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.

  • Review Article
  • Published:

Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid β-peptide

Key Points

  • Certain neurodegenerative disorders are characterized by the progressive accumulation of insoluble deposits of distinct misfolded proteins. In general, mutations causing inherited forms of the respective diseases affect the folding, aggregation and/or solubility of these proteins.

  • In Alzheimer's disease (AD), amyloid β-protein (Aβ) production by regulated intramembrane proteolysis is crucial for pathogenicity. An unusual intramembrane cleavage generates various Aβ species, only one of which (the longest variant) is particularly prone to form potentially toxic oligomers.

  • Soluble oligomeric assemblies of amyloidogenic proteins are increasingly thought to initiate disease-specific cytopathology and subsequent symptoms. Larger deposits, such as compacted Aβ plaques, seem to be relatively inert but might serve as reservoirs of diffusible oligomers.

  • Oligomeric assemblies of amyloidogenic peptides from distinct neurodegenerative diseases — such as Parkinson's, AD, Huntington's and prion disorders — share common structural properties.

  • Small oligomeric assemblies of Aβ have been specifically shown to impair long-term potentiation and dendritic spine structure in the hippocampus and to decrease memory in animals.

  • Oligomeric species of Aβ are active targets for therapeutic intervention. Aβ immunotherapy, Aβ-aggregation inhibitors, allosteric modulators of γ-secretase and Aβ-degrading proteases can all reduce oligomeric Aβ, consequent neuronal degeneration and behavioural deficits in mouse models of AD.

  • The first promising clinical data from AD human trials seem to support the potential of Aβ-directed immunotherapy that can neutralize oligomers.

  • Similar therapeutic approaches based on oligomeric amyloidogenic species are likely to apply to other protein-folding diseases of the nervous system.

Abstract

The distinct protein aggregates that are found in Alzheimer's, Parkinson's, Huntington's and prion diseases seem to cause these disorders. Small intermediates — soluble oligomers — in the aggregation process can confer synaptic dysfunction, whereas large, insoluble deposits might function as reservoirs of the bioactive oligomers. These emerging concepts are exemplified by Alzheimer's disease, in which amyloid β-protein oligomers adversely affect synaptic structure and plasticity. Findings in other neurodegenerative diseases indicate that a broadly similar process of neuronal dysfunction is induced by diffusible oligomers of misfolded proteins.

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: Amyloid β-protein generation by normal proteolytic processing of β-amyloid precursor protein.
Figure 2: Naturally secreted SDS-stable a myloid β-protein oligomers block hippocampal long-term potentiation in vivo.
Figure 3: Therapeutic approaches targeting amyloid β-protein production and oligomerization.

Similar content being viewed by others

References

  1. Gasser, T. Genetics of Parkinson's disease. Curr. Opin. Neurol. 18, 363–369 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Polymeropoulos, M. H. et al. Mutation in the α-synuclein gene identified in families with Parkinson's disease. Science 276, 2045–2047 (1997).

    Article  CAS  PubMed  Google Scholar 

  3. Kahle, P. J., Haass, C., Kretzschmar, H. A. & Neumann, M. Structure/function of α-synuclein in health and disease: rational development of animal models for Parkinson's and related diseases. J. Neurochem. 82, 449–457 (2002).

    Article  CAS  PubMed  Google Scholar 

  4. Spillantini, M. G. et al. α-Synuclein in Lewy bodies. Nature 388, 839–840 (1997).

    Article  CAS  PubMed  Google Scholar 

  5. Baba, M. et al. Aggregation of α-synuclein in Lewy bodies of sporadic Parkinson's disease and dementia with Lewy bodies. Am. J. Pathol. 152, 879–884 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Rosen, D. R. Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis. Nature 364, 362 (1993).

    Article  CAS  PubMed  Google Scholar 

  7. Deng, H. X. et al. Amyotrophic lateral sclerosis and structural defects in Cu, Zn superoxide dismutase. Science 261, 1047–1051 (1993).

    Article  CAS  PubMed  Google Scholar 

  8. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington's disease chromosomes. The Huntington's Disease Collaborative Research Group. Cell 72, 971–983 (1993).

  9. Alzheimer, A. Über eine eigenartige Erkrankung der Hirnrinde. Allg. Z. Psychiatrie Psychisch-Gerichtl. Med. 64, 146–148 (1907).

    Google Scholar 

  10. Glenner, G. G. & Wong, C. W. Alzheimer's disease: initial report of the purification and characterization of a novel cerebrovascular amyloid protein. Biochem. Biophys. Res. Commun. 120, 885–890 (1984).

    Article  CAS  PubMed  Google Scholar 

  11. Masters, C. L. et al. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc. Natl Acad. Sci. USA 82, 4245–4249 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Grundke-Iqbal, I. et al. Abnormal phosphorylation of the microtubule-associated protein τ (tau) in Alzheimer cytoskeletal pathology. Proc. Natl Acad. Sci. USA 83, 4913–4917 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Kosik, K. S., Joachim, C. L. & Selkoe, D. J. Microtubule-associated protein τ (tau) is a major antigenic component of paired helical filaments in Alzheimer disease. Proc. Natl Acad. Sci. USA 83, 4044–4048 (1986).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Nukina, N. & Ihara, Y. One of the antigenic determinants of paired helical filaments is related to τ protein. J. Biochem. 99, 1541–1544 (1986).

    Article  CAS  PubMed  Google Scholar 

  15. Glenner, G. G. & Wong, C. W. Alzheimer's disease and Down's syndrome: sharing of a unique cerebrovascular amyloid fibril protein. Biochem. Biophys. Res. Commun. 122, 1131–1135 (1984).

    Article  CAS  PubMed  Google Scholar 

  16. Olson, M. I. & Shaw, C. M. Presenile dementia and Alzheimer's disease in mongolism. Brain 92, 147–156 (1969).

    Article  CAS  PubMed  Google Scholar 

  17. Kang, J. et al. The precursor of Alzheimer's disease amyloid A4 protein resembles a cell-surface receptor. Nature 325, 733–736 (1987).

    Article  CAS  PubMed  Google Scholar 

  18. Levy, E. et al. Mutation of the Alzheimer's disease amyloid gene in hereditary cerebral hemorrhage, Dutch type. Science 248, 1124–1126 (1990).

    Article  CAS  PubMed  Google Scholar 

  19. 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 

  20. Chartier-Harlin, M. C. et al. Early-onset Alzheimer's disease caused by mutations at codon 717 of the β-amyloid precursor protein gene. Nature 353, 844–846 (1991).

    Article  CAS  PubMed  Google Scholar 

  21. Mullan, M. et al. A pathogenic mutation for probable Alzheimer's disease in the APP gene at the N-terminus of β-amyloid. Nature Genet. 1, 345–347 (1992).

    Article  CAS  PubMed  Google Scholar 

  22. Selkoe, D. J. The molecular pathology of Alzheimer's disease. Neuron 6, 487–498 (1991).

    Article  CAS  PubMed  Google Scholar 

  23. Hardy, J. A. & Higgins, G. A. Alzheimer's disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992). References 22 and 23 set out the amyloid cascade hypothesis, for which strong experimental evidence is now accumulating.

    Article  CAS  PubMed  Google Scholar 

  24. Hardy, J. & Selkoe, D. J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  26. Seubert, P. et al. Isolation and quantification of soluble Alzheimer's β-peptide from biological fluids. Nature 359, 325–327 (1992).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Haass, C. Take five-BACE and the γ-secretase quartet conduct Alzheimer's amyloid β-peptide generation. EMBO J. 23, 483–488 (2004). A review on APP processing by β- and γ-secretase.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Brown, M. S., Ye, J., Rawson, R. B. & Goldstein, J. L. Regulated intramembrane proteolysis: a control mechanism conserved from bacteria to humans. Cell 100, 391–398 (2000).

    Article  CAS  PubMed  Google Scholar 

  31. Weihofen, A. & Martoglio, B. Intramembrane-cleaving proteases: controlled liberation of functional proteins and peptides from membranes. Trends Cell Biol. 13, 71–78 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Mumm, J. S. & Kopan, R. Notch signaling: from the outside in. Dev. Biol. 228, 151–165 (2000).

    Article  CAS  PubMed  Google Scholar 

  33. Wolfe, M. S. et al. Two transmembrane aspartates in presenilin-1 required for presenilin endoproteolysis and γ-secretase activity. Nature 398, 513–517 (1999).

    Article  CAS  PubMed  Google Scholar 

  34. Steiner, H. et al. Glycine 384 is required for presenilin-1 function and is conserved in polytopic bacterial aspartyl proteases. Nature Cell Biol. 2, 848–851 (2000).

    Article  CAS  PubMed  Google Scholar 

  35. Kimberly, W. T. et al. γ-Secretase is a membrane protein complex comprised of presenilin, nicastrin, Aph-1, and Pen-2. Proc. Natl Acad. Sci. USA 100, 6382–6387 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Takasugi, N. et al. The role of presenilin cofactors in the γ-secretase complex. Nature 422, 438–441 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Edbauer, D. et al. Reconstitution of γ-secretase activity. Nature Cell Biol. 5, 486–488 (2003).

    Article  CAS  PubMed  Google Scholar 

  38. Sastre, M. et al. Presenilin-dependent γ-secretase processing of β-amyloid precursor protein at a site corresponding to the S3 cleavage of Notch. EMBO Rep. 2, 835–841 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Weidemann, A. et al. A novel ε-cleavage within the transmembrane domain of the Alzheimer amyloid precursor protein demonstrates homology with Notch processing. Biochemistry 41, 2825–2835 (2002).

    Article  CAS  PubMed  Google Scholar 

  40. Gu, Y. et al. Distinct intramembrane cleavage of the β-amyloid precursor protein family resembling γ-secretase-like cleavage of Notch. J. Biol. Chem. 276, 35235–35238 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Qi-Takahara, Y. et al. Longer forms of amyloid β protein: implications for the mechanism of intramembrane cleavage by γ-secretase. J. Neurosci. 25, 436–445 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Zhao, G. et al. γ-cleavage is dependent on ζ-cleavage during the proteolytic processing of amyloid precursor protein within its transmembrane domain. J. Biol. Chem. 280, 37689–37697 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Dickson, D. W. The pathogenesis of senile plaques. J. Neuropathol. Exp. Neurol. 56, 321–339 (1997).

    Article  CAS  PubMed  Google Scholar 

  44. Naslund, J. et al. Correlation between elevated levels of amyloid β-peptide in the brain and cognitive decline. JAMA 283, 1571–1577 (2000).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. 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  PubMed  Google Scholar 

  47. 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  PubMed  Google Scholar 

  48. Arrasate, M., Mitra, S., Schweitzer, E. S., Segal, M. R. & Finkbeiner, S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 431, 805–810 (2004). Evidence that small, diffusible aggregates of intracellular huntingtin can confer neurotoxicity, perhaps analogously to soluble Aβ oligomers.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  50. Cummings, C. J. et al. Mutation of the E6-AP ubiquitin ligase reduces nuclear inclusion frequency while accelerating polyglutamine-induced pathology in SCA1 mice. Neuron 24, 879–892 (1999).

    Article  CAS  PubMed  Google Scholar 

  51. Tsai, J., Grutzendler, J., Duff, K. & Gan, W. B. Fibrillar amyloid deposition leads to local synaptic abnormalities and breakage of neuronal branches. Nature Neurosci. 7, 1181–1183 (2004).

    Article  CAS  PubMed  Google Scholar 

  52. Caughey, B. & Lansbury, P. T. Protofibrils, pores, fibrils, and neurodegeneration: separating the responsible protein aggregates from the innocent bystanders. Annu. Rev. Neurosci. 26, 267–298 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Teplow, D. B. Structural and kinetic features of amyloid β-protein fibrillogenesis. Amyloid 5, 121–142 (1998).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  58. Bitan, G. et al. Amyloid β-protein (Aβ) assembly: Aβ40 and Aβ42 oligomerize through distinct pathways. Proc. Natl Acad. Sci. USA 100, 330–335 (2003).

    Article  CAS  PubMed  Google Scholar 

  59. 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 

  60. Lambert, M. P. et al. Diffusible, nonfibrillar ligands derived from Aβ1–42 are potent central nervous system neurotoxins. Proc. Natl Acad. Sci. USA 95, 6448–6453 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Lesne, S. et al. A specific amyloid-β protein assembly in the brain impairs memory. Nature 440, 352–357 (2006). Identification of a brain-derived Aβ oligomeric assembly, which impairs memory.

    Article  CAS  PubMed  Google Scholar 

  63. Podlisny, M. B. et al. Aggregation of secreted amyloid β-protein into sodium dodecyl sulfate-stable oligomers in cell culture. J. Biol. Chem. 270, 9564–9570 (1995).

    Article  CAS  PubMed  Google Scholar 

  64. Walsh, D. M., Tseng, B. P., Rydel, R. E., Podlisny, M. B. & Selkoe, D. J. The oligomerization of amyloid β-protein begins intracellularly in cells derived from human brain. Biochemistry 39, 10831–10839 (2000).

    Article  CAS  PubMed  Google Scholar 

  65. Funato, H., Enya, M., Yoshimura, M., Morishima-Kawashima, M. & Ihara, Y. Presence of sodium dodecyl sulfate-stable amyloid β-protein dimers in the hippocampus CA1 not exhibiting neurofibrillary tangle formation. Am. J. Pathol. 155, 23–28 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Enya, M. et al. Appearance of sodium dodecyl sulfate-stable amyloid β-protein (Aβ) dimer in the cortex during aging. Am. J. Pathol. 154, 271–279 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Kawarabayashi, T. et al. Dimeric amyloid β protein rapidly accumulates in lipid rafts followed by apolipoprotein E and phosphorylated τ accumulation in the Tg2576 mouse model of Alzheimer's disease. J. Neurosci. 24, 3801–3809 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Roher, A. E. et al. Morphology and toxicity of Aβ-(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 

  69. 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). Defines a synaptotoxic function for small, soluble oligomers of secreted Aβ in vivo.

    Article  CAS  PubMed  Google Scholar 

  70. Kamenetz, F. et al. APP processing and synaptic function. Neuron 37, 925–937 (2003). Demonstrates the effects of Aβ on synaptic function upon the stimulation of neuronal activity.

    Article  CAS  PubMed  Google Scholar 

  71. Townsend, M., Shankar, G. M., Mehta, T., Walsh, D. M. & Selkoe, D. J. Effects of secreted oligomers of amyloid β-protein on hippocampal synaptic plasticity: a potent role for trimers. J. Physiol. 572, 477–492 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Klyubin, I. et al. Amyloid β protein immunotherapy neutralizes Aβ oligomers that disrupt synaptic plasticity in vivo. Nature Med. 11, 556–561 (2005). LTP inhibition by soluble oligomers of human Aβ is prevented by active and passive Aβ immunotherapy.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  74. Cirrito, J. R. et al. Synaptic activity regulates interstitial fluid amyloid-β levels in vivo. Neuron 48, 913–922 (2005). An in vivo demonstration of the effects of synaptic activity on Aβ levels.

    Article  CAS  PubMed  Google Scholar 

  75. Snyder, E. M. et al. Regulation of NMDA receptor trafficking by amyloid-β. Nature Neurosci. 8, 1051–1058 (2005). A cellular mechanism that describes how Aβ lowers NMDA-evoked currents.

    Article  CAS  PubMed  Google Scholar 

  76. Gong, B. et al. Ubiquitin hydrolase Uch-L1 rescues β-amyloid-induced decreases in synaptic function and contextual memory. Cell 126, 775–788 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  78. Glabe, C. G. & Kayed, R. Common structure and toxic function of amyloid oligomers implies a common mechanism of pathogenesis. Neurology 66, S74–S78 (2006).

    Article  CAS  PubMed  Google Scholar 

  79. Kayed, R. et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–489 (2003). Describes common conformational epitopes on oligomers of completely distinct amyloidogenic proteins.

    Article  CAS  PubMed  Google Scholar 

  80. Lashuel, H. A. et al. α-Synuclein, especially the Parkinson's disease-associated mutants, forms pore-like annular and tubular protofibrils. J. Mol. Biol. 322, 1089–1102 (2002).

    Article  CAS  PubMed  Google Scholar 

  81. Conway, K. A., Harper, J. D. & Lansbury, P. T. Jr. Fibrils formed in vitro from α-synuclein and two mutant forms linked to Parkinson's disease are typical amyloid. Biochemistry 39, 2552–2563 (2000).

    Article  CAS  PubMed  Google Scholar 

  82. Conway, K. A., Harper, J. D. & Lansbury, P. T. Accelerated in vitro fibril formation by a mutant α-synuclein linked to early-onset Parkinson disease. Nature Med. 4, 1318–1320 (1998).

    Article  CAS  PubMed  Google Scholar 

  83. Conway, K. A. et al. Acceleration of oligomerization, not fibrillization, is a shared property of both α-synuclein mutations linked to early-onset Parkinson's disease: implications for pathogenesis and therapy. Proc. Natl Acad. Sci. USA 97, 571–576 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Marchut, A. J. & Hall, C. K. Spontaneous formation of annular structures observed in molecular dynamics simulations of polyglutamine peptides. Comput. Biol. Chem. 30, 215–218 (2006).

    Article  CAS  PubMed  Google Scholar 

  85. Srinivasan, R., Marchant, R. E. & Zagorski, M. G. ABri peptide associated with familial British dementia forms annular and ring-like protofibrillar structures. Amyloid 11, 10–13 (2004).

    Article  CAS  PubMed  Google Scholar 

  86. Wille, H., Drewes, G., Biernat, J., Mandelkow, E. M. & Mandelkow, E. Alzheimer-like paired helical filaments and antiparallel dimers formed from microtubule-associated protein τ in vitro. J. Cell. Biol. 118, 573–584 (1992).

    Article  CAS  PubMed  Google Scholar 

  87. Friedhoff, P., von Bergen, M., Mandelkow, E. M., Davies, P. & Mandelkow, E. A nucleated assembly mechanism of Alzheimer paired helical filaments. Proc. Natl Acad. Sci. USA 95, 15712–15717 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Neumann, M. et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314, 130–133 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Willem, M. et al. Control of peripheral nerve myelination by the β-secretase BACE1. Science 314, 664–666 (2006).

    Article  CAS  PubMed  Google Scholar 

  90. Schenk, D. et al. Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400, 173–177 (1999). Initial report of the beneficial effects of Aβ immunotherapy in a transgenic mouse model of AD.

    Article  CAS  PubMed  Google Scholar 

  91. Bard, F. et al. Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nature Med. 6, 916–919 (2000).

    Article  CAS  PubMed  Google Scholar 

  92. Orgogozo, J. M. et al. Subacute meningoencephalitis in a subset of patients with AD after Aβ42 immunization. Neurology 61, 46–54 (2003).

    Article  CAS  PubMed  Google Scholar 

  93. Hock, C. et al. Antibodies against β-amyloid slow cognitive decline in Alzheimer's disease. Neuron 38, 547–554 (2003). First report of the beneficial effects of Aβ immunotherapy in a small cohort of vaccinated patients with AD.

    Article  CAS  PubMed  Google Scholar 

  94. Gilman, S. et al. Clinical effects of Aβ immunization (AN1792) in patients with AD in an interrupted trial. Neurology 64, 1553–1562 (2005).

    Article  CAS  PubMed  Google Scholar 

  95. Nicoll, J. A. et al. Neuropathology of human Alzheimer disease after immunization with amyloid-β peptide: a case report. Nature Med. 9, 448–452 (2003). First report of the apparent removal of Aβ deposits in humans by a therapeutic agent.

    Article  CAS  PubMed  Google Scholar 

  96. Patton, R. L. et al. Amyloid-β peptide remnants in AN-1792-immunized Alzheimer's disease patients: a biochemical analysis. Am. J. Pathol. 169, 1048–1063 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Oddo, S., Billings, L., Kesslak, J. P., Cribbs, D. H. & LaFerla, F. M. Aβ immunotherapy leads to clearance of early, but not late, hyperphosphorylated τ aggregates via the proteasome. Neuron 43, 321–332 (2004). Further evidence of a linear connection between Aβ deposition and tau hyperphosphorylation in an animal model.

    Article  CAS  PubMed  Google Scholar 

  98. DeMattos, R. B., Bales, K. R., Cummins, D. J., Paul, S. M. & Holtzman, D. M. Brain to plasma amyloid-β efflux: a measure of brain amyloid burden in a mouse model of Alzheimer's disease. Science 295, 2264–2267 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  100. Janus, C. et al. A β peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease. Nature 408, 979–982 (2000).

    Article  CAS  PubMed  Google Scholar 

  101. Gelinas, D. S., DaSilva, K., Fenili, D., St George-Hyslop, P. & McLaurin, J. Immunotherapy for Alzheimer's disease. Proc. Natl Acad. Sci. USA 101, 14657–14662 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Selkoe, D. J. & Schenk, D. Alzheimer's disease: molecular understanding predicts amyloid-based therapeutics. Annu. Rev. Pharmacol. Toxicol. 43, 545–584 (2003).

    Article  CAS  PubMed  Google Scholar 

  103. McLaurin, J. et al. Therapeutically effective antibodies against amyloid-β peptide target amyloid-β residues 4–10 and inhibit cytotoxicity and fibrillogenesis. Nature Med. 8, 1263–1269 (2002). Important mechanistic insights about how Aβ immunotherapy can prevent oligomerization and cytotoxicity.

    Article  CAS  PubMed  Google Scholar 

  104. Solomon, B., Koppel, R., Hanan, E. & Katzav, T. Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer β-amyloid peptide. Proc. Natl Acad. Sci. USA 93, 452–455 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Qiu, W. Q. et al. Insulin-degrading enzyme regulates extracellular levels of amyloid β- protein by degradation. J. Biol. Chem. 273, 32730–32738 (1998).

    Article  CAS  PubMed  Google Scholar 

  106. Tucker, H. M. et al. The plasmin system is induced by and degrades amyloid-β aggregates. J. Neurosci. 20, 3937–3946 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Mueller-Steiner, S. et al. Antiamyloidogenic and neuroprotective functions of cathepsin B: implications for Alzheimer's disease. Neuron 51, 703–714 (2006).

    Article  CAS  PubMed  Google Scholar 

  108. Iwata, N. et al. Identification of the major Aβ1–42-degrading catabolic pathway in brain parenchyma: suppression leads to biochemical and pathological deposition. Nature Med. 6, 143–150 (2000).

    Article  CAS  PubMed  Google Scholar 

  109. Leissring, M. A. et al. Enhanced proteolysis of β-amyloid in APP transgenic mice prevents plaque formation, secondary pathology, and premature death. Neuron 40, 1087–1093 (2003).

    Article  CAS  PubMed  Google Scholar 

  110. Huang, S. M. et al. Neprilysin-sensitive synapse-associated Aβ oligomers impair neuronal plasticity and cognitive function. J. Biol. Chem. 281, 17941–17951 (2006).

    Article  CAS  PubMed  Google Scholar 

  111. McLaurin, J., Franklin, T., Zhang, X., Deng, J. & Fraser, P. E. Interactions of Alzheimer amyloid-β peptides with glycosaminoglycans effects on fibril nucleation and growth. Eur. J. Biochem. 266, 1101–1110 (1999).

    Article  CAS  PubMed  Google Scholar 

  112. McLaurin, J., Golomb, R., Jurewicz, A., Antel, J. P. & Fraser, P. E. Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid β peptide and inhibit aβ-induced toxicity. J. Biol. Chem. 275, 18495–18502 (2000).

    Article  CAS  PubMed  Google Scholar 

  113. McLaurin, J. et al. Cyclohexanehexol inhibitors of Aβ aggregation prevent and reverse Alzheimer phenotype in a mouse model. Nature Med. 12, 801–808 (2006). New Aβ-aggregation inhibitors show beneficial effects on plaque burden and behaviour in mice.

    Article  CAS  PubMed  Google Scholar 

  114. Weggen, S. et al. A subset of NSAIDs lower amyloidogenic Aβ42 independently of cyclooxygenase activity. Nature 414, 212–216 (2001). Discovery of certain NSAIDs as γ-secretase modulators: they lead to shorter, less amyloidogenic Aβ species.

    Article  CAS  PubMed  Google Scholar 

  115. Doerfler, P., Shearman, M. S. & Perlmutter, R. M. Presenilin-dependent γ-secretase activity modulates thymocyte development. Proc. Natl Acad. Sci. USA 98, 9312–9317 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Geling, A., Steiner, H., Willem, M., Bally-Cuif, L. & Haass, C. A γ-secretase inhibitor blocks Notch signaling in vivo and causes a severe neurogenic phenotype in zebrafish. EMBO Rep. 3, 688–694 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Scheuner, D. et al. Secreted amyloid β-protein similar to that in the senile plaques of Alzheimer's disease is increased in vivo by the presenilin 1 and 2 and APP mutations linked to familial Alzheimer's disease. Nature Med. 2, 864–870 (1996).

    Article  CAS  PubMed  Google Scholar 

  118. Suzuki, N. et al. An increased percentage of long amyloid β-protein secreted by familial amyloid β-protein precursor (βAPP717) mutants. Science 264, 1336–1340 (1994).

    Article  CAS  PubMed  Google Scholar 

  119. Burdick, D. et al. Assembly and aggregation properties of synthetic Alzheimer's A4/β amyloid peptide analogs. J. Biol. Chem. 267, 546–554 (1992).

    CAS  PubMed  Google Scholar 

  120. Jarrett, J. T., Berger, E. P. & Lansbury, P. T. Jr. The carboxy terminus of the β-amyloid protein is critical for the seeding of amyloid formation: implications for the pathogenesis of Alzheimer's disease. Biochemistry 32, 4693–4697 (1993).

    Article  CAS  PubMed  Google Scholar 

  121. Chen, Y. R. & Glabe, C. G. Distinct early folding and aggregation properties of Alzheimer amyloid-β peptides Aβ40 and Aβ42: stable trimer or tetramer formation by Aβ42. J. Biol. Chem. 281, 24414–24422 (2006).

    Article  CAS  PubMed  Google Scholar 

  122. Nilsberth, C. et al. The 'Arctic' APP mutation (E693G) causes Alzheimer's disease by enhanced Aβ protofibril formation. Nature Neurosci. 4, 887–893 (2001). A genetic explanation for the development of rare forms of AD that is strongly supportive of the amyloid hypothesis.

    Article  CAS  PubMed  Google Scholar 

  123. Iwatsubo, T. et al. Visualization of Aβ42(43) and Aβ40 in senile plaques with end-specific A β monoclonals: evidence that an initially deposited species is Aβ42(43). Neuron 13, 45–53 (1994).

    Article  CAS  PubMed  Google Scholar 

  124. Cai, X. D., Golde, T. E. & Younkin, S. G. Release of excess amyloid β protein from a mutant amyloid β protein precursor. Science 259, 514–516 (1993).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  126. Rovelet-Lecrux, A. et al. APP locus duplication causes autosomal dominant early-onset Alzheimer disease with cerebral amyloid angiopathy. Nature Genet. 38, 24–26 (2006).

    Article  CAS  PubMed  Google Scholar 

  127. Singleton, A. B. et al. α-Synuclein locus triplication causes Parkinson's disease. Science 302, 841 (2003).

    Article  CAS  PubMed  Google Scholar 

  128. McGowan, E. et al. Aβ42 is essential for parenchymal and vascular amyloid deposition in mice. Neuron 47, 191–199 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Bentahir, M. et al. Presenilin clinical mutations can affect γ-secretase activity by different mechanisms. J. Neurochem. 96, 732–742 (2006).

    Article  CAS  PubMed  Google Scholar 

  130. Deng, Y. et al. Deletion of presenilin 1 hydrophilic loop sequence leads to impaired γ-secretase activity and exacerbated amyloid pathology. J. Neurosci. 26, 3845–3854 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D. Teplow, G. Multhaup, E. Mandelkow and D. Walsh as well as the members of our laboratories for helpful discussions. We also thank M. Neumann for providing the figures that are included in Box 1.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

Christian Haass is a consultant for Boehringer Ingelheim Pharma KG. Dennis J. Selkoe is a consultant for Elan, plc.

Related links

Related links

DATABASES

OMIM

Alzheimer's disease

Down syndrome

Huntington's disease

Parkinson's disease

FURTHER INFORMATION

Christian Haass' homepage

Dennis J. Selkoe's homepage

Alzforum

Glossary

Amyloid

Tissue deposits of insoluble, proteinaceous fibrils that are rich in β-pleated sheet structure and therefore bind to the histochemical dye Congo red in a polarized manner.

Lewy body

(LB). A deposit of α-synuclein typically found in neuronal cell bodies of patients with Parkinson's disease or related disorders.

Lewy neurite

(LN). A deposit of α-synuclein typically found in swollen neuronal processes of patients with Parkinson's disease or related disorders.

Amyloid plaques

Spherical extracellular deposits predominantly composed of the amyloid β-peptide and found in the brains of all cases of Alzheimer's disease.

Neurofibrillary tangle

An intraneuronal filamentous aggregate composed of abnormally phosphorylated tau protein found in several human neurodegenerative disorders, including Alzheimer's disease.

Regulated intramembrane proteolysis

(RIP). Regulated cleavage of the luminal domain of certain membrane proteins is followed by a constitutive cleavage in the transmembrane domain. At least in some cases this process is involved in signalling pathways.

Intracellular domain

(ICD). One of the cleavage products of intramembrane proteolysis in the RIP pathway, it is liberated into the cytosol and in some cases targeted to the nucleus.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Haass, C., Selkoe, D. Soluble protein oligomers in neurodegeneration: lessons from the Alzheimer's amyloid β-peptide. Nat Rev Mol Cell Biol 8, 101–112 (2007). https://doi.org/10.1038/nrm2101

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrm2101

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