Abstract

Research Article

The Role of Tau Protein in Diseases

Bhaskar C Das*, Sribidya Pradhan, Devi Prasana Ojha, Arpita Das, Narayan S Hosmane and Sasmita Das

Published: 09 April, 2018 | Volume 2 - Issue 1 | Pages: 001-016

Amyloid-β peptide (Aβ) and tau protein deposits in the human brain are the pathological hallmarks of Alzheimer’s disease (AD). Tau is a class of proteins that are abundant in nerve cells and perform the function of stabilizing microtubules. However, in certain pathological situations, Tau proteins become defective and fail to adequately stabilize microtubules, which can result in the generation of abnormal masses that are toxic to neurons. This process occurs in a number of neurological disorders collectively known as Tauopathies. Tau protein is the major factor of the intracellular filamentous deposits that relate to a number of neurodegenerative diseases which includes the progressive supranuclear palsy (PSP), Pick’s disease, and Parkinsonism. The identification of mutations in Tau established that dysfunction or misregulation of tau protein is sufficient to cause dementia and neurodegeneration. In this review article, we discussed the etiology of the tau formation and role in AD and subsequently therapeutic approach for disassembling and Tau inhibition.

Read Full Article HTML DOI: 10.29328/journal.aac.1001010 Cite this Article Read Full Article PDF

References

  1. Wimo A, Jonsson L, Bond J, Prince M, Winblad B, et al. Alzheimer Disease international. The worldwide economic impact of dementia 2010. Alzheimers Dement. 2013; 9: 1-11. https://goo.gl/A1pLaU
  2. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol.2011; 7: 137-152. Ref.: https://goo.gl/eNPeUJ
  3. Cleveland DW, Hwo SY, Kirschner MW. Physical and chemical properties of purified tau factor and the role of tau in microtubule assembly. J Mol Bio. 1977; 116: 227-247. Ref.: https://goo.gl/KEAE1k; (b) Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol. 2017; 133: 665-704. Ref.: https://goo.gl/26pR2Z; (c) Avila J, Lucas JJ, Pérez M, Hernández F. Role of Tau Protein in both Physiological and Pathological Conditions. Physiol Rev. 2004; 84: 361-384. Ref.: https://goo.gl/v86s1E; (d) Nadimidla K., Ismail T., Kanapathipillai M., Tau peptides and tau mutant protein aggregation inhibition by cationic polyethyleneimine and polyarginine. Biopolymers. 2017; 107: 23024. Ref.: https://goo.gl/Vmmzn1
  4. Morris M, Maeda S, Vossel K, Mucke L. The Many Faces of Tau. Neuron. 2011; 70: 410-426. Ref.: https://goo.gl/1ukURr
  5. Ruben GC, Iqbal K, Grundke-Iqbal I, Wisniewski HM, Ciardelli TL, et al. The microtubule associated protein tau forms a triple-stranded left-hand helical polymer. J Biol Chem. 1991; 266: 22019-22027. Ref.: https://goo.gl/pzTXA6
  6. Cleveland DW, Hwo SY, Kirschner MW. Purification of tau, a microtubule-associated protein that induces assembly of microtubules from purified tubulin. J Mol Biol. 1977; 116: 207-225. Ref.: https://goo.gl/HyVHN9
  7. Imahori K, Uchida T. Physiology and pathology of tau protein kinases in relation to Alzheimer's disease. J Biochem. 1997; 121: 179-188. Ref.: https://goo.gl/JxvEpC
  8. Goedert M, Spillantini MG, Potier MC, Ulrich J, Crowther RA. Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain. EMBO J. 1989; 8: 393-399. Ref.: https://goo.gl/j1xdLw
  9. Drubin DG, Kirschner MW. Tau protein function in living cells. J Cell Biol. 1986; 103: 2739-2746. Ref.: https://goo.gl/Mt6JGU
  10. Lee G, Thangavel R, Sharma VM, Litersky JM, Bhaskar K, et al. Phosphorylation of tau by fyn: implications for Alzheimer’s disease. J Neurosci. 2004; 24: 2304-2312. Ref.: https://goo.gl/rxKLHg
  11. Roberson ED, Halabisky B, Yoo JW, Yao J, Chin J, et al. Amyloid‑β/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J Neurosci. 2011; 31: 700-711. Ref.: https://goo.gl/fLfrKo
  12. Cleveland DW, Hwo SY, Kirschner MW. Purification of tau, a microtubule-associated protein that induces assembly of microtubules from purified tubulin. J Mol Biol. 1977; 116: 207-225. Ref.: https://goo.gl/KaxvbQ
  13. Goedert M, Spillanti MG, Jakes R, Rutherford D, Crowther RA. Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer's disease. Neuron. 1989; 3: 519-526. Ref.: https://goo.gl/KsoMmD
  14. Himmler A. Structure of the bovine tau gene: alternatively spliced transcripts generate a protein family. Mol Cell Biol. 1989; 9: 1389-1396. Ref.: https://goo.gl/ZCX9yT
  15. Kopke E, Tung YC, Shaikh S, Alonso AC, Iqbal K, et al. Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J Biol Chem. 1993; 268: 24374-24384. Ref.: https://goo.gl/mq6KFV
  16. Kenessey A, Yen SH. The extent of phosphorylation of fetal tau is comparable to that of PHF-tau from Alzheimer paired helical filaments. Brain Res. 1993; 629: 40-46. Ref.: https://goo.gl/xRhuAJ
  17. Maccioni RB, Muñoz JP, Barbeito L. The molecular bases of Alzheimer's disease and other neurodegenerative disorders. Arch Med Res. 2001; 32: 367-381. Ref.: https://goo.gl/ukHCSD
  18. Kosik KS. The molecular and cellular biology of tau. Brain Pathol. 1993; 3: 39-43. Ref.: https://goo.gl/SZBXE6
  19. Mandelkow EM, Biernat J, Drewes G, Gustke N, Trinczek B, et al. Tau domains, phosphorylation, and interactions with microtubules. Neurobiol Aging. 1995; 16: 355-363. Ref.: https://goo.gl/4rCGDU
  20. Liu F, Iqbal K, Grundke-Iqbal I, Rossie S, Gong CX. Dephosphorylation of tau by protein phosphatase 5: impairment in Alzheimer's disease. J Biol Chem. 2005; 280: 1790-1796. Ref.: https://goo.gl/KNZjXj
  21. Avila J. Tau kinases and phosphatases: commentary. J Cell Mol Med. 2008; 12: 258-259. Ref.: https://goo.gl/Xt3StM
  22. Iqbal, Grundke-Iqbal I. Alzheimer neurofibrillary degeneration: significance, etiopathogenesis, therapeutics and prevention: Alzheimer review series. J Cell Mol Med. 2008; 12: 38-55. Ref.: https://goo.gl/C748JD
  23. Carrell RW, Gooptu B. Conformational changes and disease-serpins, prions and Alzheimer's. Curr Opin Struct Biol. 1998; 8: 799-809. Ref.: https://goo.gl/Ss93aG
  24. Fox N, Harvey RJ, Rossor MN. Protein folding, nucleation phenomena and delayed neurodegeneration in Alzheimer's disease. Rev Neurosci. 1996; 7: 21-28. Ref.: https://goo.gl/85dSQN
  25. Hyman BT, Augustinack JC, Ingelsson M. Transcriptional and conformational changes of the tau molecule in Alzheimer's disease. Biochim Biophys Acta. 2005; 1739: 150-157. Ref.: https://goo.gl/h7mMjT
  26. García-Sierra F, Ghoshal N, Quinn B, Berry RW, Bínder LI. Conformational changes and truncation of tau protein during tangle evolution in Alzheimer's disease. J Alzheimers Dis. 2003; 5: 65-77. Ref.: https://goo.gl/JxY4ZH
  27. Ghoshal N, García-Sierra F, Fu Y, Beckett LA, Mufson EJ, et al. Tau-66: evidence for a novel tau conformation in Alzheimer’s disease. J Neurochem. 2001; 77: 1372-1385. Ref.: https://goo.gl/GkAmVcg
  28. Ghoshal N, García-Sierra F, Wuu J, Leurgans S, Bennett DA, et al. Tau conformational changes correspond to impairments of episodic memory in mild cognitive impairment and Alzheimer's disease. Exp Neurol. 2002; 177: 475-493. Ref.: https://goo.gl/NC62Zm
  29. Mandelkow EM, Stamer K, Vogel R, Thies E, Mandelkow E. Clogging of axons by tau, inhibition of axonal traffic and starvation of synapses. Neurobiol Aging. 2003; 24: 1079-1085. Ref.: https://goo.gl/BAbCfL
  30. LaPointe NE, Morfini G, Pigino G, Gaisina IN, Kozikowski AP, et al. The amino terminus of tau inhibits kinesin-dependent axonal transport: implications for filament toxicity. J Neurosci Res. 2009; 87: 440-451. Ref.: https://goo.gl/JwWhxM
  31. Cuchillo-Ibanez I, Seereeram A, Byers HL, Leung KY, Ward MA, et al. Phosphorylation of tau regulates its axonal transport by controlling its binding to kinesin. FASEB J. 2008; 22: 3186-3195. Ref.: https://goo.gl/9PqPpd
  32. Goedert M, Spillantini MG, Potier MC, Ulrich J, Crowther RA. Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain. EMBO J. 1989; 8: 393-399. Ref.: https://goo.gl/14gouX
  33. Delisle MB, Murrell JR, Richardson R, Trofatter JA, Rascol O, et al. A mutation at codon 279 (N279K) in exon 10 of the Tau gene causes a tauopathy with dementia and supranuclear palsy. Acta Neuropathol. 1999; 98: 62-77. Ref.: https://goo.gl/6PDHpy;
    • Standridge JB. Pharmacotherapeutic approaches to the prevention of Alzheimer’s disease. Am J Geriatr Pharmacother. 2004; 2: 119-132. Ref.: https://goo.gl/FtcQSa; (b) XieYibin Y, Youbiao T, Xiubo Z, Du X, Liu Q. Ebselen ameliorates β-amyloid pathology, tau pathology, and cognitive impairment in triple-transgenic Alzheimer’s disease mice. J Biol Inorg Chem. 2017; 22: 851-865. Ref.: https://goo.gl/V5HQ8t ; (c) Dioli C, Patrício P, Trindade R, Pinto LG, Silva JM, et al. Tau-dependent suppression of adult neurogenesis in the stressed hippocampus. Mol Psychiatry. 2017; 22: 1110-1118. Ref.: https://goo.gl/GVLBH2
  34. Boimel M, Grigoriadis N, Lourbopoulos A, Touloumi O, Rosenmann D, et al. Statins reduce the neurofibrillary tangle burden in a mouse model of tauopathy. J Neuropathol Exp Neurol. 2009; 68: 314-325. Ref.: https://goo.gl/mFyk4e
  35. Piedrahita D, Hernandez I, Lopez-Tobon A, Fedorov D, Obara B, et al. Silencing of CDK5 reduces neurofibrillary tangles in transgenic Alzheimer’s mice. J Neurosci. 2010; 30: 13966-13976. Ref.: https://goo.gl/jY9YpV
  36. Leroy K, Ando K, Heraud C, Yilmaz Z, Authelet M, et al. Lithium treatment arrests the development of neurofibrillary tangles in mutant tau transgenic mice with advanced neurofibrillary pathology. J Alzheimers Dis. 2010; 19: 705-719. Ref.: https://goo.gl/j8Cc2M
  37. Villaflores OB, Chen YJ, Chen CP, Yeh JM, Wu TY. Curcuminoids and resveratrol as anti-Alzheimer agents. Taiwan J Obstet Gynecol. 2012; 51: 515-525. Ref.: https://goo.gl/dUtiou
  38. Lazar AN, Mourtas S, Youssef I, Parizot C, Dauphin A, et al. Curcumin-conjugated nanoliposomes with high affinity for Abeta deposits: possible applications to Alzheimer disease. Nanomedicine. 2013; 9: 712-721. Ref.: https://goo.gl/TKJuPP
  39. Hamaguchi T, Ono K, Yamada M. REVIEW: Curcumin and Alzheimer’s disease. CNS Neurosci Ther. 2010; 16: 285-297. Ref.: https://goo.gl/jz9Ykm
  40. Malm T, Mariani M, Donovan LJ, Neilson L, Landreth GE. Activation of the nuclear receptor PPARdelta is neuroprotective in a transgenic mouse model of Alzheimer’s disease through inhibition of inflammation. J Neuroinflamm. 2015; 12: 7. Ref.: https://goo.gl/Xuqsc7
  41. Olmos-Alonso A, Schetters ST, Sri S, Askew K, Mancuso R, et al. Pharmacological targeting of CSF1R inhibits microglial proliferation and prevents the progression of Alzheimer’s-like pathology. Brain. 2016; 139: 891-907. Ref.: https://goo.gl/Nb7PLm
  42. Goozee KG, Shah TM, Sohrabi HR, Rainey-Smith SR, Brown B, et al. Examining the potential clinical value of curcumin in the prevention and diagnosis of Alzheimer’s disease. Br J Nutr. 2016; 115: 449-465. Ref.: https://goo.gl/ebH1tT
  43. Prakash A, Kumar A. Implicating the role of lycopene in restoration of mitochondrial enzymes and BDNF levels in betaamyloid induced Alzheimers disease. Eur J Pharmacol. 2014; 15: 104-111. Ref.: https://goo.gl/fhMoFc
  44. Kishi Y, Schmelzer JD, Yao JK, Zollman PJ, Nickander KK, et al. Alpha-lipoic acid: effect on glucose uptake, sorbitol pathway, and energy metabolism in experimental diabetic neuropathy. Diabetes. 1999; 48: 2045-2051. Ref.: https://goo.gl/2BMD7A
  45. Mitsui Y, Schmelzer JD, Zollman PJ, Mitsui M, Tritschler HJ, et al. Alpha-lipoic acid provides neuroprotection from ischemia-reperfusion injury of peripheral nerve. J Neurol Sci. 1999; 163: 11-16. Ref.: https://goo.gl/BaUXjv
  46. Hardas SS, Sultana R, Clark AM, Beckett TL, Szweda LI, et al. Oxidative modification of lipoic acid by HNE in Alzheimer disease brain. Redox Biol. 2013; 1: 80-85. Ref.: https://goo.gl/obxMQu
  47. Grundman M. Vitamin E and Alzheimer disease: the basis for additional clinical trials. Am J Clin Nutr. 2000; 71: 630S-636S. Ref.: https://goo.gl/xRQ8px
  48. Isaac MG, Quinn R, Tabet N. Vitamin E for Alzheimer’s disease and mild cognitive impairment. Cochrane Database Syst Rev. 2008; 3: CD002854. Ref.: https://goo.gl/9KDm16
  49. Mancuso C, Bates TE, Butterfield DA, Calafato S, Cornelius C, et al. Natural antioxidants in Alzheimer’s disease. Expert Opin Investig Drugs. 2007; 16: 1921-1931. Ref.: https://goo.gl/iPHGWr
  50. Wattanapenpaiboon N, Wahlqvist MW. Phytonutrient deficiency: the place of palm fruit. Asia Pac J Clin Nutr. 2003; 12: 363-368. Ref.: https://goo.gl/4uKLRi
  51. Scali C, Giovannini MG, Prosperi C, Bellucci A, Pepeu G, et al. The selective cyclooxygenase-2 inhibitor rofecoxib suppresses brain inflammation and protects cholinergic neurons from excitotoxic degeneration in vivo. Neuroscience. 2003; 117: 909-919. Ref.: https://goo.gl/8MzFFu
  52. Christen Y. Ginkgo biloba and neurodegenerative disorders. Front Biosci. 2004; 1: 3091-3104. Ref.: https://goo.gl/HVh2aY
  53. Wang JZ, Wang ZF. Role of melatonin in Alzheimer-like neurodegeneration. Acta Pharmacol Sin. 2006; 27: 41-49. Ref.: https://goo.gl/uQ2MPK
  54. De la Monte SM. Contributions of brain insulin resistance and deficiency in amyloid-related neurodegeneration in Alzheimer’s disease. Drugs. 2012; 72: 49-66. Ref.: https://goo.gl/4bvq9R
  55. Freiherr J, Hallschmid M, Frey WH, Brunner YF, Chapman CD, et al. Intranasal insulin as a treatment for Alzheimer’s disease: a review of basic research and clinical evidence. CNS Drugs. 2013; 27: 505-514. Ref.: https://goo.gl/6D6eq4; (b) Wang T, Xie C, Yu P, Fang F, Zhu J, et al. Involvement of Insulin Signaling Disturbances in Bisphenol A-Induced Alzheimer’s Disease-like Neurotoxicity. Sci Rep. 2017; 7: 7497. Ref.: https://goo.gl/ahbMMW
  56. Alagiakrishnan K, Sankaralingam S, Ghosh M, Mereu L, Senior P. Antidiabetic drugs and their potential role in treating mild cognitive impairment and Alzheimer’s disease. Discov Med. 2013; 16: 277-286. Ref.: https://goo.gl/WwaQ93
  57. Chen Y, Zhang J, Zhang B, Gong CX. Targeting insulin signaling for the treatment of Alzheimer’s disease. Curr Top Med Chem. 2016; 16: 485-492. Ref.: https://goo.gl/GYoxpS
  58. Wendy Noble, Emmanuel Planel, Cindy Zehr, Vicki Olm, Jordana Meyerson, et al. Inhibition of glycogen synthase kinase-3 by lithium correlates with reduced tauopathy and degeneration in vivo. Proc Natl Acad Sci.2005;102: 6990-6995. Ref.: https://goo.gl/WavHmz
  59. Gruninger F. Invited review: Drug development for tauopathies. Neuropathol Appl Neurobiol. 2015; 41: 81-96. Ref.: https://goo.gl/EDseLe
  60. Anand R, Gill KD, Mahdi AA. Therapeutics of Alzheimer’s disease: past, present and future. Neuropharmacology. 2014; 76: 27-50. Ref.: https://goo.gl/LMx4TR
  61. Arai H, Suzuki H, Yoshiyama T, Lobello K, Peng Y, et al. Safety, tolerability and immunogenicity of an immunotherapeutic vaccine (vanutide cridificar [ACC-001]) and the QS-21 adjuvant in Japanese individuals with mild-to-moderate Alzheimer’s disease: a phase IIa, multicenter, randomized, adjuvant and placebo clinical trial. Alzheimer’s Dement. 2013; 9: 282. Ref.: https://goo.gl/o92bKg
  62. Gong CX, Liu F, Wu G, Rossie S, Wegiel J, et al. Dephosphorylation of microtubule‐associated protein tau by protein phosphatase 5. J Neurochem. 2004; 88: 298-310. Ref.: https://goo.gl/88hoHq
  63. Fei Liu, Khalid Iqbal, Inge Grundke-Iqbal, Sandra Rossie, Cheng-Xin Gong. Dephosphorylation of Tau by Protein Phosphatase 5 impairment in Alzheimer's disease. J Biological Chemistry. 2005; 280: 1790-1796. Ref.: https://goo.gl/ZMF5yN
  64. Wilkinson D, Windfeld K, Colding-Jorgensen E. Safety and efficacy of idalopirdine, a 5-HT6 receptor antagonist, in patients with moderate Alzheimer’s disease (LADDER): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Neurol. 2014; 13: 1092-1099. Ref.: https://goo.gl/V2zPw1
  65. Schneider LS. Idalopirdine for Alzheimer’s disease: written in the stars. Lancet Neurol. 2014; 13: 1063-1065. Ref.: https://goo.gl/6w1bz9
  66. Marcos B, Chuang TT, Gil-Bea FJ, Ramirez MJ. Effects of 5-HT6 receptor antagonism and cholinesterase inhibition in models of cognitive impairment in the rat. Br J Pharmacol. 2008; 155: 434-440. Ref.: https://goo.gl/r1zzkN
  67. Wilkinson D, Colding-Jorgensen E, Windfeld K. A clinical phase II study of LU AE58054 added to stable donepezil treatment in patients with moderate Alzheimer’s disease. Alzheimer’s Dement. 2013; 9: 529. Ref.: https://goo.gl/KoukT8
  68. Maher-Edwards G, Zvartau-Hind M, Hunter AJ, Gold M, Hopton G, et al. Double-blind, controlled phase II study of a 5-HT6 receptorantagonist, SB-742457, in Alzheimer’s disease. Curr Alzheimer Res. 2010; 7: 374-385. Ref.: https://goo.gl/KbDMRV
  69. Esbenshade TA, Browman KE, Miller TR, Krueger KM, Komater-Roderwald V, et al. Pharmacological properties and procognitive effects of ABT-288, a potent and selective histamine H3 receptor antagonist. J Pharmacol Exp Ther. 2012; 343: 233-245. Ref.: https://goo.gl/5SFdwH
  70. Othman AA, Haig G, Florian H, Locke C, Gertsik L, et al. The H3 antagonist ABT-288 is tolerated at significantly higher exposures in subjects with schizophrenia than in healthy volunteers. Br J Clin Pharmacol. 2014; 77: 965-974. Ref.: https://goo.gl/Rx2Gsc
  71. Haig GM, Pritchett Y, Meier A, Othman AA, Hall C, et al. A randomized study of H3 antagonist ABT-288 in mild-to-moderate Alzheimer’s dementia. J Alzheimers Dis. 2014; 42: 959-971. Ref.: https://goo.gl/P4EnRN
  72. Grove RA, Harrington CM, Mahler A, Beresford I, Maruff P, et al. A randomized, double-blind, placebo-controlled, 16-week study of the H3 receptor antagonist, GSK239512 as a monotherapy in subjects with mild-to-moderate Alzheimer’s disease. Curr Alzheimer Res. 2014; 11: 47-58. Ref.: https://goo.gl/SqH2B7
  73. Burstein AH, Grimes I, Galasko DR, Aisen PS, Sabbagh M, et al. Effect of TTP488 in patients with mild to moderate Alzheimer’s disease. BMC Neurol. 2014; 14: Ref.: https://goo.gl/mPxNah
  74. Prickaerts J, van Goethem NP, Chesworth R, Shapiro G, Boess FG, et al. EVP-6124, a novel and selective alpha7 nicotinic acetylcholine receptor partial agonist, improves memory performance by potentiating the acetylcholine response of alpha7 nicotinic acetylcholine receptors. Neuropharmacology. 2012; 62: 1099-1110. Ref.: https://goo.gl/TqXiGm
  75. Lawlor B, Kennelly S, O’Dwyer S, Cregg F, Walsh C, et al. NILVAD protocol: a European multicentre double-blind placebo-controlled trial of nilvadipine in mild-to-moderate Alzheimer’s disease. BMJ Open. 2014; 4. Ref.: https://goo.gl/95ZEUd
  76. Elias Akoury, Michal Gajda, Marcus Pickhardt, Jacek Biernat, Pornsuwan Soraya, et al. Inhibition of Tau Filament Formation by Conformational Modulation. J Am Chem Soc. 2013; 135: 2853-2862. Ref.: https://goo.gl/pYQwZt
  77. Pul R, Dodel R, Stangel M. Antibody-based therapy in Alzheimer's disease. Expert Opin Biol Ther. 2011; 11: 343-357. Ref.: https://goo.gl/xMvY13
  78. Bin Zhang, Arpita Maiti, Sharon Shively, Fara Lakhani, Gaye McDonald-Jones, et al. Microtubule-binding drugs offset tau sequestration by stabilizing microtubules and reversing fast axonal transport deficits in a tauopathy model. Proc Natl Acad Sci. 2005; 102: 227-231. Ref.: https://goo.gl/5tCRDW
  79. Zhang B, Maiti A, Shively S, Lakhani F, McDonald-Jones G, et al. Microtubule-binding drugs offset tau equestration by stabilizing microtubules and reversing fast axonal transport deficits in a tauopathy model. Proc Natl Acad Sci. 2005; 102: 227-231. Ref.: https://goo.gl/rF4cFo
  80. Jama JW, Launer LJ, Witteman JC, den Breeijen JH, Breteler MM, et al. Dietary antioxidants and cognitive function in a population-based sample of older persons. The Rotterdam Study. Am J Epidemiol. 1996; 144: 275-280. Ref.: https://goo.gl/jS9FQi
  81. Sonnen JA, Larson EB, Gray SL, Wilson A, Kohama SG, et al. Free radical damage to cerebral cortex in Alzheimer's disease, microvascular brain injury, and smoking. Ann Neurol. 2009; 65: 226-229. Ref.: https://goo.gl/DDHV1A
  82. Ahlemeyer B, Huhne R, Krieglstein J. Retinoic acid potentiated the protective effect of NGF against staurosporine-induced apoptosis in cultured chick neurons by increasing the trkA protein expression. J Neurosci Res. 2000; 60: 767-778. Ref.: https://goo.gl/FSNvaE
  83. Karasinska JM, Rinninger F, Lutjohann D, Ruddle P, Franciosi S, et al. Specific loss of brain ABCA1 increases brain cholesterol uptake and influences neuronal structure and function. J Neurosci. 2009; 29: 3579-3589. Ref.: https://goo.gl/9XDtn2
  84. Koldamova RP, Lefterov IM, Ikonomovic MD, Skoko J, Lefterov PI, et al. 22R-hydroxycholesterol and 9-cis-retinoic acid induce ATP-binding cassette transporter A1 expression and cholesterol efflux in brain cells and decrease amyloid-β secretion. J Biol Chem. 2003; 278: 13244-13256. Ref.: https://goo.gl/1FK1WR
  85. Satoh J, Kuroda Y. Amyloid precursor protein β-secretase (BACE) mRNA expression in human neural cell lines following induction of neuronal differentiation and exposure to cytokines and growth factors. Neuropathology. 2000; 20: 289-296. Ref.: https://goo.gl/ohftRG
  86. Yang Y, Quitschke WW, Brewer GJ. Up regulation of amyloid precursor protein gene promoter in rat primary hippocampal neurons by phorbol ester, IL-1 and retinoic acid, but not by reactive oxygen species. Molecular Brain Research. 1998; 60: 40-49. Ref.: https://goo.gl/1LQEEa
  87. Wang C, Ward ME, Chen R, Liu K, Tracy TE, et al. Scalable Production of iPSC-Derived Human Neurons to Identify Tau-Lowering Compounds by High-Content Screening. Stem Cell Reports. 2017; 9: 1221-1233. Ref.: https://goo.gl/heqDPE
  88. Guest J, Garg M, Bilgin A, Grant R. Relationship between central and peripheral fatty acids in humans. Lipids Health Dis. 2013; 12: 79. Ref.: https://goo.gl/gzJdnD
  89. Barberger-Gateau P, Samieri C, Féart C, Plourde M. Dietary omega 3 polyunsaturated fatty acids and Alzheimer’s disease: interaction with apolipoprotein E genotype. Curr Alzheimer Res. 2011; 8: 479-491. Ref.: https://goo.gl/3YmKjP
  90. Pan Y, Khalil H, Nicolazzo JA. The Impact of Docosahexaenoic Acid on Alzheimer's Disease: Is There a Role of the Blood-Brain Barrier?. Curr Clin Pharmacol. 2015; 10: 222-241. Ref.: https://goo.gl/4Y7U19
  91. Chakrabarti M, McDonald AJ, Will Reed J, Moss MA, Das BC, et al. Molecular Signaling Mechanisms of Natural and Synthetic Retinoids for Inhibition of Pathogenesis in Alzheimer's Disease. J Alzheimer’s Dis. 2016; 50: 335-352. Ref.: https://goo.gl/CeE2bd

Figures:

Figure 1

Figure 1

Figure 1

Figure 2

Figure 1

Figure 3

Figure 1

Figure 4

Figure 1

Figure 5

Figure 1

Figure 6

Figure 1

Figure 7

Figure 1

Figure 8

Figure 1

Figure 9

Figure 1

Figure 10

Figure 1

Figure 11

Similar Articles

Recently Viewed

Read More

Most Viewed

Read More

Help ?