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 at the interface between neurodegeneration and neuroinflammation

Abstract

Tau is an evolutionary conserved protein that promotes the assembly and stabilization of microtubules in neuronal axons. Complex patterns of posttranslational modifications (PTMs) dynamically regulate tau biochemical properties and consequently its functions. An imbalance in tau PTMs has been connected with a broad spectrum of neurodegenerative conditions which are collectively known as tauopathies and include Alzheimer’s disease (AD), progressive supranuclear palsy (PSP), and corticobasal degeneration (CBD) among others. The hallmark of these neurological disorders is the presence in the brain of fibrillary tangles constituted of misfolded species of hyper-phosphorylated tau. The pathological events leading to tau aggregation are still largely unknown but increasing evidence suggests that neuroinflammation plays a critical role in tangle formation. Moreover, tau aggregation itself could enhance inflammation through feed-forward mechanisms, amplifying the initial neurotoxic insults. Protective effects of tau against neuroinflammation have been also documented, adding another layer of complexity to this phenomenon. Here, we will review the current knowledge on tau regulation and function in health and disease. In particular, we will address its emerging role in connecting neurodegenerative and neuroinflammatory processes.

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

Fig. 1: Schematic representation of human MAPT genetic structure and tau protein isoforms.
Fig. 2: Alternative model for neuroinflammation in tau pathology.

Similar content being viewed by others

References

  1. Migheli A, Butler M, Brown K, Shelanski ML. Light and electron microscope localization of the microtubule-associated tau protein in rat brain. J Neurosci. 1988;8:1846–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Papasozomenos SC, Binder LI. Phosphorylation determines two distinct species of Tau in the central nervous system. Cell Motil Cytoskeleton. 1987;8:210–26.

    Article  CAS  PubMed  Google Scholar 

  3. Weingarten MD, Lockwood AH, Hwo SY, Kirschner MW. A protein factor essential for microtubule assembly. Proc Natl Acad Sci USA. 1975;72:1858–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Black MM, Slaughter T, Moshiach S, Obrocka M, Fischer I. Tau is enriched on dynamic microtubules in the distal region of growing axons. J Neurosci. 1996;16:3601–19.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;156:1051–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Caceres A, Kosik KS. Inhibition of neurite polarity by tau antisense oligonucleotides in primary cerebellar neurons. Nature. 1990;343:461–3.

    Article  CAS  PubMed  Google Scholar 

  7. Hong XP, Peng CX, Wei W, Tian Q, Liu YH, Yao XQ, et al. Essential role of tau phosphorylation in adult hippocampal neurogenesis. Hippocampus. 2010;20:1339–49.

    Article  CAS  PubMed  Google Scholar 

  8. Ahmed T, Van der Jeugd A, Blum D, Galas MC, D’Hooge R, Buee L, et al. Cognition and hippocampal synaptic plasticity in mice with a homozygous tau deletion. Neurobiol Aging. 2014;35:2474–8.

    Article  CAS  PubMed  Google Scholar 

  9. Lei P, Ayton S, Finkelstein DI, Spoerri L, Ciccotosto GD, Wright DK, et al. Tau deficiency induces parkinsonism with dementia by impairing APP-mediated iron export. Nat Med. 2012;18:291–5.

    Article  CAS  PubMed  Google Scholar 

  10. Elie A, Prezel E, Guerin C, Denarier E, Ramirez-Rios S, Serre L, et al. Tau co-organizes dynamic microtubule and actin networks. Sci Rep. 2015;5:9964.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Violet M, Delattre L, Tardivel M, Sultan A, Chauderlier A, Caillierez R, et al. A major role for Tau in neuronal DNA and RNA protection in vivo under physiological and hyperthermic conditions. Front Cell Neurosci. 2014;8:84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Caffrey TM, Wade-Martins R. Functional MAPT haplotypes: bridging the gap between genotype and neuropathology. Neurobiol Dis. 2007;27:1–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Evans W, Fung HC, Steele J, Eerola J, Tienari P, Pittman A, et al. The tau H2 haplotype is almost exclusively Caucasian in origin. Neurosci Lett. 2004;369:183–5.

    Article  CAS  PubMed  Google Scholar 

  14. Caillet-Boudin ML, Buee L, Sergeant N, Lefebvre B. Regulation of human MAPT gene expression. Mol Neurodegener. 2015;10:28.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA. Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron. 1989;3:519–26.

    Article  CAS  PubMed  Google Scholar 

  16. Chen J, Kanai Y, Cowan NJ, Hirokawa N. Projection domains of MAP2 and tau determine spacings between microtubules in dendrites and axons. Nature. 1992;360:674–7.

    Article  CAS  PubMed  Google Scholar 

  17. Brandt R, Leger J, Lee G. Interaction of tau with the neural plasma membrane mediated by tau’s amino-terminal projection domain. J Cell Biol. 1995;131:1327–40.

    Article  CAS  PubMed  Google Scholar 

  18. Kellogg EH, Hejab NMA, Poepsel S, Downing KH, DiMaio F, Nogales E. Near-atomic model of microtubule-tau interactions. Science. 2018;360:1242–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Schwalbe M, Ozenne V, Bibow S, Jaremko M, Jaremko L, Gajda M, et al. Predictive atomic resolution descriptions of intrinsically disordered hTau40 and alpha-synuclein in solution from NMR and small angle scattering. Structure. 2014;22:238–49.

    Article  CAS  PubMed  Google Scholar 

  20. Jeganathan S, von Bergen M, Brutlach H, Steinhoff HJ, Mandelkow E. Global hairpin folding of tau in solution. Biochemistry. 2006;45:2283–93.

    Article  CAS  PubMed  Google Scholar 

  21. Morris M, Knudsen GM, Maeda S, Trinidad JC, Ioanoviciu A, Burlingame AL, et al. Tau post-translational modifications in wild-type and human amyloid precursor protein transgenic mice. Nat Neurosci. 2015;18:1183–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Maphis N, Xu G, Kokiko-Cochran ON, Jiang S, Cardona A, Ransohoff RM, et al. Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain. Brain. 2015;138:1738–55.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Ising C, Venegas C, Zhang S, Scheiblich H, Schmidt SV, Vieira-Saecker A, et al. NLRP3 inflammasome activation drives tau pathology. Nature. 2019;575:669–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ryan NS, Rossor MN, Fox NC. Alzheimer’s disease in the 100 years since Alzheimer’s death. Brain. 2015;138:3816–21.

    Article  PubMed  Google Scholar 

  25. Fitzpatrick AWP, Falcon B, He S, Murzin AG, Murshudov G, Garringer HJ, et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature. 2017;547:185–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Irwin DJ, Brettschneider J, McMillan CT, Cooper F, Olm C, Arnold SE, et al. Deep clinical and neuropathological phenotyping of Pick disease. Ann Neurol. 2016;79:272–87.

    Article  CAS  PubMed  Google Scholar 

  27. Ahmed Z, Bigio EH, Budka H, Dickson DW, Ferrer I, Ghetti B, et al. Globular glial tauopathies (GGT): consensus recommendations. Acta Neuropathol. 2013;126:537–44.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Komori T, Arai N, Oda M, Nakayama H, Mori H, Yagishita S, et al. Astrocytic plaques and tufts of abnormal fibers do not coexist in corticobasal degeneration and progressive supranuclear palsy. Acta Neuropathol. 1998;96:401–8.

    Article  CAS  PubMed  Google Scholar 

  29. Falcon B, Zhang W, Murzin AG, Murshudov G, Garringer HJ, Vidal R, et al. Structures of filaments from Pick’s disease reveal a novel tau protein fold. Nature. 2018;561:137–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang W, Tarutani A, Newell KL, Murzin AG, Matsubara T, Falcon B. Novel tau filament fold in corticobasal degeneration. Nature. 2020;580:283–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Goedert M, Spillantini MG, Cairns NJ, Crowther RA. Tau proteins of Alzheimer paired helical filaments: abnormal phosphorylation of all six brain isoforms. Neuron. 1992;8:159–68.

    Article  CAS  PubMed  Google Scholar 

  32. Mendoza J, Sekiya M, Taniguchi T, Iijima KM, Wang R, Ando K. Global analysis of phosphorylation of tau by the checkpoint kinases Chk1 and Chk2 in vitro. J Proteome Res. 2013;12:2654–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Avila J. Tau kinases and phosphatases. J Cell Mol Med. 2008;12:258–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Biernat J, Gustke N, Drewes G, Mandelkow EM, Mandelkow E. Phosphorylation of Ser262 strongly reduces binding of tau to microtubules: distinction between PHF-like immunoreactivity and microtubule binding. Neuron. 1993;11:153–63.

    Article  CAS  PubMed  Google Scholar 

  35. Ando K, Oka M, Ohtake Y, Hayashishita M, Shimizu S, Hisanaga S, et al. Tau phosphorylation at Alzheimer’s disease-related Ser356 contributes to tau stabilization when PAR-1/MARK activity is elevated. Biochem Biophys Res Commun. 2016;478:929–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Zempel H, Mandelkow E. Lost after translation: missorting of Tau protein and consequences for Alzheimer disease. Trends Neurosci. 2014;37:721–32.

    Article  CAS  PubMed  Google Scholar 

  37. Wegmann S, Eftekharzadeh B, Tepper K, Zoltowska KM, Bennett RE, Dujardin S. Tau protein liquid-liquid phase separation can initiate tau aggregation. EMBO J. 2018;37:e98049

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Jeganathan S, Hascher A, Chinnathambi S, Biernat J, Mandelkow EM, Mandelkow E. Proline-directed pseudo-phosphorylation at AT8 and PHF1 epitopes induces a compaction of the paperclip folding of Tau and generates a pathological (MC-1) conformation. J Biol Chem. 2008;283:32066–76.

    Article  CAS  PubMed  Google Scholar 

  39. Arendt T, Stieler J, Strijkstra AM, Hut RA, Rudiger J, Van der Zee EA, et al. Reversible paired helical filament-like phosphorylation of tau is an adaptive process associated with neuronal plasticity in hibernating animals. J Neurosci. 2003;23:6972–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gamblin TC, Chen F, Zambrano A, Abraha A, Lagalwar S, Guillozet AL, et al. Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer’s disease. Proc Natl Acad Sci USA. 2003;100:10032–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Novak M, Kabat J, Wischik CM. Molecular characterization of the minimal protease resistant tau unit of the Alzheimer’s disease paired helical filament. EMBO J. 1993;12:365–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhang Z, Song M, Liu X, Kang SS, Kwon IS, Duong DM, et al. Cleavage of tau by asparagine endopeptidase mediates the neurofibrillary pathology in Alzheimer’s disease. Nat Med. 2014;20:1254–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Horowitz PM, Patterson KR, Guillozet-Bongaarts AL, Reynolds MR, Carroll CA, Weintraub ST, et al. Early N-terminal changes and caspase-6 cleavage of tau in Alzheimer’s disease. J Neurosci. 2004;24:7895–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rohn TT, Rissman RA, Davis MC, Kim YE, Cotman CW, Head E. Caspase-9 activation and caspase cleavage of tau in the Alzheimer’s disease brain. Neurobiol Dis. 2002;11:341–54.

    Article  CAS  PubMed  Google Scholar 

  45. Min SW, Chen X, Tracy TE, Li Y, Zhou Y, Wang C, et al. Critical role of acetylation in tau-mediated neurodegeneration and cognitive deficits. Nat Med. 2015;21:1154–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Sohn PD, Tracy TE, Son HI, Zhou Y, Leite RE, Miller BL, et al. Acetylated tau destabilizes the cytoskeleton in the axon initial segment and is mislocalized to the somatodendritic compartment. Mol Neurodegener. 2016;11:47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Cohen TJ, Guo JL, Hurtado DE, Kwong LK, Mills IP, Trojanowski JQ, et al. The acetylation of tau inhibits its function and promotes pathological tau aggregation. Nat Commun. 2011;2:252.

    Article  PubMed  Google Scholar 

  48. Dan A, Takahashi M, Masuda-Suzukake M, Kametani F, Nonaka T, Kondo H, et al. Extensive deamidation at asparagine residue 279 accounts for weak immunoreactivity of tau with RD4 antibody in Alzheimer’s disease brain. Acta Neuropathol Commun. 2013;1:54.

    Article  PubMed  PubMed Central  Google Scholar 

  49. Thomas SN, Funk KE, Wan Y, Liao Z, Davies P, Kuret J, et al. Dual modification of Alzheimer’s disease PHF-tau protein by lysine methylation and ubiquitylation: a mass spectrometry approach. Acta Neuropathol. 2012;123:105–17.

    Article  CAS  PubMed  Google Scholar 

  50. Liu F, Zaidi T, Iqbal K, Grundke-Iqbal I, Gong CX. Aberrant glycosylation modulates phosphorylation of tau by protein kinase A and dephosphorylation of tau by protein phosphatase 2A and 5. Neuroscience. 2002;115:829–37.

    Article  CAS  PubMed  Google Scholar 

  51. Liu F, Iqbal K, Grundke-Iqbal I, Hart GW, Gong CX. O-GlcNAcylation regulates phosphorylation of tau: a mechanism involved in Alzheimer’s disease. Proc Natl Acad Sci USA. 2004;101:10804–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Cook C, Carlomagno Y, Gendron TF, Dunmore J, Scheffel K, Stetler C, et al. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum Mol Genet. 2014;23:104–16.

    Article  CAS  PubMed  Google Scholar 

  53. Poorkaj P, Bird TD, Wijsman E, Nemens E, Garruto RM, Anderson L, et al. Tau is a candidate gene for chromosome 17 frontotemporal dementia. Ann Neurol. 1998;43:815–25.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  55. Spillantini MG, Murrell JR, Goedert M, Farlow MR, Klug A, Ghetti B. Mutation in the tau gene in familial multiple system tauopathy with presenile dementia. Proc Natl Acad Sci USA. 1998;95:7737–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Strang KH, Golde TE, Giasson BI. MAPT mutations, tauopathy, and mechanisms of neurodegeneration. Lab Investig. 2019;99:912–28.

    Article  PubMed  Google Scholar 

  57. Hong M, Zhukareva V, Vogelsberg-Ragaglia V, Wszolek Z, Reed L, Miller BI, et al. Mutation-specific functional impairments in distinct tau isoforms of hereditary FTDP-17. Science. 1998;282:1914–7.

    Article  CAS  PubMed  Google Scholar 

  58. D’Souza I, Poorkaj P, Hong M, Nochlin D, Lee VM, Bird TD, et al. Missense and silent tau gene mutations cause frontotemporal dementia with parkinsonism-chromosome 17 type, by affecting multiple alternative RNA splicing regulatory elements. Proc Natl Acad Sci USA. 1999;96:5598–603.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Pickering-Brown SM, Baker M, Nonaka T, Ikeda K, Sharma S, Mackenzie J, et al. Frontotemporal dementia with Pick-type histology associated with Q336R mutation in the tau gene. Brain. 2004;127:1415–26.

    Article  CAS  PubMed  Google Scholar 

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

    Article  Google Scholar 

  61. Alonso AC, Zaidi T, Grundke-Iqbal I, Iqbal K. Role of abnormally phosphorylated tau in the breakdown of microtubules in Alzheimer disease. Proc Natl Acad Sci USA. 1994;91:5562–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Yu A, Fox SG, Cavallini A, Kerridge C, O’Neill MJ, Wolak J, et al. Tau protein aggregates inhibit the protein-folding and vesicular trafficking arms of the cellular proteostasis network. J Biol Chem. 2019;294:7917–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Keck S, Nitsch R, Grune T, Ullrich O. Proteasome inhibition by paired helical filament-tau in brains of patients with Alzheimer’s disease. J Neurochem. 2003;85:115–22.

    Article  CAS  PubMed  Google Scholar 

  64. Mandelkow EM, Stamer K, Vogel R, Thies E, Mandelkow E. Clogging of axons by tau, inhibition of axonal traffic and starvation of synapses. Neurobiol Aging. 2003;24:1079–85.

    Article  CAS  PubMed  Google Scholar 

  65. Cowan CM, Mudher A. Are tau aggregates toxic or protective in tauopathies? Front Neurol. 2013;4:114.

    Article  PubMed  PubMed Central  Google Scholar 

  66. Tenreiro S, Eckermann K, Outeiro TF. Protein phosphorylation in neurodegeneration: friend or foe? Front Mol Neurosci. 2014;7:42.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. de Calignon A, Polydoro M, Suarez-Calvet M, William C, Adamowicz DH, Kopeikina KJ, et al. Propagation of tau pathology in a model of early Alzheimer’s disease. Neuron. 2012;73:685–97.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Iba M, Guo JL, McBride JD, Zhang B, Trojanowski JQ, Lee VM. Synthetic tau fibrils mediate transmission of neurofibrillary tangles in a transgenic mouse model of Alzheimer’s-like tauopathy. J Neurosci. 2013;33:1024–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Cho H, Choi JY, Hwang MS, Kim YJ, Lee HM, Lee HS, et al. In vivo cortical spreading pattern of tau and amyloid in the Alzheimer disease spectrum. Ann Neurol. 2016;80:247–58.

    Article  CAS  PubMed  Google Scholar 

  70. Clavaguera F, Bolmont T, Crowther RA, Abramowski D, Frank S, Probst A, et al. Transmission and spreading of tauopathy in transgenic mouse brain. Nat Cell Biol. 2009;11:909–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Clavaguera F, Akatsu H, Fraser G, Crowther RA, Frank S, Hench J, et al. Brain homogenates from human tauopathies induce tau inclusions in mouse brain. Proc Natl Acad Sci USA. 2013;110:9535–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Holmes BB, Furman JL, Mahan TE, Yamasaki TR, Mirbaha H, Eades WC, et al. Proteopathic tau seeding predicts tauopathy in vivo. Proc Natl Acad Sci USA. 2014;111:E4376–85.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Woerman AL, Aoyagi A, Patel S, Kazmi SA, Lobach I, Grinberg LT, et al. Tau prions from Alzheimer’s disease and chronic traumatic encephalopathy patients propagate in cultured cells. Proc Natl Acad Sci USA. 2016;113:E8187–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yamada K, Holth JK, Liao F, Stewart FR, Mahan TE, Jiang H, et al. Neuronal activity regulates extracellular tau in vivo. J Exp Med. 2014;211:387–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Rodriguez L, Mohamed NV, Desjardins A, Lippe R, Fon EA, Leclerc N. Rab7A regulates tau secretion. J Neurochem. 2017;141:592–605.

    Article  CAS  PubMed  Google Scholar 

  76. Mohamed NV, Desjardins A, Leclerc N. Tau secretion is correlated to an increase of Golgi dynamics. PLoS ONE. 2017;12:e0178288.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Dujardin S, Begard S, Caillierez R, Lachaud C, Delattre L, Carrier S, et al. Ectosomes: a new mechanism for non-exosomal secretion of tau protein. PLoS ONE. 2014;9:e100760.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Wang Y, Balaji V, Kaniyappan S, Kruger L, Irsen S, Tepper K, et al. The release and trans-synaptic transmission of Tau via exosomes. Mol Neurodegener. 2017;12:5.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  79. Asai H, Ikezu S, Tsunoda S, Medalla M, Luebke J, Haydar T, et al. Depletion of microglia and inhibition of exosome synthesis halt tau propagation. Nat Neurosci. 2015;18:1584–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Stern RA, Tripodis Y, Baugh CM, Fritts NG, Martin BM, Chaisson C, et al. Preliminary study of plasma exosomal tau as a potential biomarker for chronic traumatic encephalopathy. J Alzheimers Dis. 2016;51:1099–109.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Cope TE, Rittman T, Borchert RJ, Jones PS, Vatansever D, Allinson K, et al. Tau burden and the functional connectome in Alzheimer’s disease and progressive supranuclear palsy. Brain. 2018;141:550–67.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Hoenig MC, Bischof GN, Seemiller J, Hammes J, Kukolja J, Onur OA, et al. Networks of tau distribution in Alzheimer’s disease. Brain. 2018;141:568–81.

    Article  PubMed  Google Scholar 

  83. Rauch JN, Luna G, Guzman E, Audouard M, Challis C, Sibih YE, et al. LRP1 is a master regulator of tau uptake and spread. Nature. 2020;580:381–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Bertram L, Tanzi RE. The genetic epidemiology of neurodegenerative disease. J Clin Investig. 2005;115:1449–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Hoglinger GU, Melhem NM, Dickson DW, Sleiman PM, Wang LS, Klei L, et al. Identification of common variants influencing risk of the tauopathy progressive supranuclear palsy. Nat Genet. 2011;43:699–705.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Houlden H, Baker M, Morris HR, MacDonald N, Pickering-Brown S, Adamson J, et al. Corticobasal degeneration and progressive supranuclear palsy share a common tau haplotype. Neurology. 2001;56:1702–6.

    Article  CAS  PubMed  Google Scholar 

  87. Pastor P, Moreno F, Clarimon J, Ruiz A, Combarros O, Calero M, et al. MAPT H1 haplotype is associated with late-onset Alzheimer’s disease risk in APOEvarepsilon4 noncarriers: results from the Dementia Genetics Spanish Consortium. J Alzheimers Dis. 2016;49:343–52.

    Article  CAS  PubMed  Google Scholar 

  88. Allen M, Kachadoorian M, Quicksall Z, Zou F, Chai HS, Younkin C, et al. Association of MAPT haplotypes with Alzheimer’s disease risk and MAPT brain gene expression levels. Alzheimers Res Ther. 2014;6:39.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  89. Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, et al. Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA. 1993;90:1977–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Verghese PB, Castellano JM, Garai K, Wang Y, Jiang H, Shah A, et al. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc Natl Acad Sci USA. 2013;110:E1807–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhao N, Liu CC, Van Ingelgom AJ, Linares C, Kurti A, Knight JA, et al. APOE epsilon2 is associated with increased tau pathology in primary tauopathy. Nat Commun. 2018;9:4388.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Gale SC, Gao L, Mikacenic C, Coyle SM, Rafaels N, Murray Dudenkov T, et al. APOepsilon4 is associated with enhanced in vivo innate immune responses in human subjects. J Allergy Clin Immunol. 2014;134:127–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Vitek MP, Brown CM, Colton CA. APOE genotype-specific differences in the innate immune response. Neurobiol Aging. 2009;30:1350–60.

    Article  CAS  PubMed  Google Scholar 

  94. Murphy AJ, Akhtari M, Tolani S, Pagler T, Bijl N, Kuo CL, et al. ApoE regulates hematopoietic stem cell proliferation, monocytosis, and monocyte accumulation in atherosclerotic lesions in mice. J Clin Investig. 2011;121:4138–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Laskowitz DT, Lee DM, Schmechel D, Staats HF. Altered immune responses in apolipoprotein E-deficient mice. J Lipid Res. 2000;41:613–20.

    Article  CAS  PubMed  Google Scholar 

  96. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat Genet. 2013;45:1452–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Steele NZ, Carr JS, Bonham LW, Geier EG, Damotte V, Miller ZA, et al. Fine-mapping of the human leukocyte antigen locus as a risk factor for Alzheimer disease: a case-control study. PLoS Med. 2017;14:e1002272.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. James LM, Georgopoulos AP. Human leukocyte antigen as a key factor in preventing dementia and associated apolipoprotein E4 risk. Front Aging Neurosci. 2019;11:82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Vitale C, Romagnani C, Falco M, Ponte M, Vitale M, Moretta A, et al. Engagement of p75/AIRM1 or CD33 inhibits the proliferation of normal or leukemic myeloid cells. Proc Natl Acad Sci USA. 1999;96:15091–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Khera R, Das N. Complement Receptor 1: disease associations and therapeutic implications. Mol Immunol. 2009;46:761–72.

    Article  CAS  PubMed  Google Scholar 

  101. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer’s disease. Nat Genet. 2017;49:1373–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Cruchaga C, Kauwe JS, Harari O, Jin SC, Cai Y, Karch CM, et al. GWAS of cerebrospinal fluid tau levels identifies risk variants for Alzheimer’s disease. Neuron. 2013;78:256–68.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Bis JC, Jian X, Kunkle BW, Chen Y, Hamilton-Nelson KL, Bush WS. Whole exome sequencing study identifies novel rare and common Alzheimer’s-associated variants involved in immune response and transcriptional regulation. Mol Psychiatry. 2020;25:1859–75.

    Article  CAS  PubMed  Google Scholar 

  104. Yokoyama JS, Karch CM, Fan CC, Bonham LW, Kouri N, Ross OA, et al. Shared genetic risk between corticobasal degeneration, progressive supranuclear palsy, and frontotemporal dementia. Acta Neuropathol. 2017;133:825–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Broce I, Karch CM, Wen N, Fan CC, Wang Y, Tan CH, et al. Immune-related genetic enrichment in frontotemporal dementia: an analysis of genome-wide association studies. PLoS Med. 2018;15:e1002487.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  106. Yokoyama JS, Wang Y, Schork AJ, Thompson WK, Karch CM, Cruchaga C, et al. Association between genetic traits for immune-mediated diseases and Alzheimer disease. JAMA Neurol. 2016;73:691–7.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Li Q, Barres BA. Microglia and macrophages in brain homeostasis and disease. Nat Rev Immunol. 2018;18:225–42.

    Article  CAS  PubMed  Google Scholar 

  108. Serrano-Pozo A, Mielke ML, Gomez-Isla T, Betensky RA, Growdon JH, Frosch MP, et al. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer’s disease. Am J Pathol. 2011;179:1373–84.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Paulus W, Bancher C, Jellinger K. Microglial reaction in Pick’s disease. Neurosci Lett. 1993;161:89–92.

    Article  CAS  PubMed  Google Scholar 

  110. Gerhard A, Trender-Gerhard I, Turkheimer F, Quinn NP, Bhatia KP, Brooks DJ. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in progressive supranuclear palsy. Mov Disord. 2006;21:89–93.

    Article  PubMed  Google Scholar 

  111. Henkel K, Karitzky J, Schmid M, Mader I, Glatting G, Unger JW, et al. Imaging of activated microglia with PET and [11C]PK 11195 in corticobasal degeneration. Mov Disord. 2004;19:817–21.

    Article  PubMed  Google Scholar 

  112. Bolos M, Llorens-Martin M, Perea JR, Jurado-Arjona J, Rabano A, Hernandez F, et al. Absence of CX3CR1 impairs the internalization of Tau by microglia. Mol Neurodegener. 2017;12:59.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YE, et al. Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci. 2018;21:1049–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Morales I, Jimenez JM, Mancilla M, Maccioni RB. Tau oligomers and fibrils induce activation of microglial cells. J Alzheimers Dis. 2013;37:849–56.

    Article  CAS  PubMed  Google Scholar 

  115. Hoek RM, Ruuls SR, Murphy CA, Wright GJ, Goddard R, Zurawski SM, et al. Down-regulation of the macrophage lineage through interaction with OX2 (CD200). Science. 2000;290:1768–71.

    Article  CAS  PubMed  Google Scholar 

  116. Kim YS, Choi DH, Block ML, Lorenzl S, Yang L, Kim YJ, et al. A pivotal role of matrix metalloproteinase-3 activity in dopaminergic neuronal degeneration via microglial activation. FASEB J. 2007;21:179–87.

    Article  CAS  PubMed  Google Scholar 

  117. Tang Y, Le W. Differential roles of M1 and M2 microglia in neurodegenerative diseases. Mol Neurobiol. 2016;53:1181–94.

    Article  CAS  PubMed  Google Scholar 

  118. Molofsky AV, Krencik R, Ullian EM, Tsai HH, Deneen B, Richardson WD, et al. Astrocytes and disease: a neurodevelopmental perspective. Genes Dev. 2012;26:891–907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Li K, Li J, Zheng J, Qin S. Reactive astrocytes in neurodegenerative diseases. Aging Dis. 2019;10:664–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541:481–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Lueg G, Gross CC, Lohmann H, Johnen A, Kemmling A, Deppe M, et al. Clinical relevance of specific T-cell activation in the blood and cerebrospinal fluid of patients with mild Alzheimer’s disease. Neurobiol Aging. 2015;36:81–9.

    Article  CAS  PubMed  Google Scholar 

  122. Togo T, Akiyama H, Iseki E, Kondo H, Ikeda K, Kato M, et al. Occurrence of T cells in the brain of Alzheimer’s disease and other neurological diseases. J Neuroimmunol. 2002;124:83–92.

    Article  CAS  PubMed  Google Scholar 

  123. Ferretti MT, Merlini M, Spani C, Gericke C, Schweizer N, Enzmann G, et al. T-cell brain infiltration and immature antigen-presenting cells in transgenic models of Alzheimer’s disease-like cerebral amyloidosis. Brain Behav Immun. 2016;54:211–25.

    Article  CAS  PubMed  Google Scholar 

  124. Gate D, Saligrama N, Leventhal O, Yang AC, Unger MS, Middeldorp J, et al. Clonally expanded CD8 T cells patrol the cerebrospinal fluid in Alzheimer’s disease. Nature. 2020;577:399–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  125. Doherty CP, O’Keefe E, Wallace E, Loftus T, Keaney J, Kealy J, et al. Blood–brain barrier dysfunction as a hallmark pathology in chronic traumatic encephalopathy. J Neuropathol Exp Neurol. 2016;75:656–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Yates PA, Desmond PM, Phal PM, Steward C, Szoeke C, Salvado O, et al. Incidence of cerebral microbleeds in preclinical Alzheimer disease. Neurology. 2014;82:1266–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Haruwaka K, Ikegami A, Tachibana Y, Ohno N, Konishi H, Hashimoto A, et al. Dual microglia effects on blood brain barrier permeability induced by systemic inflammation. Nat Commun. 2019;10:5816.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Kovac A, Zilkova M, Deli MA, Zilka N, Novak M. Human truncated tau is using a different mechanism from amyloid-beta to damage the blood-brain barrier. J Alzheimers Dis. 2009;18:897–906.

    Article  CAS  PubMed  Google Scholar 

  129. Blair LJ, Frauen HD, Zhang B, Nordhues BA, Bijan S, Lin YC, et al. Tau depletion prevents progressive blood-brain barrier damage in a mouse model of tauopathy. Acta Neuropathol Commun. 2015;3:8.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Zotova E, Bharambe V, Cheaveau M, Morgan W, Holmes C, Harris S, et al. Inflammatory components in human Alzheimer’s disease and after active amyloid-beta42 immunization. Brain. 2013;136:2677–96.

    Article  PubMed  Google Scholar 

  131. Laurent C, Dorothee G, Hunot S, Martin E, Monnet Y, Duchamp M, et al. Hippocampal T cell infiltration promotes neuroinflammation and cognitive decline in a mouse model of tauopathy. Brain. 2017;140:184–200.

    Article  PubMed  Google Scholar 

  132. Browne TC, McQuillan K, McManus RM, O’Reilly JA, Mills KH, Lynch MA. IFN-gamma Production by amyloid beta-specific Th1 cells promotes microglial activation and increases plaque burden in a mouse model of Alzheimer’s disease. J Immunol. 2013;190:2241–51.

    Article  CAS  PubMed  Google Scholar 

  133. Dansokho C, Ait Ahmed D, Aid S, Toly-Ndour C, Chaigneau T, Calle V, et al. Regulatory T cells delay disease progression in Alzheimer-like pathology. Brain. 2016;139:1237–51.

    Article  PubMed  Google Scholar 

  134. Baek H, Ye M, Kang GH, Lee C, Lee G, Choi DB, et al. Neuroprotective effects of CD4+CD25+Foxp3+ regulatory T cells in a 3xTg-AD Alzheimer’s disease model. Oncotarget. 2016;7:69347–57.

    Article  PubMed  PubMed Central  Google Scholar 

  135. Mittal K, Eremenko E, Berner O, Elyahu Y, Strominger I, Apelblat D, et al. CD4 T cells induce a subset of MHCII-expressing microglia that attenuates Alzheimer pathology. iScience. 2019;16:298–311.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Hauser SL, Goodin DS. Chapter 458. Multiple sclerosis and other demyelinating diseases. In: Kasper DL, Fauci AS, Hauser SL, Longo DL, Jameson JL, Loscalzo J, editors. Harrison’s principles of internal medicine, 19th ed. New York, NY: The McGraw-Hill Education; 2015.

  137. International Multiple Sclerosis Genetics Consortium. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science. 2019;365:Science

    Article  CAS  Google Scholar 

  138. Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502–17.

    Article  CAS  PubMed  Google Scholar 

  139. Anderson JM, Hampton DW, Patani R, Pryce G, Crowther RA, Reynolds R, et al. Abnormally phosphorylated tau is associated with neuronal and axonal loss in experimental autoimmune encephalomyelitis and multiple sclerosis. Brain. 2008;131:1736–48.

    Article  CAS  PubMed  Google Scholar 

  140. Frid K, Einstein O, Friedman-Levi Y, Binyamin O, Ben-Hur T, Gabizon R. Aggregation of MBP in chronic demyelination. Ann Clin Transl Neurol. 2015;2:711–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Schneider A, Araujo GW, Trajkovic K, Herrmann MM, Merkler D, Mandelkow EM, et al. Hyperphosphorylation and aggregation of tau in experimental autoimmune encephalomyelitis. J Biol Chem. 2004;279:55833–9.

    Article  CAS  PubMed  Google Scholar 

  142. Anderson JM, Patani R, Reynolds R, Nicholas R, Compston A, Spillantini MG, et al. Evidence for abnormal tau phosphorylation in early aggressive multiple sclerosis. Acta Neuropathol. 2009;117:583–9.

    Article  CAS  PubMed  Google Scholar 

  143. Didonna A, Canto E, Shams H, Isobe N, Zhao C, Caillier SJ, et al. Sex-specific Tau methylation patterns and synaptic transcriptional alterations are associated with neural vulnerability during chronic neuroinflammation. J Autoimmun. 2019;101:56–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Kurnellas MP, Ghosn EE, Schartner JM, Baker J, Rothbard JJ, Negrin RS, et al. Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease. Proc Natl Acad Sci USA. 2015;112:15016–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2003;74:1200–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Yoshiyama Y, Higuchi M, Zhang B, Huang SM, Iwata N, Saido TC, et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron. 2007;53:337–51.

    Article  CAS  PubMed  Google Scholar 

  147. Gorlovoy P, Larionov S, Pham TT, Neumann H. Accumulation of tau induced in neurites by microglial proinflammatory mediators. FASEB J. 2009;23:2502–13.

    Article  CAS  PubMed  Google Scholar 

  148. Kitazawa M, Oddo S, Yamasaki TR, Green KN, LaFerla FM. Lipopolysaccharide-induced inflammation exacerbates tau pathology by a cyclin-dependent kinase 5-mediated pathway in a transgenic model of Alzheimer’s disease. J Neurosci. 2005;25:8843–53.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Bhaskar K, Konerth M, Kokiko-Cochran ON, Cardona A, Ransohoff RM, Lamb BT. Regulation of tau pathology by the microglial fractalkine receptor. Neuron. 2010;68:19–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Spangenberg EE, Lee RJ, Najafi AR, Rice RA, Elmore MR, Blurton-Jones M, et al. Eliminating microglia in Alzheimer’s mice prevents neuronal loss without modulating amyloid-beta pathology. Brain. 2016;139:1265–81.

    Article  PubMed  PubMed Central  Google Scholar 

  151. Zhou M, Xu R, Kaelber DC, Gurney ME. Tumor necrosis factor (TNF) blocking agents are associated with lower risk for Alzheimer’s disease in patients with rheumatoid arthritis and psoriasis. PLoS ONE. 2020;15:e0229819.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  152. Weiner HL, Lemere CA, Maron R, Spooner ET, Grenfell TJ, Mori C, et al. Nasal administration of amyloid-beta peptide decreases cerebral amyloid burden in a mouse model of Alzheimer’s disease. Ann Neurol. 2000;48:567–79.

    Article  CAS  PubMed  Google Scholar 

  153. Howard R, Zubko O, Bradley R, Harper E, Pank L, O’Brien J, et al. Minocycline at 2 different dosages vs. placebo for patients with mild Alzheimer disease: a randomized clinical trial. JAMA Neurol. 2020;77:164–74.

    Article  PubMed  Google Scholar 

  154. Gyengesi E, Munch G. In search of an anti-inflammatory drug for Alzheimer disease. Nat Rev Neurol. 2020;16:131–2.

    Article  CAS  PubMed  Google Scholar 

  155. Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, et al. Global prevalence of dementia: a Delphi consensus study. Lancet. 2005;366:2112–7.

    Article  PubMed  PubMed Central  Google Scholar 

  156. Jansen IE, Savage JE, Watanabe K, Bryois J, Williams DM, Steinberg S, et al. Genome-wide meta-analysis identifies new loci and functional pathways influencing Alzheimer’s disease risk. Nat Genet. 2019;51:404–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Im SY, Kim YE, Kim YJ. Genetics of progressive supranuclear palsy. J Mov Disord. 2015;8:122–9.

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  159. Kouri N, Ross OA, Dombroski B, Younkin CS, Serie DJ, Soto-Ortolaza A, et al. Genome-wide association study of corticobasal degeneration identifies risk variants shared with progressive supranuclear palsy. Nat Commun. 2015;6:7247.

    Article  CAS  PubMed  Google Scholar 

  160. Togasaki DM, Tanner CM. Epidemiologic aspects. Adv Neurol. 2000;82:53–9.

    CAS  PubMed  Google Scholar 

  161. Dickson DW. Pick’s disease: a modern approach. Brain Pathol. 1998;8:339–54.

    Article  CAS  PubMed  Google Scholar 

  162. Tacik P, DeTure M, Hinkle KM, Lin WL, Sanchez-Contreras M, Carlomagno Y, et al. A novel tau mutation in exon 12, p.Q336H, causes hereditary pick disease. J Neuropathol Exp Neurol. 2015;74:1042–52.

    Article  CAS  PubMed  Google Scholar 

  163. Kasuga K, Kikuchi M, Tokutake T, Nakaya A, Tezuka T, Tsukie T, et al. Systematic review and meta-analysis of Japanese familial Alzheimer’s disease and FTDP-17. J Hum Genet. 2015;60:281–3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Kovacs GG, Pittman A, Revesz T, Luk C, Lees A, Kiss E, et al. MAPT S305I mutation: implications for argyrophilic grain disease. Acta Neuropathol. 2008;116:103–18.

    Article  CAS  PubMed  Google Scholar 

  165. Rodriguez RD, Suemoto CK, Molina M, Nascimento CF, Leite RE, de Lucena Ferretti-Rebustini RE, et al. Argyrophilic grain disease: demographics, clinical, and neuropathological features from a large autopsy study. J Neuropathol Exp Neurol. 2016;75:628–35.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Ferrer I, Andres-Benito P, Zelaya MV, Aguirre MEE, Carmona M, Ausin K, et al. Familial globular glial tauopathy linked to MAPT mutations: molecular neuropathology and seeding capacity of a prototypical mixed neuronal and glial tauopathy. Acta Neuropathol. 2020;139:735–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. McKee AC, Daneshvar DH. The neuropathology of traumatic brain injury. Handb Clin Neurol. 2015;127:45–66.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Hermosura MC, Nayakanti H, Dorovkov MV, Calderon FR, Ryazanov AG, Haymer DS, et al. A TRPM7 variant shows altered sensitivity to magnesium that may contribute to the pathogenesis of two Guamanian neurodegenerative disorders. Proc Natl Acad Sci USA. 2005;102:11510–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Hof PR, Nimchinsky EA, Buee-Scherrer V, Buee L, Nasrallah J, Hottinger AF, et al. Amyotrophic lateral sclerosis/parkinsonism-dementia complex of Guam: quantitative neuropathology, immunohistochemical analysis of neuronal vulnerability, and comparison with related neurodegenerative disorders. Acta Neuropathol. 1994;88:397–404.

    Article  CAS  PubMed  Google Scholar 

  170. Morris HR, Steele JC, Crook R, Wavrant-De Vrieze F, Onstead-Cardinale L, Gwinn-Hardy K, et al. Genome-wide analysis of the parkinsonism-dementia complex of Guam. Arch Neurol. 2004;61:1889–97.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

The author is grateful to Dr Jorge R. Oksenberg and Dr Carolina Alquezar Burillo for critically reading the paper. The work was supported by FISM-Fondazione Italiana Sclerosi Multipla Senior Research Fellowships Cod. 2014/B/1 and Cod. 2017/B/3 and financed or co-financed with the “5 per mille” public funding.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Alessandro Didonna.

Ethics declarations

Conflict of interest

The author declares no conflict of interest.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Didonna, A. Tau at the interface between neurodegeneration and neuroinflammation. Genes Immun 21, 288–300 (2020). https://doi.org/10.1038/s41435-020-00113-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41435-020-00113-5

This article is cited by

Search

Quick links