720
Views
10
CrossRef citations to date
0
Altmetric
Extra View

Cellular prion protein and Alzheimer disease

Link to oligomeric amyloid-β and neuronal cell death

, &
Pages 114-116 | Received 20 Sep 2012, Accepted 11 Nov 2012, Published online: 15 Nov 2012

Abstract

Soluble oligomeric amyloid-β (Aβ) has been suggested to impair synaptic and neuronal function, leading to neurodegeneration that is clinically observed as the memory and cognitive dysfunction characteristic of Alzheimer disease, while the precise mechanism(s) whereby oligomeric Aβ causes neurotoxicity remains unknown. Recently, the cellular prion protein (PrPC) was reported to be an essential co-factor in mediating the neurotoxic effect of oligomeric Aβ. Our recent study showed that Prnp−/− mice are resistant to the neurotoxic effect of oligomeric Aβ in vivo and in vitro. Furthermore, application of an anti-PrPC antibody or PrPC peptide was able to block oligomeric Aβ-induced neurotoxicity. These findings demonstrate that PrPC may be involved in neuropathologic conditions other than conventional prion diseases, i.e., Creutzfeldt-Jakob disease.

This article refers to:

Alzheimer disease (AD) is a progressive neurodegenerative disorder representing the most common cause of dementia in the elderly. Characterized clinically by memory loss, dementia, and eventual decline in motor skills, AD is only definitively diagnosed after post-mortem demonstration of the neuropathological hallmarks of the disease. These hallmarks include intracellular aggregates of phosphorylated tau in neurofibrillary tangles, neuronal cell loss in specific populations within the brain, and extracellular deposits of the Aβ protein in senile plaques. Although oligomeric Aβ, out of all of the Aβ species, is suggested to play an important role in the neuronal loss, the mechanisms through which Aβ causes neuronal death are still unclear. The pathological relevance of oligomeric Aβ has been substantiated by its disease-specific accumulation in transgenic mouse AD modelsCitation1 and by the accumulation of structurally equivalent oligomers in the human brain and cerebrospinal fluid. There is also great interest in understanding the mechanisms whereby oligomeric Aβ affects synaptic functions involved in learning and memory. Such knowledge may provide significant insight into AD pathogenesis and potentially lead to better strategies for the prevention and/or treatment of AD.

Recent observations suggested the possibility of a connection between AD and prion diseases.Citation2Citation6 Initially, Lauren et al. identified PrPC as a high affinity receptor for oligomeric Aβ and showed that expression of PrPC was essential for oligomeric Aβ-induced synaptic toxicity as determined by loss of long-term potentiation (LTP)Citation2 and memory impairment in transgenic Alzheimer mice.Citation3 However, three independent studies failed to confirm that PrPC played a critical role either in vivo or in vitro.Citation4Citation6 Balducci et al. reported that oligomeric Aβ is responsible for cognitive impairment in AD and that PrPC is not required.Citation5 Another paper suggested that ablation or overexpression of PrPC had no effect on the impairment of hippocampal synaptic plasticity in APP/PS1 mice.Citation4 At the same time, independent studies, including one from our group, provided additional experimental support for the hypothesis that PrPC acts as a mediator of Aβ-induced toxicity. Recent studies confirmed that an anti-PrP antibody targeted to PrPC residues 93–102 blocks LTP induced by Aβ-containing AD brain extract.Citation7,Citation8 The cross-linking of PrPC by oligomeric Aβ triggers abnormal activation of cPLA2 and synapse damage.Citation9

Our recent study found that Prnp−/− mice are resistant to the neurotoxic effect of oligomeric Aβ in vivo and in in vitro hippocampal slice cultures. Furthermore, blocking the binding between PrPC and oligomeric Aβ using either an anti-PrPC antibody or a competitive PrPC peptide prevents Aβ oligomer-induced neurotoxicity.Citation10 These studies support the hypothesis that the PrPC/Aβ interaction is necessary for neuronal cell loss. Actually, other groups have also recently provided additional evidence that PrPC can act as a mediator of Aβ-induced toxicity.Citation11,Citation7 The expression of PrPC sensitizes cells to the toxic effects of other β-sheet-rich conformers, such as the yeast prion protein Sup35 or designed β-sheet peptides, in addition to Aβ.Citation11

The molecular mechanisms of neurotoxicity caused by oligomeric Aβ through PrPC remain unclear. NMDA receptor-mediated excitotoxicity might be the downstream mediator of Aβ neurotoxicity, since our data and others showed that an NMDA antagonist blocks the neurotoxicity.Citation10 Oligomeric Aβ was found to directly or indirectly bind the NMDA receptorCitation12, Citation11 and PrPC is also reported to interact with the NR2D subunit, which is a key regulatory subunit of the NMDA receptor.Citation13 Interestingly, we found that Aβ-induced neurotoxicity was significantly reduced by general pharmacological blockage of the NMDA receptor and by specifically blocking the NR2B subunit (our unpublished data). Consistent with our result, oligomeric Aβ induces early neuronal dysfunction largely by activation of NR2B-containing NMDA-receptors.Citation14, Citation15 Several studies indicate that NR2A is generally found at the synapse, whereas NR2B is predominantly localized to extrasynaptic sites.Citation16 Enhanced activation of extrasynaptic NR2B-containing NMDARs is common in AD and leads to excessive influx of Ca2+ into the cell, resulting in inappropriate activation of enzymes (such as calpains and other calcium-regulated enzymes) and mitochondrial dysfunction, culminating in apoptosis.Citation16, Citation17 Collectively, these data suggest that abnormal NMDA receptor function may contribute to the neurotoxicity of oligomeric Aβ through PrPC.

The connection between the NR2 subunit of the NMDA receptor and PrPC is far from being understood. This may involve interaction of PrPC with the protein assemblies and additional cellular factors required for signal transmission. Fyn kinase, a member of the Src family of tyrosine kinases (SFK), is a likely candidate being a signaling molecule that links the two proteins; it is a well-known kinase of the NR2 subunitCitation19,Citation18 and is activated by stimulation of PrPC in bioaminergic cells.Citation20 In addition, Fyn has been suggested to play roles in AD pathogenesis and Aβ-induced neurotoxicity.Citation22,Citation21 Since the molecular mechanisms by which Fyn contributes to neurotoxicity are unclear, it would be interesting to study the role of Fyn kinase in Aβ-induced neurotoxicity through PrPC and its mechanism. Indeed, Um et al. recently reported that the activation of Fyn kinase by Aβ and its neurotoxicity requires PrPC expression suggesting the PrPC-Fyn pathway is the main pathway of Aβ neurotoxicity.Citation23 Future study is required to delineate the connection between the NR2 subunit and PrPC.

Another important issue regarding the PrPC-Aβ interaction is to identify the binding sites on PrPC linked to neurotoxic signaling by oligomeric Aβ. Pretreatment with the antibody 6D11, which binds PrPC93–109, prevents neuronal cell death by oligomeric Aβ, while another anti-PrPC antibody, 6H4, which recognizes PrPC144–152, failed to block oligomeric Aβ-induced neuronal toxicity.Citation10 Consistent with the antibody blocking experiments, addition of the peptide corresponding to residues PrPC98–107 reduced the neurotoxicity of oligomeric Aβ in the hippocampal slice cultures, whereas the peptide corresponding to residues PrPC213–230 had no effect on Aβ-induced neurotoxicity. These data indicate that the essential region in PrPC that mediates the Aβ-PrPC interaction falls within residues 98–107. Further support for this finding is provided by a previous studyCitation2 that identified the amino acid sequence 95–105 as a binding site on PrPC for Aβ. Treatment with an antibody binding this region prevented the Aβ-PrPC interaction and Aβ-induced LTPCitation2 and improved cognitive deficits in aged AD transgenic mice.Citation24 Additional recent studies also demonstrated that an anti-PrP antibody targeted to PrPC93–102 blocks LTP induced by Aβ-containing AD brain extract.Citation7, Citation8 Furthermore, PrPN, a mutant lacking a large portion of the N-terminal domain, is impaired in both binding to Aβ and mediating its toxic effects.Citation11 Collectively, these findings strongly suggest that PrPC93–107 contains the critical amino acid sequence for oligomer Aβ-induced synaptic impairment and neuronal cell death. To further refine the binding site, a study using overlapping peptides as competitors for Aß binding would be required. Although an increasing number of observations support the role of PrPC as a receptor for Aβ-induced neuronal loss, further research is required to identify the neurotoxic mechanisms that act through PrPC.

Neurotoxic signaling through PrPC might explain, at least in part, mechanisms underlying the neurodegeneration seen in AD. In addition, it is possible that the Aß/PrPC interaction has a function in normal neuronal function that is subverted in the disease state. Future studies should provide further clarification of the important roles of PrPC in normal neuronal physiology as well as in the neurotoxic mechanism.

Acknowledgments

Work in the authors’ laboratories is supported by the National Institutes of Health (AG028679 to H.G.L.) and the Alzheimer’s Association (IIRG-11–173471 to H.G.L.).

References

  • Tomiyama T, Matsuyama S, Iso H, Umeda T, Takuma H, Ohnishi K, 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 2010; 30:4845 - 56; http://dx.doi.org/10.1523/JNEUROSCI.5825-09.2010; PMID: 20371804
  • Laurén J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature 2009; 457:1128 - 32; http://dx.doi.org/10.1038/nature07761; PMID: 19242475
  • Gimbel DA, Nygaard HB, Coffey EE, Gunther EC, Laurén J, Gimbel ZA, et al. Memory impairment in transgenic Alzheimer mice requires cellular prion protein. J Neurosci 2010; 30:6367 - 74; http://dx.doi.org/10.1523/JNEUROSCI.0395-10.2010; PMID: 20445063
  • Calella AM, Farinelli M, Nuvolone M, Mirante O, Moos R, Falsig J, et al. Prion protein and Abeta-related synaptic toxicity impairment. EMBO Mol Med 2010; 2:306 - 14; http://dx.doi.org/10.1002/emmm.201000082; PMID: 20665634
  • Balducci C, Beeg M, Stravalaci M, Bastone A, Sclip A, Biasini E, et al. Synthetic amyloid-beta oligomers impair long-term memory independently of cellular prion protein. Proc Natl Acad Sci U S A 2010; 107:2295 - 300; http://dx.doi.org/10.1073/pnas.0911829107; PMID: 20133875
  • Kessels HW, Nguyen LN, Nabavi S, Malinow R. The prion protein as a receptor for amyloid-beta. Nature 2010; 466:E3 - 4, discussion E4-5; http://dx.doi.org/10.1038/nature09217; PMID: 20703260
  • Barry AE, Klyubin I, Mc Donald JM, Mably AJ, Farrell MA, Scott M, et al. Alzheimer’s disease brain-derived amyloid-β-mediated inhibition of LTP in vivo is prevented by immunotargeting cellular prion protein. J Neurosci 2011; 31:7259 - 63; http://dx.doi.org/10.1523/JNEUROSCI.6500-10.2011; PMID: 21593310
  • Freir DB, Nicoll AJ, Klyubin I, Panico S, Mc Donald JM, Risse E, et al. Interaction between prion protein and toxic amyloid β assemblies can be therapeutically targeted at multiple sites. Nat Commun 2011; 2:336; http://dx.doi.org/10.1038/ncomms1341; PMID: 21654636
  • Bate C, Williams A. Amyloid-β-induced synapse damage is mediated via cross-linkage of cellular prion proteins. J Biol Chem 2011; 286:37955 - 63; http://dx.doi.org/10.1074/jbc.M111.248724; PMID: 21900234
  • Kudo W, et al. Cellular prion protein is essential for oligomeric amyloid-beta-induced neuronal cell death. Hum Mol Genet 2012; 21:1138 - 44; PMID: 22100763
  • Resenberger UK, Harmeier A, Woerner AC, Goodman JL, Müller V, Krishnan R, et al. The cellular prion protein mediates neurotoxic signalling of β-sheet-rich conformers independent of prion replication. EMBO J 2011; 30:2057 - 70; http://dx.doi.org/10.1038/emboj.2011.86; PMID: 21441896
  • Renner M, Lacor PN, Velasco PT, Xu J, Contractor A, Klein WL, et al. Deleterious effects of amyloid beta oligomers acting as an extracellular scaffold for mGluR5. Neuron 2010; 66:739 - 54; http://dx.doi.org/10.1016/j.neuron.2010.04.029; PMID: 20547131
  • Khosravani H, Zhang Y, Tsutsui S, Hameed S, Altier C, Hamid J, et al. Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Cell Biol 2008; 181:551 - 65; http://dx.doi.org/10.1083/jcb.200711002; PMID: 18443219
  • Li S, Jin M, Koeglsperger T, Shepardson NE, Shankar GM, Selkoe DJ. Soluble Aβ oligomers inhibit long-term potentiation through a mechanism involving excessive activation of extrasynaptic NR2B-containing NMDA receptors. J Neurosci 2011; 31:6627 - 38; http://dx.doi.org/10.1523/JNEUROSCI.0203-11.2011; PMID: 21543591
  • Rönicke R, Mikhaylova M, Rönicke S, Meinhardt J, Schröder UH, Fändrich M, et al. Early neuronal dysfunction by amyloid β oligomers depends on activation of NR2B-containing NMDA receptors. Neurobiol Aging 2011; 32:2219 - 28; http://dx.doi.org/10.1016/j.neurobiolaging.2010.01.011; PMID: 20133015
  • Fernandes HB, Raymond LA. NMDA Receptors and Huntington’s Disease. In: Van Dongen AM, ed. Biology of the NMDA Receptor. Boca Raton, FL: CRC Press; 2009:Chapter 2
  • Wang YT, Salter MW. Regulation of NMDA receptors by tyrosine kinases and phosphatases. Nature 1994; 369:233 - 5; http://dx.doi.org/10.1038/369233a0; PMID: 7514272
  • Nakazawa T, Komai S, Tezuka T, Hisatsune C, Umemori H, Semba K, et al. Characterization of Fyn-mediated tyrosine phosphorylation sites on GluR epsilon 2 (NR2B) subunit of the N-methyl-D-aspartate receptor. J Biol Chem 2001; 276:693 - 9; http://dx.doi.org/10.1074/jbc.M008085200; PMID: 11024032
  • Tezuka T, Umemori H, Akiyama T, Nakanishi S, Yamamoto T. PSD-95 promotes Fyn-mediated tyrosine phosphorylation of the N-methyl-D-aspartate receptor subunit NR2A. Proc Natl Acad Sci U S A 1999; 96:435 - 40; http://dx.doi.org/10.1073/pnas.96.2.435; PMID: 9892651
  • Mouillet-Richard S, Ermonval M, Chebassier C, Laplanche JL, Lehmann S, Launay JM, et al. Signal transduction through prion protein. Science 2000; 289:1925 - 8; http://dx.doi.org/10.1126/science.289.5486.1925; PMID: 10988071
  • Chin J, Palop JJ, Yu GQ, Kojima N, Masliah E, Mucke L. Fyn kinase modulates synaptotoxicity, but not aberrant sprouting, in human amyloid precursor protein transgenic mice. J Neurosci 2004; 24:4692 - 7; http://dx.doi.org/10.1523/JNEUROSCI.0277-04.2004; PMID: 15140940
  • Chin J, Palop JJ, Puoliväli J, Massaro C, Bien-Ly N, Gerstein H, et al. Fyn kinase induces synaptic and cognitive impairments in a transgenic mouse model of Alzheimer’s disease. J Neurosci 2005; 25:9694 - 703; http://dx.doi.org/10.1523/JNEUROSCI.2980-05.2005; PMID: 16237174
  • Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, et al. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci 2012; 15:1227 - 35; http://dx.doi.org/10.1038/nn.3178; PMID: 22820466
  • Chung E, Ji Y, Sun Y, Kascsak RJ, Kascsak RB, Mehta PD, et al. Anti-PrPC monoclonal antibody infusion as a novel treatment for cognitive deficits in an Alzheimer’s disease model mouse. BMC Neurosci 2010; 11:130; http://dx.doi.org/10.1186/1471-2202-11-130; PMID: 20946660

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.