Skip to main content

Advertisement

Log in

Argyrophilic grain disease differs from other tauopathies by lacking tau acetylation

  • Original Paper
  • Published:
Acta Neuropathologica Aims and scope Submit manuscript

Abstract

Post-translational modifications play a key role in tau protein aggregation and related neurodegeneration. Because hyperphosphorylation alone does not necessarily cause tau aggregation, other post-translational modifications have been recently explored. Tau acetylation promotes aggregation and inhibits tau’s ability to stabilize microtubules. Recent studies have shown co-localization of acetylated and phosphorylated tau in AD and some 4R tauopathies. We developed a novel monoclonal antibody against acetylated tau at lysine residue 274, which recognizes both 3R and 4R tau, and used immunohistochemistry and immunofluorescence to probe 22 cases, including AD and another eight familial or sporadic tauopathies. Acetylated tau was identified in all tauopathies except argyrophilic grain disease (AGD). AGD is an age-associated, common but atypical 4R tauopathy, not always associated with clinical progression. Pathologically, AGD is characterized by neuropil grains, pre-neurofibrillary tangles, and oligodendroglial coiled bodies, all recognized by phospho-tau antibodies. The lack of acetylated tau in these inclusions suggests that AGD represents a distinctive tauopathy. Our data converge with previous findings to raise the hypothesis that AGD could play a protective role against the spread of AD-related tau pathology. Tau acetylation as a key modification for the propagation tau toxicity deserves further investigation.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Ahmed Z, Doherty KM, Silveira-Moriyama L, Bandopadhyay R, Lashley T, Mamais A et al (2011) Globular glial tauopathies (GGT) presenting with motor neuron disease or frontotemporal dementia: an emerging group of 4-repeat tauopathies. Acta Neuropathol 122:28–415

    Article  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  3. Berg L, McKeel DW Jr, Miller JP, Storandt M, Rubin EH, Morris JC et al (1998) Clinicopathologic studies in cognitively healthy aging and Alzheimer’s disease: relation of histologic markers to dementia severity, age, sex, and apolipoprotein E genotype. Arch Neurol 55:326–335

    Article  PubMed  CAS  Google Scholar 

  4. Braak H, Braak E (1989) Cortical and subcortical argyrophilic grains characterize a disease associated with adult onset dementia. Neuropathol Appl Neurobiol 15:13–26

    Article  PubMed  CAS  Google Scholar 

  5. Braak H, Thal DR, Ghebremedhin E, Del Tredici K (2011) Stages of the pathologic process in Alzheimer disease: age categories from 1 to 100 years. J Neuropathol Exp Neurol 70:960–969

    Article  PubMed  CAS  Google Scholar 

  6. Buee L, Bussiere T, Buee-Scherrer V, Delacourte A, Hof PR (2000) Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res Rev 33:95–130

    Article  PubMed  CAS  Google Scholar 

  7. Buee L, Delacourte A (1999) Comparative biochemistry of tau in progressive supranuclear palsy, corticobasal degeneration, FTDP-17 and Pick’s disease. Brain Pathol 9:681–693

    Article  PubMed  CAS  Google Scholar 

  8. Cairns NJ, Bigio EH, Mackenzie IR, Neumann M, Lee VM, Hatanpaa KJ et al (2007) Neuropathologic diagnostic and nosologic criteria for frontotemporal lobar degeneration: consensus of the Consortium for Frontotemporal Lobar Degeneration. Acta Neuropathol 114:5–22

    Article  PubMed  Google Scholar 

  9. Cohen P, Frame S (2001) The renaissance of GSK3. Nat Rev Mol Cell Biol 2:769–776

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  Google Scholar 

  11. Duyckaerts C (2004) Looking for the link between plaques and tangles. Neurobiol Aging 25:735–739

    Article  PubMed  CAS  Google Scholar 

  12. Ferrer I, Barrachina M, Tolnay M, Rey MJ, Vidal N, M Carmona et al (2003) Phosphorylated protein kinases associated with neuronal and glial tau deposits in argyrophilic grain disease. Brain Pathol 13:62–78

    Article  PubMed  CAS  Google Scholar 

  13. Ferrer I, Santpere G, van Leeuwen FW (2008) Argyrophilic grain disease. Brain 131:1416–1432

    Article  PubMed  Google Scholar 

  14. Giannakopoulos P, Gold G, Kovari E, von Gunten A, Imhof A, Bouras C et al (2007) Assessing the cognitive impact of Alzheimer disease pathology and vascular burden in the aging brain: the Geneva experience. Acta Neuropathol 113:1–12

    Article  PubMed  Google Scholar 

  15. Goedert M (2001) The significance of tau and alpha-synuclein inclusions in neurodegenerative diseases. Curr Opin Genet Dev 11:343–351

    Article  PubMed  CAS  Google Scholar 

  16. Goedert M, Spillantini MG, Jakes R, Rutherford D, Crowther RA (1989) Multiple isoforms of human microtubule-associated protein tau: sequences and localization in neurofibrillary tangles of Alzheimer’s disease. Neuron 3:519–526

    Article  PubMed  CAS  Google Scholar 

  17. Goedert M, Spillantini MG, Potier MC, Ulrich J, Crowther RA (1989) Cloning and sequencing of the cDNA encoding an isoform of microtubule-associated protein tau containing four tandem repeats: differential expression of tau protein mRNAs in human brain. EMBO J 8:393–399

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  20. Hodges JR, Davies RR, Xuereb JH, Casey B, Broe M, Bak TH et al (2004) Clinicopathological correlates in frontotemporal dementia. Ann Neurol 56:399–406

    Article  PubMed  Google Scholar 

  21. Ilieva EV, Kichev A, Naudi A, Ferrer I, Pamplona R, Portero-Otin M (2011) Mitochondrial dysfunction and oxidative and endoplasmic reticulum stress in argyrophilic grain disease. J Neuropathol Exp Neurol 70:253–263

    Article  PubMed  CAS  Google Scholar 

  22. Irwin DJ, Cohen TJ, Grossman M, Arnold SE, Xie SX, Lee VM et al (2012) Acetylated tau, a novel pathological signature in Alzheimer’s disease and other tauopathies. Brain 135:807–818

    Article  PubMed  Google Scholar 

  23. Jicha GA, Petersen RC, Knopman DS, Boeve BF, Smith GE, Geda YE et al (2006) Argyrophilic grain disease in demented subjects presenting initially with amnestic mild cognitive impairment. J Neuropathol Exp Neurol 65:602–609

    Article  PubMed  Google Scholar 

  24. Knopman DS, Parisi JE, Salviati A, Floriach-Robert M, Boeve BF, Ivnik RJ et al (2003) Neuropathology of cognitively normal elderly. J Neuropathol Exp Neurol 62:1087–1095

    PubMed  CAS  Google Scholar 

  25. Kovacs GG, Majtenyi K, Spina S, Murrell JR, Gelpi E, Hoftberger R et al (2008) White matter tauopathy with globular glial inclusions: a distinct sporadic frontotemporal lobar degeneration. J Neuropathol Exp Neurol 67:963–975

    Article  PubMed  Google Scholar 

  26. Lee G, Neve RL, Kosik KS (1989) The microtubule binding domain of tau protein. Neuron 2:1615–1624

    Article  PubMed  CAS  Google Scholar 

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

    PubMed  Google Scholar 

  28. Maurage CA, Sergeant N, Schraen-Maschke S, Lebert F, Ruchoux MM, Sablonniere B et al (2003) Diffuse form of argyrophilic grain disease: a new variant of four-repeat tauopathy different from limbic argyrophilic grain disease. Acta Neuropathol 106:575–583

    Article  PubMed  Google Scholar 

  29. Min SW, Cho SH, Zhou Y, Schroeder S, Haroutunian V, Seeley WW et al (2010) Acetylation of tau inhibits its degradation and contributes to tauopathy. Neuron 67:953–966

    Article  PubMed  CAS  Google Scholar 

  30. Petersen RC, Parisi JE, Dickson DW, Johnson KA, Knopman DS, Boeve BF et al (2006) Neuropathologic features of amnestic mild cognitive impairment. Arch Neurol 63:665–672

    Article  PubMed  Google Scholar 

  31. Steuerwald GM, Baumann TP, Taylor KI, Mittag M, Adams H, Tolnay M et al (2007) Clinical characteristics of dementia associated with argyrophilic grain disease. Dement Geriatr Cogn Disord 24:229–234

    Article  PubMed  Google Scholar 

  32. Thal DR, Schultz C, Botez G, Del Tredici K, Mrak RE, Griffin WS et al (2005) The impact of argyrophilic grain disease on the development of dementia and its relationship to concurrent Alzheimer’s disease-related pathology. Neuropathol Appl Neurobiol 31:270–279

    Article  PubMed  CAS  Google Scholar 

  33. Tolnay M, Clavaguera F (2004) Argyrophilic grain disease: a late-onset dementia with distinctive features among tauopathies. Neuropathology 24:269–283

    Article  PubMed  Google Scholar 

  34. Tolnay M, Schwietert M, Monsch AU, Staehelin HB, Langui D, Probst A (1997) Argyrophilic grain disease: distribution of grains in patients with and without dementia. Acta Neuropathol 94:353–358

    Article  PubMed  CAS  Google Scholar 

  35. Tolnay M, Sergeant N, Ghestem A, Chalbot S, De Vos RA, Jansen Steur EN et al (2002) Argyrophilic grain disease and Alzheimer’s disease are distinguished by their different distribution of tau protein isoforms. Acta Neuropathol 104:425–434

    PubMed  CAS  Google Scholar 

Download references

Acknowledgments

We thank Jian Yang, Norbert Lee, and Stephanie Gaus for technical assistance, and our patients and their families for their invaluable contributions to neurodegenerative disease research. Funding was provided by National Institute of Health (NIH) P50 AG023501 to B.L.M. and W.W.S., Tau Consortium (to L.G. and W.W.S.), NIH R01AG030207 (to L.G.), NIH R01AG040311 to L.T.G., the John Douglas French Alzheimer’s Disease Foundation (to L.T.G. and W.W.S.), the Consortium for Frontotemporal Dementia Research (to W.W.S).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Li Gan or William W. Seeley.

Additional information

L. Gan and W. W. Seeley contributed equally.

Electronic supplementary material

Below is the link to the electronic supplementary material.

401_2013_1080_MOESM1_ESM.tif

Online resource 1 Antigen competition assay. a inferior temporal cortex of an AD case (case # 3) after immunohistochemistry with MAb 359. Note the plaques and tangles in dark brown. b higher magnification of (a). The plaques and tangles can be visualized in detail. c a parallel slide to (a) after immunohistochemistry with MAb 359 pre-incubated with the antigen used for its generation. All the other reaction steps were the same as in (a). Note that the reaction was negative, supporting MAb 359 specificity against tau acetylated at position 274. d higher magnification of (b). Scale bars represent 500 μm in a and c, and 100 μm in b and d. (TIFF 4699 kb)

401_2013_1080_MOESM2_ESM.tif

Online resource 2 Inferior temporal cortex of Pick’s disease (case 18) after immunofluorescence with CP-13 (a) and MAb 359 (b) antibodies. Pick bodies are both phosphorylated (a) and acetylated (b). Most of the inclusions have both changes and the processes are mainly phosphorylated only (c). Scale bar represents 50 μm (TIFF 4050 kb)

401_2013_1080_MOESM3_ESM.tif

Online resource 3 Across all tauopathies, neuronal processes were rarely ac-tau positive. This figure shows parallel sections of the hippocampal dentate gyrus granule cell (top of the figure) and molecular layers (bottom of the figure) in Alzheimer’s disease (case 3) immunostained with (a) CP-13 and (b) MAb 359. Note the strong phospho-tau positivity and absent ac-tau positivity in the external portion of the molecular layer. The dentate gyrus molecular layer contains abundant axons and dendrites. This figure demonstrates the remarkable discrepancy between phosphor-tau and ac-tau changes in neuronal processes. The arrow in b shows an acetylated tau-positive neurofibrillary tangle as an internal positive control. (TIFF 2268 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Grinberg, L.T., Wang, X., Wang, C. et al. Argyrophilic grain disease differs from other tauopathies by lacking tau acetylation. Acta Neuropathol 125, 581–593 (2013). https://doi.org/10.1007/s00401-013-1080-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00401-013-1080-2

Keywords

Navigation