Skip to main content

Advertisement

Log in

Phosphorylated tau in the retina correlates with tau pathology in the brain in Alzheimer’s disease and primary tauopathies

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

A Correction to this article was published on 23 December 2022

This article has been updated

Abstract

The retina is a potential source of biomarkers for the detection of neurodegenerative diseases. Accumulation of phosphorylated tau (p-tau) in the brain is a pathological feature characteristic for Alzheimer’s disease (AD) and primary tauopathies. In this study the presence of p-tau in the retina in relation to tau pathology in the brain was assessed. Post-mortem eyes and brains were collected through the Netherlands Brain Bank from donors with AD (n = 17), primary tauopathies (n = 8), α-synucleinopathies (n = 13), other neurodegenerative diseases including non-tau frontotemporal lobar degeneration (FTLD) (n = 9), and controls (n = 15). Retina cross-sections were assessed by immunohistochemistry using antibodies directed against total tau (HT7), 3R and 4R tau isoforms (RD3, RD4), and phospho-epitopes Ser202/Thr205 (AT8), Thr217 (anti-T217), Thr212/Ser214 (AT100), Thr181 (AT270), Ser396 (anti-pS396) and Ser422 (anti-pS422). Retinal tau load was compared to p-tau Ser202/Thr205 and p-tau Thr217 load in various brain regions. Total tau, 3R and 4R tau isoforms were most prominently present in the inner plexiform layer (IPL) and outer plexiform layer (OPL) of the retina and were detected in all cases and controls as a diffuse and somatodendritic signal. Total tau, p-tau Ser202/Thr205 and p-tau Thr217 was observed in amacrine and horizontal cells of the inner nuclear layer (INL). Various antibodies directed against phospho-epitopes of tau showed immunoreactivity in the IPL, OPL, and INL. P-tau Ser202/Thr205 and Thr217 showed significant discrimination between AD and other tauopathies, and non-tauopathy cases including controls. Whilst immunopositivity was observed for p-tau Thr212/Ser214, Thr181 and Ser396, there were no group differences. P-tau Ser422 did not show any immunoreactivity in the retina. The presence of retinal p-tau Ser202/Thr205 and Thr217 correlated with Braak stage for NFTs and with the presence of p-tau Ser202/Thr205 in hippocampus and cortical brain regions. Depending on the phospho-epitope, p-tau in the retina is a potential biomarker for AD and primary tauopathies.

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
Fig. 8

Similar content being viewed by others

Availability of data and materials

Most data generated or analyzed during this study are included in this published manuscript and the supplementary material. Additional data are available from the corresponding author upon reasonable request.

Change history

References

  1. Braak H, Braak E (1991) Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 82(4):239–259. https://doi.org/10.1007/BF00308809

    Article  CAS  Google Scholar 

  2. Braak H, Alafuzoff I, Arzberger T, Kretzschmar H, Del Tredici K (2006) Staging of Alzheimer disease-associated neurofibrillary pathology using paraffin sections and immunocytochemistry. Acta Neuropathol 112(4):389–404. https://doi.org/10.1007/s00401-006-0127-z

    Article  Google Scholar 

  3. Falcon B, Zhang W, Murzin AG, Murshudov G, Garringer HJ, Vidal R et al (2018) Structures of filaments from Pick’s disease reveal a novel tau protein fold. Nature 561(7721):137–140. https://doi.org/10.1038/s41586-018-0454-y

    Article  CAS  Google Scholar 

  4. Zhang W, Tarutani A, Newell KL, Murzin AG, Matsubara T, Falcon B et al (2020) Novel tau filament fold in corticobasal degeneration. Nature 580(7802):283–287. https://doi.org/10.1038/s41586-020-2043-0

    Article  CAS  Google Scholar 

  5. Crary JF, Trojanowski JQ, Schneider JA, Abisambra JF, Abner EL, Alafuzoff I et al (2014) Primary age-related tauopathy (PART): a common pathology associated with human aging. Acta Neuropathol 128(6):755–766. https://doi.org/10.1007/s00401-014-1349-0

    Article  CAS  Google Scholar 

  6. Kovacs GG, Ferrer I, Grinberg LT, Alafuzoff I, Attems J, Budka H et al (2016) Aging-related tau astrogliopathy (ARTAG): harmonized evaluation strategy. Acta Neuropathol 131(1):87–102. https://doi.org/10.1007/s00401-015-1509-x

    Article  CAS  Google Scholar 

  7. London A, Benhar I, Schwartz M (2013) The retina as a window to the brain-from eye research to CNS disorders. Nat Rev Neurol 9(1):44–53. https://doi.org/10.1038/nrneurol.2012.227

    Article  CAS  Google Scholar 

  8. Kaur C, Foulds WS, Ling EA (2008) Blood-retinal barrier in hypoxic ischaemic conditions: basic concepts, clinical features and management. Prog Retin Eye Res 27(6):622–647. https://doi.org/10.1016/j.preteyeres.2008.09.003

    Article  CAS  Google Scholar 

  9. Taylor AW, Streilein JW (1996) Inhibition of antigen-stimulated effector T cells by human cerebrospinal fluid. NeuroImmunoModulation 3(2–3):112–118. https://doi.org/10.1159/000097235

    Article  CAS  Google Scholar 

  10. Wilbanks GA, Streilein JW (1992) Fluids from immune privileged sites endow macrophages with the capacity to induce antigen-specific immune deviation via a mechanism involving transforming growth factor-beta. Eur J Immunol 22(4):1031–1036. https://doi.org/10.1002/eji.1830220423

    Article  CAS  Google Scholar 

  11. Loffler KU, Edward DP, Tso MO (1995) Immunoreactivity against tau, amyloid precursor protein, and beta-amyloid in the human retina. Invest Ophthalmol Vis Sci 36(1):24–31

    CAS  Google Scholar 

  12. Leger F, Fernagut PO, Canron MH, Leoni S, Vital C, Tison F et al (2011) Protein aggregation in the aging retina. J Neuropathol Exp Neurol 70(1):63–68. https://doi.org/10.1097/NEN.0b013e31820376cc

    Article  CAS  Google Scholar 

  13. Schon C, Hoffmann NA, Ochs SM, Burgold S, Filser S, Steinbach S et al (2012) Long-term in vivo imaging of fibrillar tau in the retina of P301S transgenic mice. PLoS ONE 7(12):e53547. https://doi.org/10.1371/journal.pone.0053547

    Article  CAS  Google Scholar 

  14. den Haan J, Morrema THJ, Verbraak FD, de Boer JF, Scheltens P, Rozemuller AJ et al (2018) Amyloid-beta and phosphorylated tau in post-mortem Alzheimer’s disease retinas. Acta Neuropathol Commun 6(1):147. https://doi.org/10.1186/s40478-018-0650-x

    Article  CAS  Google Scholar 

  15. Grimaldi A, Pediconi N, Oieni F, Pizzarelli R, Rosito M, Giubettini M et al (2019) Neuroinflammatory processes, A1 astrocyte activation and protein aggregation in the retina of Alzheimer’s disease patients, possible biomarkers for early diagnosis. Front Neurosci 13:925. https://doi.org/10.3389/fnins.2019.00925

    Article  Google Scholar 

  16. Hinton DR, Sadun AA, Blanks JC, Miller CA (1986) Optic-nerve degeneration in Alzheimer’s disease. N Engl J Med 315(8):485–487. https://doi.org/10.1056/nejm198608213150804

    Article  CAS  Google Scholar 

  17. Ho CY, Troncoso JC, Knox D, Stark W, Eberhart CG (2014) Beta-amyloid, phospho-tau and alpha-synuclein deposits similar to those in the brain are not identified in the eyes of Alzheimer’s and Parkinson’s disease patients. Brain Pathol 24(1):25–32. https://doi.org/10.1111/bpa.12070

    Article  CAS  Google Scholar 

  18. Williams EA, McGuone D, Frosch MP, Hyman BT, Laver N, Stemmer-Rachamimov A (2017) Absence of Alzheimer disease neuropathologic changes in eyes of subjects with alzheimer disease. J Neuropathol Exp Neurol 76(5):376–383. https://doi.org/10.1093/jnen/nlx020

    Article  CAS  Google Scholar 

  19. Klioueva NM, Rademaker MC, Dexter DT, Al-Sarraj S, Seilhean D, Streichenberger N et al (2015) BrainNet Europe’s code of conduct for brain banking. J Neural Transm (Vienna) 122(7):937–940. https://doi.org/10.1007/s00702-014-1353-5

    Article  Google Scholar 

  20. Alafuzoff I, Ince PG, Arzberger T, Al-Sarraj S, Bell J, Bodi I et al (2009) Staging/typing of Lewy body related alpha-synuclein pathology: a study of the BrainNet Europe consortium. Acta Neuropathol 117(6):635–652. https://doi.org/10.1007/s00401-009-0523-2

    Article  CAS  Google Scholar 

  21. Hyman BT, Phelps CH, Beach TG, Bigio EH, Cairns NJ, Carrillo MC et al (2012) National institute on aging-Alzheimer’s association guidelines for the neuropathologic assessment of Alzheimer’s disease. Alzheimers Dement 8(1):1–13. https://doi.org/10.1016/j.jalz.2011.10.007

    Article  Google Scholar 

  22. Thal DR, Rub U, Orantes M, Braak H (2002) Phases of A beta-deposition in the human brain and its relevance for the development of AD. Neurology 58(12):1791–1800. https://doi.org/10.1212/wnl.58.12.1791

    Article  Google Scholar 

  23. Mirra SS, Heyman A, McKeel D, Sumi SM, Crain BJ, Brownlee LM et al (1991) The consortium to establish a registry for Alzheimer’s disease (CERAD) part II standardization of the neuropathologic assessment of Alzheimer’s disease. Neurology 41(4):479–486. https://doi.org/10.1212/wnl.41.4.479

    Article  CAS  Google Scholar 

  24. Dickson DW, Bergeron C, Chin SS, Duyckaerts C, Horoupian D, Ikeda K et al (2002) Office of rare diseases neuropathologic criteria for corticobasal degeneration. J Neuropathol Exp Neurol 61(11):935–946. https://doi.org/10.1093/jnen/61.11.935

    Article  CAS  Google Scholar 

  25. Duyckaerts C, Braak H, Brion JP, Buee L, Del Tredici K, Goedert M et al (2015) PART is part of Alzheimer disease. Acta Neuropathol 129(5):749–756. https://doi.org/10.1007/s00401-015-1390-7

    Article  CAS  Google Scholar 

  26. Jellinger KA (2018) Multiple system atrophy: an oligodendroglioneural synucleinopathy1. J Alzheimers Dis 62(3):1141–1179. https://doi.org/10.3233/JAD-170397

    Article  CAS  Google Scholar 

  27. McKeith IG, Boeve BF, Dickson DW, Halliday G, Taylor JP, Weintraub D et al (2017) Diagnosis and management of dementia with lewy bodies: fourth consensus report of the DLB Consortium. Neurology 89(1):88–100. https://doi.org/10.1212/WNL.0000000000004058

    Article  Google Scholar 

  28. Bankhead P, Loughrey MB, Fernandez JA, Dombrowski Y, McArt DG, Dunne PD et al (2017) QuPath: open source software for digital pathology image analysis. Sci Rep 7(1):16878. https://doi.org/10.1038/s41598-017-17204-5

    Article  CAS  Google Scholar 

  29. Bussiere T, Hof PR, Mailliot C, Brown CD, Caillet-Boudin ML, Perl DP et al (1999) Phosphorylated serine422 on tau proteins is a pathological epitope found in several diseases with neurofibrillary degeneration. Acta Neuropathol 97(3):221–230. https://doi.org/10.1007/s004010050978

    Article  CAS  Google Scholar 

  30. de Silva R, Lashley T, Strand C, Shiarli AM, Shi J, Tian J et al (2006) An immunohistochemical study of cases of sporadic and inherited frontotemporal lobar degeneration using 3R-and 4R-specific tau monoclonal antibodies. Acta Neuropathol 111(4):329–340. https://doi.org/10.1007/s00401-006-0048-x

    Article  Google Scholar 

  31. Furcila D, DeFelipe J, Alonso-Nanclares L (2018) A study of amyloid-beta and phosphotau in plaques and neurons in the hippocampus of Alzheimer’s disease Patients. J Alzheimers Dis 64(2):417–435. https://doi.org/10.3233/JAD-180173

    Article  CAS  Google Scholar 

  32. Iacono D, Lee P, Edlow BL, Gray N, Fischl B, Kenney K et al (2020) Early-onset dementia in war veterans: brain polypathology and clinicopathologic complexity. J Neuropathol Exp Neurol 79(2):144–162. https://doi.org/10.1093/jnen/nlz122

    Article  Google Scholar 

  33. McMillan PJ, Strovas TJ, Baum M, Mitchell BK, Eck RJ, Hendricks N et al (2021) Pathological tau drives ectopic nuclear speckle scaffold protein SRRM2 accumulation in neuron cytoplasm in Alzheimer’s disease. Acta Neuropathol Commun 9(1):117. https://doi.org/10.1186/s40478-021-01219-1

    Article  CAS  Google Scholar 

  34. Ozaki K, Irioka T, Uchihara T, Yamada A, Nakamura A, Majima T et al (2021) Neuropathology of SCA34 showing widespread oligodendroglial pathology with vacuolar white matter degeneration: a case study. Acta Neuropathol Commun 9(1):172. https://doi.org/10.1186/s40478-021-01272-w

    Article  CAS  Google Scholar 

  35. Regalado-Reyes M, Furcila D, Hernandez F, Avila J, DeFelipe J, Leon-Espinosa G (2019) Phospho-tau changes in the human CA1 during Alzheimer’s disease progression. J Alzheimers Dis 69(1):277–288. https://doi.org/10.3233/JAD-181263

    Article  CAS  Google Scholar 

  36. B. Netherlands Brain, Wennstrom M, Janelidze S, Nilsson KPR, Serrano GE, Beach TG et al (2022) Cellular localization of p-tau217 in brain and its association with p-tau217 plasma levels. Acta Neuropathol Commun 10(1):3. https://doi.org/10.1186/s40478-021-01307-2

    Article  CAS  Google Scholar 

  37. Koronyo-Hamaoui M, Koronyo Y, Ljubimov AV, Miller CA, Ko MK, Black KL et al (2011) Identification of amyloid plaques in retinas from Alzheimer’s patients and noninvasive in vivo optical imaging of retinal plaques in a mouse model. Neuroimage 54(Suppl 1):S204–S217. https://doi.org/10.1016/j.neuroimage.2010.06.020

    Article  CAS  Google Scholar 

  38. Gupta N, Fong J, Ang LC, Yucel YH (2008) Retinal tau pathology in human glaucomas. Can J Ophthalmol 43(1):53–60. https://doi.org/10.3129/i07-185

    Article  Google Scholar 

  39. Pasteels B, Rogers J, Blachier F, Pochet R (1990) Calbindin and calretinin localization in retina from different species. Vis Neurosci 5(1):1–16. https://doi.org/10.1017/s0952523800000031

    Article  CAS  Google Scholar 

  40. Boon BDC, Hoozemans JJM, Lopuhaa B, Eigenhuis KN, Scheltens P, Kamphorst W et al (2018) Neuroinflammation is increased in the parietal cortex of atypical Alzheimer’s disease. J Neuroinflammation 15(1):170. https://doi.org/10.1186/s12974-018-1180-y

    Article  CAS  Google Scholar 

  41. Dowling JE (2009) Retina. In: Squire LR (ed) Encyclopedia of human biology. Academic Press, New York

    Google Scholar 

  42. Chidlow G, Wood JP, Manavis J, Finnie J, Casson RJ (2017) Investigations into retinal pathology in the early stages of a mouse model of Alzheimer’s disease. J Alzheimers Dis 56(2):655–675. https://doi.org/10.3233/JAD-160823

    Article  CAS  Google Scholar 

  43. Ho WL, Leung Y, Cheng SS, Lok CK, Ho YS, Baum L et al (2015) Investigating degeneration of the retina in young and aged tau P301L mice. Life Sci 124:16–23. https://doi.org/10.1016/j.lfs.2014.12.019

    Article  CAS  Google Scholar 

  44. Yan W, Peng YR, van Zyl T, Regev A, Shekhar K, Juric D et al (2020) Cell atlas of the human fovea and peripheral retina. Sci Rep 10(1):9802. https://doi.org/10.1038/s41598-020-66092-9

    Article  CAS  Google Scholar 

  45. Bussière T, Hof PR, Mailliot C, Brown CD, Caillet-Boudin ML, Perl DP et al (1999) Phosphorylated serine422 on tau proteins is a pathological epitope found in several diseases with neurofibrillary degeneration. Acta Neuropathol 97(3):221–230. https://doi.org/10.1007/s004010050978

    Article  Google Scholar 

  46. Chiasseu M, Alarcon-Martinez L, Belforte N, Quintero H, Dotigny F, Destroismaisons L et al (2017) Tau accumulation in the retina promotes early neuronal dysfunction and precedes brain pathology in a mouse model of Alzheimer’s disease. Mol Neurodegener 12(1):58. https://doi.org/10.1186/s13024-017-0199-3

    Article  CAS  Google Scholar 

  47. Ossenkoppele R, van der Kant R, Hansson O (2022) Tau biomarkers in Alzheimer’s disease: towards implementation in clinical practice and trials. Lancet Neurol. https://doi.org/10.1016/S1474-4422(22)00168-5

    Article  Google Scholar 

  48. Snyder PJ, Alber J, Alt C, Bain LJ, Bouma BE, Bouwman FH et al (2021) Retinal imaging in Alzheimer’s and neurodegenerative diseases. Alzheimers Dement 17(1):103–111. https://doi.org/10.1002/alz.12179

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to acknowledge all donors and their caregivers. We thank N.P. Smoor for his technical expertise and assistance in retinal tissue preparation. We thank Michiel Kooreman for helping with clinical data retrieval and his assistance at the Netherlands Brain Bank in collecting brain sections. We thank Jacoline B. ten Brink and Arthur A.B. Bergen for their technological support and advice on the retina. We thank Gina Gase for technical assistance with tissue preparation and immunohistochemistry. We thank A. Dijkstra for contributing to the assessment of the visual system. Illustration was created with BioRender.com.

Funding

This research was funded by the Netherlands Organisation for Scientific Research (NWO, MEDPHOT P18-26 Project 5).

Author information

Authors and Affiliations

Authors

Consortia

Contributions

All authors contributed to the study conception and design. Experiments were performed by THJM. Data collection and analysis were performed by FJHR and THJM. The first draft of the manuscript was written by FJHR and JJMH. All authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Frederique J. Hart de Ruyter or Jeroen J. M. Hoozemans.

Ethics declarations

Conflict of interest

F.J. Hart de Ruyter reports no competing interests; T.H.J. Morrema reports no competing interests; Dr. J. den Haan reports no competing interests; Prof. dr. J.W.R. Twisk reports no competing interests; Prof. dr. J.F. de Boer has acquired grant support (for the institution; Department of Physics, VU) from the Dutch Research Council (NWO) and from industry (Thorlabs, ASML, Heidelberg Engineering). He has received royalties related to IP on OCT technologies and semiconductor metrology. He has acted as an expert witness for a UK based law firm; Prof. Dr. P. Scheltens has received consultancy fees (paid to the university) from Alzheon, Brainstorm Cell and Green Valley. Within his university affiliation he is global PI of the phase 1b study of AC Immune, Phase 2b study with FUJI-film/Toyama and phase 2 study of UCB and phase 1 study with ImmunoBrain Checkpoint. He is chair of the EU steering committee of the phase 2b program of Vivoryon, the phase 2b study of Novartis Cardiology and co-chair of the phase 3 study with NOVO-Nordisk. He is also an employee of EQT Life Sciences (formerly LSP); Dr. B.D.C. Boon is supported by a research fellowship awarded by Alzheimer Nederland (#WE.15-2019-13); Prof. dr. D.R. Thal received speaker honorarium from Novartis Pharma Basel (Switzerland) and Biogen (USA), travel reimbursement from GE-Healthcare (UK), and UCB (Belgium), and collaborated with GE-Healthcare (UK), Novartis Pharma Basel (Switzerland), Probiodrug (Germany), and Janssen Pharmaceutical Companies (Belgium). He receives grants from Fonds Wetenschappelijk Onderzoek Vlaanderen (Belgium; FWO- G0F8516N, G065721N) and the Stichting Alzheimer Onderzoek (Belgium; SAO-FRA 2020/017); Prof. dr. A.J. Rozemuller reports no competing interests; Dr. F.D. Verbraak reports no competing interests; Dr. F. Bouwman performs contract research for Optina Dx and Optos, she has been an invited speaker at Roche and has been invited for expert testimony at Biogen. All funding is paid to her institution; Dr. J.J.M. Hoozemans received grants from the Dutch Research Council (ZonMW) and, Alzheimer Netherlands, performed contract research or received grants from Merck, ONO Pharmaceuticals, Janssen Prevention Center, DiscovericBio, AxonNeurosciences, Roche, Genentech, Promis, Denali, FirstBiotherapeutics, and Ensol Biosciences. All payments were made to the institution. Dr. J.J.M. Hoozemans participates in the scientific advisory board of Alzheimer Netherlands and is editor-in-chief for Acta Neuropathologica Communications.

Ethical approval

Donors signed informed consent for brain and eye autopsy and use of brain and retinal tissue and medical records for research prior to death. This study was approved by the ethical committee of the VU University Medical Center Amsterdam (VUmc).

Additional information

Publisher's Note

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

The original online version of this article was revised and the study group author name “on behalf of Netherlands Brain Bank” is removed from the author name.

Supplementary Information

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 6371 KB)

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hart de Ruyter, F.J., Morrema, T.H.J., den Haan, J. et al. Phosphorylated tau in the retina correlates with tau pathology in the brain in Alzheimer’s disease and primary tauopathies. Acta Neuropathol 145, 197–218 (2023). https://doi.org/10.1007/s00401-022-02525-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00401-022-02525-1

Keywords

Navigation