Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Tau in physiology and pathology

Key Points

  • Tauopathies are classified into several groups based on the isoform composition of tau aggregates. The splicing of the gene encoding tau is regulated by multiple factors besides tau mutations.

  • Tau is a natively unfolded protein that shows no tendency for aggregation by itself. Post-translational modifications may modify the processes of tau oligomerization, aggregation and tau-induced neurodegeneration.

  • The polarized distribution of tau into the axonal compartment of neurons is determined by multiple mechanisms. In addition, part of tau is actively released into extracellular space.

  • Besides stabilizing microtubules, regulating their dynamic instability and supporting axonal transport, tau can interact with various cell components and thus serves other functions in various other processes, including neuronal activity, neurogenesis, iron export and long-term depression.

  • Pathological tau may induce neurotoxicity owing to its loss of function, toxic gain of function or its mislocalization, which mediates amyloid-β-induced toxicity.

  • Several tau- or microtubule-based therapeutic approaches have been proposed, including tau aggregation inhibitors, inhibitors of kinases targeting tau, inhibition of tau acetylation, stabilization of microtubules, reduction of tau by antisense oligonucleotides, and immunotherapy using antibodies against tau or phosphorylated tau.

Abstract

Tau is a microtubule-associated protein that has a role in stabilizing neuronal microtubules and thus in promoting axonal outgrowth. Structurally, tau is a natively unfolded protein, is highly soluble and shows little tendency for aggregation. However, tau aggregation is characteristic of several neurodegenerative diseases known as tauopathies. The mechanisms underlying tau pathology and tau-mediated neurodegeneration are debated, but considerable progress has been made in the field of tau research in recent years, including the identification of new physiological roles for tau in the brain. Here, we review the expression, post-translational modifications and functions of tau in physiology and in pathophysiology.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The human MAPT gene and the splice isoforms of tau in the human brain.
Figure 2: Tau structure, domains, mutations and pathological phosphorylation sites.
Figure 3: Functions of tau.

Similar content being viewed by others

References

  1. Weingarten, M. D., Lockwood, A. H., Hwo, S. Y. & Kirschner, M. W. A protein factor essential for microtubule assembly. Proc. Natl Acad. Sci. USA 72, 1858–1862 (1975). This study led to the discovery of tau protein as a microtubule-associated protein.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kidd, M. Paired helical filaments in electron microscopy of Alzheimer's disease. Nature 197, 192–193 (1963). First observation and naming of 'paired helical filaments' in the brains of patients with AD.

    Article  CAS  PubMed  Google Scholar 

  3. Alzheimer, A. Über eine eigenartige Erkrankung der Hirnrinde. Allg. Z. Psychiatrie Psychisch-gerichtl. Med. 64, 146–148 (in German) (1907). Original description of the pathology of 'Alzheimer disease'.

    Google Scholar 

  4. Lee, V. M., Goedert, M. & Trojanowski, J. Q. Neurodegenerative tauopathies. Annu. Rev. Neurosci. 24, 1121–1159 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Hutton, M. et al. Association of missense and 5′-splice-site mutations in tau with the inherited dementia FTDP-17. Nature 393, 702–705 (1998). This study identifies MAPT mutations in the human brain and demonstrates that tau pathology is sufficient to cause neurodegeneration.

    Article  PubMed  CAS  Google Scholar 

  6. Roberson, E. D. et al. Reducing endogenous tau ameliorates amyloid β-induced deficits in an Alzheimer's disease mouse model. Science 316, 750–754 (2007). This study demonstrates in a mouse model that tau is necessary for Aβ-induced excitotoxicity.

    Article  CAS  PubMed  Google Scholar 

  7. Holmes, B. B. & Diamond, M. I. Prion-like properties of Tau protein: the importance of extracellular Tau as a therapeutic target. J. Biol. Chem. 289, 19855–19861 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Yoshiyama, Y., Lee, V. M. & Trojanowski, J. Q. Therapeutic strategies for tau mediated neurodegeneration. J. Neurol. Neurosurg. Psychiatry 84, 784–795 (2013).

    Article  PubMed  Google Scholar 

  9. Brettschneider, J., Del Tredici, K., Lee, V. M. & Trojanowski, J. Q. Spreading of pathology in neurodegenerative diseases: a focus on human studies. Nat. Rev. Neurosci. 16, 109–120 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Andreadis, A. Misregulation of tau alternative splicing in neurodegeneration and dementia. Prog. Mol. Subcell. Biol. 44, 89–107 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. LoPresti, P., Szuchet, S., Papasozomenos, S. C., Zinkowski, R. P. & Binder, L. I. Functional implications for the microtubule-associated protein tau: localization in oligodendrocytes. Proc. Natl Acad. Sci. USA 92, 10369–10373 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, G., Cowan, N. & Kirschner, M. The primary structure and heterogeneity of tau protein from mouse brain. Science 239, 285–288 (1988). This was the first study to sequence tau protein, highlighting its hydrophilic nature and the presence of pseudorepeats.

    Article  CAS  PubMed  Google Scholar 

  13. Dickson, D. W., Kouri, N., Murray, M. E. & Josephs, K. A. Neuropathology of frontotemporal lobar degeneration-tau (FTLD-tau). J. Mol. Neurosci. 45, 384–389 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Orozco, D. et al. Loss of fused in sarcoma (FUS) promotes pathological Tau splicing. EMBO Rep. 13, 759–764 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Smith, P. Y. et al. MicroRNA-132 loss is associated with tau exon 10 inclusion in progressive supranuclear palsy. Hum. Mol. Genet. 20, 4016–4024 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Santa-Maria, I. et al. Dysregulation of microRNA-219 promotes neurodegeneration through post-transcriptional regulation of tau. J. Clin. Invest. 125, 681–686 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Moschner, K. et al. RNA protein granules modulate tau isoform expression and induce neuronal sprouting. J. Biol. Chem. 289, 16814–16825 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bruch, J., Xu, H., De Andrade, A. & Hoglinger, G. Mitochondrial complex 1 inhibition increases 4-repeat isoform tau by SRSF2 upregulation. PLoS ONE 9, e113070 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kampers, T., Friedhoff, P., Biernat, J., Mandelkow, E. M. & Mandelkow, E. RNA stimulates aggregation of microtubule-associated protein tau into Alzheimer-like paired helical filaments. FEBS Lett. 399, 344–349 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. Violet, M. et al. A major role for Tau in neuronal DNA and RNA protection in vivo under physiological and hyperthermic conditions. Front. Cell Neurosci. 8, 84 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Goedert, M. & Jakes, R. Expression of separate isoforms of human tau protein: correlation with the tau pattern in brain and effects on tubulin polymerization. EMBO J. 9, 4225–4230 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chen, J., Kanai, Y., Cowan, N. J. & Hirokawa, N. Projection domains of MAP2 and tau determine spacings between microtubules in dendrites and axons. Nature 360, 674–677 (1992).

    Article  CAS  PubMed  Google Scholar 

  23. Frappier, T. F., Georgieff, I. S., Brown, K. & Shelanski, M. L. τ regulation of microtubule-microtubule spacing and bundling. J. Neurochem. 63, 2288–2294 (1994).

    Article  CAS  PubMed  Google Scholar 

  24. Liu, C. & Gotz, 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 8, e84849 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zhong, Q., Congdon, E. E., Nagaraja, H. N. & Kuret, J. Tau isoform composition influences rate and extent of filament formation. J. Biol. Chem. 287, 20711–20719 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kosik, K. S., Orecchio, L. D., Bakalis, S. & Neve, R. L. Developmentally regulated expression of specific tau sequences. Neuron 2, 1389–1397 (1989).

    Article  CAS  PubMed  Google Scholar 

  27. Takuma, H., Arawaka, S. & Mori, H. Isoforms changes of tau protein during development in various species. Brain Res. Dev. Brain Res. 142, 121–127 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Llorens-Martin, M. et al. Tau isoform with three microtubule binding domains is a marker of new axons generated from the subgranular zone in the hippocampal dentate gyrus: implications for Alzheimer's disease. J. Alzheimers Dis. 29, 921–930 (2012).

    Article  CAS  PubMed  Google Scholar 

  29. Goedert, M., Wischik, C. M., Crowther, R. A., Walker, J. E. & Klug, A. Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau. Proc. Natl Acad. Sci. USA 85, 4051–4055 (1988). This paper reveals the sequence of human tau protein and its presence in the core of tau filaments.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Adams, S. J. et al. Overexpression of wild-type murine tau results in progressive tauopathy and neurodegeneration. Am. J. Pathol. 175, 1598–1609 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Lee, G., Newman, S. T., Gard, D. L., Band, H. & Panchamoorthy, G. Tau interacts with src-family non-receptor tyrosine kinases. J. Cell Sci. 111, 3167–3177 (1998).

    CAS  PubMed  Google Scholar 

  32. Ittner, L. M. et al. Dendritic function of tau mediates amyloid-β toxicity in Alzheimer's disease mouse models. Cell 142, 387–397 (2010). This paper shows that dendritic tau plays a part in mediating Aβ-induced hyperexcitotoxicity, by regulating the synaptic distribution of FYN.

    Article  CAS  PubMed  Google Scholar 

  33. Brion, J. P., Passareiro, H., Nunez, J. & Flament-Durand, J. Mise en evidence immunologique de la proteine tau au nivea u des lesions de degenerescence neurofibrillaire de la maladie d'Alzheimer. Arch. Biol. (Bruxelles) 95, 229–235 (in French) (1985). This study provided the first evidence that NFTs in the AD brain contain tau protein.

    Google Scholar 

  34. Grundke-Iqbal, I. et al. Abnormal phosphorylation of the microtubule-associated protein τ (tau) in Alzheimer cytoskeletal pathology. Proc. Natl Acad. Sci. USA 83, 4913–4917 (1986). This study demonstrates that tau protein is present in AD NFTs in a hyperphosphorylated state.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Mukrasch, M. D. et al. Structural polymorphism of 441-residue tau at single residue resolution. PLoS Biol. 7, e34 (2009). A study of full-length tau by solution nuclear magnetic resonance, demonstrating that the majority of the polypeptide chain is in a flexible random-coil conformation, with very few transient secondary-structure elements.

    Article  CAS  PubMed  Google Scholar 

  36. Kadavath, H. et al. Tau stabilizes microtubules by binding at the interface between tubulin heterodimers. Proc. Natl Acad. Sci. USA 112, 7501–7506 (2015). This paper found that tau binds at the interface between α-tubulin–β-tubulin heterodimers through residues including the motifs that are essential for the pathological aggregation of tau, suggesting that there is competition between physiological interaction and pathogenic misfolding.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Gruning, C. S. et al. Alternative conformations of the Tau repeat domain in complex with an engineered binding protein. J. Biol. Chem. 289, 23209–23218 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Jeganathan, S., von Bergen, M., Brutlach, H., Steinhoff, H. J. & Mandelkow, E. Global hairpin folding of tau in solution. Biochemistry 45, 2283–2293 (2006).

    Article  CAS  PubMed  Google Scholar 

  39. Wischik, C. M. et al. Isolation of a fragment of tau derived from the core of the paired helical filament of Alzheimer disease. Proc. Natl Acad. Sci. USA 85, 4506–4510 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wegmann, S., Medalsy, I. D., Mandelkow, E. & Muller, D. J. The fuzzy coat of pathological human Tau fibrils is a two-layered polyelectrolyte brush. Proc. Natl Acad. Sci. USA 110, E313–E321 (2013).

    Article  PubMed  Google Scholar 

  41. Kanemaru, K., Takio, K., Miura, R., Titani, K. & Ihara, Y. Fetal-type phosphorylation of the tau in paired helical filaments. J. Neurochem. 58, 1667–1675 (1992).

    Article  CAS  PubMed  Google Scholar 

  42. Köpke, E. et al. Microtubule-associated protein tau. Abnormal phosphorylation of a non-paired helical filament pool in Alzheimer disease. J. Biol. Chem. 268, 24374–24384 (1993).

    PubMed  Google Scholar 

  43. Matsuo, E. S. et al. Biopsy-derived adult human brain tau is phosphorylated at many of the same sites as Alzheimer's disease paired helical filament tau. Neuron 13, 989–1002 (1994). This paper reveals that the post-mortem delay can cause the dephosphorylation of tau by phosphatases, leading to underestimation of the phosphorylation level of tau in vivo , and shows that tau is physiologically phosphorylated at many of the same sites that are phosphorylated in PHF-tau.

    Article  CAS  PubMed  Google Scholar 

  44. Morris, M. et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat. Neurosci. 18, 1183–1189 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hanger, D. P., Anderton, B. H. & Noble, W. Tau phosphorylation: the therapeutic challenge for neurodegenerative disease. Trends Mol. Med. 15, 112–119 (2009).

    Article  CAS  PubMed  Google Scholar 

  46. Rosseels, J. et al. Tau monoclonal antibody generation based on humanized yeast models: impact on Tau oligomerization and diagnostics. J. Biol. Chem. 290, 4059–4074 (2015).

    Article  CAS  PubMed  Google Scholar 

  47. Vega, I. E. et al. Increase in tau tyrosine phosphorylation correlates with the formation of tau aggregates. Brain Res. Mol. Brain Res. 138, 135–144 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gong, C. X., Singh, T. J., Grundke-Iqbal, I. & Iqbal, K. Phosphoprotein phosphatase activities in Alzheimer disease brain. J. Neurochem. 61, 921–927 (1993).

    Article  CAS  PubMed  Google Scholar 

  49. Chen, S., Li, B., Grundke-Iqbal, I. & Iqbal, K. IPP2A1 affects tau phosphorylation via association with the catalytic subunit of protein phosphatase 2A. J. Biol. Chem. 283, 10513–10521 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Sontag, E. et al. Downregulation of protein phosphatase 2A carboxyl methylation and methyltransferase may contribute to Alzheimer disease pathogenesis. J. Neuropathol. Exp. Neurol. 63, 1080–1091 (2004).

    Article  CAS  PubMed  Google Scholar 

  51. Planel, E. et al. Alterations in glucose metabolism induce hypothermia leading to tau hyperphosphorylation through differential inhibition of kinase and phosphatase activities: implications for Alzheimer's disease. J. Neurosci. 24, 2401–2411 (2004).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  52. Arendt, T. et al. Reversible paired helical filament-like phosphorylation of tau is an adaptive process associated with neuronal plasticity in hibernating animals. J. Neurosci. 23, 6972–6981 (2003). The authors found that tau hyperphosphorylation occurs physiologically and reversibly in hibernating animals without forming tau aggregates.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  53. Planel, E. et al. Anesthesia leads to tau hyperphosphorylation through inhibition of phosphatase activity by hypothermia. J. Neurosci. 27, 3090–3097 (2007). This paper shows that anaesthesia can cause tau hyperphosphorylation owing to inhibition of phosphatase activity by hypothermia.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  54. Lu, P. J., Wulf, G., Zhou, X. Z., Davies, P. & Lu, K. P. The prolyl isomerase Pin1 restores the function of Alzheimer-associated phosphorylated tau protein. Nature 399, 784–788 (1999).

    Article  CAS  PubMed  Google Scholar 

  55. Kondo, A. et al. Antibody against early driver of neurodegeneration cis P-tau blocks brain injury and tauopathy. Nature 523, 431–436 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Hoover, B. R. et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 68, 1067–1081 (2010). This study shows that the hyperphosphorylation of tau drives its entry into postsynaptic terminals, resulting in synaptic dysfunction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Thies, E. & Mandelkow, E. M. Missorting of tau in neurons causes degeneration of synapses that can be rescued by the kinase MARK2/Par-1. J. Neurosci. 27, 2896–2907 (2007). This study demonstrates that missorting of phosphorylated tau into dendrites and spines leads to transport inhibition and loss of spines.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  58. Zempel, H., Thies, E., Mandelkow, E. & Mandelkow, E. M. Aβ oligomers cause localized Ca2+ elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J. Neurosci. 30, 11938–11950 (2010).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  59. Guillozet-Bongaarts, A. L. et al. Pseudophosphorylation of tau at serine 422 inhibits caspase cleavage: in vitro evidence and implications for tangle formation in vivo. J. Neurochem. 97, 1005–1014 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Dickey, C. A. et al. The high-affinity HSP90–CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J. Clin. Invest. 117, 648–658 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ittner, L. M., Ke, Y. D. & Gotz, J. Phosphorylated Tau interacts with c-Jun N-terminal kinase-interacting protein 1 (JIP1) in Alzheimer disease. J. Biol. Chem. 284, 20909–20916 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bhaskar, K., Yen, S. H. & Lee, G. Disease-related modifications in tau affect the interaction between Fyn and Tau. J. Biol. Chem. 280, 35119–35125 (2005).

    Article  CAS  PubMed  Google Scholar 

  63. Reynolds, C. H. et al. Phosphorylation regulates tau interactions with Src homology 3 domains of phosphatidylinositol 3-kinase, phospholipase Cγ1, Grb2, and Src family kinases. J. Biol. Chem. 283, 18177–18186 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Min, S. W. et al. Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67, 953–966 (2010). This study identified the acetylation of tau and demonstrated that this post-translational modification may contribute to tau pathology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Cook, C. et al. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum. Mol. Genet. 23, 104–116 (2014).

    Article  CAS  PubMed  Google Scholar 

  66. Cohen, T. J., Friedmann, D., Hwang, A. W., Marmorstein, R. & Lee, V. M. The microtubule-associated tau protein has intrinsic acetyltransferase activity. Nat. Struct. Mol. Biol. 20, 756–762 (2013).

    Article  CAS  PubMed  Google Scholar 

  67. Irwin, D. J. et al. Acetylated tau neuropathology in sporadic and hereditary tauopathies. Am. J. Pathol. 183, 344–351 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Min, S. W. et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat. Med. 21, 1154–1162 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Martin, L., Latypova, X. & Terro, F. Post-translational modifications of tau protein: implications for Alzheimer's disease. Neurochem. Int. 58, 458–471 (2011).

    Article  CAS  PubMed  Google Scholar 

  70. Wang, J. Z., Grundke-Iqbal, I. & Iqbal, K. Glycosylation of microtubule-associated protein tau: an abnormal posttranslational modification in Alzheimer's disease. Nat. Med. 2, 871–875 (1996).

    Article  CAS  PubMed  Google Scholar 

  71. Liu, F., Zaidi, T., Iqbal, K., Grundke-Iqbal, I. & Gong, C. X. Aberrant glycosylation modulates phosphorylation of tau by protein kinase A and dephosphorylation of tau by protein phosphatase 2A and 5. Neuroscience 115, 829–837 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Liu, F., Iqbal, K., Grundke-Iqbal, I., Hart, G. W. & Gong, C. X. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer's disease. Proc. Natl Acad. Sci. USA 101, 10804–10809 (2004).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  73. Yuzwa, S. A., Cheung, A. H., Okon, M., McIntosh, L. P. & Vocadlo, D. J. O-GlcNAc modification of tau directly inhibits its aggregation without perturbing the conformational properties of tau monomers. J. Mol. Biol. 426, 1736–1752 (2014).

    Article  CAS  PubMed  Google Scholar 

  74. Yan, S. D. et al. Non-enzymatically glycated tau in Alzheimer's disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid β-peptide. Nat. Med. 1, 693–699 (1995).

    Article  CAS  PubMed  Google Scholar 

  75. Watanabe, A. et al. Molecular aging of tau: disulfide-independent aggregation and non-enzymatic degradation in vitro and in vivo. J. Neurochem. 90, 1302–1311 (2004). The authors show that site-specific deamidation or isomerization and non-enzymatic degradation of ageing tau facilitates tau aggregation and can cause the smearing of PHFs on western blot that is typical of AD-tau.

    Article  CAS  PubMed  Google Scholar 

  76. Ledesma, M. D., Bonay, P. & Avila, J. Tau protein from Alzheimer's disease patients is glycated at its tubulin-binding domain. J. Neurochem. 65, 1658–1664 (1995).

    Article  CAS  PubMed  Google Scholar 

  77. Reyes, J. F., Geula, C., Vana, L. & Binder, L. I. Selective tau tyrosine nitration in non-AD tauopathies. Acta Neuropathol. 123, 119–132 (2012).

    Article  CAS  PubMed  Google Scholar 

  78. Funk, K. E. et al. Lysine methylation is an endogenous post-translational modification of tau protein in human brain and a modulator of aggregation propensity. Biochem. J. 462, 77–88 (2014).

    Article  CAS  PubMed  Google Scholar 

  79. Shimura, H., Schwartz, D., Gygi, S. P. & Kosik, K. S. CHIP–Hsc70 complex ubiquitinates phosphorylated tau and enhances cell survival. J. Biol. Chem. 279, 4869–4876 (2004).

    Article  CAS  PubMed  Google Scholar 

  80. Petrucelli, L. et al. CHIP and Hsp70 regulate tau ubiquitination, degradation and aggregation. Hum. Mol. Genet. 13, 703–714 (2004).

    Article  CAS  PubMed  Google Scholar 

  81. Babu, J. R., Geetha, T. & Wooten, M. W. Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation. J. Neurochem. 94, 192–203 (2005).

    Article  CAS  PubMed  Google Scholar 

  82. Nathan, J. A., Kim, H. T., Ting, L., Gygi, S. P. & Goldberg, A. L. Why do cellular proteins linked to K63-polyubiquitin chains not associate with proteasomes? EMBO J. 32, 552–565 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Dorval, V. & Fraser, P. E. Small ubiquitin-like modifier (SUMO) modification of natively unfolded proteins tau and alpha-synuclein. J. Biol. Chem. 281, 9919–9924 (2006).

    Article  PubMed  CAS  Google Scholar 

  84. Pountney, D. L. et al. SUMO-1 marks the nuclear inclusions in familial neuronal intranuclear inclusion disease. Exp. Neurol. 184, 436–446 (2003).

    Article  CAS  PubMed  Google Scholar 

  85. Luo, H. B. et al. SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc. Natl Acad. Sci. USA 111, 16586–16591 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Corsetti, V. et al. NH2-truncated human tau induces deregulated mitophagy in neurons by aberrant recruitment of Parkin and UCHL-1: implications in Alzheimer's disease. Hum. Mol. Genet. 24, 3058–3081 (2015).

    Article  CAS  PubMed  Google Scholar 

  87. Garg, S., Timm, T., Mandelkow, E. M., Mandelkow, E. & Wang, Y. Cleavage of Tau by calpain in Alzheimer's disease: the quest for the toxic 17 kD fragment. Neurobiol. Aging 32, 1–14 (2011).

    Article  CAS  PubMed  Google Scholar 

  88. Derisbourg, M. et al. Role of the Tau N-terminal region in microtubule stabilization revealed by new endogenous truncated forms. Sci. Rep. 5, 9659 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Drubin, D. G., Caput, D. & Kirschner, M. W. Studies on the expression of the microtubule-associated protein, tau, during mouse brain development, with newly isolated complementary DNA probes. J. Cell Biol. 98, 1090–1097 (1984).

    Article  CAS  PubMed  Google Scholar 

  90. Papasozomenos, S. C. & Binder, L. I. Phosphorylation determines two distinct species of Tau in the central nervous system. Cell. Motil. Cytoskeleton 8, 210–226 (1987).

    Article  CAS  PubMed  Google Scholar 

  91. Sultan, A. et al. Nuclear tau, a key player in neuronal DNA protection. J. Biol. Chem. 286, 4566–4575 (2011).

    Article  CAS  PubMed  Google Scholar 

  92. Litman, P., Barg, J., Rindzoonski, L. & Ginzburg, I. Subcellular localization of tau mRNA in differentiating neuronal cell culture: implications for neuronal polarity. Neuron 10, 627–638 (1993).

    Article  CAS  PubMed  Google Scholar 

  93. Aronov, S., Aranda, G., Behar, L. & Ginzburg, I. Axonal tau mRNA localization coincides with tau protein in living neuronal cells and depends on axonal targeting signal. J. Neurosci. 21, 6577–6587 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Morita, T. & Sobue, K. Specification of neuronal polarity regulated by local translation of CRMP2 and Tau via the mTOR–p70S6K pathway. J. Biol. Chem. 284, 27734–27745 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Hirokawa, N., Funakoshi, T., Sato-Harada, R. & Kanai, Y. Selective stabilization of tau in axons and microtubule-associated protein 2C in cell bodies and dendrites contributes to polarized localization of cytoskeletal proteins in mature neurons. J. Cell Biol. 132, 667–679 (1996).

    Article  CAS  PubMed  Google Scholar 

  96. Kosik, K. S., Crandall, J. E., Mufson, E. J. & Neve, R. L. Tau in situ hybridization in normal and Alzheimer brain: localization in the somatodendritic compartment. Ann. Neurol. 26, 352–361 (1989).

    Article  CAS  PubMed  Google Scholar 

  97. Li, X. et al. Novel diffusion barrier for axonal retention of Tau in neurons and its failure in neurodegeneration. EMBO J. 30, 4825–4837 (2011). This study identifies an axonal sorting mechanism of tau at the axonal initial segment.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Feinstein, S. C. & 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 1739, 268–279 (2005).

    Article  CAS  PubMed  Google Scholar 

  99. Mandelkow, E. M. & Mandelkow, E. Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb. Perspect. Med. 2, a006247 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Stamer, K., Vogel, R., Thies, E., Mandelkow, E. & Mandelkow, E. M. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J. Cell Biol. 156, 1051–1063 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Dixit, R., Ross, J. L., Goldman, Y. E. & Holzbaur, E. L. Differential regulation of dynein and kinesin motor proteins by tau. Science 319, 1086–1089 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Vershinin, M., Carter, B. C., Razafsky, D. S., King, S. J. & Gross, S. P. Multiple-motor based transport and its regulation by Tau. Proc. Natl Acad. Sci. USA 104, 87–92 (2007).

    Article  CAS  PubMed  Google Scholar 

  103. Konzack, S., Thies, E., Marx, A., Mandelkow, E. M. & Mandelkow, E. Swimming against the tide: mobility of the microtubule-associated protein tau in neurons. J. Neurosci. 27, 9916–9927 (2007).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  104. Utton, M. A., Noble, W. J., Hill, J. E., Anderton, B. H. & Hanger, D. P. Molecular motors implicated in the axonal transport of tau and α-synuclein. J. Cell Sci. 118, 4645–4654 (2005).

    Article  CAS  PubMed  Google Scholar 

  105. Kanaan, N. M. et al. Pathogenic forms of tau inhibit kinesin-dependent axonal transport through a mechanism involving activation of axonal phosphotransferases. J. Neurosci. 31, 9858–9868 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Magnani, E. et al. Interaction of tau protein with the dynactin complex. EMBO J. 26, 4546–4554 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Yuan, A., Kumar, A., Peterhoff, C., Duff, K. & Nixon, R. A. Axonal transport rates in vivo are unaffected by tau deletion or overexpression in mice. J. Neurosci. 28, 1682–1687 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Caceres, A. & Kosik, K. S. Inhibition of neurite polarity by tau antisense oligonucleotides in primary cerebellar neurons. Nature 343, 461–463 (1990).

    Article  CAS  PubMed  Google Scholar 

  109. Knops, J. et al. Overexpression of tau in a nonneuronal cell induces long cellular processes. J. Cell Biol. 114, 725–733 (1991).

    Article  CAS  PubMed  Google Scholar 

  110. Dawson, H. N. et al. Inhibition of neuronal maturation in primary hippocampal neurons from tau deficient mice. J. Cell Sci. 114, 1179–1187 (2001).

    CAS  PubMed  Google Scholar 

  111. Harada, A. et al. Altered microtubule organization in small-calibre axons of mice lacking tau protein. Nature 369, 488–491 (1994). This study established the first tau-knockout transgenic mouse line and found that the deficiency of tau does not cause overt pathology, owing to the compensatory expression of MAP1A.

    Article  CAS  PubMed  Google Scholar 

  112. Tai, H. C. et al. Frequent and symmetric deposition of misfolded tau oligomers within presynaptic and postsynaptic terminals in Alzheimer inverted question marks disease. Acta Neuropathol. Commun. 2, 146 (2014).

    PubMed  PubMed Central  Google Scholar 

  113. Mondragon-Rodriguez, S. et al. Interaction of endogenous tau protein with synaptic proteins is regulated by N-methyl-d-aspartate receptor-dependent tau phosphorylation. J. Biol. Chem. 287, 32040–32053 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Frandemiche, M. L. et al. Activity-dependent tau protein translocation to excitatory synapse is disrupted by exposure to amyloid-β oligomers. J. Neurosci. 34, 6084–6097 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  115. Loomis, P. A., Howard, T. H., Castleberry, R. P. & Binder, L. I. Identification of nuclear tau isoforms in human neuroblastoma cells. Proc. Natl Acad. Sci. USA 87, 8422–8426 (1990).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Sjoberg, M. K., Shestakova, E., Mansuroglu, Z., Maccioni, R. B. & Bonnefoy, E. Tau protein binds to pericentromeric DNA: a putative role for nuclear tau in nucleolar organization. J. Cell Sci. 119, 2025–2034 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Fernandez-Nogales, M. et al. Huntington's disease is a four-repeat tauopathy with tau nuclear rods. Nat. Med. 20, 881–885 (2014).

    Article  CAS  PubMed  Google Scholar 

  118. Gheyara, A. L. et al. Tau reduction prevents disease in a mouse model of Dravet syndrome. Ann. Neurol. 76, 443–456 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Holth, J. K. et al. Tau loss attenuates neuronal network hyperexcitability in mouse and Drosophila genetic models of epilepsy. J. Neurosci. 33, 1651–1659 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Leroy, K. et al. Lack of tau proteins rescues neuronal cell death and decreases amyloidogenic processing of APP in APP/PS1 mice. Am. J. Pathol. 181, 1928–1940 (2012).

    Article  CAS  PubMed  Google Scholar 

  121. DeVos, S. L. et al. Antisense reduction of tau in adult mice protects against seizures. J. Neurosci. 33, 12887–12897 (2013). This paper demonstrates that treatment with antisense oligonucleotides can reduce tau-induced pathology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Lei, P. et al. Motor and cognitive deficits in aged tau knockout mice in two background strains. Mol. Neurodegener. 9, 29 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Hong, X. P. et al. Essential role of tau phosphorylation in adult hippocampal neurogenesis. Hippocampus 20, 1339–1349 (2010).

    Article  CAS  PubMed  Google Scholar 

  124. Fuster-Matanzo, A. et al. Function of tau protein in adult newborn neurons. FEBS Lett. 583, 3063–3068 (2009).

    Article  CAS  PubMed  Google Scholar 

  125. Lei, P. et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat. Med. 18, 291–295 (2012). This study shows that tau deficiency may cause neurodegeneration by impairing APP-mediated iron export.

    Article  CAS  PubMed  Google Scholar 

  126. Kimura, T. et al. Microtubule-associated protein tau is essential for long-term depression in the hippocampus. Phil. Trans. R. Soc. B 369, 20130144 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Ahmed, T. et al. Cognition and hippocampal synaptic plasticity in mice with a homozygous tau deletion. Neurobiol. Aging 35, 2474–2478 (2014).

    Article  CAS  PubMed  Google Scholar 

  128. Biernat, J. et al. Protein kinase MARK/PAR-1 is required for neurite outgrowth and establishment of neuronal polarity. Mol. Biol. Cell 13, 4013–4028 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Whiteman, I. T. et al. Activated actin-depolymerizing factor/cofilin sequesters phosphorylated microtubule-associated protein during the assembly of Alzheimer-like neuritic cytoskeletal striations. J. Neurosci. 29, 12994–13005 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Kouri, N. et al. Novel mutation in MAPT exon 13 (p. N410H) causes corticobasal degeneration. Acta Neuropathol. 127, 271–282 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Coppola, G. et al. Evidence for a role of the rare p. A152T variant in MAPT in increasing the risk for FTD-spectrum and Alzheimer's diseases. Hum. Mol. Genet. 21, 3500–3512 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  132. Barghorn, S. et al. Structure, microtubule interactions, and paired helical filament aggregation by tau mutants of frontotemporal dementias. Biochemistry 39, 11714–11721 (2000).

    Article  CAS  PubMed  Google Scholar 

  133. Hong, M. et al. Mutation-specific functional impairments in distinct tau isoforms of hereditary FTDP-17. Science 282, 1914–1917 (1998). The authors demonstrate that FTDP-17 mutants of tau show reduced microtubule binding and accelerated aggregation.

    Article  PubMed  CAS  Google Scholar 

  134. Crary, J. F. et al. Primary age-related tauopathy (PART): a common pathology associated with human aging. Acta Neuropathol. 128, 755–766 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Jellinger, K. A. et al. PART, a distinct tauopathy, different from classical sporadic Alzheimer disease. Acta Neuropathol. 129, 757–762 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  136. Braak, H. & Del Tredici, K. Are cases with tau pathology occurring in the absence of Aβ deposits part of the AD-related pathological process? Acta Neuropathol. 128, 767–772 (2014).

    Article  PubMed  Google Scholar 

  137. von Bergen, M. et al. Assembly of τ protein into Alzheimer paired helical filaments depends on a local sequence motif (306VQIVYK311) forming β structure. Proc. Natl Acad. Sci. USA 97, 5129–5134 (2000). This study identifies two hexapeptide motifs (VQIINK and VQIVYK) in tau R2 and R3, with high propensity for β-sheet structure responsible for tau aggregation.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Sawaya, M. R. et al. Atomic structures of amyloid cross-β spines reveal varied steric zippers. Nature 447, 453–457 (2007). This paper reveals that 4–7-residue sequence motifs of neurodegenerative disease-related proteins, including tau, are sufficient to form fibrils composed of steric zippers, formed by two tightly interdigitated β-sheets.

    Article  CAS  PubMed  Google Scholar 

  139. Khlistunova, I. et al. Inducible expression of Tau repeat domain in cell models of tauopathy: aggregation is toxic to cells but can be reversed by inhibitor drugs. J. Biol. Chem. 281, 1205–1214 (2006).

    Article  CAS  PubMed  Google Scholar 

  140. Goedert, M. et al. Assembly of microtubule-associated protein tau into Alzheimer-like filaments induced by sulphated glycosaminoglycans. Nature 383, 550–553 (1996). This study demonstrates that tau can be induced to aggregate in vitro with polyanions such as heparan sulfate.

    Article  CAS  PubMed  Google Scholar 

  141. Wille, H., Drewes, G., Biernat, J., Mandelkow, E. M. & Mandelkow, E. Alzheimer-like paired helical filaments and antiparallel dimers formed from microtubule-associated protein tau in vitro. J. Cell Biol. 118, 573–584 (1992).

    Article  CAS  PubMed  Google Scholar 

  142. Hernandez, F., Cuadros, R. & Avila, J. Zeta 14-3-3 protein favours the formation of human tau fibrillar polymers. Neurosci. Lett. 357, 143–146 (2004).

    Article  CAS  PubMed  Google Scholar 

  143. Giustiniani, J. et al. Immunophilin FKBP52 induces Tau-P301L filamentous assembly in vitro and modulates its activity in a model of tauopathy. Proc. Natl Acad. Sci. USA 111, 4584–4589 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Braak, E., Braak, H. & Mandelkow, E. M. A sequence of cytoskeleton changes related to the formation of neurofibrillary tangles and neuropil threads. Acta Neuropathol. 87, 554–567 (1994).

    Article  PubMed  CAS  Google Scholar 

  145. Alonso, A., Zaidi, T., Novak, M., Grundke-Iqbal, I. & Iqbal, K. Hyperphosphorylation induces self-assembly of tau into tangles of paired helical filaments/straight filaments. Proc. Natl Acad. Sci. USA 98, 6923–6928 (2001).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  146. Schneider, A., Biernat, J., von Bergen, M., Mandelkow, E. & Mandelkow, E. M. Phosphorylation that detaches tau protein from microtubules (Ser262, Ser214) also protects it against aggregation into Alzheimer paired helical filaments. Biochemistry 38, 3549–3558 (1999).

    Article  CAS  PubMed  Google Scholar 

  147. Tepper, K. et al. Oligomer formation of tau protein hyperphosphorylated in cells. J. Biol. Chem. 289, 34389–34407 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Wang, Y. & Mandelkow, E. Degradation of tau protein by autophagy and proteasomal pathways. Biochem. Soc. Trans. 40, 644–652 (2012).

    Article  CAS  PubMed  Google Scholar 

  149. Zilka, N. et al. Truncated tau from sporadic Alzheimer's disease suffices to drive neurofibrillary degeneration in vivo. FEBS Lett. 580, 3582–3588 (2006).

    Article  CAS  PubMed  Google Scholar 

  150. de Calignon, A. et al. Caspase activation precedes and leads to tangles. Nature 464, 1201–1204 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  151. Zhang, Z. et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer's disease. Nat. Med. 20, 1254–1262 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Wang, Y. P., Biernat, J., Pickhardt, M., Mandelkow, E. & Mandelkow, E. M. Stepwise proteolysis liberates tau fragments that nucleate the Alzheimer-like aggregation of full-length tau in a neuronal cell model. Proc. Natl Acad. Sci. USA 104, 10252–10257 (2007). The authors show that truncation of tau can accelerate its aggregation in cells and that pro-aggregant truncated tau can seed the aggregation of endogenous tau.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Fatouros, C. et al. Inhibition of tau aggregation in a novel Caenorhabditis elegans model of tauopathy mitigates proteotoxicity. Hum. Mol. Genet. 21, 3587–3603 (2012).

    Article  CAS  PubMed  Google Scholar 

  154. Clavaguera, F. et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat. Cell Biol. 11, 909–913 (2009). The authors of this study demonstrate the spreading of tau pathology in a mouse model after injection of an AD brain extract.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Peeraer, E. et al. Intracerebral injection of preformed synthetic tau fibrils initiates widespread tauopathy and neuronal loss in the brains of tau transgenic mice. Neurobiol. Dis. 73, 83–95 (2015).

    Article  CAS  PubMed  Google Scholar 

  156. Clavaguera, F. et al. Peripheral administration of tau aggregates triggers intracerebral tauopathy in transgenic mice. Acta Neuropathol. 127, 299–301 (2014).

    Article  PubMed  Google Scholar 

  157. Sanders, D. W. et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288 (2014). This paper shows that tau may form different prion-like strains, leading to distinct seeding and spreading pattern of tau pathology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  158. Zhukareva, V. et al. Loss of brain tau defines novel sporadic and familial tauopathies with frontotemporal dementia. Ann. Neurol. 49, 165–175 (2001).

    Article  CAS  PubMed  Google Scholar 

  159. Gomez-Isla, T. et al. Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer's disease. Ann. Neurol. 41, 17–24 (1997).

    Article  CAS  PubMed  Google Scholar 

  160. Morsch, R., Simon, W. & Coleman, P. D. Neurons may live for decades with neurofibrillary tangles. J. Neuropathol. Exp. Neurol. 58, 188–197 (1999).

    Article  CAS  PubMed  Google Scholar 

  161. Andorfer, C. et al. Cell-cycle reentry and cell death in transgenic mice expressing nonmutant human tau isoforms. J. Neurosci. 25, 5446–5454 (2005).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  162. Spires-Jones, T. L. et al. In vivo imaging reveals dissociation between caspase activation and acute neuronal death in tangle-bearing neurons. J. Neurosci. 28, 862–867 (2008).

    Article  PubMed Central  PubMed  Google Scholar 

  163. Santacruz, K. et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science 309, 476–481 (2005). This study describes a regulatable transgenic mouse line of tauopathy, showing that switching off the expression of the transgenic human tau can rescue cognitive deficits despite the continuing presence of tau aggregates, and implying that tau aggregates are not sufficient for neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Van der Jeugd, A. et al. Cognitive defects are reversible in inducible mice expressing pro-aggregant full-length human Tau. Acta Neuropathol. 123, 787–805 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Sydow, A. et al. Tau-induced defects in synaptic plasticity, learning, and memory are reversible in transgenic mice after switching off the toxic Tau mutant. J. Neurosci. 31, 2511–2525 (2011).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  166. Alonso Adel, C., Li, B., Grundke-Iqbal, I. & Iqbal, K. Polymerization of hyperphosphorylated tau into filaments eliminates its inhibitory activity. Proc. Natl Acad. Sci. USA 103, 8864–8869 (2006).

    Article  CAS  PubMed  Google Scholar 

  167. Walsh, D. M. & Selkoe, D. J. Aβ oligomers — a decade of discovery. J. Neurochem. 101, 1172–1184 (2007).

    Article  CAS  PubMed  Google Scholar 

  168. Lasagna-Reeves, C. A. et al. Identification of oligomers at early stages of tau aggregation in Alzheimer's disease. FASEB J. 26, 1946–1959 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Maeda, S. et al. Granular tau oligomers as intermediates of tau filaments. Biochemistry 46, 3856–3861 (2007).

    Article  PubMed  CAS  Google Scholar 

  170. Tian, H. et al. Trimeric tau is toxic to human neuronal cells at low nanomolar concentrations. Int. J. Cell Biol. 2013, 260787 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Flach, K. et al. Tau oligomers impair artificial membrane integrity and cellular viability. J. Biol. Chem. 287, 43223–43233 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Rizzu, P. et al. High prevalence of mutations in the microtubule-associated protein tau in a population study of frontotemporal dementia in the Netherlands. Am. J. Hum. Genet. 64, 414–421 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Mocanu, M. M. et al. The potential for β-structure in the repeat domain of tau protein determines aggregation, synaptic decay, neuronal loss, and coassembly with endogenous Tau in inducible mouse models of tauopathy. J. Neurosci. 28, 737–748 (2008). This paper demonstrates that the propensity for β-structure in one hexapeptide motif of pro-aggregant tau can cause neurodegeneration that can be reversed by blocking tau expression or by blocking aggregation using anti-aggregant tau.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  174. Eckermann, K. et al. The β-propensity of Tau determines aggregation and synaptic loss in inducible mouse models of tauopathy. J. Biol. Chem. 282, 31755–31765 (2007).

    Article  CAS  PubMed  Google Scholar 

  175. Sarkar, M., Kuret, J. & Lee, G. Two motifs within the tau microtubule-binding domain mediate its association with the hsc70 molecular chaperone. J. Neurosci. Res. 86, 2763–2773 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  176. Wang, Y. et al. Tau fragmentation, aggregation and clearance: the dual role of lysosomal processing. Hum. Mol. Genet. 18, 4153–4170 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Fulga, T. A. et al. Abnormal bundling and accumulation of F-actin mediates tau-induced neuronal degeneration in vivo. Nat. Cell Biol. 9, 139–148 (2007).

    Article  CAS  PubMed  Google Scholar 

  178. Guthrie, C. R., Schellenberg, G. D. & Kraemer, B. C. SUT-2 potentiates tau-induced neurotoxicity in Caenorhabditis elegans. Hum. Mol. Genet. 18, 1825–1838 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Kraemer, B. C. & Schellenberg, G. D. SUT-1 enables tau-induced neurotoxicity in C. elegans. Hum. Mol. Genet. 16, 1959–1971 (2007).

    Article  CAS  PubMed  Google Scholar 

  180. Zempel, H. et al. Amyloid-β oligomers induce synaptic damage via Tau-dependent microtubule severing by TTLL6 and spastin. EMBO J. 32, 2920–2937 (2013). This study shows that Aβ oligomers can induce dendritic missorting of tau, which causes loss of spines and loss of microtubules by the activation of spastin.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Seward, M. E. et al. Amyloid-β signals through tau to drive ectopic neuronal cell cycle re-entry in Alzheimer's disease. J. Cell Sci. 126, 1278–1286 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  182. Oddo, S. et al. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron 39, 409–421 (2003). This authors of this study generated a mouse model with enhanced aggregation of both Aβ and tau, allowing the study of the interplay between these two types of proteins.

    Article  CAS  PubMed  Google Scholar 

  183. Decker, J. M. et al. Pro-aggregant Tau impairs mossy fiber plasticity due to structural changes and Ca++ dysregulation. Acta Neuropathol. Commun. 3, 23 (2015). This paper demonstrates that tau pathology may cause presynaptic dysfunction at an early stage of neurodegeneration.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  184. Maphis, N. et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain 138, 1738–1755 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  185. Morimoto, R. I. & Cuervo, A. M. Proteostasis and the aging proteome in health and disease. J. Gerontol. A Biol. Sci. Med. Sci. 69 (Suppl. 1), 33–38 (2014).

    Article  CAS  Google Scholar 

  186. Pooler, A. M., Phillips, E. C., Lau, D. H., Noble, W. & Hanger, D. P. Physiological release of endogenous tau is stimulated by neuronal activity. EMBO Rep. 14, 389–394 (2013). This study demonstrates that the physiological release of tau can be stimulated by neuronal activity.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  187. Yamada, K. et al. Neuronal activity regulates extracellular tau in vivo. J. Exp. Med. 211, 387–393 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Johnson, G. V. et al. The tau protein in human cerebrospinal fluid in Alzheimer's disease consists of proteolytically derived fragments. J. Neurochem. 68, 430–433 (1997).

    Article  CAS  PubMed  Google Scholar 

  189. Yamada, K. et al. In vivo microdialysis reveals age-dependent decrease of brain interstitial fluid tau levels in P301S human tau transgenic mice. J. Neurosci. 31, 13110–13117 (2011). This paper demonstrates the release of tau by neurons, arguing that extracellular tau may be in equilibrium with intracellular tau.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Takamori, S. et al. Molecular anatomy of a trafficking organelle. Cell 127, 831–846 (2006). The authors report the protein and lipid composition of synaptic vesicles. Notably, tau is not among them, arguing against a release of axonal tau via synaptic vesicles.

    Article  CAS  PubMed  Google Scholar 

  191. Karch, C. M., Jeng, A. T. & Goate, A. M. Extracellular Tau levels are influenced by variability in Tau that is associated with tauopathies. J. Biol. Chem. 287, 42751–42762 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  192. Nickel, W. & Rabouille, C. Mechanisms of regulated unconventional protein secretion. Nat. Rev. Mol. Cell Biol. 10, 148–155 (2009).

    Article  CAS  PubMed  Google Scholar 

  193. Gomez-Ramos, A., Diaz-Hernandez, M., Rubio, A., Miras-Portugal, M. T. & Avila, J. Extracellular tau promotes intracellular calcium increase through M1 and M3 muscarinic receptors in neuronal cells. Mol. Cell. Neurosci. 37, 673–681 (2008).

    Article  CAS  PubMed  Google Scholar 

  194. Gomez-Ramos, A. et al. Characteristics and consequences of muscarinic receptor activation by tau protein. Eur. Neuropsychopharmacol. 19, 708–717 (2009).

    Article  CAS  PubMed  Google Scholar 

  195. Guo, J. L. & Lee, V. M. Seeding of normal Tau by pathological Tau conformers drives pathogenesis of Alzheimer-like tangles. J. Biol. Chem. 286, 15317–15331 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Michel, C. H. et al. Extracellular monomeric tau protein is sufficient to initiate the spread of tau protein pathology. J. Biol. Chem. 289, 956–967 (2014).

    Article  CAS  PubMed  Google Scholar 

  197. Holmes, B. B. et al. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proc. Natl Acad. Sci. USA 110, E3138–E3147 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  198. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991). This paper established the staging of AD on the basis of tau pathology, which is now widely accepted.

    Article  CAS  PubMed  Google Scholar 

  199. Braak, H. & Braak, E. Development of Alzheimer-related neurofibrillary changes in the neocortex inversely recapitulates cortical myelogenesis. Acta Neuropathol. 92, 197–201 (1996).

    Article  CAS  PubMed  Google Scholar 

  200. Yan, M. H., Wang, X. & Zhu, X. Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic. Biol. Med. 62, 90–101 (2013).

    Article  CAS  PubMed  Google Scholar 

  201. Nave, K. A. & Werner, H. B. Myelination of the nervous system: mechanisms and functions. Annu. Rev. Cell Dev. Biol. 30, 503–533 (2014).

    Article  CAS  PubMed  Google Scholar 

  202. Hyman, B. T., Van Hoesen, G. W., Damasio, A. R. & Barnes, C. L. Alzheimer's disease: cell-specific pathology isolates the hippocampal formation. Science 225, 1168–1170 (1984).

    Article  CAS  PubMed  Google Scholar 

  203. Hochgrafe, K. et al. Preventive methylene blue treatment preserves cognition in mice expressing full-length pro-aggregant human Tau. Acta Neuropathol. Commun. 3, 25 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Wischik, C. M. et al. Tau aggregation inhibitor therapy: an exploratory phase 2 study in mild or moderate Alzheimer's disease. J. Alzheimers Dis. 44, 705–720 (2015).

    Article  CAS  PubMed  Google Scholar 

  205. Sievers, S. A. et al. Structure-based design of non-natural amino-acid inhibitors of amyloid fibril formation. Nature 475, 96–100 (2011). This paper identifies several d-amino acid peptides that can inhibit tau aggregation by interacting with steric zippers formed by the hexapeptide.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  206. Lovestone, S. et al. A phase II trial of tideglusib in Alzheimer's disease. J. Alzheimers Dis. 45, 75–88 (2015).

    Article  CAS  PubMed  Google Scholar 

  207. Hoglinger, G. U. et al. Tideglusib reduces progression of brain atrophy in progressive supranuclear palsy in a randomized trial. Mov. Disord. 29, 479–487 (2014).

    Article  CAS  PubMed  Google Scholar 

  208. Boxer, A. L. et al. Davunetide in patients with progressive supranuclear palsy: a randomised, double-blind, placebo-controlled phase 2/3 trial. Lancet Neurol. 13, 676–685 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  209. Brunden, K. R. et al. Epothilone D improves microtubule density, axonal integrity, and cognition in a transgenic mouse model of tauopathy. J. Neurosci. 30, 13861–13866 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. US National Library of Medicine. ClinicalTrials.gov [online], (2015).

  211. Blair, L. J., Sabbagh, J. J. & Dickey, C. A. Targeting Hsp90 and its co-chaperones to treat Alzheimer's disease. Expert Opin. Ther. Targets 18, 1219–1232 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Karagoz, G. E. et al. Hsp90–Tau complex reveals molecular basis for specificity in chaperone action. Cell 156, 963–974 (2014). This is a structural study that demonstrates the interaction between HSP90 and tau.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  213. Ozcelik, S. et al. Rapamycin attenuates the progression of tau pathology in P301S tau transgenic mice. PLoS ONE 8, e62459 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Kruger, U., Wang, Y., Kumar, S. & Mandelkow, E. M. Autophagic degradation of tau in primary neurons and its enhancement by trehalose. Neurobiol. Aging 33, 2291–2305 (2012).

    Article  CAS  PubMed  Google Scholar 

  215. Berger, Z. et al. Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum. Mol. Genet. 15, 433–442 (2006).

    Article  CAS  PubMed  Google Scholar 

  216. Sigurdsson, E. M. Tau immunotherapy and imaging. Neurodegener. Dis. 13, 103–106 (2014).

    Article  CAS  PubMed  Google Scholar 

  217. Asuni, A. A., Boutajangout, A., Quartermain, D. & Sigurdsson, E. M. Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J. Neurosci. 27, 9115–9129 (2007). This paper demonstrates that a tau-based active immunotherapy reduces tau pathology and slows progression of behavioural impairment in a line of tau transgenic mice.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  218. Golde, T. E. Open questions for Alzheimer's disease immunotherapy. Alzheimers Res. Ther. 6, 3 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  219. Congdon, E. E., Gu, J., Sait, H. B. & Sigurdsson, E. M. Antibody uptake into neurons occurs primarily via clathrin-dependent Fcγ receptor endocytosis and is a prerequisite for acute tau protein clearance. J. Biol. Chem. 288, 35452–35465 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  220. Collin, L. et al. Neuronal uptake of tau/pS422 antibody and reduced progression of tau pathology in a mouse model of Alzheimer's disease. Brain 137, 2834–2846 (2014).

    Article  PubMed  Google Scholar 

  221. d'Abramo, C., Acker, C. M., Jimenez, H. T. & Davies, P. Tau passive immunotherapy in mutant P301L mice: antibody affinity versus specificity. PLoS ONE 8, e62402 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  222. Castillo-Carranza, D. L. et al. Passive immunization with Tau oligomer monoclonal antibody reverses tauopathy phenotypes without affecting hyperphosphorylated neurofibrillary tangles. J. Neurosci. 34, 4260–4272 (2014).

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  223. Yanamandra, K. et al. Anti-tau antibodies that block tau aggregate seeding in vitro markedly decrease pathology and improve cognition in vivo. Neuron 80, 402–414 (2013). This study presents a new concept of immunotherapy: targeting extracellular tau seeds with tau-specific antibodies to prevent the spreading of tau pathology.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Neve, R. L., Harris, P., Kosik, K. S., Kurnit, D. M. & Donlon, T. A. Identification of cDNA clones for the human microtubule-associated protein tau and chromosomal localization of the genes for tau and microtubule-associated protein 2. Brain Res. 387, 271–280 (1986).

    CAS  PubMed  Google Scholar 

  225. Trabzuni, D. et al. MAPT expression and splicing is differentially regulated by brain region: relation to genotype and implication for tauopathies. Hum. Mol. Genet. 21, 4094–4103 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Boutajangout, A., Boom, A., Leroy, K. & Brion, J. P. Expression of tau mRNA and soluble tau isoforms in affected and non-affected brain areas in Alzheimer's disease. FEBS Lett. 576, 183–189 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank E.-M. Mandelkow for critical reading of and insightful suggestions for the manuscript, and thank L. Krueger for fruitful discussions. The project was supported in part by the German Center for Neurodegenerative Diseases (DZNE), the Max Planck Society (MPG), the Tau Consortium and the Wellcome Trust/MRC Alzheimer Consortium.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Yipeng Wang or Eckhard Mandelkow.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

FURTHER INFORMATION

Alzforum

PowerPoint slides

Glossary

Paired helical filaments

(PHFs). Fibrous polymers of tau that resemble twisted filaments (originally considered to be pairs of filaments, hence the name) of 20 nm width and 80 nm crossover periodicity. Variants of PHFs are the 'straight filaments', which do not have a twisted structure.

Neurofibrillary tangles

(NFTs). Bundles of paired helical filaments in the cytosol of neurons.

Tau aggregation

An interaction between tau molecules that leads to the generation of fibrous polymers with a periodic structure.

Projection domain

The amino-terminal half of tau that projects away from microtubules when tau binds to microtubules.

Assembly domain

The carboxy-terminal portion of tau that is responsible for binding to microtubules.

Lys48 linkages

The binding of a ubiquitin molecule to another ubiquitin via the Lys48 of the seven Lys residues of ubiquitin.

Nucleation–elongation mechanism

A model that postulates that the rate-limiting step for protein aggregation is the formation of an initial oligomeric nucleus for aggregation. Once formed, polymerization can proceed via elongation, whereby protein subunits are directly added to the growing ends of the fibre.

Template-assisted model

A model developed to explain aggregation of prion protein. It proposes that the infectious scrapie prion protein serves as a template that catalyses conformational changes of normal PrPC to PrPSc, leading to PrPSc aggregation.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, Y., Mandelkow, E. Tau in physiology and pathology. Nat Rev Neurosci 17, 22–35 (2016). https://doi.org/10.1038/nrn.2015.1

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn.2015.1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing