.doi.org/10.1523/ JNEUROSCI.3448-06.2006]. [PMID: 17167099].
[217]
Stackman, R.W.; Eckenstein, F.; Frei, B.; Kulhanek, D.; Nowlin, J.; Quinn, J.F. Prevention of age-related spatial memory deficits in a transgenic mouse model of Alzheimer’s disease by chronic Ginkgo biloba treatment. Exp. Neurol., 2003, 184(1), 510-520. [http://dx.doi.org/10.1016/S0014-4886(03)00399-6]. [PMID: 14637120].
[218]
Quinn, J.F.; Bussiere, J.R.; Hammond, R.S.; Montine, T.J.; Henson, E.; Jones, R.E.; Stackman, R.W. Jr Chronic dietary alpha-lipoic acid reduces deficits in hippocampal memory of aged Tg2576 mice. Neurobiol. Aging, 2007, 28(2), 213-225. [http://dx.doi.org/10.1016/j.neurobiolaging.2005.12.014]. [PMID: 16448723].
[219]
Oken, B.S.; Storzbach, D.M.; Kaye, J.A. The efficacy of Ginkgo biloba on cognitive function in Alzheimer disease. Arch. Neurol., 1998, 55(11), 1409-1415. [http://dx.doi.org/10.1001/ archneur.55.11.1409]. [PMID: 9823823].
[220]
Le Bars, P.L.; Kieser, M.; Itil, K.Z. A 26-week analysis of a double-blind, placebo-controlled trial of the Ginkgo biloba extract EGb 761 in dementia. Dement. Geriatr. Cogn. Disord., 2000, 11(4), 230-237. [http://dx.doi.org/10.1159/000017242]. [PMID: 10867450].
[221]
Kanowski, S.; Hoerr, R. Ginkgo biloba extract EGb 761 in dementia: intent-to-treat analyses of a 24-week, multi-center, double-blind, placebo-controlled, randomized trial. Pharmacopsychiatry, 2003, 36(6), 297-303. [http://dx.doi.org/10.1055/s-2003-45117]. [PMID: 14663654].
[222]
Snitz, B.E.; O’Meara, E.S.; Carlson, M.C.; Arnold, A.M.; Ives, D.G.; Rapp, S.R.; Saxton, J.; Lopez, O.L.; Dunn, L.O.; Sink, K.M.; DeKosky, S.T. Ginkgo biloba for preventing cognitive decline in older adults: a randomized trial. JAMA, 2009, 302(24), 2663-2670. [http://dx.doi.org/10.1001/jama.2009.1913]. [PMID: 20040554].
[223]
DeKosky, S.T.; Williamson, J.D.; Fitzpatrick, A.L.; Kronmal, R.A.; Ives, D.G.; Saxton, J.A.; Lopez, O.L.; Burke, G.; Carlson, M.C.; Fried, L.P.; Kuller, L.H.; Robbins, J.A.; Tracy, R.P.; Woolard, N.F.; Dunn, L.; Snitz, B.E.; Nahin, R.L.; Furberg, C.D. Ginkgo biloba for prevention of dementia: a randomized controlled trial. JAMA, 2008, 300(19), 2253-2262. [http://dx.doi.org/ 10.1001/jama.2008.683]. [PMID: 19017911].
[224]
Russo, A.; Borrelli, F. Bacopa monniera, a reputed nootropic plant: an overview. Phytomedicine, 2005, 12(4), 305-317. [http://dx.doi.org/10.1016/j.phymed.2003.12.008]. [PMID: 15898709].
[225]
Shinomol, G.K. Muralidhara; Bharath, M.M. Exploring the Role of “Brahmi” (Bacopa monnieri and Centella asiatica) in Brain Function and Therapy. Recent Pat. Endocr. Metab. Immune Drug Discov., 2011, 5(1), 33-49. [http://dx.doi.org/ 10.2174/187221411794351833]. [PMID: 22074576].
[226]
Dhanasekaran, M.; Tharakan, B.; Holcomb, L.A.; Hitt, A.R.; Young, K.A.; Manyam, B.V. Neuroprotective mechanisms of ayurvedic antidementia botanical Bacopa monniera. Phytother. Res., 2007, 21(10), 965-969. [http://dx.doi.org/10.1002/ptr.2195]. [PMID: 17604373].
[227]
Uabundit, N.; Wattanathorn, J.; Mucimapura, S.; Ingkaninan, K. Cognitive enhancement and neuroprotective effects of Bacopa monnieri in Alzheimer’s disease model. J. Ethnopharmacol., 2010, 127(1), 26-31. [http://dx.doi.org/10.1016/j.jep.2009.09.056]. [PMID: 19808086].
[228]
Bhattacharya, S.K.; Bhattacharya, A.; Kumar, A.; Ghosal, S. Antioxidant activity of Bacopa monniera in rat frontal cortex, striatum and hippocampus. Phytother. Res., 2000, 14(3), 174-179. [http://dx.doi.org/10.1002/(SICI)1099-1573(200005) [PMID: 10815010]
[229]
Limpeanchob, N.; Jaipan, S.; Rattanakaruna, S.; Phrompittayarat, W.; Ingkaninan, K. Neuroprotective effect of Bacopa monnieri on beta-amyloid-induced cell death in primary cortical culture. J. Ethnopharmacol., 2008, 120(1), 112-117. [http://dx.doi.org/ 10.1016/j.jep.2008.07.039]. [PMID: 18755259].
[230]
Sadhu, A.; Upadhyay, P.; Agrawal, A.; Ilango, K.; Karmakar, D.; Singh, G.P.; Dubey, G.P. Management of cognitive determinants in senile dementia of Alzheimer’s type: therapeutic potential of a novel polyherbal drug product. Clin. Drug Investig., 2014, 34(12), 857-869. [http://dx.doi.org/10.1007/s40261-014-0235-9]. [PMID: 25316430].
[231]
Gohil, K.J.; Patel, J.A.; Gajjar, A.K. Pharmacological Review on Centella asiatica: A Potential Herbal Cure-all. Indian J. Pharm. Sci., 2010, 72(5), 546-556. [http://dx.doi.org/10.4103/0250-474X.78519]. [PMID: 21694984].
[232]
Veerendra Kumar, M.H.; Gupta, Y.K. Effect of different extracts of Centella asiatica on cognition and markers of oxidative stress in rats. J. Ethnopharmacol., 2002, 79(2), 253-260. [http://dx.doi.org/ 10.1016/S0378-8741(01)00394-4]. [PMID: 11801389].
[233]
Brinkhaus, B.; Lindner, M.; Schuppan, D.; Hahn, E.G. Chemical, pharmacological and clinical profile of the East Asian medical plant Centella asiatica. Phytomedicine, 2000, 7(5), 427-448. [http://dx.doi.org/10.1016/S0944-7113(00)80065-3]. [PMID: 11081995].
[234]
Veerendra Kumar, M.H.; Gupta, Y.K. Effect of Centella asiatica on cognition and oxidative stress in an intracerebroventricular streptozotocin model of Alzheimer’s disease in rats. Clin. Exp. Pharmacol. Physiol., 2003, 30(5-6), 336-342. [http://dx.doi.org/10.1046/ j.1440-1681.2003.03842.x]. [PMID: 12859423].
[235]
Sharma, J.; Sharma, R. Radioprotection of Swiss albino mouse by Centella asiatica extract. Phytother. Res., 2002, 16(8), 785-786. [http://dx.doi.org/10.1002/ptr.1069]. [PMID: 12458490].
[236]
Huang, S.S.; Chiu, C.S.; Chen, H.J.; Hou, W.C.; Sheu, M.J.; Lin, Y.C.; Shie, P.H.; Huang, G.J. Antinociceptive activities and the mechanisms of anti-inflammation of asiatic Acid in mice. Evid. Based Complement. Alternat. Med., 2011, 2011, 895857. [http://dx.doi.org/10.1155/2011/895857]. [PMID: 21584194].
[237]
Allegra, C. [Comparative capillaroscopic study of certain bioflavonoids and total triterpenic fractions of Centella asiatica in venous insufficiency]. Clin. Ter., 1984, 110(6), 555-559. [Comparative capillaroscopic study of certain bioflavonoids and total triterpenic fractions of Centella asiatica in venous insufficiency]. [PMID: 6238770]
[238]
Chen, C.L.; Tsai, W.H.; Chen, C.J.; Pan, T.M. Centella asiatica extract protects against amyloid β1-40-induced neurotoxicity in neuronal cells by activating the antioxidative defence system. J. Tradit. Complement. Med., 2015, 6(4), 362-369. [http://dx.doi.org/ 10.1016/j.jtcme.2015.07.002]. [PMID: 27774420].
[239]
Borek, C. Antioxidant health effects of aged garlic extract. J. Nutr., 2001, 131(3s), 1010S-1015S. [http://dx.doi.org/ 10.1093/jn/ 131.3.1010S]. [PMID: 11238807].
[240]
Borek, C. Garlic reduces dementia and heart-disease risk. J. Nutr., 2006, 136(3)(Suppl.), 810S-812S. [http://dx.doi.org/ 10.1093/ jn/136.3.810S]. [PMID: 16484570].
[241]
Qu, Z.; Mossine, V.V.; Cui, J.; Sun, G.Y.; Gu, Z. Protective Effects of AGE and Its Components on Neuroinflammation and Neurodegeneration. Neuromolecular Med., 2016, 18(3), 474-482. [http://dx.doi.org/10.1007/s12017-016-8410-1]. [PMID: 27263111].
[242]
Nillert, N.; Pannangrong, W.; Welbat, J.U.; Chaijaroonkhanarak, W.; Sripanidkulchai, K.; Sripanidkulchai, B. Neuroprotective Effects of aged garlic extract on cognitive dysfunction and neuroinflammation induced by β-amyloid in rats. Nutrients, 2017, 9(1), 24. [http://dx.doi.org/10.3390/nu9010024]. [PMID: 28054940].
[243]
Wu, H.; Devaraj, N.K. Advances in tetrazine bioorthogonal chemistry driven by the synthesis of novel tetrazines and dienophiles. Acc. Chem. Res., 2018, 51(5), 1249-1259. [http://dx.doi.org/10.1021/acs.accounts.8b00062]. [PMID: 29638113].