Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

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

Review Article

Tau Protein Aggregation in Alzheimer's Disease: Recent Advances in the Development of Novel Therapeutic Agents

Author(s): Kadja L.C. Monteiro, Marcone G. dos S. Alcântara, Thiago M. de Aquino and Edeildo F. da Silva-Júnior*

Volume 26, Issue 15, 2020

Page: [1682 - 1692] Pages: 11

DOI: 10.2174/1381612826666200414164038

Price: $65

Abstract

Major research in Alzheimer’s disease (AD) related to disease-modifying agents is concentrated on pharmacological approaches related to diagnostic markers, neurofibrillary tangles and amyloid plaques. Although most studies focus on anti-amyloid strategies, investigations on tau protein have produced significant advances in the modulation of the pathophysiology of several neurodegenerative diseases. Since the discovery of phenothiazines as tau protein aggregation inhibitors (TAGIs), many additional small molecule inhibitors have been discovered and characterized in biological model systems, which exert their interaction effects by covalent and noncovalent means. In this paper, we summarize the latest advances in the discovery and development of tau aggregation inhibitors using a specialized approach in their chemical classes. The design of new TAGIs and their encouraging use in in vivo and clinical trials support their potential therapeutic use in AD.

Keywords: Alzheimer`s disease, tau protein, aggregation inhibitors, neurodegeneration, phenothiazines, molecule inhibitors.

[1]
Fernández-Ruiz J. The biomedical challenge of neurodegenerative disorders: an opportunity for cannabinoid-based therapies to improve on the poor current therapeutic outcomes. Br J Pharmacol 2019; 176(10): 1370-83.
[http://dx.doi.org/10.1111/bph.14382] [PMID: 29856067]
[2]
Cacciatore I, Baldassarre L, Fornasari E, Mollica A, Pinnen F. Recent advances in the treatment of neurodegenerative diseases based on GSH delivery systems. Oxid Med Cell Longev 2012; 2012
[http://dx.doi.org/10.1155/2012/240146]
[3]
Baig MH, Ahmad K, Saeed M, et al. Peptide based therapeutics and their use for the treatment of neurodegenerative and other diseases. Biomed Pharmacother 2018; 103: 574-81.
[http://dx.doi.org/10.1016/j.biopha.2018.04.025] [PMID: 29677544]
[4]
Shah RS, Lee HG, Xiongwei Z, Perry G, Smith MA, Castellani RJ. Current approaches in the treatment of Alzheimer’s disease. Biomed Pharmacother 2008; 62(4): 199-207.
[http://dx.doi.org/10.1016/j.biopha.2008.02.005] [PMID: 18407457]
[5]
Götz J, Ittner A, Ittner LM. Tau-targeted treatment strategies in Alzheimer’s disease. Br J Pharmacol 2012; 165(5): 1246-59.
[http://dx.doi.org/10.1111/j.1476-5381.2011.01713.x] [PMID: 22044248]
[6]
Jouanne M, Rault S, Voisin-Chiret AS. Tau protein aggregation in Alzheimer’s disease: An attractive target for the development of novel therapeutic agents. Eur J Med Chem 2017; 139: 153-67.
[http://dx.doi.org/10.1016/j.ejmech.2017.07.070] [PMID: 28800454]
[7]
Gong C-X, Iqbal K. Hyperphosphorylation of microtubule-associated protein tau: a promising therapeutic target for Alzheimer disease. Curr Med Chem 2008; 15(23): 2321-8.
[http://dx.doi.org/10.2174/092986708785909111] [PMID: 18855662]
[8]
Cummings J, Lee G, Ritter A, Zhong K. Alzheimer’s disease drug development pipeline: 2018. Alzheimers Dement 2018; 4: 195-214.
[http://dx.doi.org/10.1016/j.trci.2018.03.009] [PMID: 29955663]
[9]
Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement 2019; 5: 272-93.
[http://dx.doi.org/10.1016/j.trci.2019.05.008] [PMID: 31334330]
[10]
Hanger DP, Anderton BH, Noble W. Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol Med 2009; 15(3): 112-9.
[http://dx.doi.org/10.1016/j.molmed.2009.01.003] [PMID: 19246243]
[11]
Sierra-Fonseca JA, Gosselink KL. Tauopathy and neurodegeneration: A role for stress. Neurobiol Stress 2018; 9: 105-12.
[http://dx.doi.org/10.1016/j.ynstr.2018.08.009] [PMID: 30450376]
[12]
Kuca K, Soukup O, Maresova P, et al. Current approaches against Alzheimer’s disease in clinical trials. J Braz Chem Soc 2016; 27: 641-9.
[http://dx.doi.org/10.5935/0103-5053.20160048]
[13]
Kumar A, Singh A, Ekavali . A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep 2015; 67(2): 195-203.
[http://dx.doi.org/10.1016/j.pharep.2014.09.004] [PMID: 25712639]
[14]
Chen HSV, Pellegrini JW, Aggarwal SK, et al. Open-channel block of N-methyl-D-aspartate (NMDA) responses by memantine: therapeutic advantage against NMDA receptor-mediated neurotoxicity. J Neurosci 1992; 12(11): 4427-36.
[http://dx.doi.org/10.1523/JNEUROSCI.12-11-04427.1992] [PMID: 1432103]
[15]
Alam S, Lingenfelter KS, Bender AM, Lindsley CW. Classics in chemical neuroscience: memantine. ACS Chem Neurosci 2017; 8(9): 1823-9.
[http://dx.doi.org/10.1021/acschemneuro.7b00270] [PMID: 28737885]
[16]
Madav Y, Wairkar S, Prabhakar B. Recent therapeutic strategies targeting beta amyloid and tauopathies in Alzheimer’s disease. Brain Res Bull 2019; 146: 171-84.
[http://dx.doi.org/10.1016/j.brainresbull.2019.01.004] [PMID: 30634016]
[17]
Bhounsule AS, Bhatt LK, Prabhavalkar KS, Oza M. Cyclin dependent kinase 5: A novel avenue for Alzheimer’s disease. Brain Res Bull 2017; 132: 28-38.
[http://dx.doi.org/10.1016/j.brainresbull.2017.05.006] [PMID: 28526617]
[18]
Journal E, Chemistry M, Chemistry P. Recent advances in microtubule-stabilizing agents. Eur J Med Chem 2015; 97: 786-815.
[19]
Panza F, Solfrizzi V, Seripa D, et al. Tau-centric targets and drugs in clinical development for the treatment of Alzheimer’s disease. BioMed Res Int 2016; 2016
[http://dx.doi.org/10.1155/2016/3245935]
[20]
Cisek K, Cooper GL, Huseby CJ, Kuret J. Structure and mechanism of action of tau aggregation inhibitors. Curr Alzheimer Res 2014; 11(10): 918-27.
[http://dx.doi.org/10.2174/1567205011666141107150331] [PMID: 25387336]
[21]
Casamenti F, Grossi C, Rigacci S, Pantano D, Luccarini I, Stefani M. Oleuropein aglycone: a possible drug against degenerative conditions. In Vivo evidence of its effectiveness against alzheimer’s disease. J Alzheimers Dis 2015; 45(3): 679-88.
[http://dx.doi.org/10.3233/JAD-142850] [PMID: 25649656]
[22]
Monti MC, Margarucci L, Riccio R, Casapullo A. Modulation of tau protein fibrillization by oleocanthal. J Nat Prod 2012; 75(9): 1584-8.
[http://dx.doi.org/10.1021/np300384h] [PMID: 22988908]
[23]
Panza F, Seripa D, Solfrizzi V, et al. Tau aggregation inhibitors: the future of Alzheimer’s pharmacotherapy? Expert Opin Pharmacother 2016; 17(4): 457-61.
[http://dx.doi.org/10.1517/14656566.2016.1146686] [PMID: 26809554]
[24]
Crowe A, James MJ, Lee VM, et al. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J Biol Chem 2013; 288(16): 11024-37.
[http://dx.doi.org/10.1074/jbc.M112.436006] [PMID: 23443659]
[25]
Ahmad B, Lapidus LJ. Curcumin prevents aggregation in α-synuclein by increasing reconfiguration rate. J Biol Chem 2012; 287(12): 9193-9.
[http://dx.doi.org/10.1074/jbc.M111.325548] [PMID: 22267729]
[26]
Akoury E, Gajda M, Pickhardt M, et al. Inhibition of tau filament formation by conformational modulation. J Am Chem Soc 2013; 135(7): 2853-62.
[http://dx.doi.org/10.1021/ja312471h] [PMID: 23360400]
[27]
Schafer KN, Cisek K, Huseby CJ, Chang E, Kuret J. Structural determinants of Tau aggregation inhibitor potency. J Biol Chem 2013; 288(45): 32599-611.
[http://dx.doi.org/10.1074/jbc.M113.503474] [PMID: 24072703]
[28]
Taniguchi S, Suzuki N, Masuda M, et al. Inhibition of heparin-induced tau filament formation by phenothiazines, polyphenols, and porphyrins. J Biol Chem 2005; 280(9): 7614-23.
[http://dx.doi.org/10.1074/jbc.M408714200] [PMID: 15611092]
[29]
Buchholz K, Schirmer RH, Eubel JK, et al. Interactions of methylene blue with human disulfide reductases and their orthologues from Plasmodium falciparum. Antimicrob Agents Chemother 2008; 52(1): 183-91.
[http://dx.doi.org/10.1128/AAC.00773-07] [PMID: 17967916]
[30]
Schirmer RH, Adler H, Pickhardt M, Mandelkow E. “Lest we forget you--methylene blue...”. Neurobiol Aging 2011; 32(12): e7-2325.
[http://dx.doi.org/10.1016/j.neurobiolaging.2010.12.012] [PMID: 21316815]
[31]
Harvey BH, Duvenhage I, Viljoen F, et al. Role of monoamine oxidase, nitric oxide synthase and regional brain monoamines in the antidepressant-like effects of methylene blue and selected structural analogues. Biochem Pharmacol 2010; 80(10): 1580-91.
[http://dx.doi.org/10.1016/j.bcp.2010.07.037] [PMID: 20699087]
[32]
Oz M, Lorke DE, Petroianu GA. Methylene blue and Alzheimer’s disease. Biochem Pharmacol 2009; 78(8): 927-32.
[http://dx.doi.org/10.1016/j.bcp.2009.04.034] [PMID: 19433072]
[33]
Jadhav S, Avila J, Schöll M, et al. A walk through tau therapeutic strategies. Acta Neuropathol Commun 2019; 7(1): 22.
[http://dx.doi.org/10.1186/s40478-019-0664-z] [PMID: 30767766]
[34]
Akoury E, Pickhardt M, Gajda M, Biernat J, Mandelkow E, Zweckstetter M. Mechanistic basis of phenothiazine-driven inhibition of Tau aggregation. Angew Chem Int Ed Engl 2013; 52(12): 3511-5.
[http://dx.doi.org/10.1002/anie.201208290] [PMID: 23401175]
[35]
Jinwal UK, Miyata Y, Koren J III, et al. Chemical manipulation of hsp70 ATPase activity regulates tau stability. J Neurosci 2009; 29(39): 12079-88.
[http://dx.doi.org/10.1523/JNEUROSCI.3345-09.2009] [PMID: 19793966]
[36]
Necula M, Kayed R, Milton S, Glabe CG. Small molecule inhibitors of aggregation indicate that amyloid β oligomerization and fibrillization pathways are independent and distinct. J Biol Chem 2007; 282(14): 10311-24.
[http://dx.doi.org/10.1074/jbc.M608207200] [PMID: 17284452]
[37]
Lo Cascio F, Kayed R, Azure C. Azure C targets and modulates toxic tau oligomers. ACS Chem Neurosci 2018; 9(6): 1317-26.
[http://dx.doi.org/10.1021/acschemneuro.7b00501] [PMID: 29378132]
[38]
Fish PV, Steadman D, Bayle ED, Whiting P. New approaches for the treatment of Alzheimer’s disease. Bioorg Med Chem Lett 2019; 29(2): 125-33.
[http://dx.doi.org/10.1016/j.bmcl.2018.11.034] [PMID: 30501965]
[39]
Iqbal K, Liu F, Gong CX. Recent developments with tau-based drug discovery. Expert Opin Drug Discov 2018; 13(5): 399-410.
[http://dx.doi.org/10.1080/17460441.2018.1445084] [PMID: 29493301]
[40]
Baddeley TC, McCaffrey J, Storey JMD, et al. Complex disposition of methylthioninium redox forms determines efficacy in tau aggregation inhibitor therapy for Alzheimer’s disease. J Pharmacol Exp Ther 2015; 352(1): 110-8.
[http://dx.doi.org/10.1124/jpet.114.219352] [PMID: 25320049]
[41]
Melis V, Magbagbeolu M, Rickard JE, et al. Effects of oxidized and reduced forms of methylthioninium in two transgenic mouse tauopathy models. Behav Pharmacol 2015; 26(4): 353-68.
[http://dx.doi.org/10.1097/FBP.0000000000000133] [PMID: 25769090]
[42]
Gauthier S, Feldman HH, Schneider LS, et al. Efficacy and safety of tau-aggregation inhibitor therapy in patients with mild or moderate Alzheimer’s disease: a randomised, controlled, double-blind, parallel-arm, phase 3 trial. Lancet 2016; 388(10062): 2873-84.
[http://dx.doi.org/10.1016/S0140-6736(16)31275-2] [PMID: 27863809]
[43]
Novak P, Schmidt R, Kontsekova E, et al. Safety and immunogenicity of the tau vaccine AADvac1 in patients with Alzheimer’s disease: a randomised, double-blind, placebo-controlled, phase 1 trial. Lancet Neurol 2017; 16(2): 123-34.
[http://dx.doi.org/10.1016/S1474-4422(16)30331-3] [PMID: 27955995]
[44]
Medina M. An Overview on the clinical development of tau-based therapeutics. Int J Mol Sci 2018; 19(4): 19.
[http://dx.doi.org/10.3390/ijms19041160] [PMID: 29641484]
[45]
National Library of Medicine. A study to evaluate the efficacy and safety of RO7105705 in patients with prodromal to mild alzheimer’s disease clinicaltrials Available at:. https://clinicaltrials.gov/ct2/show/NCT03289143
[46]
West T, Hu Y, Verghese PB, et al. Preclinical and clinical development of ABBV-8E12, a Humanized anti-tau antibody, for treatment of alzheimer’s disease and other tauopathies. J Prev Alzheimers Dis 2017; 4(4): 236-41.
[PMID: 29181488]
[47]
Budur K, Budur K, West T, et al. Be, D.M.H. Results of a Phase 1, single ascending dose, placebo-controlled study of ABBV-8E12 in patients with progressive supranuclear palsy and phase 2 study design in early alzheimer’s disease. Alzheimer’s Dement J Alzheimer’s Assoc 2017; 13: 599-600.
[http://dx.doi.org/10.1016/j.jalz.2017.07.241]
[48]
von Bergen M, Friedhoff P, Biernat J, Heberle J, Mandelkow EM, Mandelkow E. Assembly of τ protein into Alzheimer paired helical filaments depends on a local sequence motif ((306)VQIVYK(311)) forming β structure. Proc Natl Acad Sci USA 2000; 97(10): 5129-34.
[http://dx.doi.org/10.1073/pnas.97.10.5129] [PMID: 10805776]
[49]
Wang CK, Northfield SE, Huang YH, Ramos MC, Craik DJ. Inhibition of tau aggregation using a naturally-occurring cyclic peptide scaffold. Eur J Med Chem 2016; 109: 342-9.
[http://dx.doi.org/10.1016/j.ejmech.2016.01.006] [PMID: 26807864]
[50]
Zheng J, Liu C, Sawaya MR, et al. Macrocyclic β-sheet peptides that inhibit the aggregation of a tau-protein-derived hexapeptide. J Am Chem Soc 2011; 133(9): 3144-57.
[http://dx.doi.org/10.1021/ja110545h] [PMID: 21319744]
[51]
Sievers SA, Karanicolas J, Chang HW, et al. Structure-based design of non-natural amino-acid inhibitors of amyloid fibril formation. Nature 2011; 475(7354): 96-100.
[http://dx.doi.org/10.1038/nature10154] [PMID: 21677644]
[52]
Shariff L, Zhu Y, Cowper B, Di WL, Macmillan D. Sunflower trypsin inhibitor (SFTI-1) analogues of synthetic and biological origin via N→S Acyl transfer: potential inhibitors of human Kallikrein-5 (KLK5). Tetrahedron 2014; 70: 7675-80.
[http://dx.doi.org/10.1016/j.tet.2014.06.059]
[53]
Colgrave ML, Craik DJ. Thermal, chemical, and enzymatic stability of the cyclotide kalata B1: the importance of the cyclic cystine knot. Biochemistry 2004; 43(20): 5965-75.
[http://dx.doi.org/10.1021/bi049711q] [PMID: 15147180]
[54]
Pickhardt M, Gazova Z, von Bergen M, et al. Anthraquinones inhibit tau aggregation and dissolve Alzheimer’s paired helical filaments in vitro and in cells. J Biol Chem 2005; 280(5): 3628-35.
[http://dx.doi.org/10.1074/jbc.M410984200] [PMID: 15525637]
[55]
Pickhardt M, Lawatscheck C, Borner HG, Mandelkow E. Inhibition of tau protein aggregation by rhodanine-based compounds solubilized via specific formulation additives to improve bioavailability and cell viability. Curr Alzheimer Res 2017; 14(7): 742-52.
[PMID: 28155596]
[56]
Pickhardt M, von Bergen M, Gazova Z, et al. Screening for inhibitors of tau polymerization. Curr Alzheimer Res 2005; 2(2): 219-26.
[http://dx.doi.org/10.2174/1567205053585891] [PMID: 15974921]
[57]
Bulic B, Pickhardt M, Khlistunova I, et al. Rhodanine-based tau aggregation inhibitors in cell models of tauopathy. Angew Chem Int Ed Engl 2007; 46(48): 9215-9.
[http://dx.doi.org/10.1002/anie.200704051] [PMID: 17985339]
[58]
Waters ML. Aromatic interactions in model systems. Curr Opin Chem Biol 2002; 6(6): 736-41.
[http://dx.doi.org/10.1016/S1367-5931(02)00359-9] [PMID: 12470725]
[59]
Fuse S, Matsumura K, Fujita Y, Sugimoto H, Takahashi T. Development of dual targeting inhibitors against aggregations of amyloid-β and tau protein. Eur J Med Chem 2014; 85: 228-34.
[http://dx.doi.org/10.1016/j.ejmech.2014.07.095] [PMID: 25086914]
[60]
Chegaev K, Federico A, Marini E, et al. NO-donor thiacarbocyanines as multifunctional agents for Alzheimer’s disease. Bioorg Med Chem 2015; 23(15): 4688-98.
[http://dx.doi.org/10.1016/j.bmc.2015.05.050] [PMID: 26078011]
[61]
Chugunova EA, Burilov AR. Novel structural hybrids on the base of benzofuroxans and furoxans. Mini-Review. Curr Top Med Chem 2017; 17(9): 986-1005.
[http://dx.doi.org/10.2174/1568026616666160927145822] [PMID: 27697059]
[62]
Necula M, Chirita CN, Kuret J. Cyanine dye N744 inhibits tau fibrillization by blocking filament extension: implications for the treatment of tauopathic neurodegenerative diseases. Biochemistry 2005; 44(30): 10227-37.
[http://dx.doi.org/10.1021/bi050387o] [PMID: 16042400]
[63]
Miyasaka T, Xie C, Yoshimura S, et al. Curcumin improves tau-induced neuronal dysfunction of nematodes. Neurobiol Aging 2016; 39: 69-81.
[http://dx.doi.org/10.1016/j.neurobiolaging.2015.11.004] [PMID: 26923403]
[64]
Ono K, Hasegawa K, Naiki H, Yamada M. Curcumin has potent anti-amyloidogenic effects for Alzheimer’s β-amyloid fibrils in vitro. J Neurosci Res 2004; 75(6): 742-50.
[http://dx.doi.org/10.1002/jnr.20025] [PMID: 14994335]
[65]
Bijari N, Balalaie S, Akbari V, et al. Effective suppression of the modified PHF6 peptide/1N4R Tau amyloid aggregation by intact curcumin, not its degradation products: another evidence for the pigment as preventive/therapeutic “functional food”. Int J Biol Macromol 2018; 120(Pt. A): 1009-22.
[http://dx.doi.org/10.1016/j.ijbiomac.2018.08.175] [PMID: 30172816]
[66]
Okuda M, Hijikuro I, Fujita Y, et al. Design and synthesis of curcumin derivatives as tau and amyloid β dual aggregation inhibitors. Bioorg Med Chem Lett 2016; 26(20): 5024-8.
[http://dx.doi.org/10.1016/j.bmcl.2016.08.092] [PMID: 27624076]
[67]
Freyssin A, Page G, Fauconneau B, Rioux Bilan A. Natural polyphenols effects on protein aggregates in Alzheimer’s and Parkinson’s prion-like diseases. Neural Regen Res 2018; 13(6): 955-61.
[http://dx.doi.org/10.4103/1673-5374.233432] [PMID: 29926816]
[68]
Cascella M, Bimonte S, Muzio MR, Schiavone V, Cuomo A. The efficacy of Epigallocatechin-3-gallate (green tea) in the treatment of Alzheimer’s disease: an overview of pre-clinical studies and translational perspectives in clinical practice. Infect Agent Cancer 2017; 12: 36.
[http://dx.doi.org/10.1186/s13027-017-0145-6] [PMID: 28642806]
[69]
Wobst HJ, Sharma A, Diamond MI, Wanker EE, Bieschke J. The green tea polyphenol (-)-epigallocatechin gallate prevents the aggregation of tau protein into toxic oligomers at substoichiometric ratios. FEBS Lett 2015; 589(1): 77-83.
[http://dx.doi.org/10.1016/j.febslet.2014.11.026] [PMID: 25436420]
[70]
Wang DM, Li SQ, Wu WL, Zhu XY, Wang Y, Yuan HY. Effects of long-term treatment with quercetin on cognition and mitochondrial function in a mouse model of Alzheimer’s disease. Neurochem Res 2014; 39(8): 1533-43.
[http://dx.doi.org/10.1007/s11064-014-1343-x] [PMID: 24893798]
[71]
Belen Tejada-Romero Jean-Michel Carter, Yuliana Mihaylova,B.N. and A.A. Aboobaker. The flavonoid quercetin ameliorates alzheimer’s disease pathology and protects cognitive and emotional function in aged triple transgenic alzheimer’s disease model mice 2015. In press
[72]
Chen J, Deng X, Liu N, et al. Quercetin attenuates tau hyperphosphorylation and improves cognitive disorder via suppression of er stress in a manner dependent on AMPK pathway. J Funct Foods 2016; 22: 463-76.
[http://dx.doi.org/10.1016/j.jff.2016.01.036]
[73]
Kumar S, Krishnakumar VG, Morya V, Gupta S, Datta B. Nanobiocatalyst facilitated aglycosidic quercetin as a potent inhibitor of tau protein aggregation. Int J Biol Macromol 2019; 138: 168-80.
[http://dx.doi.org/10.1016/j.ijbiomac.2019.07.081] [PMID: 31306707]
[74]
de Andrade Teles RB, Diniz TC, Costa Pinto TC, et al. Flavonoids as therapeutic agents in alzheimer’s and parkinson’s diseases: a systematic review of preclinical evidences. Oxid Med Cell Longev 2018; 2018
[http://dx.doi.org/10.1155/2018/7043213]
[75]
Cuanalo-Contreras K, Park KW, Mukherjee A, Millán-Pérez Peña L, Soto C. Delaying aging in Caenorhabditis elegans with protein aggregation inhibitors. Biochem Biophys Res Commun 2017; 482(1): 62-7.
[http://dx.doi.org/10.1016/j.bbrc.2016.10.143] [PMID: 27810360]
[76]
Cornejo A, Aguilar Sandoval F, Caballero L, et al. Rosmarinic acid prevents fibrillization and diminishes vibrational modes associated to β sheet in tau protein linked to Alzheimer’s disease. J Enzyme Inhib Med Chem 2017; 32(1): 945-53.
[http://dx.doi.org/10.1080/14756366.2017.1347783] [PMID: 28701064]
[77]
Ono K, Yoshiike Y, Takashima A, Hasegawa K, Naiki H, Yamada M. Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: implications for the prevention and therapeutics of Alzheimer’s disease. J Neurochem 2003; 87(1): 172-81.
[http://dx.doi.org/10.1046/j.1471-4159.2003.01976.x] [PMID: 12969264]
[78]
Lunven L, Bonnet H, Yahiaoui S, et al. Disruption of fibers from the tau model AcPHF6 by naturally occurring aurones and synthetic analogues. ACS Chem Neurosci 2016; 7(7): 995-1003.
[http://dx.doi.org/10.1021/acschemneuro.6b00102] [PMID: 27225823]
[79]
Onishi T, Iwashita H, Uno Y, et al. A Novel glycogen synthase Kinase-3 Inhibitor 2-Methyl-5-(3-4-[(S)- Methylsulfinyl]Phenyl-1-Benzofuran-5-Yl)-1,3,4-Oxadiazole decreases tau phosphorylation and ameliorates cognitive deficits in a transgenic model of alzheimer’s disease. J Neurochem 2011; 119: 1330-40.
[http://dx.doi.org/10.1111/j.1471-4159.2011.07532.x] [PMID: 21992552]
[80]
Vicidomini C, Cioffi F, Broersen K, et al. Benzodifurans for biomedical applications: BZ4, a selective anti-proliferative and anti-amyloid lead compound. Future Med Chem 2019; 11: 285-302.
[http://dx.doi.org/10.4155/fmc-2018-0473] [PMID: 30801198]
[81]
Teng ZH, Zhou SY, Yang RT, et al. Quantitation assay for absorption and first-pass metabolism of emodin in isolated rat small intestine using liquid chromatography-tandem mass spectrometry. Biol Pharm Bull 2007; 30(9): 1628-33.
[http://dx.doi.org/10.1248/bpb.30.1628] [PMID: 17827711]
[82]
Russo EB. Cannabis therapeutics and the future of neurology. Front Integr Nuerosci 2018; 12: 51.
[http://dx.doi.org/10.3389/fnint.2018.00051] [PMID: 30405366]
[83]
Esposito G, De Filippis D, Carnuccio R, Izzo AA, Iuvone T. The marijuana component cannabidiol inhibits β-amyloid-induced tau protein hyperphosphorylation through Wnt/β-catenin pathway rescue in PC12 cells. J Mol Med (Berl) 2006; 84(3): 253-8.
[http://dx.doi.org/10.1007/s00109-005-0025-1] [PMID: 16389547]
[84]
Paranjape SR, Chiang YM, Sanchez JF, et al. Inhibition of Tau aggregation by three Aspergillus nidulans secondary metabolites: 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde. Planta Med 2014; 80(1): 77-85.
[http://dx.doi.org/10.1055/s-0033-1360180] [PMID: 24414310]
[85]
Cornejo A, Salgado F, Caballero J, Vargas R, Simirgiotis M, Areche C. Secondary metabolites in ramalina terebrata detected by UHPLC/ESI/MS/MS and identification of parietin as tau protein inhibitor. Int J Mol Sci 2016; 17(8): 1-13.
[http://dx.doi.org/10.3390/ijms17081303] [PMID: 27548142]
[86]
Areche C, Zapata F, González M, et al. Anthraquinone derivative reduces tau oligomer progression by inhibiting cysteine-cysteine interaction. ChemistryOpen 2019; 8(5): 554-9.
[http://dx.doi.org/10.1002/open.201800222] [PMID: 31065505]
[87]
Habtemariam S. Natural products in Alzheimer’s disease therapy: would old therapeutic approaches fix the broken promise of modern medicines? Molecules 2019; 24(8): 1-17.
[http://dx.doi.org/10.3390/molecules24081519] [PMID: 30999702]
[88]
Di Pietro O, Pérez-Areales, F. Javiere etal.Tetrahydrobenzo[h][1,6]naphthyridine-6-chlorotacrine hybrids as a new family of anti-Alzheimer agents targeting β-amyloid, tau, and cholinesterase pathologies. Eur J Med Chem 2014; 84: 107-17.
[http://dx.doi.org/10.1016/j.ejmech.2014.07.021]
[89]
Lv P, Xia CL, Wang N, Liu ZQ, Huang ZS, Huang SL. Synthesis and evaluation of 1,2,3,4-tetrahydro-1-acridone analogues as potential dual inhibitors for amyloid-beta and tau aggregation. Bioorg Med Chem 2018; 26(16): 4693-705.
[http://dx.doi.org/10.1016/j.bmc.2018.08.007] [PMID: 30107970]
[90]
Larbig G, Pickhardt M, Lloyd DG, Schmidt B, Mandelkow E. Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: a ligand based approach results in successful scaffold hopping. Curr Alzheimer Res 2007; 4(3): 315-23.
[http://dx.doi.org/10.2174/156720507781077250] [PMID: 17627489]
[91]
Pickhardt M, Larbig G, Khlistunova I, et al. Phenylthiazolyl-hydrazide and its derivatives are potent inhibitors of τ aggregation and toxicity in vitro and in cells. Biochemistry 2007; 46(35): 10016-23.
[http://dx.doi.org/10.1021/bi700878g] [PMID: 17685560]
[92]
Taghavi A, Nasir S, Pickhardt M, et al. N′-benzylidene-benzohydrazides as novel and selective tau-PHF ligands. J Alzheimers Dis 2011; 27(4): 835-43.
[http://dx.doi.org/10.3233/JAD-2011-111238] [PMID: 21891864]
[93]
Crowe A, Huang W, Ballatore C, et al. Identification of aminothienopyridazine inhibitors of tau assembly by quantitative high-throughput screening. Biochemistry 2009; 48(32): 7732-45.
[http://dx.doi.org/10.1021/bi9006435] [PMID: 19580328]
[94]
Ballatorea C, Brundenb KR, Piscitellia F, et al. A.B.S. Discovery of brain-penetrant, orally bioavailable aminothienopyridazine inhibitors of tau aggregation. J Med Chem 2011; 23: 3739-47.
[95]
Ballatore C, Crowe A, Piscitelli F, et al. Aminothienopyridazine inhibitors of tau aggregation: evaluation of structure-activity relationship leads to selection of candidates with desirable in vivo properties. Bioorg Med Chem 2012; 20(14): 4451-61.
[http://dx.doi.org/10.1016/j.bmc.2012.05.027] [PMID: 22717239]
[96]
Moir M, Chua SW, Reekie T, et al. Ring-opened aminothienopyridazines as novel tau aggregation inhibitors. MedChemComm 2017; 8(6): 1275-82.
[http://dx.doi.org/10.1039/C6MD00306K] [PMID: 30108838]

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