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.

  • Article
  • Published:

Caspase-2 cleavage of tau reversibly impairs memory

Abstract

In Alzheimer's disease (AD) and other tauopathies, the tau protein forms fibrils, which are believed to be neurotoxic. However, fibrillar tau has been dissociated from neuron death and network dysfunction, suggesting the involvement of nonfibrillar species. Here we describe a novel pathological process in which caspase-2 cleavage of tau at Asp314 impairs cognitive and synaptic function in animal and cellular models of tauopathies by promoting the missorting of tau to dendritic spines. The truncation product, Δtau314, resists fibrillation and is present at higher levels in brains from cognitively impaired mice and humans with AD. The expression of tau mutants that resisted caspase-2 cleavage prevented tau from infiltrating spines, dislocating glutamate receptors and impairing synaptic function in cultured neurons, and it prevented memory deficits and neurodegeneration in mice. Decreasing the levels of caspase-2 restored long-term memory in mice that had existing deficits. Our results suggest an overall treatment strategy for re-establishing synaptic function and restoring memory in patients with AD by preventing tau from accumulating in dendritic spines.

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: Levels of the tau cleavage product TCP35 correlate with memory deficits.
Figure 2: Caspase-2 cleaves tau and generates Δtau314.
Figure 3: Reducing caspase-2 levels restores memory function.
Figure 4: Effects of cleavage at Asp314 on tau mislocalization to dendritic spines, synaptic transmission and glutamate receptor localization.
Figure 5: Mutating Asp314 to Glu314 prevents tauP301L from causing cognitive deficits and neurodegeneration.
Figure 6: Missorting of Δtau314 to dendritic spines is not sufficient to disrupt synaptic or memory function.

Similar content being viewed by others

References

  1. Spillantini, M.G. & Goedert, M. Tau pathology and neurodegeneration. Lancet Neurol. 12, 609–622 (2013).

    CAS  PubMed  Google Scholar 

  2. Ballatore, C., Lee, V.M. & Trojanowski, J.Q. Tau-mediated neurodegeneration in Alzheimer's disease and related disorders. Nat. Rev. Neurosci. 8, 663–672 (2007).

    CAS  PubMed  Google Scholar 

  3. Ittner, L.M. et al. Dendritic function of tau mediates amyloid-β toxicity in Alzheimer's disease mouse models. Cell 142, 387–397 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Fath, T., Eidenmüller, J. & Brandt, R. Tau-mediated cytotoxicity in a pseudohyperphosphorylation model of Alzheimer's disease. J. Neurosci. 22, 9733–9741 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gomez-Isla, T. et al. Clinical and pathological correlates of apolipoprotein E–ɛ4 in Alzheimer's disease. Ann. Neurol. 39, 62–70 (1996).

    CAS  PubMed  Google Scholar 

  9. Rocher, A.B. et al. Structural and functional changes in tau-mutant mice neurons are not linked to the presence of NFTs. Exp. Neurol. 223, 385–393 (2010).

    CAS  PubMed  Google Scholar 

  10. Menkes-Caspi, N. et al. Pathological tau disrupts ongoing network activity. Neuron 85, 959–966 (2015).

    CAS  PubMed  Google Scholar 

  11. Santacruz, K. et al. Tau suppression in a neurodegenerative mouse model improves memory function. Science 309, 476–481 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Ramsden, M. et al. Age-dependent neurofibrillary tangle formation, neuron loss and memory impairment in a mouse model of human tauopathy (P301L). J. Neurosci. 25, 10637–10647 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Kopeikina, K.J. et al. Synaptic alterations in the rTg4510 mouse model of tauopathy. J. Comp. Neurol. 521, 1334–1353 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Rawlings, N.D., Waller, M., Barrett, A.J. & Bateman, A. MEROPS: the database of proteolytic enzymes, their substrates and inhibitors. Nucleic Acids Res. 42, D503–D509 (2014).

    CAS  PubMed  Google Scholar 

  15. Hoover, B.R. et al. Tau mislocalization to dendritic spines mediates synaptic dysfunction independently of neurodegeneration. Neuron 68, 1067–1081 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Miller, E.C. et al. Tau phosphorylation and tau mislocalization mediate soluble Aβ oligomer-induced AMPA glutamate receptor signaling deficits. Eur. J. Neurosci. 39, 1214–1224 (2014).

    PubMed  PubMed Central  Google Scholar 

  18. Fasulo, L. et al. The neuronal microtubule-associated protein tau is a substrate for caspase-3 and an effector of apoptosis. J. Neurochem. 75, 624–633 (2000).

    CAS  PubMed  Google Scholar 

  19. Cotman, C.W., Poon, W.W., Rissman, R.A. & Blurton-Jones, M. The role of caspase cleavage of tau in Alzheimer disease neuropathology. J. Neuropathol. Exp. Neurol. 64, 104–112 (2005).

    CAS  PubMed  Google Scholar 

  20. Bergeron, L. et al. Defects in regulation of apoptosis in caspase-2-deficient mice. Genes Dev. 12, 1304–1314 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Isaac, J.T., Nicoll, R.A. & Malenka, R.C. Evidence for silent synapses: implications for the expression of LTP. Neuron 15, 427–434 (1995).

    CAS  PubMed  Google Scholar 

  22. Liao, D., Hessler, N.A. & Malinow, R. Activation of postsynaptically silent synapses during pairing-induced LTP in CA1 region of hippocampal slice. Nature 375, 400–404 (1995).

    CAS  PubMed  Google Scholar 

  23. Shalini, S., Dorstyn, L., Dawar, S. & Kumar, S. Old, new and emerging functions of caspases. Cell Death Differ. 22, 526–539 (2015).

    CAS  PubMed  Google Scholar 

  24. Kumar, S., Kinoshita, M., Noda, M., Copeland, N.G. & Jenkins, N.A. Induction of apoptosis by the mouse Nedd2 gene, which encodes a protein similar to the product of the Caenorhabditis elegans cell death gene ced-3 and the mammalian IL-1β-converting enzyme. Genes Dev. 8, 1613–1626 (1994).

    CAS  PubMed  Google Scholar 

  25. Tiwari, M. et al. A non-apoptotic role for CASP2/caspase-2: modulation of autophagy. Autophagy 10, 1054–1070 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Shalini, S. et al. Caspase-2 protects against oxidative stress in vivo. Oncogene 34, 4995–5002 (2015).

    CAS  PubMed  Google Scholar 

  27. Upton, J.P. et al. IRE-1α cleaves select microRNAs during ER stress to de-repress translation of pro-apoptotic caspase-2. Science 338, 818–822 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Pozueta, J. et al. Caspase-2 is required for dendritic spine and behavioral alterations in J20 APP transgenic mice. Nat. Commun. 4, 1939 (2013).

    PubMed  PubMed Central  Google Scholar 

  29. Carroll, J.B. et al. Mice lacking caspase-2 are protected from behavioral changes, but not pathology, in the YAC128 model of Huntington disease. Mol. Neurodegener. 6, 59 (2011).

    PubMed  PubMed Central  Google Scholar 

  30. Lee, S. & Shea, T.B. Caspase-mediated truncation of tau potentiates aggregation. Int. J. Alzheimers Dis. 2012, 731063 (2012).

    PubMed  PubMed Central  Google Scholar 

  31. Wang, Y., Garg, S., Mandelkow, E.M. & Mandelkow, E. Proteolytic processing of tau. Biochem. Soc. Trans. 38, 955–961 (2010).

    CAS  PubMed  Google Scholar 

  32. Dolan, P.J. & Johnson, G.V. A caspase-cleaved form of tau is preferentially degraded through the autophagy pathway. J. Biol. Chem. 285, 21978–21987 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Gamblin, T.C. et al. Caspase cleavage of tau: linking amyloid and neurofibrillary tangles in Alzheimer's disease. Proc. Natl. Acad. Sci. USA 100, 10032–10037 (2003).

    CAS  PubMed  Google Scholar 

  34. Spires-Jones, T.L., Stoothoff, W.H., de Calignon, A., Jones, P.B. & Hyman, B.T. Tau pathophysiology in neurodegeneration: a tangled issue. Trends Neurosci. 32, 150–159 (2009).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Abraha, A. et al. C-terminal inhibition of tau assembly in vitro and in Alzheimer's disease. J. Cell Sci. 113, 3737–3745 (2000).

    CAS  PubMed  Google Scholar 

  37. Yoshiyama, Y. et al. Synapse loss and microglial activation precede tangles in a P301S tauopathy mouse model. Neuron 53, 337–351 (2007).

    CAS  PubMed  Google Scholar 

  38. Davies, P. Characterization and use of monoclonal antibodies to tau and paired-helical-filament tau. Methods Mol. Med. 32, 361–373 (2000).

    CAS  PubMed  Google Scholar 

  39. Hunsberger, H.C. et al. Effect size of memory deficits in mice with adult-onset P301L tau expression. Behav. Brain Res. 272, 181–195 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Westerman, M.A. et al. The relationship between Aβ and memory in the Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 22, 1858–1867 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Wolfer, D.P., Madani, R., Valenti, P. & Lipp, H.P. Extended analysis of path data from mutant mice using the public domain software Wintrack. Physiol. Behav. 73, 745–753 (2001).

    CAS  PubMed  Google Scholar 

  42. Lesné, S. et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature 440, 352–357 (2006).

    PubMed  Google Scholar 

  43. Wang, X. et al. Loss of sorting nexin 27 contributes to excitatory synaptic dysfunction by modulating glutamate receptor recycling in Down's syndrome. Nat. Med. 19, 473–480 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Sources of funding for this study include the US National Institutes of Health (NIH) (EUREKA grant no. R01-NS63214 (K.H.A.) and R01-NS79374 (K.H.A.)), the T. and P. Grossman Family Foundation (K.H.A.), B. Grossman (K.H.A.) and K. Moe (K.H.A.). We thank P. Davies (Albert Einstein College of Medicine) for the antibodies Alz-50, MC-1 and CP-13, as well as B. Roth (University of North Carolina at Chapel Hill) for the pAAV-CaMKIIa-EGFP vector. We are grateful to the participants of the Memory and Aging Project, as well as the staff and director (D. Bennett) of the Rush Alzheimer's Disease Center (grant no. R01-AG17917 (D. Bennett)). We thank L. Kemper, K. Leinonen and E. Steuer for excellent assistance with mouse breeding, genotyping and behavioral tests, M. Kuskowski for help with statistical analyses, and C. Wilmot, D. Liao, M. Koob and M. Walters for valuable advice and encouragement.

Author information

Authors and Affiliations

Authors

Contributions

K.H.A. conceived and supervised the project, designed the experiments and wrote the manuscript; X.Z. designed and performed the experiments and wrote the manuscript; L.A.K. designed and performed the behavioral experiments; B.S. performed the electrophysiological experiments and prepared the postsynaptic fractions; C.H. performed the behavioral experiments; K.Z. performed hippocampal volume quantification; M.R. contributed to the initial identification of Δtau314; and J.C. contributed to the behavioral experiments.

Corresponding author

Correspondence to Karen H Ashe.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text, Figures and Tables

Supplementary Figures 1–12 and Supplementary Table 1 (PDF 2550 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, X., Kotilinek, L., Smith, B. et al. Caspase-2 cleavage of tau reversibly impairs memory. Nat Med 22, 1268–1276 (2016). https://doi.org/10.1038/nm.4199

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4199

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