Skip to main content
Log in

β-amyloid Peptides and Amyloid Plaques in Alzheimer’s Disease

  • Review
  • Published:
Neurotherapeutics

Abstract

Many lines of evidence support that β-amyloid (Aβ) peptides play an important role in Alzheimer’s disease (AD), the most common cause of dementia. But despite much effort the molecular mechanisms of how Aβ contributes to AD remain unclear. While Aβ is generated from its precursor protein throughout life, the peptide is best known as the main component of amyloid plaques, the neuropathological hallmark of AD. Reduction in Aβ has been the major target of recent experimental therapies against AD. Unfortunately, human clinical trials targeting Aβ have not shown the hoped-for benefits. Thus, doubts have been growing about the role of Aβ as a therapeutic target. Here we review evidence supporting the involvement of Aβ in AD, highlight the importance of differentiating between various forms of Aβ, and suggest that a better understanding of Aβ’s precise pathophysiological role in the disease is important for correctly targeting it for potential future therapy.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Chávez-Gutiérrez L, Bammens L, Benilova I, et al. The mechanism of γ-secretase dysfunction in familial Alzheimer disease. EMBO J 2012;31:2261-2274.

    Article  PubMed Central  PubMed  Google Scholar 

  2. Haass C, Kaether C, Thinakaran G, Sisodia S. Trafficking and Proteolytic processing of APP. Cold Spring Harb Perspect Med 2012;2:a006270-a006270.

    Article  PubMed Central  PubMed  Google Scholar 

  3. Buchhave P, Minthon L, Zetterberg H, Wallin ÅK, Blennow K, Hansson O. Cerebrospinal fluid levels of β-amyloid 1-42, but not of tau, are fully changed already 5 to 10 years before the onset of Alzheimer dementia. Arch Gen Psychiatry 2012;69:98-106.

    Article  CAS  PubMed  Google Scholar 

  4. Bateman RJ, Xiong C, Benzinger TLS, et al. Clinical and biomarker changes in dominantly inherited Alzheimer's disease. N Engl J Med 2012;367:795-804.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  5. Takahashi RH. Oligomerization of Alzheimer's—amyloid within processes and synapses of cultured neurons and brain. J Neurosci 2004;24:3592-3599.

    Article  CAS  PubMed  Google Scholar 

  6. Gouras GK, Tampellini D, Takahashi RH, Capetillo-Zarate E. Intraneuronal β-amyloid accumulation and synapse pathology in Alzheimer’s disease. Acta Neuropathol 2010;119:523-541.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  7. Mucke L, Masliah E, Yu G-Q, et al. High-level neuronal expression of Aβ1-42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci 2000;20:4050-4058.

    CAS  PubMed  Google Scholar 

  8. Hong S, Ostaszewski BL, Yang T, et al. Soluble Ab oligomers are rapidly sequestered from brain ISF in vivo and bind GM1 ganglioside on cellular membranes. Neuron 2014;82:1-12.

    Article  Google Scholar 

  9. Upadhaya AR, Lungrin I, Yamaguchi H, Fändrich M, Thal DR. High-molecular weight Aβ oligomers and protofibrils are the predominant Aβ species in the native soluble protein fraction of the AD brain. J Cell Mol Med 2012;16:287-295.

    Article  CAS  PubMed  Google Scholar 

  10. Schöll M, Wall A, Thordardottir S, et al. Low PiB PET retention in presence of pathologic CSF biomarkers in Arctic APP mutation carriers. Neurology 2012;79:229-236.

    Article  PubMed  Google Scholar 

  11. Shimada H, Ataka S, Tomiyama T, Takechi H, Mori H, Miki T. Clinical course of patients with familial early-onset Alzheimer’s disease potentially lacking senile plaques bearing the E693Δ mutation in amyloid precursor protein. Dement Geriatr Cogn Disord 2011;32:45-54.

    Article  PubMed  Google Scholar 

  12. Tomiyama T, Matsuyama S, Iso H, et al. A mouse model of amyloid oligomers: their contribution to synaptic alteration, abnormal tau phosphorylation, glial activation, and neuronal loss in vivo. J Neurosci 2010;30:4845-4856.

    Article  CAS  PubMed  Google Scholar 

  13. Pozueta J, Lefort R, Shelanski ML. Synaptic changes in Alzheimer's disease and its models. Neuroscience 2013;251:51-65.

    Article  CAS  PubMed  Google Scholar 

  14. Mucke L, Selkoe DJ. Neurotoxicity of amyloid β-protein: synaptic and network dysfunction. Cold Spring Harb Perspect Med 2012;2:a006338.

    Article  PubMed Central  PubMed  Google Scholar 

  15. Mohajeri MH, Saini K, Schultz JG, et al. Passive immunization against beta -amyloid peptide protects central nervous system (CNS) neurons from increased vulnerability associated with an Alzheimer's disease-causing mutation. J Biol Chem 2002;277:33012-33017.

    Article  CAS  PubMed  Google Scholar 

  16. Tampellini D, Capetillo-Zarate E, Dumont M, et al. Effects of synaptic modulation on beta-amyloid, synaptophysin, and memory performance in Alzheimer's disease transgenic mice. J Neurosci 2010;30:14299-14304.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  17. Kim J, Chakrabarty P, Hanna A, et al. Normal cognition in transgenic BRI2-Aβ mice. Mol Neurodegeneration 2013;8:15.

    Article  CAS  Google Scholar 

  18. Dodart J-C, Bales KR, Gannon KS, et al. Immunization reverses memory deficits without reducing brain Aβ burden in Alzheimer's disease model. Nat Neurosci 2002;5:452-457.

    CAS  PubMed  Google Scholar 

  19. Holmes C, Boche D, Wilkinson D, et al. Long-term effects of Abeta42 immunisation in Alzheimer's disease: follow-up of a randomised, placebo-controlled phase I trial. Lancet 2008;372:216-223.

    Article  CAS  PubMed  Google Scholar 

  20. Esbjörner EK, Chan F, Rees E, et al. Direct observations of amyloid β self-assembly in live cells provide insights into differences in the kinetics of Ab(1–40) and Ab(1–42) aggregation. Chem Biol 2014;21:732-742.

    Article  PubMed Central  PubMed  Google Scholar 

  21. Takahashi RH, Milner TA, Li F, et al. Intraneuronal Alzheimer Aβ42 accumulates in multivesicular bodies and is associated with synaptic pathology. Am J Pathol 2002;161:1869-1879.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Holtzman DM, Herz J, Bu G. Apolipoprotein E and Apolipoprotein E receptors: normal biology and roles in Alzheimer disease. Cold Spring Harb Perspect Med 2012;2:a006312-a006312.

    PubMed Central  PubMed  Google Scholar 

  23. Kuszczyk MA, Sanchez S, Pankiewicz J, et al. Blocking the interaction between apolipoprotein E and Aβ reduces intraneuronal accumulation of Aβ and inhibits synaptic degeneration. Am J Pathol 2013;182:1750-1768.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  24. Hardy J, Bogdanovic N, Winblad B, et al. Pathways to Alzheimer's disease. J Inter Med 2014;275:296-303.

    Article  CAS  Google Scholar 

  25. Small S, Gandy S. Sorting through the cell biology of Alzheimer's disease: intracellular pathways to pathogenesis. Neuron 2006;52:15-31.

    Article  CAS  PubMed  Google Scholar 

  26. Restituito S, Khatri L, Ninan I, et al. Synaptic autoregulation by metalloproteases and γ-secretase. J Neurosci 2011;31:12083-12093.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  27. Bittner T, Fuhrmann M, Burgold S, et al. Gamma-secretase inhibition reduces spine density in vivo via an amyloid precursor protein-dependent pathway. J Neurosci 2009;29:10405-10409.

    Article  CAS  PubMed  Google Scholar 

  28. Saura CA, Chen G, Malkani S, et al. Conditional inactivation of presenilin 1 prevents amyloid accumulation and temporarily rescues contextual and spatial working memory impairments in amyloid precursor protein transgenic mice. J Neurosci 2005;25:6755-6764.

    Article  CAS  PubMed  Google Scholar 

  29. Harrison SM, Harper AJ, Hawkins J, et al. BACE1 (β-secretase) transgenic and knockout mice: identification of neurochemical deficits and behavioral changes. Mol Cell Neurosci 2003;24:646-655.

    Article  CAS  PubMed  Google Scholar 

  30. Ohno M, Sametsky EA, Younkin LH, et al. BACE1 deficiency rescues memory deficits and cholinergic dysfunction in a mouse model of Alzheimer's disease. Neuron 2004;41:27-33.

    Article  CAS  PubMed  Google Scholar 

  31. Postina R, Schroeder A, Dewachter I, et al. A disintegrin-metalloproteinase prevents amyloid plaque formation and hippocampal defects in an Alzheimer disease mouse model. J Clin Invest 2004;113:1456-1464.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  32. Gouras GK, Xu H, Jovanovic JN, et al. Generation and regulation of beta-amyloid peptide variants by neurons. J Neurochem 1998;71:1920-1925.

    Article  CAS  PubMed  Google Scholar 

  33. DeBoer SR, Dolios G, Wang R, Sisodia SS. Differential release of β-amyloid from dendrite- versus axon-targeted APP. J Neurosci 2014;34:12313-12327.

    Article  CAS  PubMed  Google Scholar 

  34. Le Gall SM, Bobé P, Reiss K, et al. ADAMs 10 and 17 represent differentially regulated components of a general shedding machinery for membrane proteins such as transforming growth factor alpha, L-selectin, and tumor necrosis factor alpha. Mol Biol Cell 2009;20:1785-1794.

    Article  PubMed Central  PubMed  Google Scholar 

  35. Musardo S, Marcello E, Gardoni F, Di Luca M. ADAM10 in synaptic physiology and pathology. Neurodegener Dis 2014;13:72-74.

    Article  CAS  PubMed  Google Scholar 

  36. Dobrowolska JA, Michener MS, Wu G, et al. CNS amyloid-β, soluble APP-α and -β kinetics during BACE inhibition. J Neurosci 2014;34:8336-8346.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  37. Aisen PS, Gauthier S, Ferris SH, et al. Tramiprosate in mild-to-moderate Alzheimer's disease—a randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch Med Sci 2011;7:102-111.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  38. Salloway S, Sperling R, Keren R, et al. A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology 2011;77:1253-1262.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  39. Adlard PA, Bica L, White AR, et al. Metal ionophore treatment restores dendritic spine density and synaptic protein levels in a mouse model of Alzheimer's disease. PLoS ONE 2011;6:e17669.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  40. Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 1999;400:173-177.

    Article  CAS  PubMed  Google Scholar 

  41. Bacskai BJ, Kajdasz ST, McLellan ME, et al. Non-Fc-mediated mechanisms are involved in clearance of amyloid-beta in vivo by immunotherapy. J Neurosci 2002;22:7873-7878.

    CAS  PubMed  Google Scholar 

  42. DeMattos RB. Brain to plasma amyloid-beta efflux: a Measure of brain amyloid burden in a mouse model of Alzheimer's disease. Science 2002;295:2264-2267.

    Article  CAS  PubMed  Google Scholar 

  43. Yamada K, Yabuki C, Seubert P, et al. Abeta immunotherapy: intracerebral sequestration of Abeta by an anti-Abeta monoclonal antibody 266 with high affinity to soluble Abeta. J Neurosci 2009;29:11393-11398.

    Article  CAS  PubMed  Google Scholar 

  44. Tampellini D, Magrané J, Takahashi RH, et al. Internalized antibodies to the Abeta domain of APP reduce neuronal Abeta and protect against synaptic alterations. J Biol Chem 2007;282:18895-18906.

    Article  CAS  PubMed  Google Scholar 

  45. Morgan D. Immunotherapy for Alzheimer’s disease. J Intern Med 2010;269:54-63.

    Article  Google Scholar 

  46. Nalivaeva NN, Belyaev ND, Kerridge C, Turner AJ. Amyloid-clearing proteins and their epigenetic regulation as a therapeutic target in Alzheimer's disease. Front Aging Neurosci 2014;6:235.

    Article  PubMed Central  PubMed  Google Scholar 

  47. Pardossi-Piquard R, Petit A, Kawarai T, et al. Presenilin-dependent transcriptional control of the Abeta-degrading enzyme neprilysin by intracellular domains of betaAPP and APLP. Neuron 2005;46:541-554.

    Article  CAS  PubMed  Google Scholar 

  48. Hama E, Saido TC. Etiology of sporadic Alzheimer's disease: somatostatin, neprilysin, and amyloid beta peptide. Med Hypotheses 2005;65:498-500.

    Article  CAS  PubMed  Google Scholar 

  49. Belyaev ND, Nalivaeva NN, Makova NZ, Turner AJ. Neprilysin gene expression requires binding of the amyloid precursor protein intracellular domain to its promoter: implications for Alzheimer disease. EMBO Rep 2009;10:94-100.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  50. Grimm MOW, Mett J, Stahlmann CP, Haupenthal VJ, Zimmer VC, Hartmann T. Neprilysin and Aβ clearance: impact of the APP intracellular domain in NEP regulation and implications in Alzheimer's disease. Front Aging Neurosci 2013;5:98.

    Article  PubMed Central  PubMed  Google Scholar 

  51. Ihara Y, Morishima-Kawashima M, Nixon R. The ubiquitin-proteasome system and the autophagic-lysosomal system in Alzheimer disease. Cold Spring Harb Perspect Med 2012;2:a006361-a006361.

    Article  PubMed Central  PubMed  Google Scholar 

  52. Gouras GK. Convergence of synapses, endosomes, and prions in the biology of neurodegenerative diseases. Int J Cell Biol 2013;2013:1-6.

    Article  Google Scholar 

  53. Lacor PN, Buniel MC, Chang L, et al. Synaptic targeting by Alzheimer's-related amyloid beta oligomers. J Neurosci 2004;24:10191-10200.

    Article  CAS  PubMed  Google Scholar 

  54. Nitsch RM, Slack BE, Wurtman RJ, Growdon JH. Release of Alzheimer amyloid precursor derivatives stimulated by activation of muscarinic acetylcholine receptors. Science 1992;258:304-307.

    Article  CAS  PubMed  Google Scholar 

  55. Kamenetz F, Tomita T, Hsieh H, et al. APP processing and synaptic function. Neuron 2003;37:925-937.

    Article  CAS  PubMed  Google Scholar 

  56. Tesseur I, Pimenova AA, Lo AC, et al. Chronic 5-HT4 receptor activation decreases Aβ production and deposition in hAPP/PS1 mice. Neurobiol Aging 2013;34:1779-1789.

    Article  CAS  PubMed  Google Scholar 

  57. Myers N, Pasquini L, Gottler J, et al. Within-patient correspondence of amyloid- and intrinsic network connectivity in Alzheimer's disease. Brain 2014;137:2052-2064.

    Article  PubMed Central  PubMed  Google Scholar 

  58. Billioti de Gage S, Moride Y, Ducruet T, et al. Benzodiazepine use and risk of Alzheimer's disease: case-control study. BMJ 2014;349:5205-5205.

    Article  Google Scholar 

  59. Shaked GM, Kummer MP, Lu DC, Galvan V, Bredesen DE, Koo EH. Abeta induces cell death by direct interaction with its cognate extracellular domain on APP (APP 597-624). FASEB J 2006;20:1254-1256.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  60. Tampellini D, Rahman N, Gallo EF, et al. Synaptic activity reduces intraneuronal Abeta, promotes APP transport to synapses, and protects against Abeta-related synaptic alterations. J Neurosci 2009;29:9704-9713.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  61. Asuni AA, Guridi M, Pankiewicz JE, Sanchez S, Sadowski MJ. Modulation of amyloid precursor protein expression reduces β-amyloid deposition in a mouse model. Ann Neurol 2014;75:684-699.

    Article  CAS  PubMed  Google Scholar 

  62. Harrison DE, Strong R, Sharp ZD, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature 2009;460:392-395.

    PubMed Central  CAS  PubMed  Google Scholar 

  63. Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molecular interplay between mammalian target of rapamycin (mTOR), amyloid-beta, and Tau: effects on cognitive impairments. J Biol Chem 2010;285:13107-13120.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  64. Spilman P, Podlutskaya N, Hart MJ, et al. Inhibition of mTOR by rapamycin abolishes cognitive deficits and reduces amyloid-beta levels in a mouse model of Alzheimer's disease. PLoS ONE 2010;5:e9979.

    Article  PubMed Central  PubMed  Google Scholar 

  65. Nixon RA, Wegiel J, Kumar A, et al. Extensive involvement of autophagy in Alzheimer disease: an immuno-electron microscopy study. J Neuropathol Exp Neurol 2005;64:113-122.

    PubMed  Google Scholar 

  66. Yu WH, Cuervo AM, Kumar A, et al. Macroautophagy—a novel β-amyloid peptide-generating pathway activated in Alzheimer's disease. J Cell Biol 2005;171:87-98.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  67. Nilsson P, Loganathan K, Sekiguchi M, et al. Aβ secretion and plaque formation depend on autophagy. Cell Rep 2013;5:61-69.

    Article  CAS  PubMed  Google Scholar 

  68. Puzzo D, Privitera L, Fa M, et al. Endogenous amyloid-β is necessary for hippocampal synaptic plasticity and memory. Ann Neurol 2011;69:819-830.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  69. Fogel H, Frere S, Segev O, et al. APP homodimers transducean amyloid-β-mediated increasein release probability at excitatory synapses. Cell Rep 2014;7:1560-1576.

    Article  CAS  PubMed  Google Scholar 

  70. Jonsson T, Atwal JK, Steinberg S, et al. A mutation in APP protects against Alzheimer's disease and age-related cognitive decline. Nature 2012;488:96-99.

    Article  CAS  PubMed  Google Scholar 

  71. Irwin DJ, Lee VMY, Trojanowski JQ. Parkinson's disease dementia: convergence of α-synuclein, tau and amyloid-β pathologies. Nat Rev Neurosci 2013;14:626-636.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  72. Irwin DJ, Abrams JY, Schonberger LB, et al. Evaluation of potential infectivity of Alzheimer and Parkinson disease proteins in recipients of cadaver-derived human growth hormone. JAMA Neurol 2013;70:462-468.

    Article  PubMed Central  PubMed  Google Scholar 

  73. Brundin P, Melki R, Kopito R. Prion-like transmission of protein aggregates in neurodegenerative diseases. Nat Rev Mol Cell Biol 2010;11:301-307.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  74. Aguzzi A, Rajendran L. The transcellular spread of cytosolic amyloids, prions, and prionoids. Neuron 2009;64:783-790.

    Article  CAS  PubMed  Google Scholar 

  75. Nussbaum JM, Seward ME, Bloom GS. Alzheimer disease: a tale of two prions. Prion 2013;7:14-19.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  76. Lu JX, Qiang W, Yau WM, Schwieters CD, Meredith SC. Molecular structure of β-amyloid fibrils in Alzheimer’s disease brain tissue. Cell 2013;154:1257-1268.

    Article  CAS  PubMed  Google Scholar 

  77. Nhan HS, Chiang K, Koo EH. The multifaceted nature of amyloid precursor protein and its proteolytic fragments: friends and foes. Acta Neuropathol 2014 Oct 7 [Epub ahead of print].

  78. Dawkins E, Small DH. Insights into the physiological function of the β-amyloid precursor protein: beyond Alzheimer's disease. J Neurochem 2014;129:756-769.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  79. Born HA, Kim J-Y, Savjani RR, et al. Genetic suppression of transgenic APP rescues hypersynchronous network activity in a mouse model of Alzeimer's disease. J Neurosci 2014;34:3826-3840.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  80. Melnikova T, Fromholt S, Kim H, et al. Reversible pathologic and cognitive phenotypes in an inducible model of Alzheimer-amyloidosis. J Neurosci 2013;33:3765-3779.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  81. O'Neill C, Kiely AP, Coakley MF, Manning S, Long-Smith CM. Insulin and IGF-1 signalling: longevity, protein homoeostasis and Alzheimer's disease. Biochem Soc Trans 2012;40:721-727.

    Article  PubMed  Google Scholar 

  82. Dumont M, Lin MT, Beal MF. Mitochondria and antioxidant targeted therapeutic strategies for Alzheimer's disease. J Alzheimers Dis 2010;20(Suppl. 2):S633–S643.

    PubMed Central  PubMed  Google Scholar 

  83. Roberson ED, Scearce-Levie K, Palop JJ, et al. Reducing endogenous tau ameliorates amyloid-induced deficits in an Alzheimer's disease mouse model. Science 2007;316:750-754.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

We are grateful for funding support from MultiPark, the Swedish Research Council, and the European Research Council.

Required Author Forms

Disclosure forms provided by the authors are available with the online version of this article.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Gunnar K. Gouras or Oskar Hansson.

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

(PDF 1224 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gouras, G.K., Olsson, T.T. & Hansson, O. β-amyloid Peptides and Amyloid Plaques in Alzheimer’s Disease. Neurotherapeutics 12, 3–11 (2015). https://doi.org/10.1007/s13311-014-0313-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13311-014-0313-y

Key Words

Navigation