GSK-3 and Tau: Comparison
Please note this is a comparison between Version 2 by Conner Chen and Version 1 by Carmen Laura Sayas.

Glycogen synthase kinase-3 (GSK-3) is a ubiquitously expressed serine/threonine kinase with a plethora of substrates. As a modulator of several cellular processes, GSK-3 has a central position in cell metabolism and signaling, with important roles both in physiological and pathological conditions. GSK-3 has been associated with a number of human disorders, such as neurodegenerative diseases including Alzheimer’s disease (AD). GSK-3 contributes to the hyperphosphorylation of tau protein, the main component of neurofibrillary tangles (NFTs), one of the hallmarks of AD. GSK-3 is further involved in the regulation of different neuronal processes that are dysregulated during AD pathogenesis, such as the generation of amyloid-β (Aβ) peptide or Aβ-induced cell death, axonal transport, cholinergic function, and adult neurogenesis or synaptic function. In this review, we will summarize recent data about GSK-3 involvement in these processes contributing to AD pathology, mostly focusing on the crucial interplay between GSK-3 and tau protein. We further discuss the current development of potential AD therapies targeting GSK-3 or GSK-3 phosphorylated tau. 

  • GSK-3
  • tau phosphorylation
  • neurodegeneration
  • Alzheimer’s disease
Please wait, diff process is still running!

References

  1. Rylatt, D.B.; Aitken, A.; Bilham, T.; Condon, G.D.; Embi, N.; Cohen, P. Glycogen synthase from rabbit skeletal muscle. Amino acid sequence at the sites phosphorylated by glycogen synthase kinase-3, and extension of the N-terminal sequence containing the site phosphorylated by phosphorylase kinase. Eur. J. Biochem. 1980, 107, 529–537.
  2. Alessi, D.R.; Caudwell, F.B.; Andjelkovic, M.; Hemmings, B.A.; Cohen, P. Molecular basis for the substrate specificity of protein kinase B; comparison with MAPKAP kinase-1 and p70 S6 kinase. FEBS Lett. 1996, 399, 333–338.
  3. Hart, M.J.; de los Santos, R.; Albert, I.N.; Rubinfeld, B.; Polakis, P. Downregulation of beta-catenin by human Axin and its association with the APC tumor suppressor, beta-catenin and GSK3 beta. Curr. Biol. 1998, 8, 573–581.
  4. Cohen, P. The Croonian Lecture 1998. Identification of a protein kinase cascade of major importance in insulin signal transduction. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci 1999, 354, 485–495.
  5. Hanger, D.P.; Hughes, K.; Woodgett, J.R.; Brion, J.P.; Anderton, B.H. Glycogen synthase kinase-3 induces Alzheimer’s disease-like phosphorylation of tau: Generation of paired helical filament epitopes and neuronal localisation of the kinase. Neurosci. Lett. 1992, 147, 58–62.
  6. Lucas, F.R.; Goold, R.G.; Gordon-Weeks, P.R.; Salinas, P.C. Inhibition of GSK-3beta leading to the loss of phosphorylated MAP-1B is an early event in axonal remodelling induced by WNT-7a or lithium. J. Cell Sci. 1998, 111 Pt 1, 1351–1361.
  7. Wittmann, T.; Waterman-Storer, C.M. Spatial regulation of CLASP affinity for microtubules by Rac1 and GSK3beta in migrating epithelial cells. J. Cell Biol. 2005, 169, 929–939.
  8. Ruel, L.; Bourouis, M.; Heitzler, P.; Pantesco, V.; Simpson, P. Drosophila shaggy kinase and rat glycogen synthase kinase-3 have conserved activities and act downstream of Notch. Nature 1993, 362, 557–560.
  9. Itoh, K.; Tang, T.L.; Neel, B.G.; Sokol, S.Y. Specific modulation of ectodermal cell fates in Xenopus embryos by glycogen synthase kinase. Development 1995, 121, 3979–3988.
  10. Kim, L.; Liu, J.; Kimmel, A.R. The novel tyrosine kinase ZAK1 activates GSK3 to direct cell fate specification. Cell 1999, 99, 399–408.
  11. Woodgett, J.R. Molecular cloning and expression of glycogen synthase kinase-3/factor A. EMBO J. 1990, 9, 2431–2438.
  12. Frame, S.; Cohen, P. GSK3 takes centre stage more than 20 years after its discovery. Biochem. J. 2001, 359, 1–16.
  13. Soutar, M.P.M.; Kim, W.-Y.; Williamson, R.; Peggie, M.; Hastie, C.J.; McLauchlan, H.; Snider, W.D.; Gordon-Weeks, P.R.; Sutherland, C. Evidence that glycogen synthase kinase-3 isoforms have distinct substrate preference in the brain. J. Neurochem. 2010, 115, 974–983.
  14. Lee, S.J.; Chung, Y.H.; Joo, K.M.; Lim, H.C.; Jeon, G.S.; Kim, D.; Lee, W.B.; Kim, Y.S.; Cha, C.I. Age-related changes in glycogen synthase kinase 3beta (GSK3beta) immunoreactivity in the central nervous system of rats. Neurosci. Lett. 2006, 409, 134–139.
  15. Yao, H.-B.; Shaw, P.-C.; Wong, C.-C.; Wan, D.C.-C. Expression of glycogen synthase kinase-3 isoforms in mouse tissues and their transcription in the brain. J. Chem. Neuroanat. 2002, 23, 291–297.
  16. Lau, K.F.; Miller, C.C.; Anderton, B.H.; Shaw, P.C. Expression analysis of glycogen synthase kinase-3 in human tissues. J. Pept. Res. 1999, 54, 85–91.
  17. Schaffer, B.; Wiedau-Pazos, M.; Geschwind, D.H. Gene structure and alternative splicing of glycogen synthase kinase 3 beta (GSK-3beta) in neural and non-neural tissues. Gene 2003, 302, 73–81.
  18. Kwok, J.B.J.; Hallupp, M.; Loy, C.T.; Chan, D.K.Y.; Woo, J.; Mellick, G.D.; Buchanan, D.D.; Silburn, P.A.; Halliday, G.M.; Schofield, P.R. GSK3B polymorphisms alter transcription and splicing in Parkinson’s disease. Ann. Neurol. 2005, 58, 829–839.
  19. Mukai, F.; Ishiguro, K.; Sano, Y.; Fujita, S.C. Alternative splicing isoform of tau protein kinase I/glycogen synthase kinase 3beta. J. Neurochem. 2002, 81, 1073–1083.
  20. Wood-Kaczmar, A.; Kraus, M.; Ishiguro, K.; Philpott, K.L.; Gordon-Weeks, P.R. An alternatively spliced form of glycogen synthase kinase-3beta is targeted to growing neurites and growth cones. Mol. Cell. Neurosci. 2009, 42, 184–194.
  21. Sutherland, C.; Leighton, I.A.; Cohen, P. Inactivation of glycogen synthase kinase-3 beta by phosphorylation: New kinase connections in insulin and growth-factor signalling. Biochem. J. 1993, 296 Pt 1, 15–19.
  22. Stambolic, V.; Woodgett, J.R. Mitogen inactivation of glycogen synthase kinase-3 beta in intact cells via serine 9 phosphorylation. Biochem. J. 1994, 303 Pt 3, 701–704.
  23. Svenningsson, P.; Tzavara, E.T.; Carruthers, R.; Rachleff, I.; Wattler, S.; Nehls, M.; McKinzie, D.L.; Fienberg, A.A.; Nomikos, G.G.; Greengard, P. Diverse psychotomimetics act through a common signaling pathway. Science 2003, 302, 1412–1415.
  24. Cole, A.; Frame, S.; Cohen, P. Further evidence that the tyrosine phosphorylation of glycogen synthase kinase-3 (GSK3) in mammalian cells is an autophosphorylation event. Biochem. J. 2004, 377, 249–255.
  25. Lochhead, P.A.; Kinstrie, R.; Sibbet, G.; Rawjee, T.; Morrice, N.; Cleghon, V. A chaperone-dependent GSK3beta transitional intermediate mediates activation-loop autophosphorylation. Mol. Cell 2006, 24, 627–633.
  26. Lesort, M.; Jope, R.S.; Johnson, G.V. Insulin transiently increases tau phosphorylation: Involvement of glycogen synthase kinase-3beta and Fyn tyrosine kinase. J. Neurochem. 1999, 72, 576–584.
  27. Hartigan, J.A.; Xiong, W.C.; Johnson, G.V. Glycogen synthase kinase 3beta is tyrosine phosphorylated by PYK2. Biochem. Biophys. Res. Commun. 2001, 284, 485–489.
  28. Sayas, C.L.; Ariaens, A.; Ponsioen, B.; Moolenaar, W.H. GSK-3 is activated by the tyrosine kinase Pyk2 during LPA1-mediated neurite retraction. Mol. Biol. Cell 2006, 17, 1834–1844.
  29. Lin, C.-F.; Chen, C.-L.; Chiang, C.-W.; Jan, M.-S.; Huang, W.-C.; Lin, Y.-S. GSK-3beta acts downstream of PP2A and the PI 3-kinase-Akt pathway, and upstream of caspase-2 in ceramide-induced mitochondrial apoptosis. J. Cell Sci. 2007, 120, 2935–2943.
  30. Moon, R.T.; Kohn, A.D.; De Ferrari, G.V.; Kaykas, A. WNT and beta-catenin signalling: Diseases and therapies. Nat. Rev. Genet. 2004, 5, 691–701.
  31. Price, M.A.; Kalderon, D. Proteolysis of the Hedgehog signaling effector Cubitus interruptus requires phosphorylation by Glycogen Synthase Kinase 3 and Casein Kinase 1. Cell 2002, 108, 823–835.
  32. Bijur, G.N.; Jope, R.S. Proapoptotic stimuli induce nuclear accumulation of glycogen synthase kinase-3 beta. J. Biol. Chem. 2001, 276, 37436–37442.
  33. Meares, G.P.; Mines, M.A.; Beurel, E.; Eom, T.-Y.; Song, L.; Zmijewska, A.A.; Jope, R.S. Glycogen synthase kinase-3 regulates endoplasmic reticulum (ER) stress-induced CHOP expression in neuronal cells. Exp. Cell Res. 2011, 317, 1621–1628.
  34. Goñi-Oliver, P.; Lucas, J.J.; Avila, J.; Hernández, F. N-terminal cleavage of GSK-3 by calpain: A new form of GSK-3 regulation. J. Biol. Chem. 2007, 282, 22406–22413.
  35. Kandasamy, A.D.; Schulz, R. Glycogen synthase kinase-3beta is activated by matrix metalloproteinase-2 mediated proteolysis in cardiomyoblasts. Cardiovasc. Res. 2009, 83, 698–706.
  36. Sayas, C.L.; Moreno-Flores, M.T.; Avila, J.; Wandosell, F. The neurite retraction induced by lysophosphatidic acid increases Alzheimer’s disease-like Tau phosphorylation. J. Biol. Chem. 1999, 274, 37046–37052.
  37. Eickholt, B.J.; Walsh, F.S.; Doherty, P. An inactive pool of GSK-3 at the leading edge of growth cones is implicated in Semaphorin 3A signaling. J. Cell Biol. 2002, 157, 211–217.
  38. Pérez, M.; Rojo, A.I.; Wandosell, F.; Díaz-Nido, J.; Avila, J. Prion peptide induces neuronal cell death through a pathway involving glycogen synthase kinase 3. Biochem. J. 2003, 372, 129–136.
  39. Takashima, A.; Honda, T.; Yasutake, K.; Michel, G.; Murayama, O.; Murayama, M.; Ishiguro, K.; Yamaguchi, H. Activation of tau protein kinase I/glycogen synthase kinase-3beta by amyloid beta peptide (25–35) enhances phosphorylation of tau in hippocampal neurons. Neurosci. Res. 1998, 31, 317–323.
  40. Fuster-Matanzo, A.; Llorens-Martín, M.; Sirerol-Piquer, M.S.; García-Verdugo, J.M.; Avila, J.; Hernández, F. Dual effects of increased glycogen synthase kinase-3β activity on adult neurogenesis. Hum. Mol. Genet. 2013, 22, 1300–1315.
  41. Garrido, J.J.; Simón, D.; Varea, O.; Wandosell, F. GSK3 alpha and GSK3 beta are necessary for axon formation. FEBS Lett. 2007, 581, 1579–1586.
  42. Jiang, H.; Guo, W.; Liang, X.; Rao, Y. Both the establishment and the maintenance of neuronal polarity require active mechanisms: Critical roles of GSK-3beta and its upstream regulators. Cell 2005, 120, 123–135.
  43. Kim, W.-Y.; Zhou, F.-Q.; Zhou, J.; Yokota, Y.; Wang, Y.-M.; Yoshimura, T.; Kaibuchi, K.; Woodgett, J.R.; Anton, E.S.; Snider, W.D. Essential roles for GSK-3s and GSK-3-primed substrates in neurotrophin-induced and hippocampal axon growth. Neuron 2006, 52, 981–996.
  44. Castaño, Z.; Gordon-Weeks, P.R.; Kypta, R.M. The neuron-specific isoform of glycogen synthase kinase-3beta is required for axon growth. J. Neurochem. 2010, 113, 117–130.
  45. Morgan-Smith, M.; Wu, Y.; Zhu, X.; Pringle, J.; Snider, W.D. GSK-3 signaling in developing cortical neurons is essential for radial migration and dendritic orientation. eLife 2014, 3, e02663.
  46. Lucas, F.R.; Salinas, P.C. WNT-7a induces axonal remodeling and increases synapsin I levels in cerebellar neurons. Dev. Biol. 1997, 192, 31–44.
  47. Kaidanovich-Beilin, O.; Woodgett, J.R. GSK-3: Functional Insights from Cell Biology and Animal Models. Front. Mol. Neurosci. 2011, 4, 40.
  48. Hoeflich, K.P.; Luo, J.; Rubie, E.A.; Tsao, M.S.; Jin, O.; Woodgett, J.R. Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation. Nature 2000, 406, 86–90.
  49. Kerkela, R.; Kockeritz, L.; Macaulay, K.; Zhou, J.; Doble, B.W.; Beahm, C.; Greytak, S.; Woulfe, K.; Trivedi, C.M.; Woodgett, J.R.; et al. Deletion of GSK-3beta in mice leads to hypertrophic cardiomyopathy secondary to cardiomyoblast hyperproliferation. J. Clin. Investig. 2008, 118, 3609–3618.
  50. O’Brien, W.T.; Harper, A.D.; Jové, F.; Woodgett, J.R.; Maretto, S.; Piccolo, S.; Klein, P.S. Glycogen synthase kinase-3beta haploinsufficiency mimics the behavioral and molecular effects of lithium. J. Neurosci. 2004, 24, 6791–6798.
  51. Beaulieu, J.-M.; Zhang, X.; Rodriguiz, R.M.; Sotnikova, T.D.; Cools, M.J.; Wetsel, W.C.; Gainetdinov, R.R.; Caron, M.G. Role of GSK3 beta in behavioral abnormalities induced by serotonin deficiency. Proc. Natl. Acad. Sci. USA 2008, 105, 1333–1338.
  52. Kimura, T.; Yamashita, S.; Nakao, S.; Park, J.-M.; Murayama, M.; Mizoroki, T.; Yoshiike, Y.; Sahara, N.; Takashima, A. GSK-3beta is required for memory reconsolidation in adult brain. PLoS ONE 2008, 3, e3540.
  53. Omata, N.; Chiu, C.-T.; Moya, P.R.; Leng, Y.; Wang, Z.; Hunsberger, J.G.; Leeds, P.; Chuang, D.-M. Lentivirally mediated GSK-3β silencing in the hippocampal dentate gyrus induces antidepressant-like effects in stressed mice. Int. J. Neuropsychopharmacol. 2011, 14, 711–717.
  54. Prickaerts, J.; Moechars, D.; Cryns, K.; Lenaerts, I.; van Craenendonck, H.; Goris, I.; Daneels, G.; Bouwknecht, J.A.; Steckler, T. Transgenic mice overexpressing glycogen synthase kinase 3beta: A putative model of hyperactivity and mania. J. Neurosci. 2006, 26, 9022–9029.
  55. MacAulay, K.; Doble, B.W.; Patel, S.; Hansotia, T.; Sinclair, E.M.; Drucker, D.J.; Nagy, A.; Woodgett, J.R. Glycogen synthase kinase 3alpha-specific regulation of murine hepatic glycogen metabolism. Cell Metab. 2007, 6, 329–337.
  56. Bhattacharjee, R.; Goswami, S.; Dey, S.; Gangoda, M.; Brothag, C.; Eisa, A.; Woodgett, J.; Phiel, C.; Kline, D.; Vijayaraghavan, S. Isoform-specific requirement for GSK3α in sperm for male fertility. Biol. Reprod. 2018, 99, 384–394.
  57. Kaidanovich-Beilin, O.; Lipina, T.V.; Takao, K.; van Eede, M.; Hattori, S.; Laliberté, C.; Khan, M.; Okamoto, K.; Chambers, J.W.; Fletcher, P.J.; et al. Abnormalities in brain structure and behavior in GSK-3alpha mutant mice. Mol. Brain 2009, 2, 35.
  58. Draffin, J.E.; Sánchez-Castillo, C.; Fernández-Rodrigo, A.; Sánchez-Sáez, X.; Ávila, J.; Wagner, F.F.; Esteban, J.A. GSK3α, not GSK3β, drives hippocampal NMDAR-dependent LTD via tau-mediated spine anchoring. EMBO J. 2021, 40, e105513.
  59. Braak, H.; de Vos, R.A.; Jansen, E.N.; Bratzke, H.; Braak, E. Neuropathological hallmarks of Alzheimer’s and Parkinson’s diseases. Prog. Brain Res. 1998, 117, 267–285.
  60. Avila, J.; Lucas, J.J.; Pérez, M.; Hernández, F. Role of Tau Protein in Both Physiological and Pathological Conditions. Physiol. Rev. 2004, 84, 361–384.
  61. Hernández, F.; Gómez de Barreda, E.; Fuster-Matanzo, A.; Lucas, J.J.; Avila, J. GSK3: A possible link between beta amyloid peptide and tau protein. Exp. Neurol. 2010, 223, 322–325.
  62. Terwel, D.; Muyllaert, D.; Dewachter, I.; Borghgraef, P.; Croes, S.; Devijver, H.; Van Leuven, F. Amyloid activates GSK-3beta to aggravate neuronal tauopathy in bigenic mice. Am. J. Pathol. 2008, 172, 786–798.
  63. Takashima, A.; Noguchi, K.; Michel, G.; Mercken, M.; Hoshi, M.; Ishiguro, K.; Imahori, K. Exposure of rat hippocampal neurons to amyloid beta peptide (25–35) induces the inactivation of phosphatidyl inositol-3 kinase and the activation of tau protein kinase I/glycogen synthase kinase-3 beta. Neurosci. Lett. 1996, 203, 33–36.
  64. Takashima, A. GSK-3 is essential in the pathogenesis of Alzheimer’s disease. J. Alzheimer’s Dis. 2006, 9, 309–317.
  65. Cai, Z.; Zhao, Y.; Zhao, B. Roles of glycogen synthase kinase 3 in Alzheimer’s disease. Curr. Alzheimer Res. 2012, 9, 864–879.
  66. Uemura, K.; Kuzuya, A.; Shimozono, Y.; Aoyagi, N.; Ando, K.; Shimohama, S.; Kinoshita, A. GSK3beta activity modifies the localization and function of presenilin 1. J. Biol. Chem. 2007, 282, 15823–15832.
  67. Mines, M.A.; Beurel, E.; Jope, R.S. Regulation of cell survival mechanisms in Alzheimer’s disease by glycogen synthase kinase-3. Int. J. Alzheimer’s Dis. 2011, 2011, 861072.
  68. Fiorentini, A.; Rosi, M.C.; Grossi, C.; Luccarini, I.; Casamenti, F. Lithium improves hippocampal neurogenesis, neuropathology and cognitive functions in APP mutant mice. PLoS ONE 2010, 5, e14382.
  69. Ly, P.T.T.; Wu, Y.; Zou, H.; Wang, R.; Zhou, W.; Kinoshita, A.; Zhang, M.; Yang, Y.; Cai, F.; Woodgett, J.; et al. Inhibition of GSK3β-mediated BACE1 expression reduces Alzheimer-associated phenotypes. J. Clin. Investig. 2013, 123, 224–235.
  70. Toledo, E.M.; Inestrosa, N.C. Activation of Wnt signaling by lithium and rosiglitazone reduced spatial memory impairment and neurodegeneration in brains of an APPswe/PSEN1DeltaE9 mouse model of Alzheimer’s disease. Mol. Psychiatry 2010, 15, 272–285.
  71. Koh, S.-H.; Noh, M.Y.; Kim, S.H. Amyloid-beta-induced neurotoxicity is reduced by inhibition of glycogen synthase kinase-3. Brain Res. 2008, 1188, 254–262.
  72. Serenó, L.; Coma, M.; Rodríguez, M.; Sánchez-Ferrer, P.; Sánchez, M.B.; Gich, I.; Agulló, J.M.; Pérez, M.; Avila, J.; Guardia-Laguarta, C.; et al. A novel GSK-3beta inhibitor reduces Alzheimer’s pathology and rescues neuronal loss in vivo. Neurobiol. Dis. 2009, 35, 359–367.
  73. Lucas, J.J.; Hernández, F.; Gómez-Ramos, P.; Morán, M.A.; Hen, R.; Avila, J. Decreased nuclear beta-catenin, tau hyperphosphorylation and neurodegeneration in GSK-3beta conditional transgenic mice. EMBO J. 2001, 20, 27–39.
  74. Hernández, F.; Borrell, J.; Guaza, C.; Avila, J.; Lucas, J.J. Spatial learning deficit in transgenic mice that conditionally over-express GSK-3beta in the brain but do not form tau filaments. J. Neurochem. 2002, 83, 1529–1533.
  75. Engel, T.; Hernández, F.; Avila, J.; Lucas, J.J. Full reversal of Alzheimer’s disease-like phenotype in a mouse model with conditional overexpression of glycogen synthase kinase-3. J. Neurosci. 2006, 26, 5083–5090.
  76. Gómez de Barreda, E.; Pérez, M.; Gómez Ramos, P.; de Cristobal, J.; Martín-Maestro, P.; Morán, A.; Dawson, H.N.; Vitek, M.P.; Lucas, J.J.; Hernández, F.; et al. Tau-knockout mice show reduced GSK3-induced hippocampal degeneration and learning deficits. Neurobiol. Dis. 2010, 37, 622–629.
  77. Amaral, A.C.; Perez-Nievas, B.G.; Siao Tick Chong, M.; Gonzalez-Martinez, A.; Argente-Escrig, H.; Rubio-Guerra, S.; Commins, C.; Muftu, S.; Eftekharzadeh, B.; Hudry, E.; et al. Isoform-selective decrease of glycogen synthase kinase-3-beta (GSK-3β) reduces synaptic tau phosphorylation, transcellular spreading, and aggregation. iScience 2021, 24, 102058.
  78. Leroy, K.; Yilmaz, Z.; Brion, J.-P. Increased level of active GSK-3beta 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.
  79. Jin, N.; Yin, X.; Yu, D.; Cao, M.; Gong, C.-X.; Iqbal, K.; Ding, F.; Gu, X.; Liu, F. Truncation and activation of GSK-3β by calpain I: A molecular mechanism links to tau hyperphosphorylation in Alzheimer’s disease. Sci. Rep. 2015, 5, 8187.
  80. Ahmad, S.; Orellana, A.; Kohler, I.; Frölich, L.; de Rojas, I.; Gil, S.; Boada, M.; Hernández, I.; Hausner, L.; Bakker, M.H.M.; et al. Association of lysophosphatidic acids with cerebrospinal fluid biomarkers and progression to Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 124.
  81. McLimans, K.E.; Willette, A.A. Autotaxin is Related to Metabolic Dysfunction and Predicts Alzheimer’s Disease Outcomes. J. Alzheimer’s Dis. 2017, 56, 403–413.
  82. Ramesh, S.; Govindarajulu, M.; Suppiramaniam, V.; Moore, T.; Dhanasekaran, M. Autotaxin−Lysophosphatidic Acid Signaling in Alzheimer’s Disease. Int. J. Mol. Sci. 2018, 19, 1827.
  83. Weingarten, M.D.; Lockwood, A.H.; Hwo, S.Y.; Kirschner, M.W. A protein factor essential for microtubule assembly. Proc. Natl. Acad. Sci. USA 1975, 72, 1858–1862.
  84. Witman, G.B.; Cleveland, D.W.; Weingarten, M.D.; Kirschner, M.W. Tubulin requires tau for growth onto microtubule initiating sites. Proc. Natl. Acad. Sci. USA 1976, 73, 4070–4074.
  85. Binder, L.I.; Frankfurter, A.; Rebhun, L.I. The distribution of tau in the mammalian central nervous system. J. Cell Biol. 1985, 101, 1371–1378.
  86. Barbier, P.; Zejneli, O.; Martinho, M.; Lasorsa, A.; Belle, V.; Smet-Nocca, C.; Tsvetkov, P.O.; Devred, F.; Landrieu, I. Role of Tau as a Microtubule-Associated Protein: Structural and Functional Aspects. Front. Aging Neurosci. 2019, 11, 204.
  87. Castle, B.T.; McKibben, K.M.; Rhoades, E.; Odde, D.J. Tau Avoids the GTP Cap at Growing Microtubule Plus-Ends. iScience 2020, 23, 101782.
  88. Qiang, L.; Sun, X.; Austin, T.O.; Muralidharan, H.; Jean, D.C.; Liu, M.; Yu, W.; Baas, P.W. Tau Does Not Stabilize Axonal Microtubules but Rather Enables Them to Have Long Labile Domains. Curr. Biol. 2018, 28, 2181–2189.e4.
  89. Baas, P.W.; Qiang, L. Tau: It’s Not What You Think. Trends Cell Biol. 2019, 29, 452–461.
  90. Lee, G.; Cowan, N.; Kirschner, M. The primary structure and heterogeneity of tau protein from mouse brain. Science 1988, 239, 285–288.
  91. Himmler, A.; Drechsel, D.; Kirschner, M.W.; Martin, D.W.J. Tau consists of a set of proteins with repeated C-terminal microtubule-binding domains and variable N-terminal domains. Mol. Cell. Biol. 1989, 9, 1381–1388.
  92. Goedert, M.; Spillantini, M.G.; Potier, M.C.; Ulrich, J.; Crowther, R.A. 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.
  93. Goedert, M.; Spillantini, M.G.; Jakes, R.; Rutherford, D.; Crowther, R.A. Multiple isoforms of human microtubule-associated protein tau: Sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron 1989, 3, 519–526.
  94. Kosik, K.S.; Orecchio, L.D.; Bakalis, S.; Neve, R.L. Developmentally regulated expression of specific tau sequences. Neuron 1989, 2, 1389–1397.
  95. McMillan, P.; Korvatska, E.; Poorkaj, P.; Evstafjeva, Z.; Robinson, L.; Greenup, L.; Leverenz, J.; Schellenberg, G.D.; D’Souza, I. Tau isoform regulation is region- and cell-specific in mouse brain. J. Comp. Neurol. 2008, 511, 788–803.
  96. Liu, F.; Gong, C.-X. Tau exon 10 alternative splicing and tauopathies. Mol. Neurodegener. 2008, 3, 8.
  97. Nelson, P.T.; Stefansson, K.; Gulcher, J.; Saper, C.B. Molecular evolution of tau protein: Implications for Alzheimer’s disease. J. Neurochem. 1996, 67, 1622–1632.
  98. Hirokawa, N.; Shiomura, Y.; Okabe, S. Tau proteins: The molecular structure and mode of binding on microtubules. J. Cell Biol. 1988, 107, 1449–1459.
  99. 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.
  100. Hernández, F.; Merchán-Rubira, J.; Vallés-Saiz, L.; Rodríguez-Matellán, A.; Avila, J. Differences Between Human and Murine Tau at the N-terminal End. Front. Aging Neurosci. 2020, 12, 11.
  101. Sayas, C.L.; Medina, M.; Cuadros, R.; Ollá, I.; García, E.; Pérez, M.; Ferrer, I.; Hernández, F.; Avila, J. Role of tau N-terminal motif in the secretion of human tau by End Binding proteins. PLoS ONE 2019, 14, e0210864.
  102. Jeganathan, S.; von Bergen, M.; Brutlach, H.; Steinhoff, H.-J.; Mandelkow, E. Global hairpin folding of tau in solution. Biochemistry 2006, 45, 2283–2293.
  103. Lindwall, G.; Cole, R.D. Phosphorylation affects the ability of tau protein to promote microtubule assembly. J. Biol. Chem. 1984, 259, 5301–5305.
  104. Mandelkow, E.M.; Biernat, J.; Drewes, G.; Gustke, N.; Trinczek, B.; Mandelkow, E. Tau domains, phosphorylation, and interactions with microtubules. Neurobiol. Aging 1995, 16, 353–355.
  105. Ramkumar, A.; Jong, B.Y.; Ori-McKenney, K.M. ReMAPping the microtubule landscape: How phosphorylation dictates the activities of microtubule-associated proteins. Dev. Dyn. Off. Publ. Am. Assoc. Anat. 2018, 247, 138–155.
  106. Noble, W.; Hanger, D.P.; Miller, C.C.J.; Lovestone, S. The importance of tau phosphorylation for neurodegenerative diseases. Front. Neurol. 2013, 4, 83.
  107. Grundke-Iqbal, I.; Iqbal, K.; Tung, Y.C.; Quinlan, M.; Wisniewski, H.M.; Binder, L.I. Abnormal phosphorylation of the microtubule-associated protein tau (tau) in Alzheimer cytoskeletal pathology. Proc. Natl. Acad. Sci. USA 1986, 83, 4913–4917.
  108. Hernandez, F.; Lucas, J.J.; Avila, J. GSK3 and tau: Two convergence points in Alzheimer’s disease. J. Alzheimer’s Dis. 2013, 33, S141–S144.
  109. Sun, W.; Qureshi, H.Y.; Cafferty, P.W.; Sobue, K.; Agarwal-Mawal, A.; Neufield, K.D.; Paudel, H.K. Glycogen synthase kinase-3beta is complexed with tau protein in brain microtubules. J. Biol. Chem. 2002, 277, 11933–11940.
  110. Yuan, Z.; Agarwal-Mawal, A.; Paudel, H.K. 14-3-3 binds to and mediates phosphorylation of microtubule-associated tau protein by Ser9-phosphorylated glycogen synthase kinase 3beta in the brain. J. Biol. Chem. 2004, 279, 26105–26114.
  111. Sadik, G.; Tanaka, T.; Kato, K.; Yamamori, H.; Nessa, B.N.; Morihara, T.; Takeda, M. Phosphorylation of tau at Ser214 mediates its interaction with 14-3-3 protein: Implications for the mechanism of tau aggregation. J. Neurochem. 2009, 108, 33–43.
  112. Cuchillo-Ibanez, I.; Seereeram, A.; Byers, H.L.; Leung, K.-Y.; Ward, M.A.; Anderton, B.H.; Hanger, D.P. Phosphorylation of tau regulates its axonal transport by controlling its binding to kinesin. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2008, 22, 3186–3195.
  113. Llorens-Martín, M.; López-Doménech, G.; Soriano, E.; Avila, J. GSK3β is involved in the relief of mitochondria pausing in a Tau-dependent manner. PLoS ONE 2011, 6, e27686.
  114. Ittner, L.M.; Ke, Y.D.; Götz, J. Phosphorylated Tau interacts with c-Jun N-terminal kinase-interacting protein 1 (JIP1) in Alzheimer disease. J. Biol. Chem. 2009, 284, 20909–20916.
  115. LaPointe, N.E.; Morfini, G.; Pigino, G.; Gaisina, I.N.; Kozikowski, A.P.; Binder, L.I.; Brady, S.T. The amino terminus of tau inhibits kinesin-dependent axonal transport: Implications for filament toxicity. J. Neurosci. Res. 2009, 87, 440–451.
  116. Xiong, L.; Duan, L.; Xu, W.; Wang, Z. Nerve growth factor metabolic dysfunction contributes to sevoflurane-induced cholinergic degeneration and cognitive impairments. Brain Res. 2019, 1707, 107–116.
  117. Mellott, T.J.; Huleatt, O.M.; Shade, B.N.; Pender, S.M.; Liu, Y.B.; Slack, B.E.; Blusztajn, J.K. Correction: Perinatal Choline Supplementation Reduces Amyloidosis and Increases Choline Acetyltransferase Expression in the Hippocampus of the APPswePS1dE9 Alzheimer’s Disease Model Mice. PLoS ONE 2017, 12, e0174875.
  118. Zhao, L.; Chu, C.-B.; Li, J.-F.; Yang, Y.-T.; Niu, S.-Q.; Qin, W.; Hao, Y.-G.; Dong, Q.; Guan, R.; Hu, W.-L.; et al. Glycogen synthase kinase-3 reduces acetylcholine level in striatum via disturbing cellular distribution of choline acetyltransferase in cholinergic interneurons in rats. Neuroscience 2013, 255, 203–211.
  119. Wang, Y.; Tian, Q.; Liu, E.-J.; Zhao, L.; Song, J.; Liu, X.-A.; Ren, Q.-G.; Jiang, X.; Zeng, J.; Yang, Y.-T.; et al. Activation of GSK-3 disrupts cholinergic homoeostasis in nucleus basalis of Meynert and frontal cortex of rats. J. Cell. Mol. Med. 2017, 21, 3515–3528.
  120. Hoshi, M.; Takashima, A.; Noguchi, K.; Murayama, M.; Sato, M.; Kondo, S.; Saitoh, Y.; Ishiguro, K.; Hoshino, T.; Imahori, K. Regulation of mitochondrial pyruvate dehydrogenase activity by tau protein kinase I/glycogen synthase kinase 3beta in brain. Proc. Natl. Acad. Sci. USA 1996, 93, 2719–2723.
  121. Hawkes, C.; Jhamandas, J.H.; Kar, S. Selective loss of basal forebrain cholinergic neurons by 192 IgG-saporin is associated with decreased phosphorylation of Ser glycogen synthase kinase-3beta. J. Neurochem. 2005, 95, 263–272.
  122. Massa, F.; Koehl, M.; Wiesner, T.; Grosjean, N.; Revest, J.-M.; Piazza, P.-V.; Abrous, D.N.; Oliet, S.H.R. Conditional reduction of adult neurogenesis impairs bidirectional hippocampal synaptic plasticity. Proc. Natl. Acad. Sci. USA 2011, 108, 6644–6649.
  123. Dupret, D.; Revest, J.-M.; Koehl, M.; Ichas, F.; De Giorgi, F.; Costet, P.; Abrous, D.N.; Piazza, P.V. Spatial relational memory requires hippocampal adult neurogenesis. PLoS ONE 2008, 3, e1959.
  124. Kempermann, G.; Kuhn, H.G.; Gage, F.H. Experience-induced neurogenesis in the senescent dentate gyrus. J. Neurosci. 1998, 18, 3206–3212.
  125. Christian, K.M.; Song, H.; Ming, G. Functions and dysfunctions of adult hippocampal neurogenesis. Annu. Rev. Neurosci. 2014, 37, 243–262.
  126. Moreno-Jiménez, E.P.; Flor-García, M.; Terreros-Roncal, J.; Rábano, A.; Cafini, F.; Pallas-Bazarra, N.; Ávila, J.; Llorens-Martín, M. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 2019, 25, 554–650.
  127. Zang, J.; Liu, Y.; Li, W.; Xiao, D.; Zhang, Y.; Luo, Y.; Liang, W.; Liu, F.; Wei, W. Voluntary exercise increases adult hippocampal neurogenesis by increasing GSK-3β activity in mice. Neuroscience 2017, 354, 122–135.
  128. Morales-Garcia, J.A.; Luna-Medina, R.; Alonso-Gil, S.; Sanz-Sancristobal, M.; Palomo, V.; Gil, C.; Santos, A.; Martinez, A.; Perez-Castillo, A. Glycogen synthase kinase 3 inhibition promotes adult hippocampal neurogenesis in vitro and in vivo. ACS Chem. Neurosci. 2012, 3, 963–971.
  129. Pérez-Domper, P.; Palomo, V.; Gradari, S.; Gil, C.; de Ceballos, M.L.; Martínez, A.; Trejo, J.L. The GSK-3-inhibitor VP2.51 produces antidepressant effects associated with adult hippocampal neurogenesis. Neuropharmacology 2017, 116, 174–187.
  130. Kisoh, K.; Hayashi, H.; Itoh, T.; Asada, M.; Arai, M.; Yuan, B.; Tanonaka, K.; Takagi, N. Involvement of GSK-3β Phosphorylation Through PI3-K/Akt in Cerebral Ischemia-Induced Neurogenesis in Rats. Mol. Neurobiol. 2017, 54, 7917–7927.
  131. Eom, T.-Y.; Jope, R.S. Blocked inhibitory serine-phosphorylation of glycogen synthase kinase-3alpha/beta impairs in vivo neural precursor cell proliferation. Biol. Psychiatry 2009, 66, 494–502.
  132. Sirerol-Piquer, M.; Gomez-Ramos, P.; Hernández, F.; Perez, M.; Morán, M.A.; Fuster-Matanzo, A.; Lucas, J.J.; Avila, J.; García-Verdugo, J.M. GSK3β overexpression induces neuronal death and a depletion of the neurogenic niches in the dentate gyrus. Hippocampus 2011, 21, 910–922.
  133. Llorens-Martín, M.; Fuster-Matanzo, A.; Teixeira, C.M.; Jurado-Arjona, J.; Ulloa, F.; Defelipe, J.; Rábano, A.; Hernández, F.; Soriano, E.; Ávila, J. GSK-3β overexpression causes reversible alterations on postsynaptic densities and dendritic morphology of hippocampal granule neurons in vivo. Mol. Psychiatry 2013, 18, 451–460.
  134. King, M.K.; Pardo, M.; Cheng, Y.; Downey, K.; Jope, R.S.; Beurel, E. Glycogen synthase kinase-3 inhibitors: Rescuers of cognitive impairments. Pharmacol. Ther. 2014, 141, 1–12.
  135. Pallas-Bazarra, N.; Jurado-Arjona, J.; Navarrete, M.; Esteban, J.A.; Hernández, F.; Ávila, J.; Llorens-Martín, M. Novel function of Tau in regulating the effects of external stimuli on adult hippocampal neurogenesis. EMBO J. 2016, 35, 1417–1436.
  136. Dioli, C.; Patrício, P.; Trindade, R.; Pinto, L.G.; Silva, J.M.; Morais, M.; Ferreiro, E.; Borges, S.; Mateus-Pinheiro, A.; Rodrigues, A.J.; et al. Tau-dependent suppression of adult neurogenesis in the stressed hippocampus. Mol. Psychiatry 2017, 22, 1110–1118.
  137. Hanger, D.P.; Anderton, B.H.; Noble, W. Tau phosphorylation: The therapeutic challenge for neurodegenerative disease. Trends Mol. Med. 2009, 15, 112–119.
  138. Bolós, M.; Pallas-Bazarra, N.; Terreros-Roncal, J.; Perea, J.R.; Jurado-Arjona, J.; Ávila, J.; Llorens-Martín, M. Soluble Tau has devastating effects on the structural plasticity of hippocampal granule neurons. Transl. Psychiatry 2017, 7, 1–16.
  139. Musardo, S.; Marcello, E. Synaptic dysfunction in Alzheimer’s disease: From the role of amyloid β-peptide to the α-secretase ADAM10. Eur. J. Pharmacol. 2017, 817, 30–37.
  140. Koffie, R.M.; Hyman, B.T.; Spires-Jones, T.L. Alzheimer’s disease: Synapses gone cold. Mol. Neurodegener. 2011, 6, 63.
  141. Zhu, L.-Q.; Wang, S.-H.; Liu, D.; Yin, Y.-Y.; Tian, Q.; Wang, X.-C.; Wang, Q.; Chen, J.-G.; Wang, J.-Z. Activation of glycogen synthase kinase-3 inhibits long-term potentiation with synapse-associated impairments. J. Neurosci. 2007, 27, 12211–12220.
  142. Zhu, L.-Q.; Liu, D.; Hu, J.; Cheng, J.; Wang, S.-H.; Wang, Q.; Wang, F.; Chen, J.-G.; Wang, J.-Z. GSK-3 beta inhibits presynaptic vesicle exocytosis by phosphorylating P/Q-type calcium channel and interrupting SNARE complex formation. J. Neurosci. 2010, 30, 3624–3633.
  143. Clayton, E.L.; Sue, N.; Smillie, K.J.; O’Leary, T.; Bache, N.; Cheung, G.; Cole, A.R.; Wyllie, D.J.; Sutherland, C.; Robinson, P.J.; et al. Dynamin I phosphorylation by GSK3 controls activity-dependent bulk endocytosis of synaptic vesicles. Nat. Neurosci. 2010, 13, 845–851.
  144. Fan, X.; Zhao, Z.; Wang, D.; Xiao, J. Glycogen synthase kinase-3 as a key regulator of cognitive function. Acta Biochim. Biophys. Sin. 2020, 52, 219–230.
  145. Magee, J.C.; Grienberger, C. Synaptic Plasticity Forms and Functions. Annu. Rev. Neurosci. 2020, 43, 95–117.
  146. Citri, A.; Malenka, R.C. Synaptic plasticity: Multiple forms, functions, and mechanisms. Neuropsychopharmacology 2008, 33, 18–41.
  147. Collingridge, G.L.; Peineau, S.; Howland, J.G.; Wang, Y.T. Long-term depression in the CNS. Nat. Rev. Neurosci. 2010, 11, 459–473.
  148. Peineau, S.; Bradley, C.; Taghibiglou, C.; Doherty, A.; Bortolotto, Z.A.; Wang, Y.T.; Collingridge, G.L. The role of GSK-3 in synaptic plasticity. Br. J. Pharmacol. 2008, 153 (Suppl. S1), S428–S437.
  149. Hooper, C.; Markevich, V.; Plattner, F.; Killick, R.; Schofield, E.; Engel, T.; Hernandez, F.; Anderton, B.; Rosenblum, K.; Bliss, T.; et al. Glycogen synthase kinase-3 inhibition is integral to long-term potentiation. Eur. J. Neurosci. 2007, 25, 81–86.
  150. Peineau, S.; Taghibiglou, C.; Bradley, C.; Wong, T.P.; Liu, L.; Lu, J.; Lo, E.; Wu, D.; Saule, E.; Bouschet, T.; et al. LTP inhibits LTD in the hippocampus via regulation of GSK3beta. Neuron 2007, 53, 703–717.
  151. Vyklicky, V.; Korinek, M.; Smejkalova, T.; Balik, A.; Krausova, B.; Kaniakova, M.; Lichnerova, K.; Cerny, J.; Krusek, J.; Dittert, I.; et al. Structure, function, and pharmacology of NMDA receptor channels. Physiol. Res. 2014, 63, S191–S203.
  152. Chen, P.; Gu, Z.; Liu, W.; Yan, Z. Glycogen synthase kinase 3 regulates N-methyl-D-aspartate receptor channel trafficking and function in cortical neurons. Mol. Pharmacol. 2007, 72, 40–51.
  153. Chater, T.E.; Goda, Y. The role of AMPA receptors in postsynaptic mechanisms of synaptic plasticity. Front. Cell Neurosci. 2014, 8, 401.
  154. Wei, J.; Liu, W.; Yan, Z. Regulation of AMPA receptor trafficking and function by glycogen synthase kinase 3. J. Biol. Chem. 2010, 285, 26369–26376.
  155. Amici, M.; Lee, Y.; Pope, R.J.P.; Bradley, C.A.; Cole, A.; Collingridge, G.L. GSK-3β regulates the synaptic expression of NMDA receptors via phosphorylation of phosphatidylinositol 4 kinase type IIα. Eur. J. Neurosci. 2020.
  156. Robinson, J.W.; Leshchyns’ka, I.; Farghaian, H.; Hughes, W.E.; Sytnyk, V.; Neely, G.G.; Cole, A.R. PI4KIIα phosphorylation by GSK3 directs vesicular trafficking to lysosomes. Biochem. J. 2014, 464, 145–156.
  157. Nelson, C.D.; Kim, M.J.; Hsin, H.; Chen, Y.; Sheng, M. Phosphorylation of threonine-19 of PSD-95 by GSK-3β is required for PSD-95 mobilization and long-term depression. J. Neurosci. 2013, 33, 12122–12135.
  158. Yagishita, S.; Murayama, M.; Ebihara, T.; Maruyama, K.; Takashima, A. Glycogen Synthase Kinase 3β-mediated Phosphorylation in the Most C-terminal Region of Protein Interacting with C Kinase 1 (PICK1) Regulates the Binding of PICK1 to Glutamate Receptor Subunit GluA2. J. Biol. Chem. 2015, 290, 29438–29448.
  159. Daw, M.I.; Chittajallu, R.; Bortolotto, Z.A.; Dev, K.K.; Duprat, F.; Henley, J.M.; Collingridge, G.L.; Isaac, J.T. PDZ proteins interacting with C-terminal GluR2/3 are involved in a PKC-dependent regulation of AMPA receptors at hippocampal synapses. Neuron 2000, 28, 873–886.
  160. Kim, C.H.; Chung, H.J.; Lee, H.K.; Huganir, R.L. Interaction of the AMPA receptor subunit GluR2/3 with PDZ domains regulates hippocampal long-term depression. Proc. Natl. Acad. Sci. USA 2001, 98, 11725–11730.
  161. Du, J.; Wei, Y.; Liu, L.; Wang, Y.; Khairova, R.; Blumenthal, R.; Tragon, T.; Hunsberger, J.G.; Machado-Vieira, R.; Drevets, W.; et al. A kinesin signaling complex mediates the ability of GSK-3beta to affect mood-associated behaviors. Proc. Natl. Acad. Sci. USA 2010, 107, 11573–11578.
  162. Hu, H.; Real, E.; Takamiya, K.; Kang, M.-G.; Ledoux, J.; Huganir, R.L.; Malinow, R. Emotion enhances learning via norepinephrine regulation of AMPA-receptor trafficking. Cell 2007, 131, 160–173.
  163. Kessels, H.W.; Malinow, R. Synaptic AMPA receptor plasticity and behavior. Neuron 2009, 61, 340–350.
  164. Carlezon, W.A.J.; Nestler, E.J. Elevated levels of GluR1 in the midbrain: A trigger for sensitization to drugs of abuse? Trends Neurosci. 2002, 25, 610–615.
  165. Kimura, T.; Whitcomb, D.J.; Jo, J.; Regan, P.; Piers, T.; Heo, S.; Brown, C.; Hashikawa, T.; Murayama, M.; Seok, H.; et al. Microtubule-associated protein tau is essential for long-term depression in the hippocampus. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci. 2014, 369, 20130144.
  166. Van der Jeugd, A.; Ahmed, T.; Burnouf, S.; Belarbi, K.; Hamdame, M.; Grosjean, M.-E.; Humez, S.; Balschun, D.; Blum, D.; Buée, L.; et al. Hippocampal tauopathy in tau transgenic mice coincides with impaired hippocampus-dependent learning and memory, and attenuated late-phase long-term depression of synaptic transmission. Neurobiol. Learn. Mem. 2011, 95, 296–304.
  167. Marciniak, E.; Leboucher, A.; Caron, E.; Ahmed, T.; Tailleux, A.; Dumont, J.; Issad, T.; Gerhardt, E.; Pagesy, P.; Vileno, M.; et al. Tau deletion promotes brain insulin resistance. J. Exp. Med. 2017, 214, 2257–2269.
  168. Yi, J.H.; Brown, C.; Whitehead, G.; Piers, T.; Lee, Y.S.; Perez, C.M.; Regan, P.; Whitcomb, D.J.; Cho, K. Glucocorticoids activate a synapse weakening pathway culminating in tau phosphorylation in the hippocampus. Pharmacol. Res. 2017, 121, 42–51.
  169. Ahmed, T.; Van der Jeugd, A.; Blum, D.; Galas, M.-C.; D’Hooge, R.; Buee, L.; Balschun, D. Cognition and hippocampal synaptic plasticity in mice with a homozygous tau deletion. Neurobiol. Aging 2014, 35, 2474–2478.
  170. Parsons, M.P.; Raymond, L.A. Extrasynaptic NMDA receptor involvement in central nervous system disorders. Neuron 2014, 82, 279–293.
  171. Massey, P.V.; Johnson, B.E.; Moult, P.R.; Auberson, Y.P.; Brown, M.W.; Molnar, E.; Collingridge, G.L.; Bashir, Z.I. Differential roles of NR2A and NR2B-containing NMDA receptors in cortical long-term potentiation and long-term depression. J. Neurosci. 2004, 24, 7821–7828.
  172. Kollen, M.; Dutar, P.; Jouvenceau, A. The magnitude of hippocampal long-term depression depends on the synaptic location of activated NR2-containing N-methyl-D-aspartate receptors. Neuroscience 2008, 154, 1308–1317.
  173. Papouin, T.; Ladépêche, L.; Ruel, J.; Sacchi, S.; Labasque, M.; Hanini, M.; Groc, L.; Pollegioni, L.; Mothet, J.-P.; Oliet, S.H.R. Synaptic and extrasynaptic NMDA receptors are gated by different endogenous coagonists. Cell 2012, 150, 633–646.
  174. Pallas-Bazarra, N.; Draffin, J.; Cuadros, R.; Antonio Esteban, J.; Avila, J. Tau is required for the function of extrasynaptic NMDA receptors. Sci. Rep. 2019, 9, 7452.
  175. Regan, P.; Piers, T.; Yi, J.-H.; Kim, D.-H.; Huh, S.; Park, S.J.; Ryu, J.H.; Whitcomb, D.J.; Cho, K. Tau phosphorylation at serine 396 residue is required for hippocampal LTD. J. Neurosci. 2015, 35, 4804–4812.
  176. Lee, G.; Newman, S.T.; Gard, D.L.; Band, H.; Panchamoorthy, G. Tau interacts with src-family non-receptor tyrosine kinases. J. Cell Sci. 1998, 111 Pt 2, 3167–3177.
  177. Lee, G.; Thangavel, R.; Sharma, V.M.; Litersky, J.M.; Bhaskar, K.; Fang, S.M.; Do, L.H.; Andreadis, A.; Van Hoesen, G.; Ksiezak-Reding, H. Phosphorylation of tau by fyn: Implications for Alzheimer’s disease. J. Neurosci. 2004, 24, 2304–2312.
  178. Ittner, L.M.; Ke, Y.D.; Delerue, F.; Bi, M.; Gladbach, A.; van Eersel, J.; Wölfing, H.; Chieng, B.C.; Christie, M.J.; Napier, I.A.; et al. Dendritic function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell 2010, 142, 387–397.
  179. Braun, N.J.; Yao, K.R.; Alford, P.W.; Liao, D. Mechanical injuries of neurons induce tau mislocalization to dendritic spines and tau-dependent synaptic dysfunction. Proc. Natl. Acad. Sci. USA 2020, 117, 29069–29079.
  180. Zempel, H.; Thies, E.; Mandelkow, E.; Mandelkow, E.-M. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J. Neurosci. 2010, 30, 11938–11950.
  181. Zempel, H.; Mandelkow, E. Lost after translation: Missorting of Tau protein and consequences for Alzheimer disease. Trends Neurosci. 2014, 37, 721–732.
  182. Bejanin, A.; Schonhaut, D.R.; La Joie, R.; Kramer, J.H.; Baker, S.L.; Sosa, N.; Ayakta, N.; Cantwell, A.; Janabi, M.; Lauriola, M.; et al. Tau pathology and neurodegeneration contribute to cognitive impairment in Alzheimer’s disease. Brain 2017, 140, 3286–3300.
  183. Lowe, V.J.; Wiste, H.J.; Senjem, M.L.; Weigand, S.D.; Therneau, T.M.; Boeve, B.F.; Josephs, K.A.; Fang, P.; Pandey, M.K.; Murray, M.E.; et al. Widespread brain tau and its association with ageing, Braak stage and Alzheimer’s dementia. Brain 2018, 141, 271–287.
  184. Alonso, A.C.; Grundke-Iqbal, I.; Iqbal, K. Alzheimer’s disease hyperphosphorylated tau sequesters normal tau into tangles of filaments and disassembles microtubules. Nat. Med. 1996, 2, 783–787.
  185. Rippin, I.; Eldar-Finkelman, H. Mechanisms and Therapeutic Implications of GSK-3 in Treating Neurodegeneration. Cells 2021, 10, 262.
  186. Khan, I.; Tantray, M.A.; Alam, M.S.; Hamid, H. Natural and synthetic bioactive inhibitors of glycogen synthase kinase. Eur. J. Med. Chem. 2017, 125, 464–477.
  187. Rippin, I.; Khazanov, N.; Joseph, S.B.; Kudinov, T.; Berent, E.; Arciniegas Ruiz, S.M.; Marciano, D.; Levy, L.; Gruzman, A.; Senderowitz, H.; et al. Discovery and Design of Novel Small Molecule GSK-3 Inhibitors Targeting the Substrate Binding Site. Int. J. Mol. Sci. 2020, 21, 8709.
  188. Eldar-Finkelman, H.; Martinez, A. GSK-3 Inhibitors: Preclinical and Clinical Focus on CNS. Front. Mol. Neurosci. 2011, 4, 32.
  189. Domínguez, J.M.; Fuertes, A.; Orozco, L.; del Monte-Millán, M.; Delgado, E.; Medina, M. Evidence for irreversible inhibition of glycogen synthase kinase-3β by tideglusib. J. Biol. Chem. 2012, 287, 893–904.
  190. Lovestone, S.; Boada, M.; Dubois, B.; Hüll, M.; Rinne, J.O.; Huppertz, H.-J.; Calero, M.; Andrés, M.V.; Gómez-Carrillo, B.; León, T.; et al. A phase II trial of tideglusib in Alzheimer’s disease. J. Alzheimer’s Dis. 2015, 45, 75–88.
  191. Congdon, E.E.; Sigurdsson, E.M. Tau-targeting therapies for Alzheimer disease. Nat. Rev. Neurol. 2018, 14, 399–415.
  192. Sayas, C.L. Chapter 10—Tau-based therapies for Alzheimer’s disease: Promising novel neuroprotective approaches. In Neuroprotection in Autism, Schizophrenia and Alzheimer’s Disease; Gozes, I., Levine, J., Eds.; Academic Press: Cambridge, MA, USA, 2020; pp. 245–272.
  193. Hoskin, J.L.; Sabbagh, M.N.; Al-Hasan, Y.; Decourt, B. Tau immunotherapies for Alzheimer’s disease. Expert Opin. Investig. Drugs 2019, 28, 545–554.
  194. Boutajangout, A.; Ingadottir, J.; Davies, P.; Sigurdsson, E.M. Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. J. Neurochem. 2011, 118, 658–667.
  195. Liu, W.; Zhao, L.; Blackman, B.; Parmar, M.; Wong, M.Y.; Woo, T.; Yu, F.; Chiuchiolo, M.J.; Sondhi, D.; Kaminsky, S.M.; et al. Vectored Intracerebral Immunization with the Anti-Tau Monoclonal Antibody PHF1 Markedly Reduces Tau Pathology in Mutant Tau Transgenic Mice. J. Neurosci. 2016, 36, 12425–12435.
  196. Chai, X.; Wu, S.; Murray, T.K.; Kinley, R.; Cella, C.V.; Sims, H.; Buckner, N.; Hanmer, J.; Davies, P.; O’Neill, M.J.; et al. Passive immunization with anti-Tau antibodies in two transgenic models: Reduction of Tau pathology and delay of disease progression. J. Biol. Chem. 2011, 286, 34457–34467.
  197. D’Abramo, C.; Acker, C.M.; Jimenez, H.T.; Davies, P. Tau passive immunotherapy in mutant P301L mice: Antibody affinity versus specificity. PLoS ONE 2013, 8, e62402.
  198. Van Kolen, K.; Malia, T.J.; Theunis, C.; Nanjunda, R.; Teplyakov, A.; Ernst, R.; Wu, S.-J.; Luo, J.; Borgers, M.; Vandermeeren, M.; et al. Discovery and Functional Characterization of hPT3, a Humanized Anti-Phospho Tau Selective Monoclonal Antibody. J. Alzheimer’s Dis. 2020, 77, 1397–1416.
  199. Sankaranarayanan, S.; Barten, D.M.; Vana, L.; Devidze, N.; Yang, L.; Cadelina, G.; Hoque, N.; DeCarr, L.; Keenan, S.; Lin, A.; et al. Passive immunization with phospho-tau antibodies reduces tau pathology and functional deficits in two distinct mouse tauopathy models. PLoS ONE 2015, 10, e0125614.
  200. Neddens, J.; Temmel, M.; Flunkert, S.; Kerschbaumer, B.; Hoeller, C.; Loeffler, T.; Niederkofler, V.; Daum, G.; Attems, J.; Hutter-Paier, B. Phosphorylation of different tau sites during progression of Alzheimer’s disease. Acta Neuropathol. Commun. 2018, 6, 52.
  201. Bittar, A.; Bhatt, N.; Kayed, R. Advances and considerations in AD tau-targeted immunotherapy. Neurobiol. Dis. 2020, 134, 104707.
  202. Galpern, W.R.; Mercken, M.; Van Kolen, K.; Timmers, M.; Haeverans, K.; Janssens, L.; Triana-Baltzer, G.; Kolb, H.C.; Jacobs, T.; Nandy, P.; et al. P1-052: A single ascending dose study to evaluate the safety, tolerability, pharmacokinetics, and pharmacodynamics of the anti-phospho-tau antibody jnj-63733657 in healthy subjects. Alzheimer’s Dement. 2019, 15, P252–P253.
  203. Hickman, D.T.; López-Deber, M.P.; Ndao, D.M.; Silva, A.B.; Nand, D.; Pihlgren, M.; Giriens, V.; Madani, R.; St-Pierre, A.; Karastaneva, H.; et al. Sequence-independent control of peptide conformation in liposomal vaccines for targeting protein misfolding diseases. J. Biol. Chem. 2011, 286, 13966–13976.
  204. Theunis, C.; Crespo-Biel, N.; Gafner, V.; Pihlgren, M.; López-Deber, M.P.; Reis, P.; Hickman, D.T.; Adolfsson, O.; Chuard, N.; Ndao, D.M.; et al. Efficacy and safety of a liposome-based vaccine against protein Tau, assessed in tau.P301L mice that model tauopathy. PLoS ONE 2013, 8, e72301.
More
Video Production Service