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.

  • Perspective
  • Published:

Three dimensions of the amyloid hypothesis: time, space and 'wingmen'

Abstract

The amyloid hypothesis, which has been the predominant framework for research in Alzheimer's disease (AD), has been the source of considerable controversy. The amyloid hypothesis postulates that amyloid-β peptide (Aβ) is the causative agent in AD. It is strongly supported by data from rare autosomal dominant forms of AD. However, the evidence that Aβ causes or contributes to age-associated sporadic AD is more complex and less clear, prompting criticism of the hypothesis. We provide an overview of the major arguments for and against the amyloid hypothesis. We conclude that Aβ likely is the key initiator of a complex pathogenic cascade that causes AD. However, we argue that Aβ acts primarily as a trigger of other downstream processes, particularly tau aggregation, which mediate neurodegeneration. Aβ appears to be necessary, but not sufficient, to cause AD. Its major pathogenic effects may occur very early in the disease process.

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: An updated framework of the amyloid hypothesis.

Similar content being viewed by others

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Bettens, K., Sleegers, K. & Van Broeckhoven, C. Genetic insights in Alzheimer's disease. Lancet Neurol. 12, 92–104 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. 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 

  4. 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 

  5. Tsubuki, S., Takaki, Y. & Saido, T.C. Dutch, Flemish, Italian, and Arctic mutations of APP and resistance of Abeta to physiologically relevant proteolytic degradation. Lancet 361, 1957–1958 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Tomiyama, T. et al. A new amyloid beta variant favoring oligomerization in Alzheimer's-type dementia. Ann. Neurol. 63, 377–387 (2008).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Sleegers, K. et al. APP duplication is sufficient to cause early onset Alzheimer's dementia with cerebral amyloid angiopathy. Brain 129, 2977–2983 (2006).

    Article  PubMed  Google Scholar 

  9. Cabrejo, L. et al. Phenotype associated with APP duplication in five families. Brain 129, 2966–2976 (2006).

    Article  PubMed  Google Scholar 

  10. Prasher, V.P. et al. Molecular mapping of Alzheimer-type dementia in Down's syndrome. Ann. Neurol. 43, 380–383 (1998).

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. 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 

  13. Chávez-Gutiérrez, L. et al. The mechanism of gamma-Secretase dysfunction in familial Alzheimer disease. EMBO J. 31, 2261–2274 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Shepherd, C., McCann, H. & Halliday, G.M. Variations in the neuropathology of familial Alzheimer's disease. Acta Neuropathol. 118, 37–52 (2009).

    Article  CAS  PubMed  Google Scholar 

  15. Bateman, R.J. et al. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N. Engl. J. Med. 367, 795–804 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Ryman, D.C. et al. Symptom onset in autosomal dominant Alzheimer disease: a systematic review and meta-analysis. Neurology 83, 253–260 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Corder, E.H. et al. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science 261, 921–923 (1993).

    Article  CAS  PubMed  Google Scholar 

  18. Chiang, G.C. et al. Hippocampal atrophy rates and CSF biomarkers in elderly APOE2 normal subjects. Neurology 75, 1976–1981 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Verghese, P.B., Castellano, J.M. & Holtzman, D.M. Apolipoprotein E in Alzheimer's disease and other neurological disorders. Lancet Neurol. 10, 241–252 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Castellano, J.M. et al. Human apoE isoforms differentially regulate brain amyloid-beta peptide clearance. Sci. Transl. Med. 3, 89ra57 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fagan, A.M. et al. Human and murine ApoE markedly alters A beta metabolism before and after plaque formation in a mouse model of Alzheimer's disease. Neurobiol. Dis. 9, 305–318 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Hudry, E. et al. Gene transfer of human Apoe isoforms results in differential modulation of amyloid deposition and neurotoxicity in mouse brain. Sci. Transl. Med. 5, 212ra161 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Morris, J.C. et al. APOE predicts amyloid-beta but not tau Alzheimer pathology in cognitively normal aging. Ann. Neurol. 67, 122–131 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Vemuri, P. et al. Effect of apolipoprotein E on biomarkers of amyloid load and neuronal pathology in Alzheimer disease. Ann. Neurol. 67, 308–316 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Verghese, P.B. et al. ApoE influences amyloid-beta (Abeta) clearance despite minimal apoE/Abeta association in physiological conditions. Proc. Natl. Acad. Sci. USA 110, E1807–E1816 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Bales, K.R. et al. Lack of apolipoprotein E dramatically reduces amyloid beta-peptide deposition. Nat. Genet. 17, 263–264 (1997).

    Article  CAS  PubMed  Google Scholar 

  27. Bien-Ly, N., Gillespie, A.K., Walker, D., Yoon, S.Y. & Huang, Y. Reducing human apolipoprotein E levels attenuates age-dependent Abeta accumulation in mutant human amyloid precursor protein transgenic mice. J. Neurosci. 32, 4803–4811 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kim, J. et al. Haploinsufficiency of human APOE reduces amyloid deposition in a mouse model of amyloid-beta amyloidosis. J. Neurosci. 31, 18007–18012 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kim, J. et al. Anti-apoE immunotherapy inhibits amyloid accumulation in a transgenic mouse model of Abeta amyloidosis. J. Exp. Med. 209, 2149–2156 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Sunderland, T. et al. Cerebrospinal fluid beta-amyloid1–42 and tau in control subjects at risk for Alzheimer's disease: the effect of APOE epsilon4 allele. Biol. Psychiatry 56, 670–676 (2004).

    Article  CAS  PubMed  Google Scholar 

  31. Jonsson, T. et al. A mutation in APP protects against Alzheimer's disease and age-related cognitive decline. Nature 488, 96–99 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Braak, H. & Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 82, 239–259 (1991).

    Article  CAS  PubMed  Google Scholar 

  33. Serrano-Pozo, A., Frosch, M.P., Masliah, E. & Hyman, B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 1, a006189 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Gómez-Isla, T. et al. Neuronal loss correlates with but exceeds neurofibrillary tangles in Alzheimer's disease. Ann. Neurol. 41, 17–24 (1997).

    Article  PubMed  Google Scholar 

  35. Arriagada, P.V., Marzloff, K. & Hyman, B.T. Distribution of Alzheimer-type pathologic changes in nondemented elderly individuals matches the pattern in Alzheimer's disease. Neurology 42, 1681–1688 (1992).

    Article  CAS  PubMed  Google Scholar 

  36. Arriagada, P.V., Growdon, J.H., Hedley-Whyte, E.T. & Hyman, B.T. Neurofibrillary tangles but not senile plaques parallel duration and severity of Alzheimer's disease. Neurology 42, 631–639 (1992).

    Article  CAS  PubMed  Google Scholar 

  37. Price, J.L., Davis, P.B., Morris, J.C. & White, D.L. The distribution of tangles, plaques and related immunohistochemical markers in healthy aging and Alzheimer's disease. Neurobiol. Aging 12, 295–312 (1991).

    Article  CAS  PubMed  Google Scholar 

  38. Braak, H. & Del Tredici, K. The pathological process underlying Alzheimer's disease in individuals under thirty. Acta Neuropathol. 121, 171–181 (2011).

    Article  PubMed  Google Scholar 

  39. Price, J.L. & Morris, J.C. Tangles and plaques in nondemented aging and “preclinical” Alzheimer's disease. Ann. Neurol. 45, 358–368 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Elobeid, A., Soininen, H. & Alafuzoff, I. Hyperphosphorylated tau in young and middle-aged subjects. Acta Neuropathol. 123, 97–104 (2012).

    Article  CAS  PubMed  Google Scholar 

  41. Knopman, D.S. et al. Neuropathology of cognitively normal elderly. J. Neuropathol. Exp. Neurol. 62, 1087–1095 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Petersen, R.C. et al. Neuropathologic features of amnestic mild cognitive impairment. Arch. Neurol. 63, 665–672 (2006).

    Article  PubMed  Google Scholar 

  43. Gómez-Isla, T. et al. Profound loss of layer II entorhinal cortex neurons occurs in very mild Alzheimer's disease. J. Neurosci. 16, 4491–4500 (1996).

    Article  PubMed  PubMed Central  Google Scholar 

  44. West, M.J., Coleman, P.D., Flood, D.G. & Troncoso, J.C. Differences in the pattern of hippocampal neuronal loss in normal ageing and Alzheimer's disease. Lancet 344, 769–772 (1994).

    Article  CAS  PubMed  Google Scholar 

  45. Tiraboschi, P., Hansen, L.A., Thal, L.J. & Corey-Bloom, J. The importance of neuritic plaques and tangles to the development and evolution of AD. Neurology 62, 1984–1989 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Reed, L.A. et al. Autosomal dominant dementia with widespread neurofibrillary tangles. Ann. Neurol. 42, 564–572 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Lindquist, S.G. et al. Alzheimer disease-like clinical phenotype in a family with FTDP-17 caused by a MAPT R406W mutation. Eur. J. Neurol. 15, 377–385 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Kauwe, J.S. et al. Variation in MAPT is associated with cerebrospinal fluid tau levels in the presence of amyloid-beta deposition. Proc. Natl. Acad. Sci. USA 105, 8050–8054 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ferreira, A., Lu, Q., Orecchio, L. & Kosik, K.S. Selective phosphorylation of adult tau isoforms in mature hippocampal neurons exposed to fibrillar A beta. Mol. Cell. Neurosci. 9, 220–234 (1997).

    Article  CAS  PubMed  Google Scholar 

  50. Zempel, H., Thies, E., Mandelkow, E. & Mandelkow, E.M. Abeta oligomers cause localized Ca(2+) elevation, missorting of endogenous Tau into dendrites, Tau phosphorylation, and destruction of microtubules and spines. J. Neurosci. 30, 11938–11950 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rapoport, M., Dawson, H.N., Binder, L.I., Vitek, M.P. & Ferreira, A. Tau is essential to beta -amyloid-induced neurotoxicity. Proc. Natl. Acad. Sci. USA 99, 6364–6369 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Jin, M. et al. Soluble amyloid beta-protein dimers isolated from Alzheimer cortex directly induce Tau hyperphosphorylation and neuritic degeneration. Proc. Natl. Acad. Sci. USA 108, 5819–5824 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Choi, S.H. et al. A three-dimensional human neural cell culture model of Alzheimer's disease. Nature 515, 274–278 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hurtado, D.E. et al. A{beta} accelerates the spatiotemporal progression of tau pathology and augments tau amyloidosis in an Alzheimer mouse model. Am. J. Pathol. 177, 1977–1988 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Lewis, J. et al. Enhanced neurofibrillary degeneration in transgenic mice expressing mutant tau and APP. Science 293, 1487–1491 (2001).

    Article  CAS  PubMed  Google Scholar 

  56. Götz, J., Chen, F., van Dorpe, J. & Nitsch, R.M. Formation of neurofibrillary tangles in P301l tau transgenic mice induced by Abeta 42 fibrils. Science 293, 1491–1495 (2001).

    Article  PubMed  Google Scholar 

  57. Roberson, E.D. et al. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer's disease mouse model. Science 316, 750–754 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Roberson, E.D. et al. Amyloid-beta/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer's disease. J. Neurosci. 31, 700–711 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Lippa, C.F. et al. Lewy bodies contain altered alpha-synuclein in brains of many familial Alzheimer's disease patients with mutations in presenilin and amyloid precursor protein genes. Am. J. Pathol. 153, 1365–1370 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Hashimoto, M. & Masliah, E. Alpha-synuclein in Lewy body disease and Alzheimer's disease. Brain Pathol. 9, 707–720 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Masliah, E. et al. beta-amyloid peptides enhance alpha-synuclein accumulation and neuronal deficits in a transgenic mouse model linking Alzheimer's disease and Parkinson's disease. Proc. Natl. Acad. Sci. USA 98, 12245–12250 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Larson, M.E. et al. Soluble alpha-synuclein is a novel modulator of Alzheimer's disease pathophysiology. J. Neurosci. 32, 10253–10266 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Josephs, K.A. et al. TDP-43 is a key player in the clinical features associated with Alzheimer's disease. Acta Neuropathol. 127, 811–824 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Jack, C.R. Jr. et al. Tracking pathophysiological processes in Alzheimer's disease: an updated hypothetical model of dynamic biomarkers. Lancet Neurol. 12, 207–216 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Jack, C.R. Jr. et al. Evidence for ordering of Alzheimer disease biomarkers. Arch. Neurol. 68, 1526–1535 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Roe, C.M. et al. Amyloid imaging and CSF biomarkers in predicting cognitive impairment up to 7.5 years later. Neurology 80, 1784–1791 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Vos, S.J. et al. Preclinical Alzheimer's disease and its outcome: a longitudinal cohort study. Lancet Neurol. 12, 957–965 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Villemagne, V.L. et al. Amyloid beta deposition, neurodegeneration, and cognitive decline in sporadic Alzheimer's disease: a prospective cohort study. Lancet Neurol. 12, 357–367 (2013).

    Article  CAS  PubMed  Google Scholar 

  69. Chen, X. et al. Pittsburgh compound B retention and progression of cognitive status–a meta-analysis. Eur. J. Neurol. 21, 1060–1067 (2014).

    Article  CAS  PubMed  Google Scholar 

  70. Villemagne, V.L. et al. Longitudinal assessment of Abeta and cognition in aging and Alzheimer disease. Ann. Neurol. 69, 181–192 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Knopman, D.S. et al. Short-term clinical outcomes for stages of NIA-AA preclinical Alzheimer disease. Neurology 78, 1576–1582 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Tarawneh, R. et al. Visinin-like protein-1: diagnostic and prognostic biomarker in Alzheimer disease. Ann. Neurol. 70, 274–285 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Donohue, M.C. et al. The preclinical Alzheimer cognitive composite: measuring amyloid-related decline. JAMA Neurol. 71, 961–970 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chételat, G. et al. Accelerated cortical atrophy in cognitively normal elderly with high beta-amyloid deposition. Neurology 78, 477–484 (2012).

    Article  PubMed  CAS  Google Scholar 

  75. Shankar, G.M. et al. Amyloid-beta protein dimers isolated directly from Alzheimer's brains impair synaptic plasticity and memory. Nat. Med. 14, 837–842 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. 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  PubMed  Google Scholar 

  77. Mucke, L. & Selkoe, D.J. Neurotoxicity of amyloid beta-protein: synaptic and network dysfunction. Cold Spring Harb. Perspect. Med. 2, a006338 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Esparza, T.J. et al. Amyloid-beta oligomerization in Alzheimer dementia versus high-pathology controls. Ann. Neurol. 73, 104–119 (2013).

    Article  CAS  PubMed  Google Scholar 

  79. Tomic, J.L., Pensalfini, A., Head, E. & Glabe, C.G. Soluble fibrillar oligomer levels are elevated in Alzheimer's disease brain and correlate with cognitive dysfunction. Neurobiol. Dis. 35, 352–358 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. 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 

  81. Lesné, S.E. et al. Brain amyloid-beta oligomers in ageing and Alzheimer's disease. Brain 136, 1383–1398 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  82. Handoko, M. et al. Correlation of specific amyloid-beta oligomers with tau in cerebrospinal fluid from cognitively normal older adults. JAMA Neurol. 70, 594–599 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  83. Zhang, Y. et al. A lifespan observation of a novel mouse model: in vivo evidence supports abeta oligomer hypothesis. PLoS ONE 9, e85885 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Tomiyama, T. et al. A mouse model of amyloid beta oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. J. Neurosci. 30, 4845–4856 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  86. Ma, Q.L. et al. Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: suppression by omega-3 fatty acids and curcumin. J. Neurosci. 29, 9078–9089 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  88. Haass, C. & Mandelkow, E. Fyn-tau-amyloid: a toxic triad. Cell 142, 356–358 (2010).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Martin, L. et al. Tau protein kinases: involvement in Alzheimer's disease. Ageing Res. Rev. 12, 289–309 (2013).

    Article  CAS  PubMed  Google Scholar 

  91. 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 

  92. Sanders, D.W. et al. Distinct tau prion strains propagate in cells and mice and define different tauopathies. Neuron 82, 1271–1288 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. de Calignon, A. et al. Propagation of tau pathology in a model of early Alzheimer's disease. Neuron 73, 685–697 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Liu, L. et al. Trans-synaptic spread of tau pathology in vivo. PLoS ONE 7, e31302 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Guo, J.L. et al. Distinct alpha-synuclein strains differentially promote tau inclusions in neurons. Cell 154, 103–117 (2013).

    Article  CAS  PubMed  Google Scholar 

  96. Giasson, B.I. et al. Initiation and synergistic fibrillization of tau and alpha-synuclein. Science 300, 636–640 (2003).

    Article  CAS  PubMed  Google Scholar 

  97. Dasuri, K., Zhang, L. & Keller, J.N. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radic. Biol. Med. 62, 170–185 (2013).

    Article  CAS  PubMed  Google Scholar 

  98. De Strooper, B. Proteases and proteolysis in Alzheimer disease: a multifactorial view on the disease process. Physiol. Rev. 90, 465–494 (2010).

    Article  CAS  PubMed  Google Scholar 

  99. Nixon, R.A. & Yang, D.S. Autophagy failure in Alzheimer's disease–locating the primary defect. Neurobiol. Dis. 43, 38–45 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Taylor, R.C. & Dillin, A. Aging as an event of proteostasis collapse. Cold Spring Harb. Perspect. Biol. 3, a004440 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Lu, T. et al. REST and stress resistance in ageing and Alzheimer's disease. Nature 507, 448–454 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Praticò, D., Uryu, K., Leight, S., Trojanoswki, J.Q. & Lee, V.M. Increased lipid peroxidation precedes amyloid plaque formation in an animal model of Alzheimer amyloidosis. J. Neurosci. 21, 4183–4187 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  103. Benzing, W.C. et al. Evidence for glial-mediated inflammation in aged APP(SW) transgenic mice. Neurobiol. Aging 20, 581–589 (1999).

    Article  CAS  PubMed  Google Scholar 

  104. Calkins, M.J., Manczak, M., Mao, P., Shirendeb, U. & Reddy, P.H. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer's disease. Hum. Mol. Genet. 20, 4515–4529 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Guix, F.X. et al. Modification of gamma-secretase by nitrosative stress links neuronal ageing to sporadic Alzheimer's disease. EMBO Mol. Med. 4, 660–673 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Wahlster, L. et al. Presenilin-1 adopts pathogenic conformation in normal aging and in sporadic Alzheimer's disease. Acta Neuropathol. 125, 187–199 (2013).

    Article  CAS  PubMed  Google Scholar 

  107. Kukreja, L., Kujoth, G.C., Prolla, T.A., Van Leuven, F. & Vassar, R. Increased mtDNA mutations with aging promotes amyloid accumulation and brain atrophy in the APP/Ld transgenic mouse model of Alzheimer's disease. Mol. Neurodegener. 9, 16 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Liu, Y. et al. IKKbeta deficiency in myeloid cells ameliorates Alzheimer's disease-related symptoms and pathology. J. Neurosci. 34, 12982–12999 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  109. Durazzo, T.C., Mattsson, N. & Weiner, M.W. Smoking and increased Alzheimer's disease risk: a review of potential mechanisms. Alzheimers Dement. 10, S122–S145 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  110. Moreno-Gonzalez, I., Estrada, L.D., Sanchez-Mejias, E. & Soto, C. Smoking exacerbates amyloid pathology in a mouse model of Alzheimer's disease. Nat. Commun. 4, 1495 (2013).

    Article  PubMed  CAS  Google Scholar 

  111. Ulrich, J.D. et al. Altered microglial response to Abeta plaques in APPPS1–21 mice heterozygous for TREM2. Mol. Neurodegener. 9, 20 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Cady, J. et al. TREM2 variant p.R47H as a risk factor for sporadic amyotrophic lateral sclerosis. JAMA Neurol. 71, 449–453 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  113. Rayaprolu, S. et al. TREM2 in neurodegeneration: evidence for association of the p.R47H variant with frontotemporal dementia and Parkinson's disease. Mol. Neurodegener. 8, 19 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Melnikova, T. et al. Reversible pathologic and cognitive phenotypes in an inducible model of Alzheimer-amyloidosis. J. Neurosci. 33, 3765–3779 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Kim, J. et al. Normal cognition in transgenic BRI2-Abeta mice. Mol. Neurodegener. 8, 15 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Berger-Sweeney, J. et al. Impairments in learning and memory accompanied by neurodegeneration in mice transgenic for the carboxyl-terminus of the amyloid precursor protein. Brain Res. Mol. Brain Res. 66, 150–162 (1999).

    Article  CAS  PubMed  Google Scholar 

  117. Gao, Y. & Pimplikar, S.W. The gamma -secretase-cleaved C-terminal fragment of amyloid precursor protein mediates signaling to the nucleus. Proc. Natl. Acad. Sci. USA 98, 14979–14984 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. 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 

  119. Zheng, H. & Koo, E.H. Biology and pathophysiology of the amyloid precursor protein. Mol. Neurodegener. 6, 27 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Bero, A.W. et al. Neuronal activity regulates the regional vulnerability to amyloid-beta deposition. Nat. Neurosci. 14, 750–756 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Kang, J.E. et al. Amyloid-beta dynamics are regulated by orexin and the sleep-wake cycle. Science 326, 1005–1007 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  122. Xie, L. et al. Sleep drives metabolite clearance from the adult brain. Science 342, 373–377 (2013).

    Article  CAS  PubMed  Google Scholar 

  123. Siegel, S.J., Bieschke, J., Powers, E.T. & Kelly, J.W. The oxidative stress metabolite 4-hydroxynonenal promotes Alzheimer protofibril formation. Biochemistry 46, 1503–1510 (2007).

    Article  CAS  PubMed  Google Scholar 

  124. Head, E. et al. Oxidation of Abeta and plaque biogenesis in Alzheimer's disease and Down syndrome. Neurobiol. Dis. 8, 792–806 (2001).

    Article  CAS  PubMed  Google Scholar 

  125. Kress, B.T. et al. Impairment of paravascular clearance pathways in the aging brain. Ann. Neurol. 76, 845–861 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Zhao, W., Zhang, J., Davis, E.G. & Rebeck, G.W. Aging reduces glial uptake and promotes extracellular accumulation of Abeta from a lentiviral vector. Front. Aging Neurosci. 6, 210 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  127. Mawuenyega, K.G. et al. Decreased clearance of CNS beta-amyloid in Alzheimer's disease. Science 330, 1774 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Hipp, M.S., Park, S.H. & Hartl, F.U. Proteostasis impairment in protein-misfolding and -aggregation diseases. Trends Cell Biol. 24, 506–514 (2014).

    Article  CAS  PubMed  Google Scholar 

  129. Holmes, C. et al. Long-term effects of Abeta42 immunization in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 372, 216–223 (2008).

    Article  CAS  PubMed  Google Scholar 

  130. Boche, D. et al. Reduction of aggregated Tau in neuronal processes but not in the cell bodies after Abeta42 immunisation in Alzheimer's disease. Acta Neuropathol. 120, 13–20 (2010).

    Article  CAS  PubMed  Google Scholar 

  131. Serrano-Pozo, A. et al. Beneficial effect of human anti-amyloid-beta active immunization on neurite morphology and tau pathology. Brain 133, 1312–1327 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  132. Vellas, B. et al. Long-term follow-up of patients immunized with AN1792: reduced functional decline in antibody responders. Curr. Alzheimer Res. 6, 144–151 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Mills, S.M. et al. Preclinical trials in autosomal dominant AD: implementation of the DIAN-TU trial. Rev. Neurol. (Paris) 169, 737–743 (2013).

    Article  CAS  Google Scholar 

  134. Reiman, E.M. et al. Alzheimer's Prevention Initiative: a plan to accelerate the evaluation of presymptomatic treatments. J. Alzheimers Dis. 26 (suppl. 3), 321–329 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

E.S.M. is supported by NINDS grant K08NS079405 and Alzheimer's Association grant NIRG-305476. D.M.H. is supported by NIH grants R01 NS090934, R01 AG047644, P01 NS074969, P01 NS080675, PO1-AG03991, R01 NS034467, P01-AG026276, and U01 AG032438 and from the Tau Consortium and Cure Alzheimer's Fund.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to David M Holtzman.

Ethics declarations

Competing interests

D.M.H. is a co-founder of C2N Diagnostics, LLC, a member of the scientific advisory board of C2N Diagnostics, and a consultant for Genentech, AstraZeneca, Neurophage and Eli Lilly. Washington University receives grants for the laboratory of D.M.H. from C2N Diagnostics, Eli Lilly and Janssen.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Musiek, E., Holtzman, D. Three dimensions of the amyloid hypothesis: time, space and 'wingmen'. Nat Neurosci 18, 800–806 (2015). https://doi.org/10.1038/nn.4018

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4018

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