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:

Intracellular amyloid-β in Alzheimer's disease

Key Points

  • This Review examines the current evidence supporting a role for intracellular amyloid-β (Aβ) in the pathogenesis of Alzheimer's disease. Intracellular Aβ accumulation appears to be an early event in the disease, and may be a source for extracellular deposits as the disease progresses.

  • Aβ can be produced within cells or taken up from extracellular sources through receptor binding and subsequent internalization. This leads to accumulation within endosomes, multivesicular bodies, lysosomes, mitochondria, the endoplasmic reticulum and trans-Golgi network, and the cytosol.

  • Oligomeric Aβ assembly states have been implicated as the critical pathological species. Aβ oligomerization may commence within cells, possibly through membrane interactions, and these Aβ oligomers may act intracellularly to cause neuronal dysfunction, or pass into the extracellular space.

  • Intracellular Aβ in transgenic mouse models of Alzheimer's disease is associated with cognitive deficits, tau phosphorylation, neuronal loss and synaptic dysfunction.

  • Various treatments and paradigms affect intracellular Aβ, including aging, docosahexaenoic acid, insulin, learning, stress and the Alzheimer's disease risk factor allele APOE*ε4.

Abstract

The primal role that the amyloid-β (Aβ) peptide has in the development of Alzheimer's disease is now almost universally accepted. It is also well recognized that Aβ exists in multiple assembly states, which have different physiological or pathophysiological effects. Although the classical view is that Aβ is deposited extracellularly, emerging evidence from transgenic mice and human patients indicates that this peptide can also accumulate intraneuronally, which may contribute to disease progression.

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: APP proteolysis.
Figure 2: Sites of cellular Aβ production.
Figure 3: Pathological effects of intraneuronal Aβ.

Similar content being viewed by others

References

  1. Alzheimer A. Über eine eigenartige Erkrankung der Hirnrinde. Allgemeine Zeitschrift für Psychiatrie und Psychisch-Gerichtliche Medizine 64, 146–148 (1907).

    Google Scholar 

  2. 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 

  3. 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 

  4. Goedert, M., Wischik, C. M., Crowther, R. A., Walker, J. E. & Klug, A. Cloning and sequencing of the cDNA encoding a core protein of the paired helical filament of Alzheimer disease: identification as the microtubule-associated protein tau. Proc. Natl Acad. Sci. USA 85, 4051–4055 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Ihara, Y., Nukina, N., Miura, R. & Ogawara, M. Phosphorylated tau protein is integrated into paired helical filaments in Alzheimer's disease. J. Biochem. (Tokyo) 99, 1807–1810 (1986).

    Article  CAS  Google Scholar 

  7. Kosik, K. S., Joachim, C. L. & Selkoe, D. J. Microtubule-associated protein tau (τ) 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 

  8. Rozemuller, J. M., Eikelenboom, P. & Stam, F. C. Role of microglia in plaque formation in senile dementia of the Alzheimer type. An immunohistochemical study. Virchows Arch. B Cell Pathol. 51, 247–254 (1986).

    Article  CAS  Google Scholar 

  9. Wyss-Coray, T. Inflammation in Alzheimer disease: driving force, bystander or beneficial response? Nature Med. 12, 1005–1015 (2006).

    CAS  PubMed  Google Scholar 

  10. Markesbery, W. R. Oxidative stress hypothesis in Alzheimer's disease. Free Radic. Biol. Med. 23, 134–147 (1997).

    Article  CAS  PubMed  Google Scholar 

  11. McGeer, P. L., Rogers, J. & McGeer, E. G. Inflammation, anti-inflammatory agents and Alzheimer disease: the last 12 years. J. Alzheimers Dis. 9, 271–276 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Allinson, T. M., Parkin, E. T., Turner, A. J. & Hooper, N. M. ADAMs family members as amyloid precursor protein α-secretases. J. Neurosci. Res. 74, 342–352 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Vassar, R. et al. β-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286, 735–741 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Hussain, I. et al. Identification of a novel aspartic protease (Asp 2) as β-secretase. Mol. Cell. Neurosci. 14, 419–427 (1999).

    Article  CAS  PubMed  Google Scholar 

  15. Sinha, S. et al. Purification and cloning of amyloid precursor protein β-secretase from human brain. Nature 402, 537–540 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. 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 

  17. Steiner, H. et al. PEN-2 is an integral component of the γ-secretase complex required for coordinated expression of presenilin and nicastrin. J. Biol. Chem. 277, 39062–39065 (2002).

    Article  CAS  PubMed  Google Scholar 

  18. Francis, R. et al. aph-1 and pen-2 are required for Notch pathway signaling, γ-secretase cleavage of βAPP, and presenilin protein accumulation. Dev. Cell 3, 85–97 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Levitan, D. et al. PS1 N- and C-terminal fragments form a complex that functions in APP processing and Notch signaling. Proc. Natl Acad. Sci. USA 98, 12186–12190 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yu, G. et al. Nicastrin modulates presenilin-mediated notch/glp-1 signal transduction and βAPP processing. Nature 407, 48–54 (2000).

    Article  CAS  PubMed  Google Scholar 

  21. Kojro, E. & Fahrenholz, F. The non-amyloidogenic pathway: structure and function of α-secretases. Subcell. Biochem. 38, 105–127 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Haass, C., Hung, A. Y., Schlossmacher, M. G., Teplow, D. B. & Selkoe, D. J. β-Amyloid peptide and a 3-kDa fragment are derived by distinct cellular mechanisms. J. Biol. Chem. 268, 3021–3024 (1993).

    Article  CAS  PubMed  Google Scholar 

  23. 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 

  24. Younkin, S. G. The role of Aβ 42 in Alzheimer's disease. J. Physiol. Paris 92, 289–292 (1998).

    Article  CAS  PubMed  Google Scholar 

  25. St George-Hyslop, P. H. & Petit, A. Molecular biology and genetics of Alzheimer's disease. C. R. Biol. 328, 119–130 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Haass, C. et al. The Swedish mutation causes early-onset Alzheimer's disease by β-secretase cleavage within the secretory pathway. Nature Med. 1, 1291–1296 (1995).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  28. Guo, Q. et al. Increased vulnerability of hippocampal neurons to excitotoxic necrosis in presenilin-1 mutant knock-in mice. Nature Med. 5, 101–106 (1999).

    Article  CAS  PubMed  Google Scholar 

  29. Jankowsky, J. L. et al. Mutant presenilins specifically elevate the levels of the 42 residue β-amyloid peptide in vivo: evidence for augmentation of a 42-specific γ secretase. Hum. Mol. Genet. 13, 159–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. 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 

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

    Article  PubMed  Google Scholar 

  32. Gyure, K. A., Durham, R., Stewart, W. F., Smialek, J. E. & Troncoso, J. C. Intraneuronal Aβ-amyloid precedes development of amyloid plaques in Down syndrome. Arch. Pathol. Lab. Med. 125, 489–492 (2001).

    Article  CAS  PubMed  Google Scholar 

  33. Mori, C. et al. Intraneuronal Aβ42 accumulation in Down syndrome brain. Amyloid 9, 88–102 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Grundke-Iqbal, I. et al. Amyloid protein and neurofibrillary tangles coexist in the same neuron in Alzheimer disease. Proc. Natl Acad. Sci. USA 86, 2853–2857 (1989).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Blurton-Jones, M. & Laferla, F. M. Pathways by which Aβ facilitates tau pathology. Curr. Alzheimer Res. 3, 437–448 (2006).

    Article  CAS  PubMed  Google Scholar 

  36. Gouras, G. K. et al. Intraneuronal Aβ42 accumulation in human brain. Am. J. Pathol. 156, 15–20 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Takahashi, R. H. et al. Intraneuronal Alzheimer Aβ42 accumulates in multivesicular bodies and is associated with synaptic pathology. Am. J. Pathol. 161, 869–1879 (2002). Using electron microscope analysis, this paper shows that in AD brains, intraneuronal Aβ accumulates within the multivesicular bodies.

    Article  Google Scholar 

  38. D'Andrea, M. R. et al. The use of formic acid to embellish amyloid plaque detection in Alzheimer's disease tissues misguides key observations. Neurosci. Lett. 342, 114–118 (2003).

    Article  CAS  PubMed  Google Scholar 

  39. Ohyagi, Y. et al. Intraneuronal amyloid β42 enhanced by heating but counteracted by formic acid. J. Neurosci. Methods 159, 134–138 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Chui, D. H. et al. Transgenic mice with Alzheimer presenilin 1 mutations show accelerated neurodegeneration without amyloid plaque formation. Nature Med. 5, 560–564 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Knobloch, M., Konietzko, U., Krebs, D. C. & Nitsch, R. M. Intracellular Aβ and cognitive deficits precede β-amyloid deposition in transgenic arcAβ mice. Neurobiol. Aging, 31 July 2006 (doi:10.1016/j.neurobiolaging.2006.06.019). Provides evidence linking intraneuronal Aβ accumulation to cognitive deficits in transgenic mice.

    Article  CAS  PubMed  Google Scholar 

  42. Kuo, Y. M. et al. The evolution of Aβ peptide burden in the APP23 transgenic mice: implications for Aβ deposition in Alzheimer disease. Mol. Med. 7, 609–618 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Li, Q. X. et al. Intracellular accumulation of detergent-soluble amyloidogenic Aβ fragment of Alzheimer's disease precursor protein in the hippocampus of aged transgenic mice. J. Neurochem. 72, 2479–2487 (1999).

    Article  CAS  PubMed  Google Scholar 

  44. Lord, A. et al. The Arctic Alzheimer mutation facilitates early intraneuronal Aβ aggregation and senile plaque formation in transgenic mice. Neurobiol. Aging 27, 67–77 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Oakley, H. et al. Intraneuronal β-amyloid aggregates, neurodegeneration, and neuron loss in transgenic mice with five familial Alzheimer's disease mutations: potential factors in amyloid plaque formation. J. Neurosci. 26, 10129–10140 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Oddo, S. et al. Triple-transgenic model of Alzheimer's disease with plaques and tangles: intracellular Aβ and synaptic dysfunction. Neuron 39, 409–421 (2003). Provides the earliest in vivo evidence that links intraneuronal Aβ to synaptic dysfunction, showing that intraneuronal Aβ accumulation leads to a profound LTP deficit in 3xTg-AD mice.

    Article  CAS  PubMed  Google Scholar 

  47. Wirths, O. et al. Intraneuronal Aβ accumulation precedes plaque formation in β-amyloid precursor protein and presenilin-1 double-transgenic mice. Neurosci. Lett. 306, 116–120 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Oddo, S., Caccamo, A., Smith, I. F., Green, K. N. & LaFerla, F. M. A dynamic relationship between intracellular and extracellular pools of Aβ. Am. J. Pathol. 168, 184–194 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Wegiel, J. et al. Intraneuronal Aβ immunoreactivity is not a predictor of brain amyloidosis-β or neurofibrillary degeneration. Acta Neuropathol. (Berl) 113, 389–402 (2007).

    Article  CAS  Google Scholar 

  50. Kinoshita, A. et al. Demonstration by FRET of BACE interaction with the amyloid precursor protein at the cell surface and in early endosomes. J. Cell Sci. 116, 3339–3346 (2003).

    Article  CAS  PubMed  Google Scholar 

  51. Breen, K. C., Bruce, M. & Anderton, B. H. β amyloid precursor protein mediates neuronal cell–cell and cell–surface adhesion. J. Neurosci. Res. 28, 90–100 (1991).

    Article  CAS  PubMed  Google Scholar 

  52. Sabo, S. L., Ikin, A. F., Buxbaum, J. D. & Greengard, P. The Alzheimer amyloid precursor protein (APP) and FE65, an APP-binding protein, regulate cell movement. J. Cell Biol. 153, 1403–1414 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Xu, H., Greengard, P. & Gandy, S. Regulated formation of Golgi secretory vesicles containing Alzheimer β-amyloid precursor protein. J. Biol. Chem. 270, 23243–23245 (1995).

    Article  CAS  PubMed  Google Scholar 

  54. Mizuguchi, M., Ikeda, K. & Kim, S. U. Differential distribution of cellular forms of β-amyloid precursor protein in murine glial cell cultures. Brain Res. 584, 219–225 (1992).

    Article  CAS  PubMed  Google Scholar 

  55. Wertkin, A. M. et al. Human neurons derived from a teratocarcinoma cell line express solely the 695-amino acid amyloid precursor protein and produce intracellular β-amyloid or A4 peptides. Proc. Natl Acad. Sci. USA 90, 9513–9517 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Martin, B. L. et al. Intracellular accumulation of β-amyloid in cells expressing the Swedish mutant amyloid precursor protein. J. Biol. Chem. 270, 26727–26730 (1995).

    Article  CAS  PubMed  Google Scholar 

  57. Rogaeva, E. et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nature Genet. 39, 168–177 (2007).

    Article  CAS  PubMed  Google Scholar 

  58. Golde, T. E., Estus, S., Younkin, L. H., Selkoe, D. J. & Younkin, S. G. Processing of the amyloid protein precursor to potentially amyloidogenic derivatives. Science 255, 728–730 (1992).

    Article  CAS  PubMed  Google Scholar 

  59. Koo, E. H. & Squazzo, S. L. Evidence that production and release of amyloid β-protein involves the endocytic pathway. J. Biol. Chem. 269, 17386–17389 (1994).

    Article  CAS  PubMed  Google Scholar 

  60. Perez, R. G. et al. Mutagenesis identifies new signals for β-amyloid precursor protein endocytosis, turnover, and the generation of secreted fragments, including Aβ42. J. Biol. Chem. 274, 18851–18856 (1999).

    Article  CAS  PubMed  Google Scholar 

  61. Cam, J. A. et al. The low density lipoprotein receptor-related protein 1B retains β-amyloid precursor protein at the cell surface and reduces amyloid-β peptide production. J. Biol. Chem. 279, 29639–29646 (2004).

    Article  CAS  PubMed  Google Scholar 

  62. 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). Provides strong evidence that Aβ in neurons is generated by the secretory pathway.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Cook, D. G. et al. Alzheimer's A β(1–42) is generated in the endoplasmic reticulum/intermediate compartment of NT2N cells. Nature Med. 3, 1021–1023 (1997).

    Article  CAS  PubMed  Google Scholar 

  64. Lee, S. J. et al. A detergent-insoluble membrane compartment contains A β in vivo. Nature Med. 4, 730–734 (1998).

    Article  CAS  PubMed  Google Scholar 

  65. Skovronsky, D. M., Doms, R. W. & Lee, V. M. Detection of a novel intraneuronal pool of insoluble amyloid β protein that accumulates with time in culture. J. Cell Biol. 141, 1031–1039 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Wild-Bode, C. et al. Intracellular generation and accumulation of amyloid beta-peptide terminating at amino acid 42. J. Biol. Chem. 272, 16085–16088 (1997).

    Article  CAS  PubMed  Google Scholar 

  67. Hartmann, T. et al. Distinct sites of intracellular production for Alzheimer's disease A β40/42 amyloid peptides. Nature Med. 3, 1016–10120 (1997). Shows that both Aβ 40 and Aβ 42 are generated intracellularly but at different sites.

    Article  CAS  PubMed  Google Scholar 

  68. Bu, G., Cam, J. & Zerbinatti, C. LRP in amyloid-β production and metabolism. Ann. N. Y. Acad. Sci. 1086, 35–53 (2006).

    Article  CAS  PubMed  Google Scholar 

  69. Deane, R. et al. RAGE mediates amyloid-β peptide transport across the blood–brain barrier and accumulation in brain. Nature Med. 9, 907–913 (2003).

    Article  CAS  PubMed  Google Scholar 

  70. Nagele, R. G., D'Andrea, M. R., Anderson, W. J. & Wang, H. Y. Intracellular accumulation of β-amyloid1–42 in neurons is facilitated by the α7 nicotinic acetylcholine receptor in Alzheimer's disease. Neuroscience 110, 199–211 (2002). Shows that Aβ binds to α7nAChR and suggests that it is subsequently internalized, thus providing a mechanism for intraneuronal Aβ accumulation.

    Article  CAS  PubMed  Google Scholar 

  71. Yazawa, H. et al. β amyloid peptide (Aβ42) is internalized via the G-protein-coupled receptor FPRL1 and forms fibrillar aggregates in macrophages. FASEB J. 15, 2454–2462 (2001).

    Article  CAS  PubMed  Google Scholar 

  72. Clifford, P. M. et al. Aβ peptides can enter the brain through a defective blood–brain barrier and bind selectively to neurons. Brain Res. 1142, 223–236 (2007).

    Article  CAS  PubMed  Google Scholar 

  73. Wang, H. Y. et al. β-Amyloid1–42 binds to α7 nicotinic acetylcholine receptor with high affinity. Implications for Alzheimer's disease pathology. J. Biol. Chem. 275, 5626–5632 (2000).

    Article  CAS  PubMed  Google Scholar 

  74. Oddo, S. et al. Chronic nicotine administration exacerbates tau pathology in a transgenic model of Alzheimer's disease. Proc. Natl Acad. Sci. USA 102, 3046–3051 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Zerbinatti, C. V. et al. Apolipoprotein E and low density lipoprotein receptor-related protein facilitate intraneuronal Aβ42 accumulation in amyloid model mice. J. Biol. Chem. 281, 36180–36186 (2006).

    Article  CAS  PubMed  Google Scholar 

  76. Yan, S. D. et al. RAGE and amyloid-β peptide neurotoxicity in Alzheimer's disease. Nature 382, 685–691 (1996).

    Article  CAS  PubMed  Google Scholar 

  77. Sasaki, N. et al. Immunohistochemical distribution of the receptor for advanced glycation end products in neurons and astrocytes in Alzheimer's disease. Brain Res. 888, 256–262 (2001).

    Article  CAS  PubMed  Google Scholar 

  78. Du Yan, S. et al. Amyloid-β peptide-receptor for advanced glycation endproduct interaction elicits neuronal expression of macrophage-colony stimulating factor: a proinflammatory pathway in Alzheimer disease. Proc. Natl Acad. Sci. USA 94, 5296–5301 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Iribarren, P., Zhou, Y., Hu, J., Le, Y. & Wang, J. M. Role of formyl peptide receptor-like 1 (FPRL1/FPR2) in mononuclear phagocyte responses in Alzheimer disease. Immunol. Res. 31, 165–176 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Snyder, E. M. et al. Regulation of NMDA receptor trafficking by amyloid-β. Nature Neurosci. 8, 1051–1058 (2005).

    Article  CAS  PubMed  Google Scholar 

  81. Bi, X., Gall, C. M., Zhou, J. & Lynch, G. Uptake and pathogenic effects of amyloid β peptide 1–42 are enhanced by integrin antagonists and blocked by NMDA receptor antagonists. Neuroscience 112, 827–840 (2002).

    Article  CAS  PubMed  Google Scholar 

  82. Reisberg, B. et al. Memantine in moderate-to-severe Alzheimer's disease. N. Engl. J. Med. 348, 1333–1341 (2003).

    Article  CAS  PubMed  Google Scholar 

  83. Minkeviciene, R., Banerjee, P. & Tanila, H. Memantine improves spatial learning in a transgenic mouse model of Alzheimer's disease. J. Pharmacol. Exp. Ther. 311, 677–682 (2004).

    Article  CAS  PubMed  Google Scholar 

  84. 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 

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

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  87. 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). Using cells from the human brain, this paper shows that Aβ oligomerization starts intraneuronally.

    Article  CAS  PubMed  Google Scholar 

  88. Oddo, S. et al. Temporal profile of amyloid-β (Aβ) oligomerization in an in vivo model of Alzheimer disease. A link between Aβ and tau pathology. J. Biol. Chem. 281, 1599–1604 (2006).

    Article  CAS  PubMed  Google Scholar 

  89. Takahashi, R. H. et al. Oligomerization of Alzheimer's β-amyloid within processes and synapses of cultured neurons and brain. J. Neurosci. 24, 3592–3599 (2004). Shows that Aβ oligomers accumulate intraneuronally in AD brain.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Kim, S. I., Yi, J. S. & Ko, Y. G. Amyloid β oligomerization is induced by brain lipid rafts. J. Cell Biochem. 99, 878–889 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Waschuk, S. A., Elton, E. A., Darabie, A. A., Fraser, P. E. & McLaurin, J. A. Cellular membrane composition defines Aβ-lipid interactions. J. Biol. Chem. 276, 33561–33568 (2001).

    Article  CAS  PubMed  Google Scholar 

  93. Matsuzaki, K. et al. Inhibitors of amyloid β-protein aggregation mediated by GM1-containing raft-like membranes. Biochim. Biophys. Acta 1768, 122–130 (2007).

    Article  CAS  PubMed  Google Scholar 

  94. Wakabayashi, M., Okada, T., Kozutsumi, Y. & Matsuzaki, K. GM1 ganglioside-mediated accumulation of amyloid β-protein on cell membranes. Biochem. Biophys. Res. Commun. 328, 1019–1023 (2005).

    Article  CAS  PubMed  Google Scholar 

  95. Yamamoto, N. et al. Accelerated Aβ aggregation in the presence of GM1-ganglioside-accumulated synaptosomes of aged apoE4-knock-in mouse brain. FEBS Lett. 569, 135–139 (2004).

    Article  CAS  PubMed  Google Scholar 

  96. D'Andrea, M. R., Nagele, R. G., Wang, H. Y., Peterson, P. A. & Lee, D. H. Evidence that neurones accumulating amyloid can undergo lysis to form amyloid plaques in Alzheimer's disease. Histopathology 38, 120–134 (2001). Provides evidence suggesting that in the brains of patients with AD, intracellular Aβ accumulation may lead to neuronal death, and that after being released in the extracellular compartment, it can contribute to the formation of plaques.

    Article  CAS  PubMed  Google Scholar 

  97. Bahr, B. A. et al. Amyloid β protein is internalized selectively by hippocampal field CA1 and causes neurons to accumulate amyloidogenic carboxyterminal fragments of the amyloid precursor protein. J. Comp. Neurol. 397, 139–147 (1998).

    Article  CAS  PubMed  Google Scholar 

  98. Schenk, D. et al. Immunization with amyloid-β attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400, 173–177 (1999).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  100. Oddo, S., Billings, L., Kesslak, J. P., Cribbs, D. H. & LaFerla, F. M. Aβ immunotherapy leads to clearance of early, but not late, hyperphosphorylated tau aggregates via the proteasome. Neuron 43, 321–332 (2004). Shows that Aβ immunotherapy clears intraneuronal Aβ accumulation, which leads to the removal of tau deposits, thus linking intracellular Aβ and tau pathology.

    Article  CAS  PubMed  Google Scholar 

  101. Gouras, G. K., Almeida, C. G. & Takahashi, R. H. Intraneuronal Aβ accumulation and origin of plaques in Alzheimer's disease. Neurobiol. Aging 26, 1235–1244 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Tseng, B. P., Kitazawa, M. & LaFerla, F. M. Amyloid β-peptide: the inside story. Curr. Alzheimer Res. 1, 231–239 (2004).

    Article  CAS  PubMed  Google Scholar 

  103. Langui, D. et al. Subcellular topography of neuronal Aβ peptide in APPxPS1 transgenic mice. Am. J. Pathol. 165, 1465–1477 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Almeida, C. G., Takahashi, R. H. & Gouras, G. K. β-amyloid accumulation impairs multivesicular body sorting by inhibiting the ubiquitin–proteasome system. J. Neurosci. 26, 4277–4288 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Gregori, L., Fuchs, C., Figueiredo-Pereira, M. E., Van Nostrand, W. E. & Goldgaber, D. Amyloid β-protein inhibits ubiquitin-dependent protein degradation in vitro. J. Biol. Chem. 270, 19702–19708 (1995).

    Article  CAS  PubMed  Google Scholar 

  106. Oh, S. et al. Amyloid peptide attenuates the proteasome activity in neuronal cells. Mech. Ageing Dev. 126, 1292–1299 (2005).

    Article  CAS  PubMed  Google Scholar 

  107. Tseng, B. P., Green, K. N., Chan, J., Blurton-Jones, M. & LaFerla, F. Aβ inhibits the proteasome and enhances amyloid and tau accumulation. Neurobiol. Aging (in the press).

  108. Manczak, M. et al. Mitochondria are a direct site of Aβ accumulation in Alzheimer's disease neurons: implications for free radical generation and oxidative damage in disease progression. Hum. Mol. Genet. 15, 1437–1449 (2006).

    Article  CAS  PubMed  Google Scholar 

  109. Hansson, C. A. et al. Nicastrin, presenilin, APH-1, and PEN-2 form active γ-secretase complexes in mitochondria. J. Biol. Chem. 279, 51654–51660 (2004).

    Article  CAS  PubMed  Google Scholar 

  110. Caspersen, C. et al. Mitochondrial Aβ: a potential focal point for neuronal metabolic dysfunction in Alzheimer's disease. FASEB J. 19, 2040–2041 (2005).

    Article  CAS  PubMed  Google Scholar 

  111. Keil, U. et al. Mitochondrial dysfunction induced by disease relevant AβPP and tau protein mutations. J. Alzheimers Dis. 9, 139–146 (2006).

    Article  PubMed  Google Scholar 

  112. Billings, L. M., Oddo, S., Green, K. N., McGaugh, J. L. & LaFerla, F. M. Intraneuronal Aβ causes the onset of early Alzheimer's disease-related cognitive deficits in transgenic mice. Neuron 45, 675–688 (2005). Shows that intraneuronal Aβ accumulation is responsible for the onset of cognitive decline in the 3xTg-AD.

    Article  CAS  PubMed  Google Scholar 

  113. Morris, R. G., Anderson, E., Lynch, G. S. & Baudry, M. Selective impairment of learning and blockade of long-term potentiation by an N-methyl-D-aspartate receptor antagonist, AP5. Nature 319, 774–776 (1986).

    Article  CAS  PubMed  Google Scholar 

  114. 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 

  115. Green, K. N. et al. Dietary docosahexaenoic acid and docosapentaenoic acid ameliorate amyloid-β and tau pathology via a mechanism involving presenilin 1 levels. J. Neurosci. 27, 4385–4395 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Hashimoto, M. et al. Chronic administration of docosahexaenoic acid ameliorates the impairment of spatial cognition learning ability in amyloid β-infused rats. J. Nutr. 135, 549–555 (2005).

    Article  CAS  PubMed  Google Scholar 

  117. Lim, G. P. et al. A diet enriched with the omega-3 fatty acid docosahexaenoic acid reduces amyloid burden in an aged Alzheimer mouse model. J. Neurosci. 25, 3032–3040 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Gasparini, L. et al. Stimulation of β-amyloid precursor protein trafficking by insulin reduces intraneuronal β-amyloid and requires mitogen-activated protein kinase signaling. J. Neurosci. 21, 2561–2570 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Billings, L. M., Green, K. N., McGaugh, J. L. & LaFerla, F. M. Learning decreases Aβ*56 and tau pathology and ameliorates behavioral decline in 3xTg-AD mice. J. Neurosci. 27, 751–761 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Green, K. N., Billings, L. M., Roozendaal, B., McGaugh, J. L. & LaFerla, F. M. Glucocorticoids increase amyloid-β and tau pathology in a mouse model of Alzheimer's disease. J. Neurosci. 26, 9047–9056 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Sparks, D. L. et al. Induction of Alzheimer-like β-amyloid immunoreactivity in the brains of rabbits with dietary cholesterol. Exp. Neurol. 126, 88–94 (1994).

    Article  CAS  PubMed  Google Scholar 

  122. Misonou, H., Morishima-Kawashima, M. & Ihara, Y. Oxidative stress induces intracellular accumulation of amyloid β-protein (Aβ) in human neuroblastoma cells. Biochemistry 39, 6951–6959 (2000).

    Article  CAS  PubMed  Google Scholar 

  123. Hasegawa, T. et al. Homocysteic acid induces intraneuronal accumulation of neurotoxic Aβ42: implications for the pathogenesis of Alzheimer's disease. J. Neurosci. Res. 80, 869–876 (2005).

    Article  CAS  PubMed  Google Scholar 

  124. D'Andrea, M. R., Nagele, R. G., Wang, H. Y. & Lee, D. H. Consistent immunohistochemical detection of intracellular β-amyloid42 in pyramidal neurons of Alzheimer's disease entorhinal cortex. Neurosci. Lett. 333, 163–166 (2002).

    Article  CAS  PubMed  Google Scholar 

  125. D'Andrea, M. R. et al. Lipofuscin and Aβ42 exhibit distinct distribution patterns in normal and Alzheimer's disease brains. Neurosci. Lett. 323, 45–49 (2002).

    Article  CAS  PubMed  Google Scholar 

  126. LaFerla, F. M., Troncoso, J. C., Strickland, D. K., Kawas, C. H. & Jay, G. Neuronal cell death in Alzheimer's disease correlates with apoE uptake and intracellular Aβ stabilization. J. Clin. Invest. 100, 310–320 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  127. Mochizuki, A., Tamaoka, A., Shimohata, A., Komatsuzaki, Y. & Shoji, S. Aβ42-positive non-pyramidal neurons around amyloid plaques in Alzheimer's disease. Lancet 355, 42–43 (2000).

    Article  CAS  PubMed  Google Scholar 

  128. Mendell, J. R., Sahenk, Z., Gales, T. & Paul, L. Amyloid filaments in inclusion body myositis. Novel findings provide insight into nature of filaments. Arch. Neurol. 48, 1229–1234 (1991).

    Article  CAS  PubMed  Google Scholar 

  129. Sarkozi, E., Askanas, V., Johnson, S. A., Engel, W. K. & Alvarez, R. B. β-Amyloid precursor protein mRNA is increased in inclusion-body myositis muscle. Neuroreport 4, 815–818 (1993).

    Article  CAS  PubMed  Google Scholar 

  130. Askanas, V., Engel, W. K. & Alvarez, R. B. Enhanced detection of congo-red-positive amyloid deposits in muscle fibers of inclusion body myositis and brain of Alzheimer's disease using fluorescence technique. Neurology 43, 1265–1267 (1993).

    Article  CAS  PubMed  Google Scholar 

  131. Kimura, N. et al. Age-related changes of intracellular Aβ in cynomolgus monkey brains. Neuropathol. Appl. Neurobiol. 31, 170–180 (2005).

    Article  CAS  PubMed  Google Scholar 

  132. Martin, L. J., Pardo, C. A., Cork, L. C. & Price, D. L. Synaptic pathology and glial responses to neuronal injury precede the formation of senile plaques and amyloid deposits in the aging cerebral cortex. Am. J. Pathol. 145, 1358–1381 (1994).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Cummings, B. J., Su, J. H., Cotman, C. W., White, R. & Russell, M. J. β-amyloid accumulation in aged canine brain: a model of early plaque formation in Alzheimer's disease. Neurobiol. Aging 14, 547–560 (1993).

    Article  CAS  PubMed  Google Scholar 

  134. Torp, R. et al. Ultrastructural evidence of fibrillar β-amyloid associated with neuronal membranes in behaviorally characterized aged dog brains. Neuroscience 96, 495–506 (2000).

    Article  CAS  PubMed  Google Scholar 

  135. Cruz, J. C. et al. p25/cyclin-dependent kinase 5 induces production and intraneuronal accumulation of amyloid β in vivo. J. Neurosci. 26, 10536–10541 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  136. Casas, C. et al. Massive CA1/2 neuronal loss with intraneuronal and N-terminal truncated Aβ42 accumulation in a novel Alzheimer transgenic model. Am. J. Pathol. 165, 1289–1300 (2004). Provides in vivo evidence that intraneuronal Aβ accumulation is toxic and leads to cell death.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  137. Van Broeck, B. et al. Intraneuronal amyloid β and reduced brain volume in a novel APP T714I mouse model for Alzheimer's disease. Neurobiol. Aging 16 Nov 2006 (doi:10.1016/j.neurobiolaging.2006.10.016).

    Article  CAS  PubMed  Google Scholar 

  138. Shie, F. S., LeBoeuf, R. C. & Jin, L. W. Early intraneuronal Aβ deposition in the hippocampus of APP transgenic mice. Neuroreport 14, 123–129 (2003).

    Article  CAS  PubMed  Google Scholar 

  139. Magrane, J. et al. Intraneuronal β-amyloid expression downregulates the Akt survival pathway and blunts the stress response. J. Neurosci. 25, 10960–10969 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Suo, Z. et al. GRK5 deficiency leads to early Alzheimer-like pathology and working memory impairment. Neurobiol. Aging 2 October 2006 (doi: 10.1016/j.neurobiolaging.2006.08.013).

    Article  CAS  PubMed  Google Scholar 

  141. Sugarman, M. C. et al. Inclusion body myositis-like phenotype induced by transgenic overexpression of βAPP in skeletal muscle. Proc. Natl Acad. Sci. USA 99, 6334–6339 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Kitazawa, M., Green, K. N., Caccamo, A. & LaFerla, F. M. Genetically augmenting Aβ42 levels in skeletal muscle exacerbates inclusion body myositis-like pathology and motor deficits in transgenic mice. Am. J. Pathol. 168, 1986–1997 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Moussa, C. E. et al. Transgenic expression of β-APP in fast-twitch skeletal muscle leads to calcium dyshomeostasis and IBM-like pathology. FASEB J. 20, 2165–2167 (2006).

    Article  CAS  PubMed  Google Scholar 

  144. Echeverria, V. et al. Altered mitogen-activated protein kinase signaling, tau hyperphosphorylation and mild spatial learning dysfunction in transgenic rats expressing the β-amyloid peptide intracellularly in hippocampal and cortical neurons. Neuroscience 129, 583–592 (2004).

    Article  CAS  PubMed  Google Scholar 

  145. Tawil, R. & Griggs, R. C. Inclusion body myositis. Curr. Opin. Rheumatol. 14, 653–657 (2002).

    Article  CAS  PubMed  Google Scholar 

  146. Askanas, V. & Engel, W. K. Inclusion-body myositis: a myodegenerative conformational disorder associated with Aβ, protein misfolding, and proteasome inhibition. Neurology 66, S39–S48 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank M. Blurton-Jones and A. Parachikova for critically reading the manuscript and for assistance with the figures. This work was supported in part by grants from the Alzheimer's Association and the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Frank M. LaFerla.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

DATABASES

OMIM

Alzheimer's disease

Down's syndrome

Inclusion body myositis

FURTHER INFORMATION

Author's homepage

Glossary

Declarative memory

Aspect of human memory that stores facts and experiences. It can be further subdivided into episodic memory and semantic memory.

Pathognomic

A lesion or sign, the occurrence of which provides evidence that a particular disease is present.

Multivesicular body

(MVB). MVBs have a role in transporting cargo to the lysosome system for degradation, and throughout the neuron for signalling purposes, and are important in regulating and degrading transmembrane proteins.

Neoepitope

Antibody recognition site of a terminal sequence in which the same sequence of amino acids are not recognized when located internally as part of the intact protein.

Mild cognitive impairment

(MCI). A transition stage between the cognitive changes of normal aging and Alzheimer's disease.

Retromer recycling endosomes

The retromer complex has been shown to be important in recycling transmembrane receptors from endosomes to the trans-Golgi network.

Holoprotein

The full-length, native polypeptide, before proteolytic cleavage events that might occur during maturation.

Minigene

A fragment of a genomic sequence that includes the exons and introns and is cloned into a eukaryotic expression vector.

Lipid raft

A specialized membrane domain that is enriched in cholesterol.

Nucleation seed

A molecule that facilitates the assembly of a polymeric structure.

Synaptosome

Isolated synapse of a neuron obtained via homogenization of nerve tissue.

Ubiquitin–proteasome system

Proteasomes are large protein complexes that degrade damaged or superfluous proteins that are tagged by a small protein called ubiquitin.

Rights and permissions

Reprints and permissions

About this article

Cite this article

LaFerla, F., Green, K. & Oddo, S. Intracellular amyloid-β in Alzheimer's disease. Nat Rev Neurosci 8, 499–509 (2007). https://doi.org/10.1038/nrn2168

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrn2168

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