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.

  • Protocol
  • Published:

Preparation and characterization of toxic Aβ aggregates for structural and functional studies in Alzheimer's disease research

Abstract

The amyloid cascade hypothesis, supported by strong evidence from genetics, pathology and studies using animal models, implicates amyloid-β (Aβ) oligomerization and fibrillogenesis as central causative events in the pathogenesis of Alzheimer's disease (AD). Today, significant efforts in academia, biotechnology and the pharmaceutical industry are devoted to identifying the mechanisms by which the process of Aβ aggregation contributes to neurodegeneration in AD and to the identity of the toxic Aβ species. In this paper, we describe methods and detailed protocols for reproducibly preparing Aβ aggregates of defined size distribution and morphology, including monomers, protofibrils and fibrils, using size exclusion chromatography. In addition, we describe detailed biophysical procedures for elucidating the structural features, aggregation kinetics and toxic properties of the different Aβ aggregation states, with special emphasis on protofibrillar intermediates. The information provided by this approach allows for consistent correlation between the properties of the aggregates and their toxicity toward primary neurons and/or cell lines. A better understanding of the molecular and structural basis of Aβ aggregation and toxicity is crucial for the development of effective strategies aimed at prevention and/or treatment of AD. Furthermore, the identification of specific aggregation states, which correlate with neurodegeneration in AD, could lead to the development of diagnostic tools to detect and monitor disease progression. The procedures described can be performed in as little as 1 day, or may take longer, depending on the exact toxicity assays used.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Aβ fibril formation pathway.
Figure 2
Figure 3: SEC fractionation and biophysical characterization of Aβ42 monomers and protofibrils.
Figure 4: Application to fAD-associated Aβ mutant peptides.
Figure 5: Application to synthetic and recombinant Aβ peptides.
Figure 6: Neurotoxicity of SEC-purified Aβ monomers and protofibrils.

Similar content being viewed by others

References

  1. Hebert, L.E., Scherr, P.A., Bienias, J.L., Bennett, D.A. & Evans, D.A. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Arch. Neurol. 60, 1119–1122 (2003).

    Article  PubMed  Google Scholar 

  2. Selkoe, D.J. Alzheimer's disease: genes, proteins, and therapy. Physiol. Rev. 81, 741–766 (2001).

    Article  CAS  PubMed  Google Scholar 

  3. Glenner, G.G., Wong, C.W., Quaranta, V. & Eanes, E.D. The amyloid deposits in Alzheimer's disease: their nature and pathogenesis. Appl. Pathol. 2, 357–369 (1984).

    CAS  PubMed  Google Scholar 

  4. Haass, C. et al. Amyloid beta-peptide is produced by cultured cells during normal metabolism. Nature 359, 322–325 (1992).

    Article  CAS  PubMed  Google Scholar 

  5. Busciglio, J., Gabuzda, D.H., Matsudaira, P. & Yankner, B.A. Generation of beta-amyloid in the secretory pathway in neuronal and nonneuronal cells. Proc. Natl. Acad. Sci. USA 90, 2092–2096 (1993).

    Article  CAS  PubMed  Google Scholar 

  6. Shoji, M. et al. Production of the Alzheimer amyloid beta protein by normal proteolytic processing. Science 258, 126–129 (1992).

    Article  CAS  PubMed  Google Scholar 

  7. Hardy, J.A. & Higgins, G.A. Alzheimer's disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992).

    Article  CAS  PubMed  Google Scholar 

  8. Selkoe, D.J. Alzheimer's disease: a central role for amyloid. J. Neuropathol. Exp. Neurol. 53, 438–447 (1994).

    Article  CAS  PubMed  Google Scholar 

  9. 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  PubMed  Google Scholar 

  10. Kelly, J.W. The alternative conformations of amyloidogenic proteins and their multi-step assembly pathways. Curr. Opin. Struct. Biol. 8, 101–106 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. Harper, J.D., Lieber, C.M. & Lansbury, P.T., Jr. Atomic force microscopic imaging of seeded fibril formation and fibril branching by the Alzheimer's disease amyloid-beta protein. Chem. Biol. 4, 951–959 (1997).

    Article  CAS  PubMed  Google Scholar 

  12. Harper, J.D., Wong, S.S., Lieber, C.M. & Lansbury, P.T. Observation of metastable Abeta amyloid protofibrils by atomic force microscopy. Chem. Biol. 4, 119–125 (1997).

    Article  CAS  PubMed  Google Scholar 

  13. Walsh, D.M., Lomakin, A., Benedek, G.B., Condron, M.M. & Teplow, D.B. Amyloid beta-protein fibrillogenesis. Detection of a protofibrillar intermediate. J. Biol. Chem. 272, 22364–22372 (1997).

    Article  CAS  PubMed  Google Scholar 

  14. Hartley, D.M. et al. Protofibrillar intermediates of amyloid beta-protein induce acute electrophysiological changes and progressive neurotoxicity in cortical neurons. J. Neurosci. 19, 8876–8884 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Lambert, M.P. et al. Diffusible, nonfibrillar ligands derived from Abeta1-42 are potent central nervous system neurotoxins. Proc. Natl. Acad. Sci. USA 95, 6448–6453 (1998).

    Article  CAS  PubMed  Google Scholar 

  16. Kayed, R. et al. Annular protofibrils are a structurally and functionally distinct type of amyloid oligomer. J. Biol. Chem. 284, 4230–4237 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bitan, G. et al. Amyloid beta -protein (Abeta) assembly: Abeta 40 and Abeta 42 oligomerize through distinct pathways. Proc. Natl. Acad. Sci. USA 100, 330–335 (2003).

    Article  CAS  PubMed  Google Scholar 

  18. Lashuel, H.A. et al. Mixtures of wild-type and a pathogenic (E22G) form of Abeta40 in vitro accumulate protofibrils, including amyloid pores. J. Mol. Biol. 332, 795–808 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  Google Scholar 

  20. Walsh, D.M. et al. Amyloid beta-protein fibrillogenesis. Structure and biological activity of protofibrillar intermediates. J. Biol. Chem. 274, 25945–25952 (1999).

    Article  CAS  PubMed  Google Scholar 

  21. Jan, A., Gokce, O., Luthi-Carter, R. & Lashuel, H.A. The ratio of monomeric to aggregated forms of Abeta40 and Abeta42 is an important determinant of amyloid-beta aggregation, fibrillogenesis, and toxicity. J. Biol. Chem. 283, 28176–28189 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Williams, A.D. et al. Structural properties of Abeta protofibrils stabilized by a small molecule. Proc. Natl. Acad. Sci. USA 102, 7115–7120 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Kheterpal, I. et al. Abeta protofibrils possess a stable core structure resistant to hydrogen exchange. Biochemistry 42, 14092–14098 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Stine, W.B., Jr. et al. The nanometer-scale structure of amyloid-beta visualized by atomic force microscopy. J. Protein Chem. 15, 193–203 (1996).

    Article  CAS  PubMed  Google Scholar 

  25. Serpell, L.C. Alzheimer's amyloid fibrils: structure and assembly. Biochim Biophys Acta 1502, 16–30 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Narang, H.K. High-resolution electron microscopic analysis of the amyloid fibril in Alzheimer's disease. J. Neuropathol. Exp. Neurol. 39, 621–631 (1980).

    Article  CAS  PubMed  Google Scholar 

  27. Merz, P.A. et al. Ultrastructural morphology of amyloid fibrils from neuritic and amyloid plaques. Acta Neuropathol 60, 113–124 (1983).

    Article  CAS  PubMed  Google Scholar 

  28. Gong, Y. et al. Alzheimer's disease-affected brain: presence of oligomeric A beta ligands (ADDLs) suggests a molecular basis for reversible memory loss. Proc. Natl. Acad. Sci. US A 100, 10417–10422 (2003).

    Article  CAS  Google Scholar 

  29. Lemere, C.A. et al. Sequence of deposition of heterogeneous amyloid beta-peptides and APO E in Down syndrome: implications for initial events in amyloid plaque formation. Neurobiol. Dis. 3, 16–32 (1996).

    Article  CAS  PubMed  Google Scholar 

  30. Irizarry, M.C. et al. Abeta deposition is associated with neuropil changes, but not with overt neuronal loss in the human amyloid precursor protein V717F (PDAPP) transgenic mouse. J. Neurosci. 17, 7053–7059 (1997).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  33. Citron, M. et al. Mutation of the beta-amyloid precursor protein in familial Alzheimer's disease increases beta-protein production. Nature 360, 672–674 (1992).

    Article  CAS  PubMed  Google Scholar 

  34. Eckman, C.B. et al. A new pathogenic mutation in the APP gene (I716V) increases the relative proportion of A beta 42(43). Hum. Mol. Genet. 6, 2087–2089 (1997).

    Article  CAS  PubMed  Google Scholar 

  35. Goate, A. et al. Segregation of a missense mutation in the amyloid precursor protein gene with familial Alzheimer's disease. Nature 349, 704–706 (1991).

    Article  CAS  PubMed  Google Scholar 

  36. Nilsberth, C. et al. The 'Arctic' APP mutation (E693G) causes Alzheimer's disease by enhanced Abeta protofibril formation. Nat. Neurosci. 4, 887–893 (2001).

    Article  CAS  PubMed  Google Scholar 

  37. Holcomb, L. et al. Accelerated Alzheimer-type phenotype in transgenic mice carrying both mutant amyloid precursor protein and presenilin 1 transgenes. Nat. Med. 4, 97–100 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Moechars, D. et al. Early phenotypic changes in transgenic mice that overexpress different mutants of amyloid precursor protein in brain. J. Biol. Chem. 274, 6483–6492 (1999).

    Article  CAS  PubMed  Google Scholar 

  39. Meyer-Luehmann, M. et al. Exogenous induction of cerebral beta-amyloidogenesis is governed by agent and host. Science 313, 1781–1784 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Ye, C.P., Selkoe, D.J. & Hartley, D.M. Protofibrils of amyloid beta-protein inhibit specific K+ currents in neocortical cultures. Neurobiol. Dis. 13, 177–190 (2003).

    Article  CAS  PubMed  Google Scholar 

  41. Ye, C., Walsh, D.M., Selkoe, D.J. & Hartley, D.M. Amyloid beta-protein induced electrophysiological changes are dependent on aggregation state: N-methyl-D-aspartate (NMDA) versus non-NMDA receptor/channel activation. Neurosci. Lett. 366, 320–325 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Wang, Q., Walsh, D.M., Rowan, M.J., Selkoe, D.J. & Anwyl, R. Block of long-term potentiation by naturally secreted and synthetic amyloid beta-peptide in hippocampal slices is mediated via activation of the kinases c-Jun N-terminal kinase, cyclin-dependent kinase 5, and p38 mitogen-activated protein kinase as well as metabotropic glutamate receptor type 5. J. Neurosci. 24, 3370–3378 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Lambert, M.P. et al. Vaccination with soluble Abeta oligomers generates toxicity-neutralizing antibodies. J. Neurochem. 79, 595–605 (2001).

    Article  CAS  PubMed  Google Scholar 

  44. Dodart, J.C. et al. Immunization reverses memory deficits without reducing brain Abeta burden in Alzheimer's disease model. Nat. Neurosci. 5, 452–457 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Lashuel, H.A., Hartley, D., Petre, B.M., Walz, T. & Lansbury, P.T., Jr. Neurodegenerative disease: amyloid pores from pathogenic mutations. Nature 418, 291 (2002).

    Article  CAS  PubMed  Google Scholar 

  46. Arispe, N., Rojas, E. & Pollard, H.B. Alzheimer disease amyloid beta protein forms calcium channels in bilayer membranes: blockade by tromethamine and aluminum. Proc. Natl. Acad. Sci. USA 90, 567–571 (1993).

    Article  CAS  PubMed  Google Scholar 

  47. Kayed, R. et al. Permeabilization of lipid bilayers is a common conformation-dependent activity of soluble amyloid oligomers in protein misfolding diseases. J. Biol. Chem. 279, 46363–46366 (2004).

    Article  CAS  PubMed  Google Scholar 

  48. Li, S. et al. Soluble oligomers of amyloid Beta protein facilitate hippocampal long-term depression by disrupting neuronal glutamate uptake. Neuron 62, 788–801 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Deshpande, A., Mina, E., Glabe, C. & Busciglio, J. Different conformations of amyloid beta induce neurotoxicity by distinct mechanisms in human cortical neurons. J. Neurosci. 26, 6011–6018 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Klein, A.M., Kowall, N.W. & Ferrante, R.J. Neurotoxicity and oxidative damage of beta amyloid 1–42 versus beta amyloid 1–40 in the mouse cerebral cortex. Ann. N Y Acad. Sci. 893, 314–320 (1999).

    Article  CAS  PubMed  Google Scholar 

  51. Roselli, F. et al. Soluble beta-amyloid1-40 induces NMDA-dependent degradation of postsynaptic density-95 at glutamatergic synapses. J. Neurosci. 25, 11061–11070 (2005).

    Article  CAS  PubMed  Google Scholar 

  52. Zagorski, M.G. et al. Methodological and chemical factors affecting amyloid beta peptide amyloidogenicity. Methods Enzymol. 309, 189–204 (1999).

    Article  CAS  PubMed  Google Scholar 

  53. Bitan, G. & Teplow, D.B. Preparation of aggregate-free, low molecular weight amyloid-beta for assembly and toxicity assays. Methods Mol. Biol. 299, 3–9 (2005).

    CAS  PubMed  Google Scholar 

  54. Ward, R.V. et al. Fractionation and characterization of oligomeric, protofibrillar and fibrillar forms of beta-amyloid peptide. Biochem. J. 348 (Pt 1): 137–144 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Klyubin, I. et al. Amyloid beta protein immunotherapy neutralizes Abeta oligomers that disrupt synaptic plasticity in vivo. Nat. Med. 11, 556–561 (2005).

    Article  CAS  PubMed  Google Scholar 

  56. Roher, A.E. et al. Morphology and toxicity of Abeta-(1–42) dimer derived from neuritic and vascular amyloid deposits of Alzheimer's disease. J. Biol. Chem. 271, 20631–20635 (1996).

    Article  CAS  PubMed  Google Scholar 

  57. Snyder, S.W. et al. Amyloid-beta aggregation: selective inhibition of aggregation in mixtures of amyloid with different chain lengths. Biophys. J. 67, 1216–1228 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Herzig, M.C. et al. Abeta is targeted to the vasculature in a mouse model of hereditary cerebral hemorrhage with amyloidosis. Nat. Neurosci. 7, 954–960 (2004).

    Article  CAS  PubMed  Google Scholar 

  59. Younkin, S.G. Evidence that A beta 42 is the real culprit in Alzheimer's disease. Ann. Neurol. 37, 287–288 (1995).

    Article  CAS  PubMed  Google Scholar 

  60. Zou, K. et al. Amyloid beta-protein (Abeta)1–40 protects neurons from damage induced by Abeta1–42 in culture and in rat brain. J. Neurochem. 87, 609–619 (2003).

    Article  CAS  PubMed  Google Scholar 

  61. Arimon, M., Grimminger, V., Sanz, F. & Lashuel, H.A. Hsp104 targets multiple intermediates on the amyloid pathway and suppresses the seeding capacity of Abeta fibrils and protofibrils. J. Mol. Biol. 384, 1157–1173 (2008).

    Article  CAS  PubMed  Google Scholar 

  62. Pike, C.J., Burdick, D., Walencewicz, A.J., Glabe, C.G. & Cotman, C.W. Neurodegeneration induced by beta-amyloid peptides in vitro: the role of peptide assembly state. J. Neurosci. 13, 1676–1687 (1993).

    Article  CAS  PubMed  Google Scholar 

  63. Lorenzo, A. & Yankner, B.A. Beta-amyloid neurotoxicity requires fibril formation and is inhibited by congo red. Proc. Natl. Acad. Sci. USA 91, 12243–12247 (1994).

    Article  CAS  PubMed  Google Scholar 

  64. Seilheimer, B. et al. The toxicity of the Alzheimer's beta-amyloid peptide correlates with a distinct fiber morphology. J. Struct. Biol. 119, 59–71 (1997).

    Article  CAS  PubMed  Google Scholar 

  65. Whalen, B.M., Selkoe, D.J. & Hartley, D.M. Small non-fibrillar assemblies of amyloid beta-protein bearing the Arctic mutation induce rapid neuritic degeneration. Neurobiol. Dis. 20, 254–266 (2005).

    Article  CAS  PubMed  Google Scholar 

  66. Wogulis, M. et al. Nucleation-dependent polymerization is an essential component of amyloid-mediated neuronal cell death. J. Neurosci. 25, 1071–1080 (2005).

    Article  CAS  PubMed  Google Scholar 

  67. Chromy, B.A. et al. Self-assembly of Abeta(1–42) into globular neurotoxins. Biochemistry 42, 12749–12760 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Walsh, D.M., Klyubin, I., Fadeeva, J.V., Rowan, M.J. & Selkoe, D.J. Amyloid-beta oligomers: their production, toxicity and therapeutic inhibition. Biochem. Soc. Trans. 30, 552–557 (2002).

    Article  CAS  PubMed  Google Scholar 

  69. Cleary, J.P. et al. Natural oligomers of the amyloid-beta protein specifically disrupt cognitive function. Nat. Neurosci. 8, 79–84 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  72. Lambert, M.P. et al. Monoclonal antibodies that target pathological assemblies of Abeta. J. Neurochem. 100, 23–35 (2007).

    Article  CAS  PubMed  Google Scholar 

  73. Bitan, G., Fradinger, E.A., Spring, S.M. & Teplow, D.B. Neurotoxic protein oligomers--what you see is not always what you get. Amyloid 12, 88–95 (2005).

    Article  PubMed  Google Scholar 

  74. May, P.C. et al. Beta-Amyloid peptide in vitro toxicity: lot-to-lot variability. Neurobiol. Aging 13, 605–607 (1992).

    Article  CAS  PubMed  Google Scholar 

  75. 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  PubMed  PubMed Central  Google Scholar 

  76. Finder, V.H., Vodopivec, I., Nitsch, R.M. & Glockshuber, R. The recombinant amyloid-beta peptide Abeta1–42 aggregates faster and is more neurotoxic than synthetic Abeta1–42. J. Mol. Biol. 396, 9–18.

  77. Garai, K., Crick, S.L., Mustafi, S.M. & Frieden, C. Expression and purification of amyloid-beta peptides from Escherichia coli. Protein Expr. Purif. 66, 107–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Sato, T. et al. Inhibitors of amyloid toxicity based on beta-sheet packing of Abeta40 and Abeta42. Biochemistry 45, 5503–5516 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Brewer, G.J., Torricelli, J.R., Evege, E.K. & Price, P.J. Optimized survival of hippocampal neurons in B27-supplemented Neurobasal, a new serum-free medium combination. J. Neurosci. Res. 35, 567–576 (1993).

    Article  CAS  PubMed  Google Scholar 

  80. Tseng, B.P. et al. Deposition of monomeric, not oligomeric, Abeta mediates growth of Alzheimer's disease amyloid plaques in human brain preparations. Biochemistry 38, 10424–10431 (1999).

    Article  CAS  PubMed  Google Scholar 

  81. Perczel, A., Hollosi, M., Tusnady, G. & Fasman, G.D. Convex constraint analysis: a natural deconvolution of circular dichroism curves of proteins. Protein Eng. 4, 669–679 (1991).

    Article  CAS  PubMed  Google Scholar 

  82. Perczel, A., Park, K. & Fasman, G.D. Analysis of the circular dichroism spectrum of proteins using the convex constraint algorithm: a practical guide. Anal. Biochem. 203, 83–93 (1992).

    Article  CAS  PubMed  Google Scholar 

  83. Hepler, R.W. et al. Solution state characterization of amyloid beta-derived diffusible ligands. Biochemistry 45, 15157–15167 (2006).

    Article  CAS  PubMed  Google Scholar 

  84. Brahms, S. & Brahms, J. Determination of protein secondary structure in solution by vacuum ultraviolet circular dichroism. J. Mol. Biol. 138, 149–178 (1980).

    Article  CAS  PubMed  Google Scholar 

  85. Betts, V. et al. Aggregation and catabolism of disease-associated intra-Abeta mutations: reduced proteolysis of AbetaA21G by neprilysin. Neurobiol. Dis. 31, 442–450 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Pace, C.N., Vajdos, F., Fee, L., Grimsley, G. & Gray, T. How to measure and predict the molar absorption coefficient of a protein. Protein Sci. 4, 2411–2423 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Westermark, P. et al. Amyloid: toward terminology clarification. Report from the Nomenclature Committee of the International Society of Amyloidosis. Amyloid 12, 1–4 (2005).

    Article  CAS  PubMed  Google Scholar 

  88. Dobson, C.M. The structural basis of protein folding and its links with human disease. Philos. Trans. R. Soc. Lond. 356, 133–145 (2001).

    Article  CAS  Google Scholar 

  89. Roher, A.E. et al. Oligomerizaiton and fibril asssembly of the amyloid-beta protein. Biochim. Biophys. Acta. 1502, 31–43 (2000).

    Article  CAS  PubMed  Google Scholar 

  90. Petkova, A.T. et al. Self-propagating, molecular-level polymorphism in Alzheimer's beta-amyloid fibrils. Science 307, 262–265 (2005).

    Article  CAS  PubMed  Google Scholar 

  91. Inouye, H., Fraser, P.E. & Kirschner, D.A. Structure of beta-crystallite assemblies formed by Alzheimer beta-amyloid protein analogues: analysis by x-ray diffraction. Biophys. J. 64, 502–519 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Chamberlain, A.K. et al. Ultrastructural organization of amyloid fibrils by atomic force microscopy. Biophys. J. 79, 3282–3293 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Hoshi, M. et al. Spherical aggregates of beta-amyloid (amylospheroid) show high neurotoxicity and activate tau protein kinase I/glycogen synthase kinase-3beta. Proc. Natl. Acad. Sci. USA 100, 6370–6375 (2003).

    Article  CAS  PubMed  Google Scholar 

  94. Bitan, G., Lomakin, A. & Teplow, D.B. Amyloid beta-protein oligomerization: prenucleation interactions revealed by photo-induced cross-linking of unmodified proteins. J. Biol. Chem. 276, 35176–35184 (2001).

    Article  CAS  PubMed  Google Scholar 

  95. O'Nuallain, B., Williams, A.D., Westermark, P. & Wetzel, R. Seeding specificity in amyloid growth induced by heterologous fibrils. J. Biol. Chem. 279, 17490–17499 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by the Swiss Federal Institute of Technology Lausanne (EPFL) and by grants from the Swiss National Foundation (Grant # 310000-110027), the National Institute on Aging, USA (Grant # AG19970) (D.M.H.) and from the Alzheimer's Association (D.M.H.). We thank AC Immune (S.A.), Lausanne, Switzerland for financially supporting Asad Jan. We also thank Professor Andrea Pfeifer, Dr. Andreas Muhs and Dr. Oskar Adolfsson from AC Immune (S.A.), Lausanne, Switzerland for thoughtful discussions. We also thank Professor Carl Frieden, Washington University, St. Louis, USA and Professor Rudolf Glockshuber, ETH, Zurich, Switzerland for kindly providing recombinant Aβ peptides. We gratefully acknowledge Dr. Graham Knott at Bio-Electron Microscopy Facility (CIME), EPFL, Lausanne for technical support with TEM; Dr. Harald Wutzel, Laboratory of Polymers, EPFL, Lausanne for help with dynamic light-scattering measurements; and Dr. Michel Prudent, LMNN, EPFL, Lausanne for help with mass spectrometry measurements.

Author information

Authors and Affiliations

Authors

Contributions

A.J. and H.A.L. contributed to the fractionation data and biophysical characterization of SEC fractions. D.M.H. provided the Aβ toxicity data and protocols for preparing and treating neuronal cultures in modified neurobasal media for toxicity studies. A.J., D.M.H. and H.A.L. contributed to writing the paper.

Corresponding authors

Correspondence to Dean M Hartley or Hilal A Lashuel.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figure 1

(a) Circular dichroism (CD) spectroscopy analysis of SEC fractions from Superdex 75 HR 10/30 (A1340 and A1342) in a Jasco-810 CD Spectrometer using a 10 mm pathiength cell (black line = A1342 protofibrils; red line = A1342 monomer obtained using DMSO method; blue line = A1342 monomer obtained using the 6 M guanidine-HC1 method; green line = A1340 monomer obtained using the 6 M guanidine-HC1 method). (b) A1340 (monomeric = M) and A1342 (monomeric = M and protofibriliar = PF) SEC fractions were obtained from a Superdex 75 HR 10/30 column and were electrophoresed on NuPAGE 4-12% Bis Tris gels (invitrogen, cat. No. NPO336BOX). The proteins were detected by (left) silver staining (SiiverXpress, invitrogen Cat. No. LC61 00) and also, after having been transferred on a Nitroceilulose membrane (Whatman, Cat. No. 10401396), immunobiotting with anti-A13 antibody 6E10 (Signet, Cat. No. 9320). (C) Thiofiavin-T (ThT) binding over time by protofibriliar and monomeric A1342 fractions (10 pMAI342; 96 h, 37°C without agitation). (d-e) Fibril formation byA1342 crude (stock solution obtained after DMSO solubilization method): sAI342 was solubilized using DMSO (1 mg mi-1) and incubated at 37°C with gentle agitation (4,000 rpm) (d) ThT binding and (e) representative TEM image of resultant fibrils after 24 h of incubation. (t) SEC fractionation of synthetic A1340 (Keck facility), solubilized using DMSO method (1 mg mi-1), on a Superdex 75 HR 10/30 SEC column into protofibrils and monomeric A13 fractions. A1340 was solubilized and fractionated, without incubation (red line), or fractionated after incubation at room temperature, overnight (black line). (g) SEC fractionation of recombinant rAI340 from independent research groups, solubilized using DMSO method (0.5-1 mg mi-1), on a Superdex 75 HR 10/30 SEC. rAI340 1 (black line; 0.5 mg mi-1) was a generous gift from Prof. Rudolf Giockshuber, ETH, Zurich, Switzerland and rAI340 2 (green line; 1 mg mi-1) was generously provided by Prof. Carl Frieden, Washington University, St. Louis, USA. (Abbreviations: ON = Overnight; RT= Room temperature; PF=A13 Protofibrils; M= Monomeric A13; CR= Crude A1342; a.u. = arbitrary units; the error bars in (c and d) represent STD in duplicate samples; scale bar= 200 nm). (TIFF 1575 kb)

Supplementary Figure 2

SEC sub-fractionation of A42 protofibrils on a Superose 6 HR 10130 SEC column. (a) A1342 was solubilized using DMSO method (2mg mi-i) and fractionated into protofibrillar (Fi-F4) and monomeric (F5-F6) fractions on a Superose 6 HR 10/30 SEC coiumn. (b) Anaiyticai SEC of Fi-F4 on Superose 6 pc 3.2/30 SEC coiumn. (c) CD spectroscopy anaiysis of A13 42 protofibriiiar fractions from Superose6 HR 10/30 as in Suppiementary Figure ia. (d-f) Representative TEM images of (d) Fl, (e) F3 and (f) F5. (Abbreviations: F= Fraction; scaie bar= 200 nm). (TIFF 1005 kb)

Supplementary Figure 3

Stability of crude A42 solution and SEC isolated protofibrillar and monomeric A42 fractions. (a) A1342 was solubilized (DM50 method, 1 mg mV1) and split into three aliquots (— 300 p 1/aliquot). The first aliquot was injected immediately into a Superdex 75 HR 10/30 column. The second and third aliquots were incubated at 4°C for 4 and 24 h respectively, and then injected. At each time point, the solution was centrifuged (16,000 g, 4°C, 10 mm) and 250 p1 of the supernatant were injected. (b) A42 protofibrils were obtained by SEC and the fraction (50 pM, 1 ml) was split in two aliquots (500 pI/aliquot). The first aliquot was re-injected immediately into a Superdex 75 HR 10/30 column. The second aliquot was incubated at 4°C for 24 h then re-injected as above. At each time point, the solution was centrifuged (16,000 g, 4°C, 10 mm) and 400 p1 of supernatant were injected. (c) ThT binding by crude, protofibrillar and monomeric A1342 after incubation at 4°C. (d-i) Representative TEM images: (d and g) A1342 crude soon after solubilization and after 4 h of incubation at 4°C, (e and h) A1342 protofibrils soon after fractionation and after 24 h of incubation at 4°C and (f and i) monomeric A13 42 soon after fractionation and after 24 h of incubation at 4°C. (Abbreviations: PF= A1342 Protofibrils; M= Monomeric A1342; CR= Crude A1342; a.u. = arbitrary units; the error bars in (c) represent STD in duplicate samples; scale bar= 200 nm). (TIFF 1813 kb)

Supplementary Figure 4

Comparative analysis of A concentration determination by amino acid analysis (AAA), BCA method and UV A280 nm method. (a) The protein concentration of four A13 samples, from different fractionation experiments, was determined by AAA, BCA kit (Pierce Cat. No. 23227) and UV A280 nm methods. AAA was carried out in FGCZ, ETH, Zurich, Switzerland. A3 standard curve, obtained from serial dilutions of A340 in H20, was used for BOA. UV A280 nm for different fractions was determined in a 10 mm path-length cuvette using a Cary spectrophotometer. (b) Protein concentration of eight A13 samples, from different fractionation experiments, was determined by BOA kit and UVA280 nm methods. A13 concentration by BOA method was determined using a standard curve generated from both A1340 and bovine serum albumin (BSA) serial dilutions. UV A280 nm was carried out as in (a) above. BSA was provided as part of the PIEROE BOA kit. (c) Generation of standard curve of A1340 for BOA assay. A1340 was freshly solubilized in double distilled H20 (1 mg mi-1 200 pM by net weight; 80% peptide content) and serial dilutions (5 - 200 pM) were prepared (in HO) for generating a standard curve for BOA assay. The stock (200 pM) A1340 solution was aliquoted into sterile tubes (Fisherbrand, Oat. No. 05-669-27; 200 p1/tube) and frozen at -20°O. After 24 h of freezing, one of the aliquots was thawn on ice, and serial dilutions (5-200 pM) were carried out in H20. Protein concentration for both series of dilutions was determined by the BOA kit. Synthetic A 3 (Keck facility) was used for all measurements outlined in (a-c). (TIFF 554 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Jan, A., Hartley, D. & Lashuel, H. Preparation and characterization of toxic Aβ aggregates for structural and functional studies in Alzheimer's disease research. Nat Protoc 5, 1186–1209 (2010). https://doi.org/10.1038/nprot.2010.72

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2010.72

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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