Skip to main content
The American Journal of Pathology logoLink to The American Journal of Pathology
. 2004 Nov;165(5):1461–1464. doi: 10.1016/S0002-9440(10)63404-9

Staying Connected

Synapses in Alzheimer Disease

Hyoung-gon Lee 1, Paula I Moreira 1, Xiongwei Zhu 1, Mark A Smith 1, George Perry 1
PMCID: PMC1618677  PMID: 15509517

Careful dissection is continuing to reveal the complexity and importance of synapse alterations in Alzheimer disease (AD). Synaptic alterations are the best biological correlate to the extent of cognitive loss1,2 and have become the gold standard of meaningful biomarkers of AD. Not neurofibrillary tangles (NFT), senile plaques, or even neuronal loss show such strong statistical correlation with dementia.2–4

Connections are the hallmark of neurons and without functional synapses, neurons are in “solitary.” Recent findings showing that amyloid-β (Aβ) affects and accumulates in synapses5 and that amyloid-β protein precursor (AβPP) over-expressing mice show synaptic alterations6,7 have provided additional evidence that synapses are a critical readout of the biology of AD.

In this issue, Gylys and co-workers8 provide an elegant and precise dissection of synaptic alterations in AD using synaptosomal preparations. The results are both consistent with the synaptic alteration hypothesis proposed by Terry and colleagues2–4 and refine it. In previous studies synapses have been quantitated by synaptophysin immunoreactivity,1,9 a protein belonging to a subclass of synaptic vesicles. However, when synapses were quantitated by precise ultrastructural morphometric/stereological approaches, reduction in synapses was much less striking than the reduction in synaptophysin.10 Furthermore, synapse alterations included major remodeling of morphology11 as noted in some of the earliest studies of AD.12 These established findings lead one to consider whether the alterations in synapses in AD are limited to global reduction in vesicles rather than synapse changes. Gylys and collaborators8 used a broad range of pre- and postsynaptic markers to precisely analyze synaptosomes by flow cytometry. They found that the synaptosomes in AD show no reduction in synaptophysin and only a slight reduction in the overall number of synaptosomes. To account for the undoubted major reduction in synaptophysin while synapse number and synaptic vesicle number are only slightly reduced, brings to mind the seminal findings of Suzuki and Terry13 who reported substantial axonal transport abnormalities in AD. Consistently, one finds vesicle pile-up in cell bodies similar to transport blockage models.14 Recent findings that microtubules are greatly reduced in AD15 and that AβPP/Aβ play important roles in vesicular transport16–20 further link Terry’s transport and Gylys’ synaptic hypotheses. Not answered is where the extrasynaptic synaptophysin is located. One of the most attractive possibilities is that many of the synaptophysin vesicles are located in axons; for that reason, Terry’s initial insight to focus on vesicles was particularly fruitful because it marked the reduction most specific to AD. Additional work either by cell fractionation studies, as Gylys and collaborators8 have so elegantly performed, or by immunoelectron microscopy is called for to dissect this issue more clearly.

Synaptic Degeneration by Aβ

Investigators studying the primary culprit responsible for AD have primarily focused on Aβ as the predominant factor responsible for the disease21 and, according to the amyloid cascade hypothesis, the accumulation and toxicity of Aβ leads to neuronal and synaptic loss in AD.22 Aβ is derived by proteolytic cleavage of AβPP, a protein of unknown cellular function that has the general properties of a cell surface receptor.23 The regulatory activity of three different proteolytic enzymes, α, β, and γ-secretases, at their specific cleavage sites yields a number of different products, including Aβ1–40 and Aβ1–42.24 While Aβ1–40 is the predominant product of this proteolytic pathway, Aβ1–42 is far more fibrillogenic in vitro and is the major Aβ species present in the core of senile plaques (both AD and non-AD related).25,26 Although many new questions have arisen regarding which Aβ molecule, oligomer or fibril, is the real culprit in AD pathophysiology, Aβ is inherently toxic in cell culture models.27,28 However, in vivo studies show contradictory results. AβPP transgenic mice, which possess high levels of Aβ and deposition of Aβ plaques, presented marked deficits in spatial learning by the age of 9 months.29 Although no neurotoxicity was observed in these mice, it is thought that their impaired spatial learning, which is correlated with long-term potentiation, is related to synaptic loss marked by synaptophysin. Furthermore, human studies show that, while amyloid plaque deposition is increased in brains of AD patients when compared to age-matched controls, the presence and density of amyloid deposition correlate poorly with AD symptoms and severity.30,31 In light of these findings, the amyloid cascade hypothesis has been “forced” to update, ie, instead of neuronal degeneration (neurotoxicity), Aβ induces synaptic degeneration (synaptotoxicity).22 In this updated hypothesis, it is suggested that oligomeric Aβ rather than fibrillized Aβ, may accumulates in synaptic terminals causing synaptic degeneration.5 Although no direct evidence showing whether oligomeric Aβ causes morphological and biochemical changes in synapses exists, correlation and electrophysiological studies support this updated hypothesis.

Studies performed with AβPP transgenic mouse lines showed that synaptophysin immunoreactivity is significantly decreased when compared with non-transgenic controls at 2 to 3 months old, an age where Aβ plaque formation is nonexistent in AβPP transgenic mouse lines.32 Indeed, decreases in presynaptic terminals are critically dependent on cortical Aβ levels and not on Aβ plaque burden or AβPP levels.7 In accordance, presynaptic terminals are already significantly depleted in 2- to 4-month-old AβPP transgenic mice, which present high levels of soluble Aβ levels, but have no Aβ plaque deposition. Furthermore, electrophysiological studies of young AβPP transgenic mice presenting AD-causing mutations have revealed significant deficits in basal synaptic transmission and/or long-term potentiation (LTP) in the hippocampus, well before the development of Aβ deposits. For example, transgenic mice possessing the V717F AβPP mutation show relatively smaller excitatory postsynaptic potentials and rapid decay of LTP when compared with age-matched non-transgenic mice.33 Similarly, intracerebroventricular microinjection of cell medium containing Aβ oligomers and abundant monomers, but without fibrilized Aβ, potently inhibited LTP in adult rats.34 These findings support data obtained from cell culture studies that show that oligomers of synthetic Aβ can cause acute electrophysiological changes in cultured neurons or hippocampal slices.35,36

Furthermore, it is important to emphasize the potential role of oxidative stress in synaptic degeneration, since Aβ is known to either increase or decrease oxidative stress dependent on its presentation. In fact, a recent study demonstrates that while the correlation between oxidative stress and synaptic degeneration does not reach statistical significance, the tendency of the correlation is consistent with the idea that an increase in oxidative stress may adversely affect maintenance of synaptic density.11

Good Face of Aβ: Aβ May Not Be the Cause of Synaptic Degeneration but Rather a Repair

It is important to emphasize that different types of cells such as neurons, astrocytes, neuroblastoma cells, hepatoma cells, fibroblasts, and platelets produce Aβ.37–39 These data suggest that Aβ should have an important function in normal cell development and maintenance. Recently, it has been shown that nanomolar concentrations of Aβ can block neuronal apoptosis following oxidative damage, which suggests that Aβ is essential for neuronal survival.40,41 These findings are consistent with the trophic and neuroprotective actions of Aβ at physiological concentrations in deprived conditions and neonatal cells reported during the last decade.27,42–47 Moreover, studies in vivo 48,49 show that Aβ deposition occurs after oxidative stress insults, suggesting that Aβ may have a protective role against oxidative stress.50 Indeed, we observed that in brains of AD and Down syndrome patients oxidative stress markers precede Aβ deposition and, oxidative stress seems to be inversely correlated with Aβ deposition.48,49 It is unclear whether oxidative stress also precedes synaptic degeneration and further studies are required to clarify this issue.

Together, these data provide a plausible physiological explanation for the increased generation of Aβ in AD, ie, the presence of Aβ in degenerating synapses may represent a compensatory response aimed to fight oxidative stress and promote neurite outgrowth and prevent neuronal degeneration. Therefore, in a chronological study, we can easily imagine a scenario where an initial oxidative stress event causes a neuronal defense response, such as the increase in Aβ monomer and oligomeric species,51–53 aimed to neutralize the initial cellular stress.48,49 This situation may give the “wrong” idea that Aβ is present before or in the absence of oxidative stress and, consequently, to misinterpretations that Aβ actually precedes oxidative injury and causes synaptic degeneration.

Conclusions

Synaptic degeneration is one of the most prominent events occurring in brains of AD patients. However, its causes and mechanistic basis remains unclear. While Aβ accumulation in synaptic termini may play an important role in synaptic degeneration, as suggested by previous studies, including the elegant one by Gylys et al,8 the protective role of Aβ strongly also suggests that accumulation of Aβ may be a compensatory reaction against pathological stress (eg, oxidative stress) and aimed to maintain normal synaptic functions. Furthermore, the importance of NFT in synaptic degeneration should not be ignored. Since a previous study suggested that NFT mediate starvation of synapses54 followed by synaptic degeneration, it would be extremely interesting to analyze NFT (phospho-tau) in synaptosome model, like Aβ analysis done by Gylys et al.8 Interestingly, synaptic degeneration is not only found in AD but also in other neurodegenerative diseases such as prion diseases and Huntington disease.55,56 Therefore, the information obtained from AD research may also help in understanding the mechanisms of synaptic degeneration in other neurodegenerative diseases.

Footnotes

Address reprint requests to George Perry, Ph.D., Institute of Pathology, Case Western Reserve University, 2085 Adelbert Road, Cleveland, OH 44106. E-mail: george.perry@case.edu.

References

  1. Masliah E, Terry RD, DeTeresa RM, Hansen LA. Immunohistochemical quantification of the synapse-related protein synaptophysin in Alzheimer disease. Neurosci Lett. 1989;103:234–239. doi: 10.1016/0304-3940(89)90582-x. [DOI] [PubMed] [Google Scholar]
  2. Terry RD, Masliah E, Salmon DP, Butters N, DeTeresa R, Hill R, Hansen LA, Katzman R. Physical basis of cognitive alterations in Alzheimer’s disease: synapse loss is the major correlate of cognitive impairment. Ann Neurol. 1991;30:572–580. doi: 10.1002/ana.410300410. [DOI] [PubMed] [Google Scholar]
  3. Masliah E, Terry RD. Role of synaptic pathology in the mechanisms of dementia in Alzheimer’s disease. Clin Neurosci. 1993;1:192–198. [Google Scholar]
  4. Masliah E, Terry R. The role of synaptic proteins in the pathogenesis of disorders of the central nervous system. Brain Pathol. 1993;3:77–85. doi: 10.1111/j.1750-3639.1993.tb00728.x. [DOI] [PubMed] [Google Scholar]
  5. Takahashi RH, Almeida CG, Kearney PF, Yu F, Lin MT, Milner TA, Gouras GK. Oligomerization of Alzheimer’s beta-amyloid within processes and synapses of cultured neurons and brain. J Neurosci. 2004;24:3592–3599. doi: 10.1523/JNEUROSCI.5167-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Masliah E, Abraham C, Johnson W, Forss-Petter S, Mallory M, Terry R, Mucke L. Synaptic alterations in the cortex of APP transgenic mice. J Neuropathol Exp Neurol. 1993;52:307. [Google Scholar]
  7. Mucke L, Masliah E, Yu GQ, Mallory M, Rockenstein EM, Tatsuno G, Hu K, Kholodenko D, Johnson-Wood K, McConlogue L. High-level neuronal expression of abeta 1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J Neurosci. 2000;20:4050–4058. doi: 10.1523/JNEUROSCI.20-11-04050.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Gylys KH, Fein JA, Yang F, Wiley DJ, Miller CA, Cole GM. Synaptic changes in Alzheimer’s disease: increased amyloid-β and gliosis in surviving terminals is accompanied by decreased PSD-95 fluorescence. Am J Pathol. 2004;165:1809–1817. doi: 10.1016/s0002-9440(10)63436-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Hamos JE, DeGennaro LJ, Drachman DA. Synaptic loss in Alzheimer’s disease and other dementias. Neurology. 1989;39:355–361. doi: 10.1212/wnl.39.3.355. [DOI] [PubMed] [Google Scholar]
  10. DeKosky ST, Scheff SW. Synapse loss in frontal cortex biopsies in Alzheimer’s disease: correlation with cognitive severity. Ann Neurol. 1990;27:457–464. doi: 10.1002/ana.410270502. [DOI] [PubMed] [Google Scholar]
  11. Scheff SW, Price DA. Synaptic pathology in Alzheimer’s disease: a review of ultrastructural studies. Neurobiol Aging. 2003;24:1029–1046. doi: 10.1016/j.neurobiolaging.2003.08.002. [DOI] [PubMed] [Google Scholar]
  12. Gonatas NK, Anderson W, Evangelista I. The contribution of altered synapses in the senile plaque: an electron microscopic study in Alzheimer’s dementia. J Neuropathol Exp Neurol. 1967;26:25–39. doi: 10.1097/00005072-196701000-00003. [DOI] [PubMed] [Google Scholar]
  13. Suzuki K, Terry RD. Fine structural localization of acid phosphatase in senile plaques in Alzheimer’s presenile dementia. Acta Neuropathol (Berl) 1967;8:276–284. doi: 10.1007/BF00688828. [DOI] [PubMed] [Google Scholar]
  14. Praprotnik D, Smith MA, Richey PL, Vinters HV, Perry G. Filament heterogeneity within the dystrophic neurites of senile plaques suggests blockage of fast axonal transport in Alzheimer’s disease. Acta Neuropathol (Berl) 1996;91:226–235. doi: 10.1007/s004010050420. [DOI] [PubMed] [Google Scholar]
  15. Cash AD, Aliev G, Siedlak SL, Nunomura A, Fujioka H, Zhu X, Raina AK, Vinters HV, Tabaton M, Johnson AB, Paula-Barbosa M, Avila J, Jones PK, Castellani RJ, Smith MA, Perry G. Microtubule reduction in Alzheimer’s disease and aging is independent of tau filament formation. Am J Pathol. 2003;162:1623–1627. doi: 10.1016/s0002-9440(10)64296-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Morfini G, Pigino G, Beffert U, Busciglio J, Brady ST. Fast axonal transport misregulation and Alzheimer’s disease. Neuromol Med. 2002;2:89–99. doi: 10.1385/NMM:2:2:089. [DOI] [PubMed] [Google Scholar]
  17. Stamer K, Vogel R, Thies E, Mandelkow E, Mandelkow EM. Tau blocks traffic of organelles, neurofilaments, and APP vesicles in neurons and enhances oxidative stress. J Cell Biol. 2002;156:1051–1063. doi: 10.1083/jcb.200108057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Cai D, Leem JY, Greenfield JP, Wang P, Kim BS, Wang R, Lopes KO, Kim SH, Zheng H, Greengard P, Sisodia SS, Thinakaran G, Xu H. Presenilin-1 regulates intracellular trafficking and cell surface delivery of β-amyloid precursor protein. J Biol Chem. 2003;278:3446–3454. doi: 10.1074/jbc.M209065200. [DOI] [PubMed] [Google Scholar]
  19. Pigino G, Morfini G, Pelsman A, Mattson MP, Brady ST, Busciglio J. Alzheimer’s presenilin 1 mutations impair kinesin-based axonal transport. J Neurosci. 2003;23:4499–4508. doi: 10.1523/JNEUROSCI.23-11-04499.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Tezapsidis N, Merz PA, Merz G, Hong H. Microtubular interactions of presenilin direct kinesis of Abeta peptide and its precursors. FASEB J. 2003;17:1322–1324. doi: 10.1096/fj.02-0980fje. [DOI] [PubMed] [Google Scholar]
  21. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–356. doi: 10.1126/science.1072994. [DOI] [PubMed] [Google Scholar]
  22. Selkoe DJ. Alzheimer’s disease is a synaptic failure. Science. 2002;298:789–791. doi: 10.1126/science.1074069. [DOI] [PubMed] [Google Scholar]
  23. Kang J, Lemaire HG, Unterbeck A, Salbaum JM, Masters CL, Grzeschik KH, Multhaup G, Beyreuther K, Muller-Hill B. The precursor of Alzheimer’s disease amyloid A4 protein resembles a cell-surface receptor. Nature. 1987;325:733–736. doi: 10.1038/325733a0. [DOI] [PubMed] [Google Scholar]
  24. Sisodia SS. Secretion of the beta-amyloid precursor protein. Ann NY Acad Sci. 1992;674:53–57. doi: 10.1111/j.1749-6632.1992.tb27476.x. [DOI] [PubMed] [Google Scholar]
  25. Burdick D, Soreghan B, Kwon M, Kosmoski J, Knauer M, Henschen A, Yates J, Cotman C, Glabe C. Assembly and aggregation properties of synthetic Alzheimer’s A4/beta amyloid peptide analogs. J Biol Chem. 1992;267:546–554. [PubMed] [Google Scholar]
  26. Jarrett JT, Lansbury PT., Jr Seeding “one-dimensional crystallization” of amyloid: a pathogenic mechanism in Alzheimer’s disease and scrapie? Cell. 1993;73:1055–1058. doi: 10.1016/0092-8674(93)90635-4. [DOI] [PubMed] [Google Scholar]
  27. Yankner BA, Duffy LK, Kirschner DA. Neurotrophic and neurotoxic effects of amyloid beta protein: reversal by tachykinin neuropeptides. Science. 1990;250:279–282. doi: 10.1126/science.2218531. [DOI] [PubMed] [Google Scholar]
  28. Dahlgren KN, Manelli AM, Stine WB, Jr, Baker LK, Krafft GA, LaDu MJ. Oligomeric and fibrillar species of amyloid-beta peptides differentially affect neuronal viability. J Biol Chem. 2002;277:32046–32053. doi: 10.1074/jbc.M201750200. [DOI] [PubMed] [Google Scholar]
  29. Hsiao K, Chapman P, Nilsen S, Eckman C, Harigaya Y, Younkin S, Yang F, Cole G. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science. 1996;274:99–102. doi: 10.1126/science.274.5284.99. [DOI] [PubMed] [Google Scholar]
  30. Giannakopoulos P, Herrmann FR, Bussiere T, Bouras C, Kovari E, Perl DP, Morrison JH, Gold G, Hof PR. Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer’s disease. Neurology. 2003;60:1495–1500. doi: 10.1212/01.wnl.0000063311.58879.01. [DOI] [PubMed] [Google Scholar]
  31. Guillozet AL, Weintraub S, Mash DC, Mesulam MM. Neurofibrillary tangles, amyloid, and memory in aging and mild cognitive impairment. Arch Neurol. 2003;60:729–736. doi: 10.1001/archneur.60.5.729. [DOI] [PubMed] [Google Scholar]
  32. Hsia AY, Masliah E, McConlogue L, Yu GQ, Tatsuno G, Hu K, Kholodenko D, Malenka RC, Nicoll RA, Mucke L. Plaque-independent disruption of neural circuits in Alzheimer’s disease mouse models. Proc Natl Acad Sci USA. 1999;96:3228–3233. doi: 10.1073/pnas.96.6.3228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Larson J, Lynch G, Games D, Seubert P. Alterations in synaptic transmission and long-term potentiation in hippocampal slices from young and aged PDAPP mice. Brain Res. 1999;840:23–35. doi: 10.1016/s0006-8993(99)01698-4. [DOI] [PubMed] [Google Scholar]
  34. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, Anwyl R, Wolfe MS, Rowan MJ, Selkoe DJ. Naturally secreted oligomers of amyloid beta protein potently inhibit hippocampal long-term potentiation in vivo. Nature. 2002;416:535–539. doi: 10.1038/416535a. [DOI] [PubMed] [Google Scholar]
  35. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, Morgan TE, Rozovsky I, Trommer B, Viola KL, Wals P, Zhang C, Finch CE, Krafft GA, Klein WL. Diffusible, nonfibrillar ligands derived from Abeta1–42 are potent central nervous system neurotoxins. Proc Natl Acad Sci USA. 1998;95:6448–6453. doi: 10.1073/pnas.95.11.6448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Dougherty JJ, Wu J, Nichols RA. Beta-amyloid regulation of presynaptic nicotinic receptors in rat hippocampus and neocortex. J Neurosci. 2003;23:6740–6747. doi: 10.1523/JNEUROSCI.23-17-06740.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Haass C, Schlossmacher MG, Hung AY, Vigo-Pelfrey C, Mellon A, Ostaszewski BL, Lieberburg I, Koo EH, Schenk D, Teplow DB. Amyloid beta-peptide is produced by cultured cells during normal metabolism. Nature. 1992;359:322–325. doi: 10.1038/359322a0. [DOI] [PubMed] [Google Scholar]
  38. Busciglio J, Gabuzda DH, Matsudaira P, Yankner BA. Generation of beta-amyloid in the secretory pathway in neuronal and nonneuronal cells. Proc Natl Acad Sci USA. 1993;90:2092–2096. doi: 10.1073/pnas.90.5.2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Chen M, Inestrosa NC, Ross GS, Fernandez HL. Platelets are the primary source of amyloid beta-peptide in human blood. Biochem Biophys Res Commun. 1995;213:96–103. doi: 10.1006/bbrc.1995.2103. [DOI] [PubMed] [Google Scholar]
  40. Zou K, Gong JS, Yanagisawa K, Michikawa M. A novel function of monomeric amyloid beta-protein serving as an antioxidant molecule against metal-induced oxidative damage. J Neurosci. 2002;22:4833–4841. doi: 10.1523/JNEUROSCI.22-12-04833.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Plant LD, Boyle JP, Smith IF, Peers C, Pearson HA. The production of amyloid beta peptide is a critical requirement for the viability of central neurons. J Neurosci. 2003;23:5531–5535. doi: 10.1523/JNEUROSCI.23-13-05531.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Whitson JS, Selkoe DJ, Cotman CW. Amyloid beta protein enhances the survival of hippocampal neurons in vitro. Science. 1989;243:1488–1490. doi: 10.1126/science.2928783. [DOI] [PubMed] [Google Scholar]
  43. Behl C, Davis J, Cole GM, Schubert D. Vitamin E protects nerve cells from amyloid beta protein toxicity. Biochem Biophys Res Commun. 1992;186:944–950. doi: 10.1016/0006-291x(92)90837-b. [DOI] [PubMed] [Google Scholar]
  44. Koo EH, Park L, Selkoe DJ. Amyloid beta-protein as a substrate interacts with extracellular matrix to promote neurite outgrowth. Proc Natl Acad Sci USA. 1993;90:4748–4752. doi: 10.1073/pnas.90.10.4748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Singh VK, Cheng JF, Leu SJ. Effect of substance P and protein kinase inhibitors on beta-amyloid peptide-induced proliferation of cultured brain cells. Brain Res. 1994;660:353–356. doi: 10.1016/0006-8993(94)91313-7. [DOI] [PubMed] [Google Scholar]
  46. Luo Y, Sunderland T, Roth GS, Wolozin B. Physiological levels of beta-amyloid peptide promote PC12 cell proliferation. Neurosci Lett. 1996;217:125–128. [PubMed] [Google Scholar]
  47. Kaltschmidt B, Uherek M, Wellmann H, Volk B, Kaltschmidt C. Inhibition of NF-kappaB potentiates amyloid beta-mediated neuronal apoptosis. Proc Natl Acad Sci USA. 1999;96:9409–9414. doi: 10.1073/pnas.96.16.9409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Nunomura A, Perry G, Aliev G, Hirai K, Takeda A, Balraj EK, Jones PK, Ghanbari H, Wataya T, Shimohama S, Chiba S, Atwood CS, Petersen RB, Smith MA. Oxidative damage is the earliest event in Alzheimer disease. J Neuropathol Exp Neurol. 2001;60:759–767. doi: 10.1093/jnen/60.8.759. [DOI] [PubMed] [Google Scholar]
  49. Nunomura A, Perry G, Pappolla MA, Friedland RP, Hirai K, Chiba S, Smith MA. Neuronal oxidative stress precedes amyloid-beta deposition in Down syndrome. J Neuropathol Exp Neurol. 2000;59:1011–1017. doi: 10.1093/jnen/59.11.1011. [DOI] [PubMed] [Google Scholar]
  50. Curtain CC, Ali F, Volitakis I, Cherny RA, Norton RS, Beyreuther K, Barrow CJ, Masters CL, Bush AI, Barnham KJ. Alzheimer’s disease amyloid-β binds copper and zinc to generate an allosterically ordered membrane-penetrating structure containing superoxide dismutase-like subunits. J Biol Chem. 2001;276:20466–20473. doi: 10.1074/jbc.M100175200. [DOI] [PubMed] [Google Scholar]
  51. Yan SD, Yan SF, Chen X, Fu J, Chen M, Kuppusamy P, Smith MA, Perry G, Godman GC, Nawroth P, Zweier JL, Stern D. Non-enzymatically glycated tau in Alzheimer’s disease induces neuronal oxidant stress resulting in cytokine gene expression and release of amyloid beta-peptide. Nat Med. 1995;1:693–699. doi: 10.1038/nm0795-693. [DOI] [PubMed] [Google Scholar]
  52. Misonou H, Morishima-Kawashima M, Ihara Y. Oxidative stress induces intracellular accumulation of amyloid beta-protein (Abeta) in human neuroblastoma cells. Biochemistry. 2000;39:6951–6959. doi: 10.1021/bi000169p. [DOI] [PubMed] [Google Scholar]
  53. Paola D, Domenicotti C, Nitti M, Vitali A, Borghi R, Cottalasso D, Zaccheo D, Odetti P, Strocchi P, Marinari UM, Tabaton M, Pronzato MA. Oxidative stress induces increase in intracellular amyloid beta-protein production and selective activation of betaI and betaII PKCs in NT2 cells. Biochem Biophys Res Commun. 2000;268:642–646. doi: 10.1006/bbrc.2000.2164. [DOI] [PubMed] [Google Scholar]
  54. Mandelkow EM, Stamer K, Vogel R, Thies E, Mandelkow E. Clogging of axons by tau, inhibition of axonal traffic and starvation of synapses. Neurobiol Aging. 2003;24:1079–1085. doi: 10.1016/j.neurobiolaging.2003.04.007. [DOI] [PubMed] [Google Scholar]
  55. Cunningham C, Deacon R, Wells H, Boche D, Waters S, Diniz CP, Scott H, Rawlins JN, Perry VH. Synaptic changes characterize early behavioural signs in the ME7 model of murine prion disease. Eur J Neurosci. 2003;17:2147–2155. doi: 10.1046/j.1460-9568.2003.02662.x. [DOI] [PubMed] [Google Scholar]
  56. Li JY, Plomann M, Brundin P. Huntington’s disease: a synaptopathy? Trends Mol Med. 2003;9:414–420. doi: 10.1016/j.molmed.2003.08.006. [DOI] [PubMed] [Google Scholar]

Articles from The American Journal of Pathology are provided here courtesy of American Society for Investigative Pathology

RESOURCES