Pathogenic Tau Protein Species: Promising Therapeutic Targets for Ocular Neurodegenerative Diseases

Abstract

Tau is a microtubule-associated protein, which is highly expressed in the central nervous system as well as ocular neurons and stabilizes microtubule structure. It is a phospho-protein being moderately phosphorylated under physiological conditions but its abnormal hyperphosphorylation or some post-phosphorylation modifications would result in a pathogenic condition, microtubule dissociation, and aggregation. The aggregates can induce neuroinflammation and trigger some pathogenic cascades, leading to neurodegeneration. Taking these together, targeting pathogenic tau employing tau immunotherapy may be a promising therapeutic strategy in fighting with cerebral and ocular neurodegenerative disorders.

Keywords:

Microtubule-associated Protein, Neurodegenerative Disorders, Tau, Ocular Neurons

References
1. Weingarten MD, Lockwood AH, Hwo S-Y, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci 1975;72:1858–1862.

2. Andreadis A. Misregulation of tau alternative splicing in neurodegeneration and dementia. Prog Mol Subcell Biol 2006;44:89–107.

3. LoPresti P, Szuchet S, Papasozomenos SC, Zinkowski RP, Binder LI. Functional implications for the microtubule-associated protein tau: localization in oligodendrocytes. Proc Natl Acad Sci 1995;92:10369–10373.

4. Dehmelt L, Halpain S. The MAP2/Tau family of microtubule-associated proteins. Genome Biol 2004;6:204.

5. Feinstein SC, Wilson L. Inability of tau to properly regulate neuronal microtubule dynamics: a loss-of-function mechanism by which tau might mediate neuronal cell death. Biochim Biophys Acta 2005;1739:268–279.

6. Mandelkow E-M, Mandelkow E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2012;2:a006247.

7. Khatoon S, Grundke-Iqbal I, Iqbal K. Levels of normal and abnormally phosphorylated tau in different cellular and regional compartments of Alzheimer disease and control brains. FEBS Lett 1994;351:80–84.

8. Spires-Jones TL, Stoothoff WH, de Calignon A, Jones PB, Hyman BT. Tau pathophysiology in neurodegeneration: a tangled issue. Trends Neurosci 2009;32:150–159.

9. Shahani N, Brandt R. Functions and malfunctions of the tau proteins. Cell Mol Life Sci 2002;59:1668–1680.

10. Wang Y, Mandelkow E. Tau in physiology and pathology. Nat Rev Neurosci 2016;17:22.

11. Mazzaro N, Barini E, Spillantini MG, Goedert M, Medini P, Gasparini L. Tau- driven neuronal and neurotrophic dysfunction in a mouse model of early tauopathy. J Neurosci 2016;36:2086–2100.

12. Aboelnour A, Van der Spuy J, Powner M, Jeffery G. Primate retinal cones express phosphorylated tau associated with neuronal degeneration yet survive in old age. Exp Eye Res 2017;165:105–108.

13. Gupta N, Fong J, Ang LC, Yucel YH. Retinal tau pathology in human glaucomas. Can J Ophthalmol 2008;43:53–60.

14. Liu F, Gong CX. Tau exon 10 alternative splicing and tauopathies. Mol Neurodegener 2008;3:8.

15. Lee G, Cowan N, Kirschner M. The primary structure and heterogeneity of tau protein from mouse brain. Science 1988;239:285–288.

16. Goedert M, Spillantini 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.

17. Avila J, Jiménez JS, Sayas CL, Bolós M, Zabala JC, Rivas G, et al. Tau structures. Front Aging Neurosci 2016;8:262.

18. Martin L, Latypova X, Wilson CM, Magnaudeix A, Perrin M-L, Yardin C, et al. Tau protein kinases: involvement in Alzheimer’s disease. Ageing Res Rev 2013;12:289–309.

19. Boutajangout A, Wisniewski T. Tau-based therapeutic approaches for Alzheimer’s disease-a mini-review. Gerontology 2014;60:381–385.

20. McMillan P, Korvatska E, Poorkaj P, Evstafjeva Z, Robinson L, Greenup L, et al. Tau isoform regulation is region-and cell-specific in mouse brain. J Comp Neurol 2008;511:788–803.

21. Jeganathan S, von Bergen M, Mandelkow E-M, Mandelkow E. The natively unfolded character of tau and its aggregation to Alzheimer-like paired helical filaments. Biochemistry 2008;47:10526–10539.

22. Mukrasch MD, Bibow S, Korukottu J, Jeganathan S, Biernat J, Griesinger C, et al. Structural polymorphism of 441-residue tau at single residue resolution. PLOS Biol 2009;7:e34.

23. Sillen A, Barbier P, Landrieu I, Lefebvre S, Wieruszeski J-M, Leroy A, et al. NMR investigation of the interaction between the neuronal protein tau and the microtubules. Biochemistry 2007;46:3055–3064.

24. Chen J, Kanai Y, Cowan N, Hirokawa N. Projection domains of MAP2 and tau determine spacings between microtubules in dendrites and axons. Nature 1992;360:674.

25. Liu C, Götz J. Profiling murine tau with 0N, 1N and 2N isoform-specific antibodies in brain and peripheral organs reveals distinct subcellular localization, with the 1N isoform being enriched in the nucleus. PLOS ONE 2013;8:e84849.

26. Brandt R, Léger J, Lee G. Interaction of tau with the neural plasma membrane mediated by tau’s amino-terminal projection domain. J Cell Biol 1995;131:1327–1340.

27. Gauthier-Kemper A, Weissmann C, Golovyashkina N, Sebö-Lemke Z, Drewes G, Gerke V, et al. The frontotemporal dementia mutation R406W blocks tau’s interaction with the membrane in an annexin A2–dependent manner. J Cell Biol 2011;192:647–661.

28. Magnani E, Fan J, Gasparini L, Golding M, Williams M, Schiavo G, et al. Interaction of tau protein with the dynactin complex. EMBO J 2007;26:4546–4554.

29. Liu C, Song X, Nisbet R, Götz J. Co-immunoprecipitation with tau isoform-specific antibodies reveals distinct protein interactions, and highlights a putative role for 2N tau in disease. J Biol Chem 2016;291:8173–8188.

30. Morris M, Maeda S, Vossel K, Mucke L. The many faces of tau. Neuron 2011;70:410–426.

31. Flanagan LA, Cunningham CC, Chen J, Prestwich GD, Kosik KS, Janmey PA. The structure of divalent cationinduced aggregates of PIP2 and their alteration by gelsolin and tau. Biophys J 1997;73:1440–1447.

32. Surridge CD, Burns RG. The difference in the binding of phosphatidylinositol distinguishes MAP2 from MAP2C and Tau. Biochemistry 1994;33:8051–8057.

33. Qi H, Cantrelle Fo-X, Benhelli-Mokrani H, Smet-Nocca C, Buée L, Lippens G, et al. Nuclear magnetic resonance spectroscopy characterization of interaction of Tau with DNA and its regulation by phosphorylation. Biochemistry 2015;54:1525–1533.

34. Wang XS, Wang DL, Zhao J, Qu MH, Zhou XH, He HJ, et al. The proline-rich domain and the microtubule binding domain of protein tau acting as RNA binding domains. Protein Pept Lett 2006;13:679–685.

35. Eidenmüller J, Thomas F, Thorsten M, Madeline P, Sontag E, Brandt R. Phosphorylation-mimicking glutamate clusters in the proline-rich region are sufficient to simulate the functional deficiencies of hyperphosphorylated tau protein. Biochem J 2001;357:759–767.

36. Goode BL, Denis PE, Panda D, Radeke MJ, Miller HP, Wilson L, et al. Functional interactions between the proline-rich and repeat regions of tau enhance microtubule binding and assembly. Mol Biol Cell 1997;8:353–365.

37. He HJ, Wang XS, Pan R, Wang DL, Liu MN, He RQ. The proline-rich domain of tau plays a role in interactions with actin. BMC Cell Biol 2009;10:81.

38. Gong C-X, Liu F, Grundke-Iqbal I, Iqbal K. Post-translational modifications of tau protein in Alzheimer’s disease. J Neural Transm 2005;112:813–838.

39. Martin L, Latypova X, Terro F. Post-translational modifications of tau protein: implications for Alzheimer’s disease. Neurochem Int 2011;58:458–471.

40. Min S-W, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med 2015;21:1154–1162.

41. Xu L, Zheng J, Margittai M, Nussinov R, Ma B. How does hyperphopsphorylation promote tau aggregation and modulate filament structure and stability? ACS Chem Neurosci 2016;7:565–575.

42. Iqbal K, Liu F, Gong C-X. Tau and neurodegenerative disease: the story so far. Nat Rev Neurol 2016;12:15–27.

43. Ballatore C, Lee VM-Y, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci 2007;8:663–672.

44. Zhao H, Chang R, Che H, Wang J, Yang L, Fang W, et al. Hyperphosphorylation of tau protein by calpain regulation in retina of Alzheimer’s disease transgenic mouse. Neurosci Lett 2013;551:12–16.

45. Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci 2015;18:1183–1189.

46. Rodríguez SM, Gallardo AS, Pereyra PG, Macías M, Ordaz B, Ortega FP, et al. Phosphorylation of tau protein correlates with changes in hippocampal theta oscillations and reduces hippocampal excitability in Alzheimer’s model. J Biol Chem 2018;293:8462–8472.

47. Hanger DP, Betts JC, Loviny TL, Blackstock WP, Anderton BH. New phosphorylation sites identified in hyperphosphorylated tau (paired helical filament-tau) from Alzheimer’s disease brain using nanoelectrospray mass spectrometry. J Neurochem 1998;71:2465–2476.

48. Strong M, Yang W, Strong W, Leystra-Lantz C, Jaffe H, Pant H. Tau protein hyperphosphorylation in sporadic ALS with cognitive impairment. Neurology 2006;66:1770–1771.

49. Yang W, Sopper MM, Leystra-Lantz C, Strong MJ. Microtubule-associated tau protein positive neuronal and glial inclusions in ALS. Neurology 2003;61:1766–1773.

50. Yang W, Leystra-Lantz C, Strong MJ. Upregulation of GSK3β expression in frontal and temporal cortex in ALS with cognitive impairment (ALSci). Brain Res 2008;1196:131–139.

51. Yang W, Strong MJ. Widespread neuronal and glial hyperphosphorylated tau deposition in ALS with cognitive impairment. Amyotroph Lateral Scler 2012;13:178–193.

52. Behrouzi R, Liu X, Wu D, Robinson AC, Tanaguchi- Watanabe S, Rollinson S, et al. Pathological tau deposition in Motor Neurone Disease and frontotemporal lobar degeneration associated with TDP-43 proteinopathy. Acta Neuropathol Commun 2016;4:33.

53. Reynolds CH, Betts JC, Blackstock WP, Nebreda AR, Anderton BH. Phosphorylation sites on tau identified by nanoelectrospray mass spectrometry. J Neurochem 2000;74:1587–1595.

54. Gohar M, Yang W, Strong W, Volkening K, Leystra-Lantz C, Strong MJ. Tau phosphorylation at threonine-175 leads to fibril formation and enhanced cell death: implications for amyotrophic lateral sclerosis with cognitive impairment. J Neurochem 2009;108:634–643.

55. Moszczynski AJ, Gohar M, Volkening K, Leystra-Lantz C, Strong W, Strong MJ. Thr 175-phosphorylated tau induces pathologic fibril formation via GSK3β-mediated phosphorylation of Thr 231 in vitro. Neurobiol Aging. 2015;36:1590–1599.

56. Hampel H, Blennow K, Shaw LM, Hoessler YC, Zetterberg H, Trojanowski JQ. Total and phosphorylated tau protein as biological markers of Alzheimer’s disease. Exp Gerontol 2010;45:30–40.

57. Gong C-X, Lidsky T, Wegiel J, Zuck L, Grundke-Iqbal I, Iqbal K. Phosphorylation of microtubule-associated protein tau is regulated by protein phosphatase 2A in mammalian brain implications for neurofibrillary degeneration in Alzheimer’s disease. J Biol Chem 2000;275:5535– 5544.

58. Boban M, Miskic T, Leko MB, Hof PR, Simic G. Human neuroblastoma SH-SY5Y cells treated with okadaic acid express phosphorylated high molecular weight tau immunoreactive protein species. J Neurosci Methods 2018:284265.

59. Abrahamson M, Barrett AJ, Salvesen G, Grubb A. Isolation of six cysteine proteinase inhibitors from human urine. Their physicochemical and enzyme kinetic properties and concentrations in biological fluids. J Biol Chem 1986;261:11282–11289.

60. Deng A, Irizarry MC, Nitsch RM, Growdon JH, Rebeck GW. Elevation of cystatin C in susceptible neurons in Alzheimer’s disease. Am J Pathol 2001;159:1061–1068.

61. Mathews PM, Levy E. Cystatin C in aging and in Alzheimer’s disease. Ageing Res Rev 2016;32:38–50.

62. Duan J, Marcellus KA, Qin X, Wang Y, Paudel HK. Cystatin C promotes tau protein phosphorylation and causes microtubule instability by inhibiting intracellular turnover of GSK3β in neurons. Mol Cell Neurosci 2018; 89:1–8.

63. Bramblett GT, Goedert M, Jakes R, Merrick SE, Trojanowski JQ, Lee VM. Abnormal tau phosphorylation at Ser 396 in Alzheimer’s disease recapitulates development and contributes to reduced microtubule binding. Neuron 1993;10:1089–1099.

64. Pei J-J, Braak E, Braak H, Grundke-Iqbal I, Iqbal K, Winblad B, et al. Distribution of active glycogen synthase kinase 3β (GSK-3β) in brains staged for Alzheimer disease neurofibrillary changes. J Neuropathol Exp Neurol 1999;58:1010–1019.

65. Pei J-J, Tanaka T, Tung Y-C, Braak E, Iqbal K, Grundke- Iqbal I. Distribution, levels, and activity of glycogen synthase kinase-3 in the Alzheimer disease brain. J Neuropathol Exp Neurol 1997;56:70–78.

66. Yamaguchi H, Ishiguro K, Uchida T, Takashima A, Lemere CA, Imahori K. Preferential labeling of Alzheimer neurofibrillary tangles with antisera for tau protein kinase (TPK) I/glycogen synthase kinase-3β and cyclin-dependent kinase 5, a component of TPK II. Acta Neuropathol 1996;92:232–241.

67. Hooper C, Killick R, Lovestone S. The GSK3 hypothesis of Alzheimer’s disease. J Neurochem 2008;104:1433–1439.

68. Leroy K, Yilmaz Z, Brion JP. Increased level of active GSK- 3β in Alzheimer’s disease and accumulation in argyrophilic grains and in neurones at different stages of neurofibrillary degeneration. Neuropathol Appl Neurobiol 2007;33:43–55.

69. Nikolic M, Dudek H, Kwon YT, Ramos YF, Tsai LH. The cdk5/p35 kinase is essential for neurite outgrowth during neuronal differentiation. Genes Dev 1996;10:816–825.

70. Kondo A, Shahpasand K, Mannix R, Qiu J, Moncaster J, Chen C-H, et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature 2015;523:431–436.

71. Ward SM, Himmelstein DS, Lancia JK, Binder LI. Tau oligomers and tau toxicity in neurodegenerative disease. Biochem Soc Trans 2012; 40:667–671.

72. Von Bergen M, Barghorn S, Biernat J, Mandelkow E-M, Mandelkow E. Tau aggregation is driven by a transition from random coil to beta sheet structure. Biochim Biophys Acta 2005;1739:158–166.

73. Ganguly P, Do TD, Larini L, LaPointe NE, Sercel AJ, Shade MF, et al. Tau assembly: the dominant role of PHF6 (VQIVYK) in microtubule-binding region repeat R3. J Phys Chem B 2015;119:4582–4593.

74. von Bergen M, Barghorn S, Li L, Marx A, Biernat J, Mandelkow E-M, et al. Mutations of tau protein in frontotemporal dementia promote aggregation of paired helical filaments by enhancing local β-structure. J Biol Chem 2001;276:48165–48174.

75. Peterson DW, Zhou H, Dahlquist FW, Lew J. A soluble oligomer of tau associated with fiber formation analyzed by NMR. Biochemistry 2008;47:7393–7404.

76. Meraz-Ríos MA, Lira-De León KI, Campos-Peña V, De Anda-Hernández MA, Mena-López R. Tau oligomers and aggregation in Alzheimer’s disease. J Neurochem 2010;112:1353–1367.

77. Alonso AD, Zaidi T, Novak M, Barra HS, Grundke-Iqbal I, Iqbal K. Interaction of tau isoforms with Alzheimer’s disease abnormally hyperphosphorylated tau and in vitro phosphorylation into the disease-like protein. J Biol Chem 2001;276:37967–37973.

78. Hasegawa M, Watanabe S, Kondo H, Akiyama H, Mann DM, Saito Y, et al. 3R and 4R tau isoforms in paired helical filaments in Alzheimer’s disease. Acta Neuropathol 2014;127:303–305.

79. Lewis J, McGowan E, Rockwood J, Melrose H, Nacharaju P, Van Slegtenhorst M, et al. Neurofibrillary tangles, amyotrophy and progressive motor disturbance in mice expressing mutant (P301L) tau protein. Nat Genet 2000;25:402–405.

80. Zhong Q, Congdon EE, Nagaraja HN, Kuret J. Tau isoform composition influences the rate and extent of filament formation. J Biol Chem 2012;287:20711–10719.

81. Kuret J, Chirita CN, Congdon EE, Kannanayakal T, Li G, Necula M, et al. Pathways of tau fibrillization. Biochim Biophys Acta 2005;1739:167–178.

82. Sibille N, Sillen A, Leroy A, Wieruszeski J-M, Mulloy B, Landrieu I, et al. Structural impact of heparin binding to fulllength Tau as studied by NMR spectroscopy. Biochemistry 2006;45:12560–12572.

83. Wilson DM, Binder LI. Free fatty acids stimulate the polymerization of tau and amyloid beta peptides. In vitro evidence for a common effector of pathogenesis in Alzheimer’s disease. Am J Pathol 1997;150:2181.

84. Iqbal K, Grundke-Iqbal I. Ubiquitination and abnormal phosphorylation of paired helical filaments in Alzheimer’s disease. Mol Neurobiol 1991;5:399–410.

85. Neddens J, Temmel M, Flunkert S, Kerschbaumer B, Hoeller C, Loeffler T, et al. Phosphorylation of different tau sites during progression of Alzheimer’s disease. Acta Neuropathol Commun 2018;6:52.

86. Lin YT, Cheng JT, Liang LC, Ko CY, Lo YK, Lu PJ. The binding and phosphorylation of Thr231 is critical for Tau’s hyperphosphorylation and functional regulation by glycogen synthase kinase 3β. J Neurochem 2007;103:802–813.

87. Von Bergen M, Barghorn S, Biernat J, Mandelkow E-M, Mandelkow E. Tau aggregation is driven by a transition from random coil to beta sheet structure. Biochim Biophys Acta 2005;1739:158–166.

88. Kolarova M, García-Sierra F, Bartos A, Ricny J, Ripova D. Structure and pathology of tau protein in Alzheimer disease. Int J Alzheimers Dis 2012;2012:731526.

89. de Calignon A, Polydoro M, Suárez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron 2012;73:685–697.

90. Goedert M. Alzheimer’s and Parkinson’s diseases: The prion concept in relation to assembled Aβ, tau, and α- synuclein. Science 2015;349:1255555.

91. Pooler AM, Polydoro M, Wegmann S, Nicholls SB, Spires- Jones TL, Hyman BT. Propagation of tau pathology in Alzheimer’s disease: identification of novel therapeutic targets. Alzheimers Res Ther 2013;5:49.

92. Mohamed NV, Herrou T, Plouffe V, Piperno N, Leclerc N. Spreading of tau pathology in Alzheimer’s disease by cellto- cell transmission. Eur J Neurosci 2013;37:1939-1948.

93. Wegmann S, Bennett RE, Amaral AS, Hyman BT. Studying tau protein propagation and pathology in the mouse brain using adeno-associated viruses. Methods Cell Biol 2017;141:307–322.

94. Bilkei-Gorzo A. Genetic mouse models of brain ageing and Alzheimer’s disease. Pharmacol Ther 2014;142:244–257.

95. Ojo JO, Mouzon BC, Crawford F. Repetitive head trauma, chronic traumatic encephalopathy and tau: Challenges in translating from mice to men. Exp Neurol 2016;275:389–404.

96. Kanaan NM, Pigino GF, Brady ST, Lazarov O, Binder LI, Morfini GA. Axonal degeneration in Alzheimer’s disease: when signaling abnormalities meet the axonal transport system. Exp Neurol 2013;246:44–53.

97. Hernandez F, Avila J. Tauopathies. Cell Mol Life Sci 2007;64:2219–2233.

98. Lee VM, Goedert M, Trojanowski JQ. Neurodegenerative tauopathies. Annu Rev Neurosci 2001;24:1121–1159.

99. Leyns CE, Holtzman DM. Glial contributions to neurodegeneration in tauopathies. Mol Neurodegener 2017;12:50.

100. Chen JA, Chen Z, Won H, Huang AY, Lowe JK, Wojta K, et al. Joint genome-wide association study of progressive supranuclear palsy identifies novel susceptibility loci and genetic correlation to neurodegenerative diseases. Mol Neurodegener 2018;13:41.

101. Ferrer I, García MA, Carmona M, Andrés-Benito P, Torrejón-Escribano B, Garcia-Esparcia P, et al. Involvement of oligodendrocytes in tau seeding and spreading in tauopathies. Front Aging Neurosci 2019;11:112.

102. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci 2015;18:1584.

103. Johnson VE, Stewart W, Smith DH. Widespread tau and amyloid-beta pathology many years after a single traumatic brain injury in humans. Brain Pathol 2012;22:142– 149.

104. McKee AC, Stein TD, Kiernan PT, Alvarez VE. The neuropathology of chronic traumatic encephalopathy. Brain Pathol 2015;25:350–364.

105. Blennow K, Hardy J, Zetterberg H. The neuropathology and neurobiology of traumatic brain injury. Neuron 2012;76:886–899.

106. Kandimalla R, Manczak M, Fry D, Suneetha Y, Sesaki H, Reddy PH. Reduced dynamin-related protein 1 protects against phosphorylated tau-induced mitochondrial dysfunction and synaptic damage in Alzheimer’s disease. Hum Mol Genet 2016;25:4881–4897.

107. Wang H-H, Li H-L, Liu R, Zhang Y, Liao K, Wang Q, et al. Tau overexpression inhibits cell apoptosis with the mechanisms involving multiple viability-related factors. J Alzheimers Dis 2010;21:167–179.

108. Li H-L, Wang H-H, Liu S-J, Deng Y-Q, Zhang Y-J, Tian Q, et al. Phosphorylation of tau antagonizes apoptosis by stabilizing β-catenin, a mechanism involved in Alzheimer’s neurodegeneration. Proc Natl Acad Sci 2007;104:3591–3596.

109. Chung W-S, Welsh CA, Barres BA, Stevens B. Do glia drive synaptic and cognitive impairment in disease? Nat Neurosci 2015;18:1539.

110. Rosenfeld PJ, Brown DM, Heier JS, Boyer DS, Kaiser PK, Chung CY, et al. Ranibizumab for neovascular age-related macular degeneration. N Engl J Med 2006;355:1419–1431.

111. Moszczynski AJ, Strong W, Xu K, McKee A, Brown A, Strong MJ. Pathologic Thr175 tau phosphorylation in CTE and CTE with ALS. Neurology 2018;90:e380–e387.

112. Bloom GS. Amyloid-β and tau: the trigger and bullet in Alzheimer disease pathogenesis. JAMA Neurol 2014;71:505–508.

113. Lloret A, Badia M-C, Giraldo E, Ermak G, Alonso M-D, Pallardó FV, et al. Amyloid-β toxicity and tau hyperphosphorylation are linked via RCAN1 in Alzheimer’s disease. J Alzheimers Dis 2011;27:701–709.

114. Götz J, Chen FV, Van Dorpe J, Nitsch R. Formation of neurofibrillary tangles in P301L tau transgenic mice induced by Aβ42 fibrils. Science 2001;293:1491–1495.

115. Lewis J, Dickson DW, Lin W-L, Chisholm L, Corral A, Jones G, et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 2001;293:1487–1491.

116. Badiola N, De Oliveira RM, Herrera F, Guardia-Laguarta C, Gonçalves SA, Pera M, et al. Tau enhances α-synuclein aggregation and toxicity in cellular models of synucleinopathy. PLOS ONE 2011;6:e26609.

117. Marui W, Iseki E, Uéda K, Kosaka K. Occurrence of human α-synuclein immunoreactive neurons with neurofibrillary tangle formation in the limbic areas of patients with Alzheimer’s disease. J Neurol Sci 2000;174:81–84.

118. Qureshi HY, Paudel HK. Parkinsonian neurotoxin 1-methyl- 4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) and α- synuclein mutations promote Tau protein phosphorylation at Ser262 and destabilize microtubule cytoskeleton in vitro. J Biol Chem 2011;286:5055–5068.

119. Foster NL, Wilhelmsen K, Sima AA, Jones MZ, D’Amato CJ, Gilman S, et al. Frontotemporal dementia and parkinsonism linked to chromosome 17: a consensus conference. Ann Neurol 1997;41:706–715.

120. Hutton M, Lendon CL, Rizzu P, Baker M, Froelich S, Houlden H, et al. Association of missense and 5′-splicesite mutations in tau with the inherited dementia FTDP-17. Nature 1998;393:702.

121. Varani L, Hasegawa M, Spillantini MG, Smith MJ, Murrell JR, Ghetti B, et al. Structure of tau exon 10 splicing regulatory element RNA and destabilization by mutations of frontotemporal dementia and parkinsonism linked to chromosome 17. Proc Natl Acad Sci 1999;96:8229–8234.

122. Goedert M, Spillantini M, Jakes R, Crowtherp R, Vanmechelen E, Probst A, et al. Molecular dissection of the paired helical filament. Neurobiol Aging 1995;16:325–334.

123. Ingelsson M, Ramasamy K, Cantuti-Castelvetri I, Skoglund L, Matsui T, Orne J, et al. No alteration in tau exon 10 alternative splicing in tangle-bearing neurons of the Alzheimer’s disease brain. Acta Neuropathol 2006;112:439–449.

124. Chambers CB, Lee JM, Troncoso JC, Reich S, Muma NA. Overexpression of four-repeat tau mRNA isoforms in progressive supranuclear palsy but not in Alzheimer’s disease. Ann Neurol 1999;46:325–332.

125. Ingelsson M, Ramasamy K, Russ C, Freeman SH, Orne J, Raju S, et al. Increase in the relative expression of tau with four microtubule binding repeat regions in frontotemporal lobar degeneration and progressive supranuclear palsy brains. Acta Neuropathol 2007;114:471–479.

126. Patterson KR, Remmers C, Fu Y, Brooker S, Kanaan NM, Vana L, et al. Characterization of prefibrillar tau oligomers in vitro and in Alzheimers disease. J Biol Chem 2011;286:23063–23076.

127. Lasagna-Reeves CA, L Castillo-Carranza D, R Jackson G, Kayed R. Tau oligomers as potential targets for immunotherapy for Alzheimer’s disease and tauopathies. Curr Alzheimer Res 2011;8:659–665.

128. Garwood CJ, Cooper JD, Hanger DP, Noble W. Antiinflammatory impact of minocycline in a mouse model of tauopathy. Front Psychiatry. 2010;1:136.

129. Jaworski T, Lechat B, Demedts D, Gielis L, Devijver H, Borghgraef P, et al. Dendritic degeneration, neurovascular defects, and inflammation precede neuronal loss in a mouse model for tau-mediated neurodegeneration. Am J Pathol 2011;179:2001–2015.

130. Nilson AN, English KC, Gerson JE, Barton Whittle T, Nicolas Crain C, Xue J, et al. Tau oligomers associate with inflammation in the brain and retina of tauopathy mice and in neurodegenerative diseases. J Alzheimers Dis 2017;55:1083–1099.

131. Lu Y, Li Z, Zhang X, Ming B, Jia J, Wang R, et al. Retinal nerve fiber layer structure abnormalities in early Alzheimer’s disease: evidence in optical coherence tomography. Neurosci Lett 2010;480:69–72.

132. Ho W-L, Leung Y, Tsang AW-T, So K-F, Chiu K, Chang RCC. Tauopathy in the retina and optic nerve: does it shadow pathological changes in the brain? Mol Vis 2012;18:2700– 2710.

133. Colligris P, Perez de Lara MJ, Colligris B, Pintor J. Ocular manifestations of alzheimer’s and other neurodegenerative diseases: the prospect of the eye as a tool for the early diagnosis of Alzheimer’s disease. J Ophthalmol 2018;2018:8538573.

134. Berisha F, Feke GT, Trempe CL, McMeel JW, Schepens CL. Retinal abnormalities in early Alzheimer’s disease. Invest Ophthalmol Vis Sci 2007;48:2285–2289.

135. Sadun AA, Bassi CJ. Optic nerve damage in Alzheimer’s disease. Ophthalmology 1990;97:9–17.

136. Guo L, Duggan J, Cordeiro MF. Alzheimer’s disease and retinal neurodegeneration. Curr Alzheimer Res 2010;7:3– 14.

137. Danesh-Meyer HV, Birch H, Ku JY, Carroll S, Gamble G. Reduction of optic nerve fibers in patients with Alzheimer disease identified by laser imaging. Neurology 2006;67:1852–1854.

138. Iseri PK, Altinas O, Tokay T, Yuksel N. Relationship between cognitive impairment and retinal morphological and visual functional abnormalities in Alzheimer disease. J Neuroophthalmol 2006;26:18–24.

139. Frost S, Guymer R, Zaw Aung K, Lance Macaulay S, Sohrabi HR, Bourgeat P, et al. Alzheimer’s disease and the early signs of age-related macular degeneration. Curr Alzheimer Res 2016;13:1259–1266.

140. Koronyo-Hamaoui M, Koronyo Y, Ljubimov AV, Miller CA, Ko MK, Black KL, et al. Identification of amyloid plaques in retinas from Alzheimer’s patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model. Neuroimage 2011;54:S204–S217.

141. Loeffler KU, Edward DP, Tso MO. Tau-2 immunoreactivity of corpora amylacea in the human retina and optic nerve. Invest Ophthalmol Vis Sci 1993;34:2600–2603.

142. Loffler KU, Edward DP, Tso MO. Immunoreactivity against tau, amyloid precursor protein, and beta-amyloid in the human retina. Invest Ophthalmol Vis Sci 1995;36:24–31.

143. Flaxman SR, Bourne RR, Resnikoff S, Ackland P, Braithwaite T, Cicinelli MV, et al. Global causes of blindness and distance vision impairment 1990–2020: a systematic review and meta-analysis. Lancet Glob Health 2017;5:e1221–e1234.

144. Luthert PJ. Pathogenesis of age-related macular degeneration. Diagn Histopathol 2011;17:10–16.

145. Leger F, Fernagut PO, Canron MH, Leoni S, Vital C, Tison F, et al. Protein aggregation in the aging retina. J Neuropathol Exp Neurol 2011;70:63–68.

146. Wostyn P, Audenaert K, De Deyn PP. Alzheimer’s disease and glaucoma: is there a causal relationship? Br J Ophthalmol 2009;93:1557–1559.

147. Tseng H, Kasmala L, Proia A, McKinnon S. Expression of protein markers of Alzheimer’s disease in human glaucoma eyes. Invest Ophthalmol Vis Sci 2007;48:3269–3269.

148. Chiasseu M, Cueva Vargas JL, Destroismaisons L, Vande Velde C, Leclerc N, Di Polo A. Tau accumulation, altered phosphorylation, and missorting promote neurodegeneration in glaucoma. J Neurosci 2016;36:5785–5798.

149. Oka T, Tamada Y, Nakajima E, Shearer TR, Azuma M. Presence of calpain-induced proteolysis in retinal degeneration and dysfunction in a rat model of acute ocular hypertension. J Neurosci Res 2006;83:1342–1351.

150. Branca D. Calpain-related diseases. Biochem Biophys Res Commun 2004;322:1098–1104.

151. Yoneda S, Hara H, Hirata A, Fukushima M, Inomata Y, Tanihara H. Vitreous fluid levels of β-amyloid (1–42) and tau in patients with retinal diseases. Jpn J Ophthalmol 2005;49:106–108.

152. Goedert M, Jakes R, Vanmechelen E. Monoclonal antibody AT8 recognises tau protein phosphorylated at both serine 202 and threonine 205. Neurosci Lett 1995;189:167–169.

153. Gupta N, Fong J, Ang LC, Yucel YH. Retinal tau pathology in human glaucomas. Can J Ophthalmol 2008;43:53–60.

154. Liu B, Rasool S, Yang Z, Glabe CG, Schreiber SS, Ge J, et al. Amyloid-peptide vaccinations reduce β-amyloid plaques but exacerbate vascular deposition and inflammation in the retina of Alzheimer’s transgenic mice. Am J Pathol 2009;175:2099–2110.

155. Linsenmeier RA, Padnick–Silver L. Metabolic dependence of photoreceptors on the choroid in the normal and detached retina. Invest Ophthalmol Vis Sci 2000;41:3117–3123.

156. Calaza KC, Kam JH, Hogg C, Jeffery G. Mitochondrial decline precedes phenotype development in the complement factor H mouse model of retinal degeneration but can be corrected by near infrared light. Neurobiol Aging 2015;36:2869–2876.

157. Nakayama T, Goshima Y, Misu Y, Kato T. Role of Cdk5 and Tau phosphorylation in heterotrimeric G protein–mediated retinal growth cone collapse. J Neurobiol 1999;41:326–339.

158. Allen B, Ingram E, Takao M, Smith MJ, Jakes R, Virdee K, et al. Abundant tau filaments and nonapoptotic neurodegeneration in transgenic mice expressing human P301S tau protein. J Neurosci 2002;22:9340–9351.

159. Gasparini L, Crowther RA, Martin KR, Berg N, Coleman M, Goedert M, et al. Tau inclusions in retinal ganglion cells of human P301S tau transgenic mice: effects on axonal viability. Neurobiol Aging 2011;32:419–433.

160. Mazzaro N, Barini E, Spillantini MG, Goedert M, Medini P, Gasparini L. Tau-Driven neuronal and neurotrophic dysfunction in a mouse model of early tauopathy. J Neurosci 2016;36:2086–2100.

161. Bull ND, Guidi A, Goedert M, Martin KR, Spillantini MG. Reduced axonal transport and increased excitotoxic retinal ganglion cell degeneration in mice transgenic for human mutant P301S tau. PLoS One 2012;7:e34724.

162. Spires TL, Hyman BT. Transgenic models of Alzheimer’s disease: learning from animals. NeuroRx 2005;2:423–437.

163. Ho WL, Leung Y, Cheng SS, Lok CK, Ho YS, Baum L, et al. Investigating degeneration of the retina in young and aged tau P301L mice. Life Sci 2015;124:16–23.

164. Schroer TA. Dynactin. Annu Rev Cell Dev Biol 2004;20:759–779.

165. Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol 2002;156:1051–1063.

166. Cingolani C, Rogers B, Lu L, Kachi S, Shen J, Campochiaro PA. Retinal degeneration from oxidative damage. Free Radic Biol Med 2006;40:660–669.

167. Tanito M, Masutani H, Nakamura H, Oka S-i, Ohira A, Yodoi J. Attenuation of retinal photooxidative damage in thioredoxin transgenic mice. Neurosci Lett 2002;326:142– 146.

168. Léveillard T, Mohand-Saïd S, Lorentz O, Hicks D, Fintz AC, Clérin E, et al. Identification and characterization of rodderived cone viability factor. Nat Genet 2004;36:755.

169. Sahel J-A. Saving cone cells in hereditary rod diseases: a possible role for rod-derived cone viability factor (RdCVF) therapy. Retina 2005;25:S38–S39.

170. Leveillard T, Sahel JA. Rod-derived cone viability factor for treating blinding diseases: from clinic to redox signaling. Sci Transl Med 2010;2:26ps16.

171. Fridlich R, Delalande F, Jaillard C, Lu J, Poidevin L, Cronin T, et al. The thioredoxin-like protein rod-derived cone viability factor (RdCVFL) interacts with TAU and inhibits its phosphorylation in the retina. Mol Cell Proteomics 2009;8:1206–1218.

172. Byrne LC, Dalkara D, Luna G, Fisher SK, Clérin E, Sahel JA, et al. Viral-mediated RdCVF and RdCVFL expression protects cone and rod photoreceptors in retinal degeneration. J Clin Invest 2015;125:105–116.

173. Hosokawa M, Arai T, Masuda-Suzukake M, Nonaka T, Yamashita M, Akiyama H, et al. Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice. PLOS ONE 2012;7:e52389.

174. Min S-W, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med 2015;21:1154–1162.

175. Myeku N, Clelland CL, Emrani S, Kukushkin NV, Yu WH, Goldberg AL, et al. Tau-driven 26S proteasome impairment and cognitive dysfunction can be prevented early in disease by activating cAMP-PKA signaling. Nat Med 2016;22:46–53.

176. Yanamandra K, Kfoury N, Jiang H, Mahan TE, Ma S, Maloney SE, et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 2013;80:402–414.

177. Castillo-Carranza DL, Sengupta U, Guerrero-Munoz MJ, Lasagna-Reeves CA, Gerson JE, Singh G, et al. Passive immunization with Tau oligomer monoclonal anti-hyperphosphorylated neurofibrillary tangles. J Neurosci 2014;34:4260–4272.

178. Bi M, Ittner A, Ke YD, Götz J, Ittner LM. Tau-targeted immunization impedes progression of neurofibrillary histopathology in aged P301L tau transgenic mice. PLOS ONE 2011;6:e26860.

179. Boimel M, Grigoriadis N, Lourbopoulos A, Haber E, Abramsky O, Rosenmann H. Efficacy and safety of immunization with phosphorylated tau against neurofibrillary tangles in mice. Exp Neurol 2010;224:472–485.

180. Ittner A, Bertz J, Suh LS, Stevens CH, Götz J, Ittner LM. Tau-targeting passive immunization modulates aspects of pathology in tau transgenic mice. J Neurochem 2015;132:135–145.

181. Brunden KR, Ballatore C, Crowe A, Smith AB, Lee VM-Y, Trojanowski JQ. Tau-directed drug discovery for Alzheimer’s disease and related tauopathies: a focus on tau assembly inhibitors. Exp Neurol 2010;223:304–310.

182. Mohamed T, Hoang T, Jelokhani-Niaraki M, Rao PP. Tau-derived-hexapeptide 306VQIVYK311 aggregation inhibitors: nitrocatechol moiety as a pharmacophore in drug design. ACS Chem Neurosci 2013;4:1559–1570.

183. Mohamed T, PN Rao P. Alzheimer’s disease: emerging trends in small molecule therapies. Curr Med Chem 2011;18:4299–4320.

184. Dammers C, Yolcu D, Kukuk L, Willbold D, Pickhardt M, Mandelkow E, et al. Selection and characterization of tau binding D-enantiomeric peptides with potential for therapy of Alzheimer disease. PLOS ONE 2016;11:e0167432.

185. Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-Fukae C, Li H-F, et al. A neuronal microtubule-interacting agent, NAPVSIPQ, reduces tau pathology and enhances cognitive function in a mouse model of Alzheimer’s disease. J Pharmacol Exp Ther 2008;325:146–153.

186. Ivashko-Pachima Y, Maor-Nof M, Gozes I. NAP (davunetide) preferential interaction with dynamic 3-repeat Tau explains differential protection in selected tauopathies. PLOS ONE 2019;14:e0213666.

187. Eschmann NA, Do TD, LaPointe NE, Shea J-E, Feinstein SC, Bowers MT, et al. Tau aggregation propensity engrained in its solution state. J Phys Chem B 2015;119:14421–14432.

188. Schafer KN, Cisek K, Huseby CJ, Chang E, Kuret J. Structural determinants of tau aggregation inhibitor potency. J Biol Chem 2013;288:32599–32611.

189. Levine ZA, Larini L, LaPointe NE, Feinstein SC, Shea JE. Regulation and aggregation of intrinsically disordered peptides. Proc Natl Acad Sci 2015;112:2758–2763.

190. Nadimidla K, Ismail T, Kanapathipillai M. Tau peptides and tau mutant protein aggregation inhibition by cationic polyethyleneimine and polyarginine. Biopolymers 2017;107:e23024.

191. DeVos SL, Goncharoff DK, Chen G, Kebodeaux CS, Yamada K, Stewart FR, et al. Antisense reduction of tau in adult mice protects against seizures. J Neurosci 2013;33:12887–12897.

192. Khosravi Z, Khalili MAN, Moradi S, Sajedi RH, Zeinoddini M. The molecular chaperone artemin efficiently blocks fibrillization of TAU protein in vitro. Cell J 2018;19:569–577.