PrP is a central player in toxicity mediated by soluble aggregates of neurodegeneration-causing proteins

Grant T Corbett, Zemin Wang, Wei Hong, Marti Colom-Cadena, Jamie Rose, Meichen Liao, Adhana Asfaw, Tia C Hall, Lai Ding, Alexandra DeSousa, Matthew P Frosch, John Collinge, David A Harris, Michael S Perkinton, Tara L Spires-Jones, Tracy L Young-Pearse, Andrew Billinton, Dominic M Walsh, Grant T Corbett, Zemin Wang, Wei Hong, Marti Colom-Cadena, Jamie Rose, Meichen Liao, Adhana Asfaw, Tia C Hall, Lai Ding, Alexandra DeSousa, Matthew P Frosch, John Collinge, David A Harris, Michael S Perkinton, Tara L Spires-Jones, Tracy L Young-Pearse, Andrew Billinton, Dominic M Walsh

Abstract

Neurodegenerative diseases are an enormous public health problem, affecting tens of millions of people worldwide. Nearly all of these diseases are characterized by oligomerization and fibrillization of neuronal proteins, and there is great interest in therapeutic targeting of these aggregates. Here, we show that soluble aggregates of α-synuclein and tau bind to plate-immobilized PrP in vitro and on mouse cortical neurons, and that this binding requires at least one of the same N-terminal sites at which soluble Aβ aggregates bind. Moreover, soluble aggregates of tau, α-synuclein and Aβ cause both functional (impairment of LTP) and structural (neuritic dystrophy) compromise and these deficits are absent when PrP is ablated, knocked-down, or when neurons are pre-treated with anti-PrP blocking antibodies. Using an all-human experimental paradigm involving: (1) isogenic iPSC-derived neurons expressing or lacking PRNP, and (2) aqueous extracts from brains of individuals who died with Alzheimer's disease, dementia with Lewy bodies, and Pick's disease, we demonstrate that Aβ, α-synuclein and tau are toxic to neurons in a manner that requires PrPC. These results indicate that PrP is likely to play an important role in a variety of late-life neurodegenerative diseases and that therapeutic targeting of PrP, rather than individual disease proteins, may have more benefit for conditions which involve the aggregation of more than one protein.

Keywords: Alzheimer’s disease; Aβ; Dementia with Lewy bodies; Prion protein; Tau; α-Synuclein.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Fig. 1
Fig. 1
Preparation and characterization of soluble aggregates of Aβ, α-synuclein and tau. ac Representative chromatograms depicting isolation of monomeric (a) Aβ1–42, (b) α-synuclein (αSyn), and (c) tau. Downward arrows indicate elution of globular molecular weight standards, the shaded region indicates the fraction retained for aggregation experiments and the inset SDS-PAGE/coomassie (CBB) depicts migration of the purified monomers. d–f Freshly SEC-isolated monomer was diluted to 20 µM (Aβ1-42; d) and 35 µM (αSyn; e) and 50 µM (tau; f), combined with Thioflavin-T (ThT) and aggregation monitored until ThT signals plateaued (Tmax). Aβ and αSyn were aggregated without additives while tau was aggregated in the presence of 50 and 100 µM heparin and DTT, respectively. Binding of ThT to Aβ1–42 and αSyn was monitored continuously, whereas aliquots of tau were removed, mixed with ThT and assessed at 3-day intervals. In each case, identical reactions without protein (buffer) served as negative control. Fibrils were harvested at time points indicated by downward arrows, mounted on grids and representative negative-stain electron microscope (EM) micrographs of Aβ, αSyn and tau Tmax fibrils are presented to the right of each graph. g–i Representative negative-stain EM micrographs of immersion sonicated soluble aggregates (Aβ SBAs, g; αSyn SAAs, h; tau STAs, i). Size distribution of SBAs, SAAs and STAs as determined by negative-stain EM are presented to the right of each micrograph. j–l Neat Tmax fibrils (N), the supernatant of centrifuged fibrils (S), resuspended washed fibril pellets (P), and SBAs (j), SAAs (k), and STAs (l) were used for ThT binding. Buffer alone (B) and freshly isolated monomer (M) were included as negative controls. Scale bar in micrographs from d–i = 100 nm. Data in d–f and j–l are the mean ± SD, indicate three technical replicates and are representative of at least three independent experiments. Molecular weight markers (in kDa) are indicated to the right of the inset CBB gels in a, b and c
Fig. 2
Fig. 2
Soluble aggregates of Aβ, α-Synuclein and Tau bind to the N-terminus of PrP. a–c Binding of Aβ (a), αSyn (b) and tau (c) monomers, soluble aggregates and fibrils to immobilized PrP23–231 was assessed using an ELISA-like microtiter plate assay. Data shown are the mean ± SD from a single experiment, whereas inset EC50s are from at least four independent experiments. d–f Binding of soluble protein aggregates to immobilized PrP23–231 can be inhibited by anti-PrP monoclonal antibodies (mAbs) to Sites I (MI-0131) and II (ICSM35), but not a nonspecific mAb (46–4). Data shown are the mean ± SD from a single experiment, whereas inset IC50s are from at least four independent experiments. g–i To determine the regions of PrP involved in binding soluble protein aggregates, PrP23–231, PrP91–231, and PrP119–231 were immobilized and binding of SBAs (g), SAAs (h) and STAs (i) measured. Data shown are the mean ± SD from a single experiment, whereas inset EC50s are from at least 4 independent experiments
Fig. 3
Fig. 3
SBAs, SAAs and STAs bind to primary neurons in a dose-dependent and saturable manner. a, c, e Soluble protein aggregates were added to primary mouse neurons (MPNs) and binding assessed using serial-permeabilized immunocytochemistry. Representative images SBAs (a), SAAs (c) and STAs (e) are shown. Staining for MAP2, PrP and bound proteins are shown in blue, red and yellow, respectively. b, d, f Relative dose–response binding of SBAs (a), SAAs (c) and STAs (e) to MPNs was quantified using a custom FIJI macro that identified soluble protein aggregate puncta that were at least 50% PrP colocalized. Relative values were determined by normalizing binding signals to those obtained with the highest protein concentration analyzed (3 µM). Inset images depict enlarged triple-colocalization images for neurons treated with 1 μM SPAs (from a, c and e). Scale bar in a, c and e = 50 µm, and data in b, d and f represent three independent experiments with 60 images analyzed per experiment per dose
Fig. 4
Fig. 4
SBAs, SAAs and STAs bind to primary neurons in a PrP-dependent manner. a, c and e Soluble protein aggregates were added to wild-type (WT) and PrP-null (Prnp−/−) MPNs and binding assessed using serial-permeabilized immunocytochemistry. Representative images for SBAs (a), SAAs (c) and STAs (e) are shown. Staining for MAP2, PrP and bound proteins are shown in blue, red and yellow, respectively. Enlarged triple-colocalization images, indicated by boxed regions, are presented to the right of each panel. b, d and f, Relative binding of SBAs (b), SAAs (d) and STAs (f) to WT and Prnp−/− MPNs was determined as in Fig. 3. Binding of soluble protein aggregates to Prnp−/− MPNs is expressed as % binding to WT neurons. Scale bar in a, c and e = 50 µm, and data in b, d and f are the mean ± SD and represent three independent experiments with 45–300 images analyzed per experiment per genotype
Fig. 5
Fig. 5
SBAs, SAAs and STAs are toxic to primary neurons in a manner requiring PrP. a, c and e, WT MPNs were incubated without (Media), with monomers (1 μM), or with SBAs (a), SAAs (c) and STAs (e) and neurite length measured using live-cell imaging. Each well was imaged for 6 h prior to the addition of samples, and NeuroTrack-identified neurite lengths were used to normalize values obtained across 96 h after sample addition. Each point represents the mean ± SEM of four images taken from three independent wells. b, d, f Plots of normalized neurite length for WT PMNs incubated without (Media), with monomers (1 μM), or with SBAs (b), SAAs (d) and STAs (f). Data are derived from the last 6 h of the traces shown in a, c and e, respectively. Each point represents the mean ±  SEM of 4 images from three wells for each 2 h bin. g, h, i Plots of normalized neurite length for WT and PrP-null (Prnp−/−; gray shading) MPNs incubated without (Media) or with SBAs (g), SAAs (h) and STAs (i). Data are derived from the last 6 h of treatment. Each point represents the mean ± SEM of four images from three wells for each 2 h bin from three independent experiments
Fig. 6
Fig. 6
PrP is required for SPA-mediated inhibition of LTP. Field excitatory postsynaptic potential (fEPSP) were recorded simultaneously from sets of four slices using a MED64 Quad II system. a–c Time-course traces show that SBAs (a), SAAs (b) and STAs (c) dose-dependently inhibit hippocampal LTP in wild-type (WT) slices. In a and b, aCSF control (Ctr), black; 50 nM SPAs, grey; 100 nM SPAs, blue; 250 nM SPAs, green; and 500 nM SPAs, red. In c, aCSF control (Ctr), black; 0.5 nM SPAs, gray; 1 nM SPAs, blue; 2 nM SPAs, green; and 10 nM SPAs, red. d–f Time-course traces indicate that SBAs (d; 500 nM) and SAAs (e; 500 nM) and STAs (f; 10 nM) potently inhibit LTP in WT, but not PrP-null (Prnp−/−), slices. aCSF control on WT slices (WT Ctr), black; aCSF control on Prnp−/− (KO Ctr) slices, gray; SPAs on WT slices, red; SPAs on Prnp−/− slices, blue. In a–f, the gray horizontal bar represents the duration of SPA treatment and data represent the mean ± SD of 5–6 animals per group. Statistical analysis of results are provided in Supplementary Figure 5
Fig. 7
Fig. 7
SBAs, SAAs and STAs are toxic to induced neurons in a manner requiring PrP. a–c iPSC-derived human neurons (iNs) expressing (CR-C) or lacking PrP (CR-PRNP; gray shading) were incubated without (Media) or with SBAs (a), SAAs (b) and STAs (c) and neurite length measured using live-cell imaging. The CR-PRNP line was generated using CRISPR editing (Supplementary Figure 6). Data were collected and analyzed as in Fig. 5 and represent the mean ± SEM of four images from three wells for each 2 h bin from three independent experiments. d–f To determine whether immunotargeting PrP could protect against neurotoxicity induced by soluble protein aggregates, iNs were treated with or without anti-PrP mAbs to Site I (MI-0131) or II (ICSM35) before the addition of SBAs (d), SAAs (e) and STAs (f) and neurite length measured using live-cell imaging. Data were collected and analyzed as in Fig. 5 and represent the mean ± SEM of four images from three wells for each 2 h bin from three independent experiments
Fig. 8
Fig. 8
Soluble extracts from AD, DLB and PiD brains are toxic to neurons in a manner requiring PrP. ax−42 immunoassay quantification of Aβ present in homogenates from AD1 brain after immunodepletion with AW7 (AW7 ID) or preimmune serum (Mock). b iNs were incubated without (Media) or with extracts from brain AD1 ID’ed with AW7 (AW7 ID) or preimmune serum (Mock) and neurite length measured using live-cell imaging. cx−42 immunoassay quantification of Aβ present in homogenates from AD2 brain after immunodepletion with AW7 (AW7 ID) or preimmune serum (Mock). d iNs expressing (CR-Control; circles) or lacking PrP (CR-PRNP; diamonds, grey shading) were incubated without (Media) or with extracts from brain AD2 ID’ed with AW7 or preimmune serum (Mock) and neurite length measured using live-cell imaging. e ELISA quantification of αSyn present in homogenates from DLB1 brain after ID with 2F12 (2F12 ID) or isotype control (Mock). f iNs were incubated without (Media) or with extracts from brain DLB1 ID’ed with 2F12 (2F12 ID) or isotype control (Mock) and neurite length measured using live-cell imaging. g ELISA quantification of αSyn present in homogenates from DLB2 brain after ID with 2F12 (2F12 ID) or isotype control (Mock). h CR-Control and CR-PRNP iNs were incubated without (Media) or with extracts from brain DLB2 ID’ed with 2F12 (2F12 ID) or isotype control (Mock) and neurite length measured using live-cell imaging. i ELISA quantification of tau present in homogenates from PiD1 brain after ID with Tau5 (Tau5 ID) or isotype control (Mock). j iNs were incubated without (Media) or with extracts from brain PiD1 ID’ed with Tau5 (Tau5 ID) or isotype control (Mock) and neurite length measured using live-cell imaging. k ELISA quantification of tau present in homogenates from PiD2 brain after ID with Tau5 (Tau5 ID) or isotype control (Mock). l, CR-Control and CR-PRNP iNs were incubated without (Media) or with extracts from brain PiD2 ID’ed with Tau5 (Tau5 ID) or isotype control (Mock) and neurite length measured using live-cell imaging. In a, c, e, g, i and k, data represent the mean ± SD of three technical replicates and are representative of at least two independent experiments. In b, d, f, h, j and l, data were collected and analyzed as before and represent the mean ± SEM of four images from three wells for the last 6 h of three independent experiments

References

    1. Aguzzi A, Falsig J. Prion propagation, toxicity and degradation. Nat Neurosci. 2012;15:936–939. doi: 10.1038/nn.3120.
    1. Arosio P, Knowles TP, Linse S. On the lag phase in amyloid fibril formation. Phys Chem Chem Phys. 2015;17:7606–7618. doi: 10.1039/c4cp05563b.
    1. Ayachit U (2015) The ParaView guide: a parallel visualization application. Kitware, Clifton Park
    1. 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 USA. 2010;107:2295–2300. doi: 10.1073/pnas.0911829107.
    1. Ballatore C, Lee VM, Trojanowski JQ. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci. 2007;8:663–672. doi: 10.1038/nrn2194.
    1. Baron GS, Wehrly K, Dorward DW, Chesebro B, Caughey B. Conversion of raft associated prion protein to the protease-resistant state requires insertion of PrP-res (PrP(Sc)) into contiguous membranes. EMBO J. 2002;21:1031–1040. doi: 10.1093/emboj/21.5.1031.
    1. Beland M, Roucou X. The prion protein unstructured N-terminal region is a broad-spectrum molecular sensor with diverse and contrasting potential functions. J Neurochem. 2012;120:853–868. doi: 10.1111/j.1471-4159.2011.07613.x.
    1. Benilova I, Karran E, De Strooper B. The toxic Aβ oligomer and Alzheimer’s disease: an emperor in need of clothes. Nat Neurosci. 2012;15:349–357. doi: 10.1038/nn.3028.
    1. Beraldo FH, Arantes CP, Santos TG, Machado CF, Roffe M, Hajj GN, et al. Metabotropic glutamate receptors transduce signals for neurite outgrowth after binding of the prion protein to laminin gamma1 chain. FASEB J. 2011;25:265–279. doi: 10.1096/fj.10-161653.
    1. Betts V, Leissring MA, Dolios G, Wang R, Selkoe DJ, Walsh DM. Aggregation and catabolism of disease-associated intra-Aβ mutations: reduced proteolysis of AβA21G by neprilysin. Neurobiol Dis. 2008;31:442–450. doi: 10.1016/j.nbd.2008.06.001.
    1. Biasini E, Turnbaugh JA, Unterberger U, Harris DA. Prion protein at the crossroads of physiology and disease. Trends Neurosci. 2012;35:92–103. doi: 10.1016/j.tins.2011.10.002.
    1. Bueler H, Aguzzi A, Sailer A, Greiner RA, Autenried P, Aguet M, et al. Mice devoid of PrP are resistant to scrapie. Cell. 1993;73:1339–1347. doi: 10.1016/0092-8674(93)90360-3.
    1. Bueler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, DeArmond SJ, et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature. 1992;356:577–582. doi: 10.1038/356577a0.
    1. Buell AK, Galvagnion C, Gaspar R, Sparr E, Vendruscolo M, Knowles TP, et al. Solution conditions determine the relative importance of nucleation and growth processes in alpha-synuclein aggregation. Proc Natl Acad Sci USA. 2014;111:7671–7676. doi: 10.1073/pnas.1315346111.
    1. Calella AM, Farinelli M, Nuvolone M, Mirante O, Moos R, Falsig J, et al. Prion protein and Aβ-related synaptic toxicity impairment. EMBO Mol Med. 2010;2:306–314. doi: 10.1002/emmm.201000082.
    1. Caughey B, Baron GS, Chesebro B, Jeffrey M. Getting a grip on prions: oligomers, amyloids, and pathological membrane interactions. Annu Rev Biochem. 2009;78:177–204. doi: 10.1146/annurev.biochem.78.082907.145410.
    1. Caughey B, Brown K, Raymond GJ, Katzenstein GE, Thresher W. Binding of the protease-sensitive form of PrP (prion protein) to sulfated glycosaminoglycan and congo red [corrected] J Virol. 1994;68:2135–2141.
    1. Caughey B, Lansbury PT. Protofibrils, pores, fibrils, and neurodegeneration: separating the responsible protein aggregates from the innocent bystanders. Annu Rev Neurosci. 2003;26:267–298. doi: 10.1146/annurev.neuro.26.010302.081142.
    1. Chen S, Yadav SP, Surewicz WK. Interaction between human prion protein and amyloid-beta (Aβ) oligomers: role OF N-terminal residues. J Biol Chem. 2010;285:26377–26383. doi: 10.1074/jbc.M110.145516.
    1. Cisse M, Sanchez PE, Kim DH, Ho K, Yu GQ, Mucke L. Ablation of cellular prion protein does not ameliorate abnormal neural network activity or cognitive dysfunction in the J20 line of human amyloid precursor protein transgenic mice. J Neurosci. 2011;31:10427–10431. doi: 10.1523/JNEUROSCI.1459-11.2011.
    1. Cohen SI, Linse S, Luheshi LM, Hellstrand E, White DA, Rajah L, et al. Proliferation of amyloid-beta42 aggregates occurs through a secondary nucleation mechanism. Proc Natl Acad Sci USA. 2013;110:9758–9763. doi: 10.1073/pnas.1218402110.
    1. Conway KA, Harper JD, Lansbury PT. Accelerated in vitro fibril formation by a mutant alpha-synuclein linked to early-onset Parkinson disease. Nat Med. 1998;4:1318–1320. doi: 10.1038/3311.
    1. Dohler F, Sepulveda-Falla D, Krasemann S, Altmeppen H, Schluter H, Hildebrand D, et al. High molecular mass assemblies of amyloid-beta oligomers bind prion protein in patients with Alzheimer’s disease. Brain. 2014;137:873–886. doi: 10.1093/brain/awt375.
    1. El-Agnaf OM, Jakes R, Curran MD, Middleton D, Ingenito R, Bianchi E, et al. Aggregates from mutant and wild-type alpha-synuclein proteins and NAC peptide induce apoptotic cell death in human neuroblastoma cells by formation of beta-sheet and amyloid-like filaments. FEBS Lett. 1998;440:71–75. doi: 10.1016/s0014-5793(98)01418-5.
    1. Esparza TJ, Wildburger NC, Jiang H, Gangolli M, Cairns NJ, Bateman RJ, et al. Soluble amyloid-beta aggregates from human Alzheimer’s disease brains. Sci Rep. 2016;6:38187. doi: 10.1038/srep38187.
    1. Fa M, Puzzo D, Piacentini R, Staniszewski A, Zhang H, Baltrons MA, et al. Extracellular tau oligomers produce an immediate impairment of LTP and memory. Sci Rep. 2016;6:19393. doi: 10.1038/srep19393.
    1. Falcon B, Zhang W, Murzin AG, Murshudov G, Garringer HJ, Vidal R, et al. Structures of filaments from Pick’s disease reveal a novel tau protein fold. Nature. 2018;561:137–140. doi: 10.1038/s41586-018-0454-y.
    1. Falcon B, Zivanov J, Zhang W, Murzin AG, Garringer HJ, Vidal R, et al. Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature. 2019;568:420–423. doi: 10.1038/s41586-019-1026-5.
    1. Ferreira DG, Temido-Ferreira M, Vicente Miranda H, Batalha VL, Coelho JE, Szego EM, et al. alpha-synuclein interacts with PrP(C) to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci. 2017;20:1569–1579. doi: 10.1038/nn.4648.
    1. Fitzpatrick AWP, Falcon B, He S, Murzin AG, Murshudov G, Garringer HJ, et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature. 2017;547:185–190. doi: 10.1038/nature23002.
    1. Fluharty BR, Biasini E, Stravalaci M, Sclip A, Diomede L, Balducci C, et al. An N-terminal fragment of the prion protein binds to amyloid-beta oligomers and inhibits their neurotoxicity in vivo. J Biol Chem. 2013;288:7857–7866. doi: 10.1074/jbc.M112.423954.
    1. Freir DB, Nicoll AJ, Klyubin I, Panico S, Mc Donald JM, Risse E, et al. Interaction between prion protein and toxic amyloid beta assemblies can be therapeutically targeted at multiple sites. Nat Commun. 2011;2:336. doi: 10.1038/ncomms1341.
    1. Glynn MW, McAllister AK. Immunocytochemistry and quantification of protein colocalization in cultured neurons. Nat Protoc. 2006;1:1287–1296. doi: 10.1038/nprot.2006.220.
    1. Gomes LA, Hipp SA, Rijal Upadhaya A, Balakrishnan K, Ospitalieri S, Koper MJ, Largo-Barrientos P, et al. Aβ-induced acceleration of Alzheimer-related tau-pathology spreading and its association with prion protein. Acta Neuropathol. 2019;138:913–941. doi: 10.1007/s00401-019-02053-5.
    1. Grey M, Dunning CJ, Gaspar R, Grey C, Brundin P, Sparr E, et al. Acceleration of alpha-synuclein aggregation by exosomes. J Biol Chem. 2015;290:2969–2982. doi: 10.1074/jbc.M114.585703.
    1. Grundke-Iqbal I, Iqbal K, Quinlan M, Tung YC, Zaidi MS, Wisniewski HM. Microtubule-associated protein tau. A component of Alzheimer paired helical filaments. J Biol Chem. 1986;261:6084–6089.
    1. Guix FX, Corbett GT, Cha DJ, Mustapic M, Liu W, Mengel D, Chen Z, Aikawa E, Young-Pearse T, Kapogiannis D, et al. Detection of aggregation-competent tau in neuron-derived extracellular vesicles. Int J Mol Sci. 2018 doi: 10.3390/ijms19030663.
    1. Hellstrand E, Boland B, Walsh DM, Linse S. Amyloid beta-protein aggregation produces highly reproducible kinetic data and occurs by a two-phase process. ACS Chem Neurosci. 2010;1:13–18. doi: 10.1021/cn900015v.
    1. Hepler RW, Grimm KM, Nahas DD, Breese R, Dodson EC, Acton P, et al. Solution state characterization of amyloid beta-derived diffusible ligands. Biochemistry. 2006;45:15157–15167. doi: 10.1021/bi061850f.
    1. Herms J, Tings T, Gall S, Madlung A, Giese A, Siebert H, et al. Evidence of presynaptic location and function of the prion protein. J Neurosci. 1999;19:8866–8875. doi: 10.1523/JNEUROSCI.19-20-08866.1999.
    1. Hong W, Wang Z, Liu W, O’Malley TT, Jin M, Willem M, et al. Diffusible, highly bioactive oligomers represent a critical minority of soluble Aβ in Alzheimer’s disease brain. Acta Neuropathol. 2018;136:19–40. doi: 10.1007/s00401-018-1846-7.
    1. Hu NW, Corbett GT, Moore S, Klyubin I, O’Malley TT, Walsh DM, et al. Extracellular forms of abeta and tau from iPSC models of Alzheimer’s disease disrupt synaptic plasticity. Cell Rep. 2018;23:1932–1938. doi: 10.1016/j.celrep.2018.04.040.
    1. Hu NW, Nicoll AJ, Zhang D, Mably AJ, O’Malley T, Purro SA, et al. mGlu5 receptors and cellular prion protein mediate amyloid-beta-facilitated synaptic long-term depression in vivo. Nat Commun. 2014;5:3374. doi: 10.1038/ncomms4374.
    1. Jackson GS, Hill AF, Joseph C, Hosszu L, Power A, Waltho JP, et al. Multiple folding pathways for heterologously expressed human prion protein. Biochim Biophys Acta. 1999;1431:1–13. doi: 10.1016/S0167-4838(99)00038-2.
    1. Jin M, O’Nuallain B, Hong W, Boyd J, Lagomarsino VN, O’Malley TT, et al. An in vitro paradigm to assess potential anti-Abeta antibodies for Alzheimer’s disease. Nat Commun. 2018;9:2676. doi: 10.1038/s41467-018-05068-w.
    1. Kaech S, Banker G. Culturing hippocampal neurons. Nat Protoc. 2006;1:2406–2415. doi: 10.1038/nprot.2006.356.
    1. Kanmert D, Cantlon A, Muratore CR, Jin M, O’Malley TT, Lee G, et al. C-terminally truncated forms of tau, but not full-length tau or its C-terminal fragments, are released from neurons independently of cell death. J Neurosci. 2015;35:10851–10865. doi: 10.1523/JNEUROSCI.0387-15.2015.
    1. Karpuj MV, Giles K, Gelibter-Niv S, Scott MR, Lingappa VR, Szoka FC, et al. Phosphorothioate oligonucleotides reduce PrP levels and prion infectivity in cultured cells. Mol Med. 2007;13:190–198. doi: 10.2119/2006-00073.Karpuj.
    1. Kay KR, Smith C, Wright AK, Serrano-Pozo A, Pooler AM, Koffie R, et al. Studying synapses in human brain with array tomography and electron microscopy. Nat Protoc. 2013;8:1366–1380. doi: 10.1038/nprot.2013.078.
    1. Kertesz A, Munoz D. Relationship between frontotemporal dementia and corticobasal degeneration/progressive supranuclear palsy. Dement Geriatr Cogn Disord. 2004;17:282–286. doi: 10.1159/000077155.
    1. Kessels Helmut W., Nguyen Louis N., Nabavi Sadegh, Malinow Roberto. The prion protein as a receptor for amyloid-β. Nature. 2010;466(7308):E3–E4. doi: 10.1038/nature09217.
    1. Lambert MP, Barlow AK, Chromy BA, Edwards C, Freed R, Liosatos M, et al. 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.
    1. Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-beta oligomers. Nature. 2009;457:1128–1132. doi: 10.1038/nature07761.
    1. Le NTT, Wu B, Harris DA. Prion neurotoxicity. Brain Pathol. 2019;29:263–277. doi: 10.1111/bpa.12694.
    1. Levin AA. Treating disease at the RNA level with oligonucleotides. N Engl J Med. 2019;380:57–70. doi: 10.1056/NEJMra1705346.
    1. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 2001;25:402–408. doi: 10.1006/meth.2001.1262.
    1. Lu JX, Qiang W, Yau WM, Schwieters CD, Meredith SC, Tycko R. Molecular structure of beta-amyloid fibrils in Alzheimer’s disease brain tissue. Cell. 2013;154:1257–1268. doi: 10.1016/j.cell.2013.08.035.
    1. Mallucci GR, White MD, Farmer M, Dickinson A, Khatun H, Powell AD, et al. Targeting cellular prion protein reverses early cognitive deficits and neurophysiological dysfunction in prion-infected mice. Neuron. 2007;53:325–335. doi: 10.1016/j.neuron.2007.01.005.
    1. Masters CL, Simms G, Weinman NA, Multhaup G, McDonald BL, Beyreuther K. Amyloid plaque core protein in Alzheimer disease and Down syndrome. Proc Natl Acad Sci USA. 1985;82:4245–4249. doi: 10.1073/pnas.82.12.4245.
    1. Mc Donald JM, O'Malley TT, Liu W, Mably AJ, Brinkmalm G, Portelius E, Wittbold WM 3rd, Frosch MP, Walsh DM. The aqueous phase of Alzheimer's disease brain contains assemblies built from approximately 4 and approximately 7 kDa Abeta species. Alzheimers Dement. 2015;11:1286–1305. doi: 10.1016/j.jalz.2015.01.005.
    1. Medina M, Avila J. The role of extracellular Tau in the spreading of neurofibrillary pathology. Front Cell Neurosci. 2014;8:113. doi: 10.3389/fncel.2014.00113.
    1. Mengel D, Hong W, Corbett GT, Liu W, DeSousa A, Solforosi L, et al. PrP-grafted antibodies bind certain amyloid beta-protein aggregates, but do not prevent toxicity. Brain Res. 2019;1710:125–135. doi: 10.1016/j.brainres.2018.12.038.
    1. Minikel EV, Vallabh SM, Lek M, Estrada K, Samocha KE, Sathirapongsasuti JF, et al. Quantifying prion disease penetrance using large population control cohorts. Sci Transl Med. 2016;8:322–329. doi: 10.1126/scitranslmed.aad5169.
    1. Myers JK, Pace CN, Scholtz JM. Helix propensities are identical in proteins and peptides. Biochemistry. 1997;36:10923–10929. doi: 10.1021/bi9707180.
    1. Nazor Friberg K, Hung G, Wancewicz E, Giles K, Black C, Freier S, et al. Intracerebral infusion of antisense oligonucleotides into prion-infected mice. Mol Ther Nucl Acids. 2012;1:e9. doi: 10.1038/mtna.2011.6.
    1. Nicoll AJ, Panico S, Freir DB, Wright D, Terry C, Risse E, et al. Amyloid-beta nanotubes are associated with prion protein-dependent synaptotoxicity. Nat Commun. 2013;4:2416. doi: 10.1038/ncomms3416.
    1. Ondrejcak Tomas, Klyubin Igor, Corbett Grant T., Fraser Graham, Hong Wei, Mably Alexandra J., Gardener Matthew, Hammersley Jayne, Perkinton Michael S., Billinton Andrew, Walsh Dominic M., Rowan Michael J. Cellular Prion Protein Mediates the Disruption of Hippocampal Synaptic Plasticity by Soluble Tau In Vivo. The Journal of Neuroscience. 2018;38(50):10595–10606. doi: 10.1523/JNEUROSCI.1700-18.2018.
    1. O’Dowd ST, Ardah MT, Johansson P, Lomakin A, Benedek GB, Roberts KA, et al. The ELISA-measured increase in cerebrospinal fluid tau that discriminates Alzheimer’s disease from other neurodegenerative disorders is not attributable to differential recognition of tau assembly forms. J Alzheimers Dis. 2013;33:923–928. doi: 10.3233/JAD-2012-121393.
    1. Pan KM, Baldwin M, Nguyen J, Gasset M, Serban A, Groth D, et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci USA. 1993;90:10962–10966. doi: 10.1073/pnas.90.23.10962.
    1. Probst A, Anderton BH, Ulrich J, Kohler R, Kahn J, Heitz PU. Pick’s disease: an immunocytochemical study of neuronal changes. Monoclonal antibodies show that Pick bodies share antigenic determinants with neurofibrillary tangles and neurofilaments. Acta Neuropathol. 1983;60:175–182. doi: 10.1007/BF00691864.
    1. Prusiner SB. Prions. Proc Natl Acad Sci USA. 1998;95:13363–13383. doi: 10.1073/pnas.95.23.13363.
    1. Qiang W, Yau WM, Lu JX, Collinge J, Tycko R. Structural variation in amyloid-beta fibrils from Alzheimer’s disease clinical subtypes. Nature. 2017;541:217–221. doi: 10.1038/nature20814.
    1. Resenberger UK, Harmeier A, Woerner AC, Goodman JL, Muller V, Krishnan R, et al. The cellular prion protein mediates neurotoxic signalling of beta-sheet-rich conformers independent of prion replication. EMBO J. 2011;30:2057–2070. doi: 10.1038/emboj.2011.86.
    1. Resenberger UK, Winklhofer KF, Tatzelt J. Neuroprotective and neurotoxic signaling by the prion protein. Top Curr Chem. 2011;305:101–119. doi: 10.1007/128_2011_160.
    1. Sandberg MK, Al-Doujaily H, Sharps B, De Oliveira MW, Schmidt C, Richard-Londt A, et al. Prion neuropathology follows the accumulation of alternate prion protein isoforms after infective titre has peaked. Nat Commun. 2014;5:4347. doi: 10.1038/ncomms5347.
    1. Sanjana NE, Shalem O, Zhang F. Improved vectors and genome-wide libraries for CRISPR screening. Nat Methods. 2014;11:783–784. doi: 10.1038/nmeth.3047.
    1. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nat Methods. 2012;9:676–682. doi: 10.1038/nmeth.2019.
    1. Selkoe DJ. Folding proteins in fatal ways. Nature. 2003;426:900–904. doi: 10.1038/nature02264.
    1. Selkoe DJ. State of aggregation. Nat Neurosci. 2011;14:399. doi: 10.1038/nn0411-399.
    1. Shankar GM, Li S, Mehta TH, Garcia-Munoz A, Shepardson NE, Smith I, et al. Amyloid-beta protein dimers isolated directly from Alzheimer’s brains impair synaptic plasticity and memory. Nat Med. 2008;14:837–842. doi: 10.1038/nm1782.
    1. Smith LM, Kostylev MA, Lee S, Strittmatter SM. Systematic and standardized comparison of reported amyloid-beta receptors for sufficiency, affinity, and Alzheimer’s disease relevance. J Biol Chem. 2019;294:6042–6053. doi: 10.1074/jbc.RA118.006252.
    1. Soto C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci. 2003;4:49–60. doi: 10.1038/nrn1007.
    1. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc Natl Acad Sci USA. 1998;95:6469–6473. doi: 10.1073/pnas.95.11.6469.
    1. Um JW, Nygaard HB, Heiss JK, Kostylev MA, Stagi M, Vortmeyer A, et al. Alzheimer amyloid-beta oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat Neurosci. 2012;15:1227–1235. doi: 10.1038/nn.3178.
    1. Usenovic M, Niroomand S, Drolet RE, Yao L, Gaspar RC, Hatcher NG, et al. Internalized tau oligomers cause neurodegeneration by inducing accumulation of pathogenic tau in human neurons derived from induced pluripotent stem cells. J Neurosci. 2015;35:14234–14250. doi: 10.1523/JNEUROSCI.1523-15.2015.
    1. La Vitola P, Beeg M, Balducci C, Santamaria G, Restelli E, Colombo L, et al. Cellular prion protein neither binds to alpha-synuclein oligomers nor mediates their detrimental effects. Brain. 2019;142:249–254. doi: 10.1093/brain/awy318.
    1. Walsh DM, Hartley DM, Selkoe DJ. The many faces of Aβ: structures and activity. Curr Med Chem Immunol Endocr Metab Agents. 2003;3:277–291. doi: 10.2174/1568013033483311.
    1. Walsh DM, Selkoe DJ. A critical appraisal of the pathogenic protein spread hypothesis of neurodegeneration. Nat Rev Neurosci. 2016;17:251–260. doi: 10.1038/nrn.2016.13.
    1. Walsh DM, Teplow DB. Alzheimer’s disease and the amyloid beta-protein. Prog Mol Biol Transl Sci. 2012;107:101–124. doi: 10.1016/B978-0-12-385883-2.00012-6.
    1. Wang Z, Jackson RJ, Hong W, Taylor WM, Corbett GT, Moreno A, et al. Human brain-derived abeta oligomers bind to synapses and disrupt synaptic activity in a manner that requires APP. J Neurosci. 2017;37:11947–11966. doi: 10.1523/JNEUROSCI.2009-17.2017.
    1. Williams TL, Choi JK, Surewicz K, Surewicz WK. Soluble prion protein binds isolated low molecular weight amyloid-beta oligomers causing cytotoxicity inhibition. ACS Chem Neurosci. 2015;6:1972–1980. doi: 10.1021/acschemneuro.5b00229.
    1. Yang T, Li S, Xu H, Walsh DM, Selkoe DJ. Large soluble oligomers of amyloid beta-protein from Alzheimer brain are far less neuroactive than the smaller oligomers to which they dissociate. J Neurosci. 2017;37:152–163. doi: 10.1523/JNEUROSCI.1698-16.2016.
    1. Younan ND, Sarell CJ, Davies P, Brown DR, Viles JH. The cellular prion protein traps Alzheimer’s Abeta in an oligomeric form and disassembles amyloid fibers. FASEB J. 2013;27:1847–1858. doi: 10.1096/fj.12-222588.
    1. Zahn R, von Schroetter C, Wuthrich K. Human prion proteins expressed in Escherichia coli and purified by high-affinity column refolding. FEBS Lett. 1997;417:400–404. doi: 10.1016/S0014-5793(97)01330-6.
    1. Zeng H, Guo M, Martins-Taylor K, Wang X, Zhang Z, Park JW, et al. Specification of region-specific neurons including forebrain glutamatergic neurons from human induced pluripotent stem cells. PLoS ONE. 2010;5:e11853. doi: 10.1371/journal.pone.0011853.
    1. Zhang Y, Pak C, Han Y, Ahlenius H, Zhang Z, Chanda S, et al. Rapid single-step induction of functional neurons from human pluripotent stem cells. Neuron. 2013;78:785–798. doi: 10.1016/j.neuron.2013.05.029.

Source: PubMed

3
Tilaa