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:

A molecular chaperone breaks the catalytic cycle that generates toxic Aβ oligomers

Abstract

Alzheimer's disease is an increasingly prevalent neurodegenerative disorder whose pathogenesis has been associated with aggregation of the amyloid-β peptide (Aβ42). Recent studies have revealed that once Aβ42 fibrils are generated, their surfaces effectively catalyze the formation of neurotoxic oligomers. Here we show that a molecular chaperone, a human Brichos domain, can specifically inhibit this catalytic cycle and limit human Aβ42 toxicity. We demonstrate in vitro that Brichos achieves this inhibition by binding to the surfaces of fibrils, thereby redirecting the aggregation reaction to a pathway that involves minimal formation of toxic oligomeric intermediates. We verify that this mechanism occurs in living mouse brain tissue by cytotoxicity and electrophysiology experiments. These results reveal that molecular chaperones can help maintain protein homeostasis by selectively suppressing critical microscopic steps within the complex reaction pathways responsible for the toxic effects of protein misfolding and aggregation.

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: Kinetics of Aβ42 aggregation in the presence of Brichos.
Figure 2: Inhibition by Brichos of Aβ42 surface-catalyzed secondary nucleation of oligomers.
Figure 3: Brichos interacts with fibrillar but not monomeric Aβ42.
Figure 4: Brichos inhibits the formation of Aβ42 oligomers by suppressing secondary nucleation and redirecting the reaction pathway toward elongation events.
Figure 5: Brichos reduces the toxicity associated with the aggregation of Aβ42 in mouse brain slices.
Figure 6: Brichos inhibits the catalytic cycle that generates toxic Aβ42 oligomers.

Similar content being viewed by others

References

  1. Aguzzi, A. & O'Connor, T. Protein aggregation diseases: pathogenicity and therapeutic perspectives. Nat. Rev. Drug Discov. 9, 237–248 (2010).

    Article  CAS  Google Scholar 

  2. Dobson, C.M. Protein folding and misfolding. Nature 426, 884–890 (2003).

    Article  CAS  Google Scholar 

  3. Haass, C. & Selkoe, D.J. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid β-peptide. Nat. Rev. Mol. Cell Biol. 8, 101–112 (2007).

    Article  CAS  Google Scholar 

  4. Selkoe, D.J. Folding proteins in fatal ways. Nature 426, 900–904 (2003).

    Article  CAS  Google Scholar 

  5. Tanzi, R.E. & Bertram, L. Twenty years of the Alzheimer's disease amyloid hypothesis: a genetic perspective. Cell 120, 545–555 (2005).

    Article  CAS  Google Scholar 

  6. Chiti, F. & Dobson, C.M. Protein misfolding, functional amyloid, and human disease. Annu. Rev. Biochem. 75, 333–366 (2006).

    Article  CAS  Google Scholar 

  7. Knowles, T.P.J., Vendruscolo, M. & Dobson, C.M. The amyloid state and its association with protein misfolding diseases. Nat. Rev. Mol. Cell Biol. 15, 384–396 (2014).

    Article  CAS  Google Scholar 

  8. Sipe, J.D. et al. Amyloid fibril protein nomenclature: 2012 recommendations from the nomenclature committee of the International Society of Amyloidosis. Amyloid 19, 167–170 (2012).

    Article  CAS  Google Scholar 

  9. Goldschmidt, L., Tenga, P.K., Riek, R. & Eisenberg, D. Identifying the amylome, proteins capable of forming amyloid-like fibrils. Proc. Natl. Acad. Sci. USA 107, 3487–3492 (2010).

    Article  CAS  Google Scholar 

  10. Greenwald, J. & Riek, R. Biology of amyloid: structure, function, and regulation. Structure 18, 1244–1260 (2010).

    Article  CAS  Google Scholar 

  11. Koo, E.H., Lansbury, P.T. Jr. & Kelly, J.W. Amyloid diseases: abnormal protein aggregation in neurodegeneration. Proc. Natl. Acad. Sci. USA 96, 9989–9990 (1999).

    Article  CAS  Google Scholar 

  12. Bucciantini, M. et al. Inherent toxicity of aggregates implies a common mechanism for protein misfolding diseases. Nature 416, 507–511 (2002).

    Article  CAS  Google Scholar 

  13. Kayed, R. et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–489 (2003).

    Article  CAS  Google Scholar 

  14. Walsh, D.M. et al. Naturally secreted oligomers of amyloid β protein potently inhibit hippocampal long-term potentiation in vivo. Nature 416, 535–539 (2002).

    Article  CAS  Google Scholar 

  15. Collins, S.R., Douglass, A., Vale, R.D. & Weissman, J.S. Mechanism of prion propagation: amyloid growth occurs by monomer addition. PLoS Biol. 2, e321 (2004).

    Article  Google Scholar 

  16. Jarrett, J.T. & Lansbury, P.T. Jr. Seeding “one-dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer's disease and scrapie? Cell 73, 1055–1058 (1993).

    Article  CAS  Google Scholar 

  17. Knowles, T.P.J. et al. An analytical solution to the kinetics of breakable filament assembly. Science 326, 1533–1537 (2009).

    Article  CAS  Google Scholar 

  18. Cremades, N. et al. Direct observation of the interconversion of normal and toxic forms of α-synuclein. Cell 149, 1048–1059 (2012).

    Article  CAS  Google Scholar 

  19. Cohen, S.I.A. et al. Proliferation of amyloid-beta42 aggregates occurs through a secondary nucleation mechanism. Proc. Natl. Acad. Sci. USA 110, 9758–9763 (2013).

    Article  CAS  Google Scholar 

  20. Hellstrand, E., Boland, B., Walsh, D.M. & Linse, S. Amyloid β-protein aggregation produces highly reproducible kinetic data and occurs by a two-phase process. ACS Chem. Neurosci. 1, 13–18 (2010).

    Article  CAS  Google Scholar 

  21. Lee, J., Culyba, E.K., Powers, E.T. & Kelly, J.W. Amyloid-β forms fibrils by nucleated conformational conversion of oligomers. Nat. Chem. Biol. 7, 602–609 (2011).

    Article  CAS  Google Scholar 

  22. Serio, T.R. et al. Nucleated conformational conversion and the replication of conformational information by a prion determinant. Science 289, 1317–1321 (2000).

    Article  CAS  Google Scholar 

  23. Kar, K., Jayaraman, M., Sahoo, B., Kodali, R. & Wetzel, R. Critical nucleus size for disease-related polyglutamine aggregation is repeat-length dependent. Nat. Struct. Mol. Biol. 18, 328–336 (2011).

    Article  CAS  Google Scholar 

  24. Oosawa, F. & Asakura, S. Thermodynamics of the Polymerization of Protein (Academic Press, 1975).

  25. Ferrone, F.A., Hofrichter, J. & Eaton, W.A. Kinetics of sickle hemoglobin polymerization. II. A double nucleation mechanism. J. Mol. Biol. 183, 611–631 (1985).

    Article  CAS  Google Scholar 

  26. Cohen, S.I.A., Vendruscolo, M., Dobson, C.M. & Knowles, T.P.J. Nucleated polymerization with secondary pathways. II. Determination of self-consistent solutions to growth processes described by non-linear master equations. J. Chem. Phys. 135, 065106 (2011).

    Article  Google Scholar 

  27. Ruschak, A.M. & Miranker, A.D. Fiber-dependent amyloid formation as catalysis of an existing reaction pathway. Proc. Natl. Acad. Sci. USA 104, 12341–12346 (2007).

    Article  CAS  Google Scholar 

  28. Arosio, P., Cukalevski, R., Frohm, B., Knowles, T.P.J. & Linse, S. Quantification of the concentration of Aβ42 propagons during the lag phase by an amyloid chain reaction assay. J. Am. Chem. Soc. 136, 219–225 (2014).

    Article  CAS  Google Scholar 

  29. Jeong, J.S., Ansaloni, A., Mezzenga, R., Lashuel, H.A. & Dietler, G. Novel mechanistic insight into the molecular basis of amyloid polymorphism and secondary nucleation during amyloid formation. J. Mol. Biol. 425, 1765–1781 (2013).

    Article  CAS  Google Scholar 

  30. Salvadores, N., Shahnawaz, M., Scarpini, E., Tagliavini, F. & Soto, C. Detection of misfolded Aβ oligomers for sensitive biochemical diagnosis of Alzheimer's disease. Cell Rep. 7, 261–268 (2014).

    Article  CAS  Google Scholar 

  31. Willander, H. et al. Brichos domains efficiently delay fibrillation of amyloid beta-peptide. J. Biol. Chem. 287, 31608–31617 (2012).

    Article  CAS  Google Scholar 

  32. Hartl, F.U. Molecular chaperones in cellular protein folding. Nature 381, 571–579 (1996).

    Article  CAS  Google Scholar 

  33. Hartl, F.U., Bracher, A. & Hayer-Hartl, M. Molecular chaperones in protein folding and proteostasis. Nature 475, 324–332 (2011).

    Article  CAS  Google Scholar 

  34. Kim, Y.E., Hipp, M.S., Bracher, A., Hayer-Hartl, M. & Hartl, F.U. Molecular chaperone functions in protein folding and proteostasis. Annu. Rev. Biochem. 82, 323–355 (2013).

    Article  CAS  Google Scholar 

  35. Balch, W.E., Morimoto, R.I., Dillin, A. & Kelly, J.W. Adapting proteostasis for disease intervention. Science 319, 916–919 (2008).

    Article  CAS  Google Scholar 

  36. Doyle, S.M., Genest, O. & Wickner, S. Protein rescue from aggregates by powerful molecular chaperone machines. Nat. Rev. Mol. Cell Biol. 14, 617–629 (2013).

    Article  CAS  Google Scholar 

  37. Auluck, P.K., Chan, H.Y.E., Trojanowski, J.Q., Lee, V.M.Y. & Bonini, N.M. Chaperone suppression of alpha-synuclein toxicity in a Drosophila model for Parkinson's disease. Science 295, 865–868 (2002).

    Article  CAS  Google Scholar 

  38. Bence, N.F., Sampat, R.M. & Kopito, R.R. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 292, 1552–1555 (2001).

    Article  CAS  Google Scholar 

  39. Morimoto, R.I. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 22, 1427–1438 (2008).

    Article  CAS  Google Scholar 

  40. Sakahira, H., Breuer, P., Hayer-Hartl, M.K. & Hartl, F.U. Molecular chaperones as modulators of polyglutamine protein aggregation and toxicity. Proc. Natl. Acad. Sci. USA 99 (suppl. 4), 16412–16418 (2002).

    Article  CAS  Google Scholar 

  41. Schaffar, G. et al. Cellular toxicity of polyglutamine expansion proteins: mechanism of transcription factor deactivation. Mol. Cell 15, 95–105 (2004).

    Article  CAS  Google Scholar 

  42. Månsson, C. et al. Interaction of the molecular chaperone DNAJB6 with growing amyloid-beta 42 (Aβ42) aggregates leads to sub-stoichiometric inhibition of amyloid formation. J. Biol. Chem. 289, 31066–31076 (2014).

    Article  Google Scholar 

  43. Sánchez-Pulido, L., Devosi, D. & Valencia, A. Brichos: a conserved domain in proteins associated with dementia, respiratory distress and cancer. Trends Biochem. Sci. 27, 329–332 (2002).

    Article  Google Scholar 

  44. Hedlund, J., Johansson, J. & Persson, B. Brichos: a superfamily of multidomain proteins with diverse functions. BMC Res. Notes 2, 180 (2009).

    Article  Google Scholar 

  45. Knight, S.D., Presto, J., Linse, S. & Johansson, J. The brichos domain, amyloid fibril formation, and their relationship. Biochemistry 52, 7523–7531 (2013).

    Article  CAS  Google Scholar 

  46. Willander, H., Hermansson, E., Johansson, J. & Presto, J. Brichos domain associated with lung fibrosis, dementia and cancer: a chaperone that prevents amyloid fibril formation? FEBS J. 278, 3893–3904 (2011).

    Article  CAS  Google Scholar 

  47. Nerelius, C., Gustafsson, M., Nordling, K., Larsson, A. & Johansson, J. Anti-amyloid activity of the c-terminal domain of prosp-c against amyloid beta-peptide and medin. Biochemistry 48, 3778–3786 (2009).

    Article  CAS  Google Scholar 

  48. Coomaraswamy, J. et al. Modeling familial Danish dementia in mice supports the concept of the amyloid hypothesis of Alzheimer's disease. Proc. Natl. Acad. Sci. USA 107, 7969–7974 (2010).

    Article  CAS  Google Scholar 

  49. Kim, J. et al. Bri2 (itm2b) inhibits Aβ deposition in vivo. J. Neurosci. 28, 6030–6036 (2008).

    Article  CAS  Google Scholar 

  50. Vidal, R. et al. A stop-codon mutation in the BRI gene associated with familial British dementia. Nature 399, 776–781 (1999).

    Article  CAS  Google Scholar 

  51. Willander, H. et al. High-resolution structure of a brichos domain and its implications for anti-amyloid chaperone activity on lung surfactant protein c. Proc. Natl. Acad. Sci. USA 109, 2325–2329 (2012).

    Article  CAS  Google Scholar 

  52. Cohen, S.I.A. et al. Nucleated polymerization with secondary pathways. I. Time evolution of the principal moments. J. Chem. Phys. 135, 065105 (2011).

    Article  Google Scholar 

  53. Schnitzler, A. & Gross, J. Normal and pathological oscillatory communication in the brain. Nat. Rev. Neurosci. 6, 285–296 (2005).

    Article  CAS  Google Scholar 

  54. Knowles, T.P.J. et al. Role of intermolecular forces in defining material properties of protein nanofibrils. Science 318, 1900–1903 (2007).

    Article  CAS  Google Scholar 

  55. Lue, L.F. et al. Soluble amyloid beta peptide concentration as a predictor of synaptic change in Alzheimer's disease. Am. J. Pathol. 155, 853–862 (1999).

    Article  CAS  Google Scholar 

  56. McLean, C.A. et al. Soluble pool of Aβ amyloid as a determinant of severity of neurodegeneration in Alzheimer's disease. Ann. Neurol. 46, 860–866 (1999).

    Article  CAS  Google Scholar 

  57. Näslund, J. et al. Correlation between elevated levels of amyloid beta-peptide in the brain and cognitive decline. J. Am. Med. Assoc. 283, 1571–1577 (2000).

    Article  Google Scholar 

  58. Wang, J., Dickson, D.W., Trojanowski, J.Q. & Lee, V.M. The levels of soluble versus insoluble brain Aβ distinguish Alzheimer's disease from normal and pathologic aging. Exp. Neurol. 158, 328–337 (1999).

    Article  CAS  Google Scholar 

  59. Hermansson, E. et al. The chaperone domain BRICHOS prevents CNS toxicity of amyloid-β peptide in Drosophila melanogaster. Dis. Model. Mechl. 7, 659–665 (2014).

    Article  Google Scholar 

  60. Walsh, D.M. et al. A facile method for expression and purification of the Alzheimer's disease-associated amyloid beta-peptide. FEBS J. 276, 1266–1281 (2009).

    Article  CAS  Google Scholar 

  61. Waltersson, Y., Linse, S., Brodin, P. & Grundström, T. Mutational effects on the cooperativity of Ca2 binding in calmodulin. Biochemistry 32, 7866–7871 (1993).

    Article  CAS  Google Scholar 

  62. Cedervall, T. et al. Understanding the nanoparticle-protein corona using methods to quantify exchange rates and affinities of proteins for nanoparticles. Proc. Natl. Acad. Sci. USA 104, 2050–2055 (2007).

    Article  CAS  Google Scholar 

  63. Johansson, C. et al. Biophysical studies of engineered mutant proteins based on calbindin D9k modified in the pseudo EF-hand. Eur. J. Biochem. 187, 455–460 (1990).

    Article  CAS  Google Scholar 

  64. Lindman, S. et al. Salting the charged surface: pH and salt dependence of protein G B1 stability. Biophys. J. 90, 2911–2921 (2006).

    Article  CAS  Google Scholar 

  65. Johansson, H. et al. The Brichos domain-containing C-terminal part of pro-surfactant protein C binds to an unfolded poly-val transmembrane segment. J. Biol. Chem. 281, 21032–21039 (2006).

    Article  CAS  Google Scholar 

  66. Burguillos, M.A. et al. Caspase signalling controls microglia activation and neurotoxicity. Nature 472, 319–324 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge financial support from the Schiff Foundation (S.I.A.C.), the Swedish Research Council (S.L., J.J. and J.P.) and its Linneaus Centre Organizing Molecular Matter (S.L.), the Crafoord Foundation (S.L.), Alzheimerfonden (S.L.), the Frances and Augustus Newman Foundation (P.A. and T.P.J.K.), the European Research Council (T.P.J.K. and S.L.), the Biotechnology and Biological Sciences Research Council (T.P.J.K.), the Nanometer Structure Consortium at Lund University (S.L.), the KID PhD studentship grant (F.R.K. and L.D.), the Swedish Medical Association (A.F.), the Brain Fund (A.F.), the Strategic Program in Neurosciences at the Karolinska Institutet (A.F.), the Swiss National Science Foundation (P.A.) and the Wellcome Trust (C.M.D. and T.P.J.K.). We thank T. Weaver (University of Cincinnati) for reagents.

Author information

Authors and Affiliations

Authors

Contributions

S.I.A.C., T.P.J.K., A.F. and S.L. designed the study. J.P., F.R.K., A.F., L.D., C.D., X.Y., B.F., H.B. and S. L. performed the experiments. S.I.A.C., P.A., T.P.J.K., F.R.K., C.M.D., A.F. and S.L. analyzed the data. S.I.A.C., P.A., C.M.D., T.P.J.K. and S.L. wrote the paper. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence to André Fisahn, Tuomas P J Knowles or Sara Linse.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Saturation of the effect of Brichos at high chaperone concentrations.

Kinetic reaction profiles for the aggregation of Aβ42 are shown from left to right for reactions in the absence of Brichos and with 67%, 100%, 120%, 170%, 250% and 330% Aβ42 monomer equivalents of Brichos. At high chaperone concentrations, secondary nucleation is completed inhibited and additional chaperone has no effect on the kinetics of aggregation. The inhibitory effect reaches this saturation at a critical Brichos concentration which corresponds to approximately 100% of Aβ42 monomer equivalents.

Supplementary Figure 2 Kinetic analysis of the mode of action of Brichos.

(a) Kinetic reaction profiles for the aggregation of Aβ42 are shown from left to right for reactions in the absence of Brichos and with 10%, 15%, 22%, 35%, 50% and 75% Aβ42 monomer equivalents of Brichos. Continuous lines represent fits of the integrated rate for Aβ42 aggregation, Eq. 1, to the data19. (b) Decrease of the effective microscopic rates for secondary nucleation (k2) and fibril elongation (k+) with increasing chaperone concentration evaluated from the fits. The microscopic rate constants are normalised relative to the values in the absence of Brichos. The Brichos concentration is given in Aβ42 monomer equivalents. The analysis indicates that Brichos affects specifically the secondary nucleation pathway. To further quantify the behaviour as a function of chaperone concentration, we also performed a first-principles kinetic analysis (see Supplementary Note for a derivation) that introduces explicitly into the reaction scheme the reversible binding of the chaperone along the fibril surface by considering a Langmuir-type adsorption, resulting in the fits in Fig. 1c (thin dotted lines).

Supplementary Figure 3 Control experiments with other proteins.

Kinetic reaction profiles with 3µM Aβ42 and four control proteins at varying concentration (0.45µM pale blue; 1.5µM cyan; 3µM green). The proteins are calbindin D9k, calmodulin, protein G B1-domain and fatty-acid free human serum albumin (HSA). Calbindin D9k and PGB1 have a more negative net charge and are more hydrophilic than Brichos, and calmodulin is significantly more negatively charged. (a-c) Calbindin, PGB1 and calmodulin have no effect on Aβ42 aggregation in this concentration range. (d) HSA affects the aggregation reaction but does not match the prediction for selective inhibition of secondary nucleation (green dashed line), implying that the mechanism of inhibition is different to that identified for Brichos. Four replicates of each condition are shown.

Supplementary Figure 4 Cell viability and cytotoxicity assays.

(a) Cell viability as measured by the MTS assay shows that Brichos removes toxicity even in reactions containing pre-formed fibrils. (b, c) Cell apoptosis as measured by caspase assay for two cell lines: (b) more sensitive to perturbations of the medium, and (c) less sensitive to perturbations of the medium. For both cell lines, the change in the caspase signal relative to the control that is induced by Aβ42 aggregation is abolished by Brichos. The change in caspase signal induced by Aβ42 toxicity after a fixed time for these two cell lines differs in direction since the measured signal depends on the time of the readout relative to the characteristic time of cell death (see Supplementary Note). In both cases the change in caspase signal indicates increased toxicity, and is suppressed by Brichos. The concentration of monomeric peptide was 1µM in (a) and (b) and 10µM in (c); the concentration of pre-formed fibrils was 10 nM in (a) and (b) and 100 nM in (c). The average and standard deviations shown are over four replicates of each condition in (a) and (b) and six replicates of each condition in (c).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Note (PDF 1350 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Cohen, S., Arosio, P., Presto, J. et al. A molecular chaperone breaks the catalytic cycle that generates toxic Aβ oligomers. Nat Struct Mol Biol 22, 207–213 (2015). https://doi.org/10.1038/nsmb.2971

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2971

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