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.

  • Article
  • Published:

α-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B

Abstract

Synucleinopathies, such as Parkinson's disease and dementia with Lewy bodies, are neurodegenerative disorders that are characterized by the accumulation of α-synuclein (aSyn) in intracellular inclusions known as Lewy bodies. Prefibrillar soluble aSyn oligomers, rather than larger inclusions, are currently considered to be crucial species underlying synaptic dysfunction. We identified the cellular prion protein (PrPC) as a key mediator in aSyn-induced synaptic impairment. The aSyn-associated impairment of long-term potentiation was blocked in Prnp null mice and rescued following PrPC blockade. We found that extracellular aSyn oligomers formed a complex with PrPC that induced the phosphorylation of Fyn kinase via metabotropic glutamate receptors 5 (mGluR5). aSyn engagement of PrPC and Fyn activated NMDA receptor (NMDAR) and altered calcium homeostasis. Blockade of mGluR5-evoked phosphorylation of NMDAR in aSyn transgenic mice rescued synaptic and cognitive deficits, supporting the hypothesis that a receptor-mediated mechanism, independent of pore formation and membrane leakage, is sufficient to trigger early synaptic damage induced by extracellular aSyn.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: PrPC mediates synaptic impairment induced by extracellular aSyn oligomers.
Figure 2: PrPC dependent-toxic effects of aSyn oligomers are mediated by Src family kinases.
Figure 3: aSyn oligomers, but not monomers, increase intracellular Ca2+ levels in primary neuronal cultures in a PrPC/NMDAR2B-dependent mechanism.
Figure 4: PrPC blockade rescues LTP impairment induced by extracellular aSyn oligomers through a mechanism dependent on mGluR5.
Figure 5: In vivo treatment of Thy1-aSyn (aSyn Tg) mice with KW-6002 rescues aSyn-associated cognitive deficits.
Figure 6: aSyn-associated synaptic and NMDAR dysfunction are rescued by KW-6002 in vivo treatment.

Similar content being viewed by others

References

  1. Yang, W. & Yu, S. Synucleinopathies: common features and hippocampal manifestations. Cell. Mol. Life Sci. 74, 1485–1501 (2017).

    Article  CAS  PubMed  Google Scholar 

  2. Lee, H.-J., Bae, E.-J. & Lee, S.-J. Extracellular α-synuclein: a novel and crucial factor in Lewy body diseases. Nat. Rev. Neurol. 10, 92–98 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Braak, H., Rüb, U., Jansen Steur, E.N.H., Del Tredici, K. & de Vos, R.A. Cognitive status correlates with neuropathologic stage in Parkinson disease. Neurology 64, 1404–1410 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. Goldman, J.G., Williams-Gray, C., Barker, R.A., Duda, J.E. & Galvin, J.E. The spectrum of cognitive impairment in Lewy body diseases. Mov. Disord. 29, 608–621 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Diógenes, M.J. et al. Extracellular alpha-synuclein oligomers modulate synaptic transmission and impair LTP via NMDA-receptor activation. J. Neurosci. 32, 11750–11762 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ferreira, D.G. et al. Adenosine A2A receptors modulate α-synuclein aggregation and toxicity. Cereb. Cortex 27, 718–730 (2017).

    PubMed  Google Scholar 

  7. Resenberger, U.K. et al. The cellular prion protein mediates neurotoxic signalling of β-sheet-rich conformers independent of prion replication. EMBO J. 30, 2057–2070 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Laurén, J., Gimbel, D.A., Nygaard, H.B., Gilbert, J.W. & Strittmatter, S.M. Cellular prion protein mediates impairment of synaptic plasticity by amyloid-β oligomers. Nature 457, 1128–1132 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wulf, M.-A., Senatore, A. & Aguzzi, A. The biological function of the cellular prion protein: an update. BMC Biol. 15, 34 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Um, J.W. et al. Alzheimer amyloid-β oligomer bound to postsynaptic prion protein activates Fyn to impair neurons. Nat. Neurosci. 15, 1227–1235 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Um, J.W. & Strittmatter, S.M. Amyloid-β induced signaling by cellular prion protein and Fyn kinase in Alzheimer disease. Prion 7, 37–41 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Williamson, R., Usardi, A., Hanger, D.P. & Anderton, B.H. Membrane-bound beta-amyloid oligomers are recruited into lipid rafts by a fyn-dependent mechanism. FASEB J. 22, 1552–1559 (2008).

    Article  CAS  PubMed  Google Scholar 

  13. Grant, S.G. et al. Impaired long-term potentiation, spatial learning, and hippocampal development in fyn mutant mice. Science 258, 1903–1910 (1992).

    Article  CAS  PubMed  Google Scholar 

  14. Suzuki, T. & Okumura-Noji, K. NMDA receptor subunits epsilon 1 (NR2A) and epsilon 2 (NR2B) are substrates for Fyn in the postsynaptic density fraction isolated from the rat brain. Biochem. Biophys. Res. Commun. 216, 582–588 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Nakazawa, T. 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. 276, 693–699 (2001).

    Article  CAS  PubMed  Google Scholar 

  16. Salter, M.W. & Kalia, L.V. Src kinases: a hub for NMDA receptor regulation. Nat. Rev. Neurosci. 5, 317–328 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Collins, M.O. et al. Molecular characterization and comparison of the components and multiprotein complexes in the postsynaptic proteome. J. Neurochem. 97 (Suppl. 1), 16–23 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Chesselet, M.-F. et al. A progressive mouse model of Parkinson's disease: the Thy1-aSyn (“Line 61”) mice. Neurotherapeutics 9, 297–314 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Magen, I. et al. Cognitive deficits in a mouse model of pre-manifest Parkinson's disease. Eur. J. Neurosci. 35, 870–882 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Caetano, F.A. et al. Amyloid-beta oligomers increase the localization of prion protein at the cell surface. J. Neurochem. 117, 538–553 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Um, J.W. et al. Metabotropic glutamate receptor 5 is a coreceptor for Alzheimer aβ oligomer bound to cellular prion protein. Neuron 79, 887–902 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Emes, R.D. et al. Evolutionary expansion and anatomical specialization of synapse proteome complexity. Nat. Neurosci. 11, 799–806 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Sarantis, K., Tsiamaki, E., Kouvaros, S., Papatheodoropoulos, C. & Angelatou, F. Adenosine A2A receptors permit mGluR5-evoked tyrosine phosphorylation of NR2B (Tyr1472) in rat hippocampus: a possible key mechanism in NMDA receptor modulation. J. Neurochem. 135, 714–726 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Batalha, V.L. et al. Adenosine A(2A) receptor blockade reverts hippocampal stress-induced deficits and restores corticosterone circadian oscillation. Mol. Psychiatry 18, 320–331 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Coelho, J.E. et al. Overexpression of adenosine A2A Receptors in rats: effects on depression, locomotion, and anxiety. Front. Psychiatry 5, 67 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Batalha, V.L. et al. The caffeine-binding adenosine A2A receptor induces age-like HPA-axis dysfunction by targeting glucocorticoid receptor function. Sci. Rep. 6, 31493 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yang, M. et al. Characterization of the potency, selectivity, and pharmacokinetic profile for six adenosine A2A receptor antagonists. Naunyn Schmiedebergs Arch. Pharmacol. 375, 133–144 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Schulz-Schaeffer, W.J. The synaptic pathology of α-synuclein aggregation in dementia with Lewy bodies, Parkinson's disease and Parkinson's disease dementia. Acta Neuropathol. 120, 131–143 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Kayed, R. et al. Common structure of soluble amyloid oligomers implies common mechanism of pathogenesis. Science 300, 486–489 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Outeiro, T.F. et al. Formation of toxic oligomeric α-synuclein species in living cells. PLoS One 3, e1867 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Marques, O. & Outeiro, T.F. Alpha-synuclein: from secretion to dysfunction and death. Cell Death Dis. 3, e350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Martin, Z.S. et al. α-Synuclein oligomers oppose long-term potentiation and impair memory through a calcineurin-dependent mechanism: relevance to human synucleopathic diseases. J. Neurochem. 120, 440–452 (2012).

    Article  CAS  PubMed  Google Scholar 

  33. Beraldo, F. H. et al. Regulation of Amyloid β oligomer binding to neurons and neurotoxicity by the complex prion protein/mGluR5. J. Biol. Chem. 291, 21945–21955 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Schmitz, M. et al. Loss of prion protein leads to age-dependent behavioral abnormalities and changes in cytoskeletal protein expression. Mol. Neurobiol. 50, 923–936 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Gimbel, D.A. et al. Memory impairment in transgenic Alzheimer mice requires cellular prion protein. J. Neurosci. 30, 6367–6374 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Khosravani, H. et al. Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J. Cell Biol. 181, 551–565 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Urrea, L. et al. Involvement of cellular prion protein in α-synuclein transport in neurons. Mol. Neurobiol. http://dx.doi.org/10.1007/s12035-017-0451-4 (2017).

  38. Linden, R. et al. Physiology of the prion protein. Physiol. Rev. 88, 673–728 (2008).

    Article  CAS  PubMed  Google Scholar 

  39. Sorgato, M.C. & Bertoli, A. From cell protection to death: may Ca2+ signals explain the chameleonic attributes of the mammalian prion protein? Biochem. Biophys. Res. Commun. 379, 171–174 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Larson, M. et al. The complex PrP(c)-Fyn couples human oligomeric Aβ with pathological tau changes in Alzheimer's disease. J. Neurosci. 32, 16857–71a (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. De Mario, A. et al. The prion protein constitutively controls neuronal store-operated Ca2+ entry through Fyn kinase. Front. Cell. Neurosci. 9, 416 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Criado, J.R. et al. Mice devoid of prion protein have cognitive deficits that are rescued by reconstitution of PrP in neurons. Neurobiol. Dis. 19, 255–265 (2005).

    Article  CAS  PubMed  Google Scholar 

  43. Collinge, J. et al. Prion protein is necessary for normal synaptic function. Nature 370, 295–297 (1994).

    Article  CAS  PubMed  Google Scholar 

  44. Curtis, J., Errington, M., Bliss, T., Voss, K. & MacLeod, N. Age-dependent loss of PTP and LTP in the hippocampus of PrP-null mice. Neurobiol. Dis. 13, 55–62 (2003).

    Article  PubMed  Google Scholar 

  45. Katamine, S. et al. Impaired motor coordination in mice lacking prion protein. Cell. Mol. Neurobiol. 18, 731–742 (1998).

    Article  CAS  PubMed  Google Scholar 

  46. Tebano, M.T. et al. Adenosine A2A receptors and metabotropic glutamate 5 receptors are co-localized and functionally interact in the hippocampus: a possible key mechanism in the modulation of N-methyl-D-aspartate effects. J. Neurochem. 95, 1188–1200 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Dungo, R. & Deeks, E.D. Istradefylline: first global approval. Drugs 73, 875–882 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Kachroo, A. & Schwarzschild, M.A. Adenosine A2A receptor gene disruption protects in an α-synuclein model of Parkinson's disease. Ann. Neurol. 71, 278–282 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Kasai, T. et al. Increased α-synuclein levels in the cerebrospinal fluid of patients with Creutzfeldt-Jakob disease. J. Neurol. 261, 1203–1209 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Di Scala, C. et al. Common molecular mechanism of amyloid pore formation by Alzheimer's β-amyloid peptide and α-synuclein. Sci. Rep. 6, 28781 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rockenstein, E. et al. Differential neuropathological alterations in transgenic mice expressing alpha-synuclein from the platelet-derived growth factor and Thy-1 promoters. J. Neurosci. Res. 68, 568–578 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Büeler, H. et al. Normal development and behaviour of mice lacking the neuronal cell-surface PrP protein. Nature 356, 577–582 (1992).

    Article  PubMed  Google Scholar 

  53. Morris, R.G., Garrud, P., Rawlins, J.N. & O'Keefe, J. Place navigation impaired in rats with hippocampal lesions. Nature 297, 681–683 (1982).

    Article  CAS  PubMed  Google Scholar 

  54. Laurent, C. et al. A2A adenosine receptor deletion is protective in a mouse model of tauopathy. Mol. Psychiatry 21, 97–107 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Pedersen, W.A., Wan, R., Zhang, P. & Mattson, M.P. Urocortin, but not urocortin II, protects cultured hippocampal neurons from oxidative and excitotoxic cell death via corticotropin-releasing hormone receptor type I. J. Neurosci. 22, 404–412 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Valadas, J.S. et al. Neuroprotection afforded by adenosine A2A receptor blockade is modulated by corticotrophin-releasing factor (CRF) in glutamate injured cortical neurons. J. Neurochem. 123, 1030–1040 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. Knot, H.J. et al. Twenty years of calcium imaging: cell physiology to dye for. Mol. Interv. 5, 112–127 (2005).

    Article  CAS  PubMed  Google Scholar 

  58. Barhoumi, R., Qian, Y., Burghardt, R.C. & Tiffany-Castiglioni, E. Image analysis of Ca2+ signals as a basis for neurotoxicity assays: promises and challenges. Neurotoxicol. Teratol. 32, 16–24 (2010).

    Article  CAS  PubMed  Google Scholar 

  59. Garman, R.H. Histology of the central nervous system. Toxicol. Pathol. 39, 22–35 (2011).

    Article  PubMed  Google Scholar 

  60. Vicente Miranda, H. et al. Heat-mediated enrichment of α-synuclein from cells and tissue for assessing post-translational modifications. J. Neurochem. 126, 673–684 (2013).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank L. Gros for figure layout design and C. Fahlbusch (University Medical Center Göttingen), A. Margarida Nascimento and J. Rino (Instituto de Medicina Molecular (iMM) Bioimaging facility), I. Moreira and J. Marques (iMM Rodent facility), and the iMM Histology and Comparative Pathology laboratory for technical assistance. M.T.F., H.V.M. and J.E.C. were supported by individual grants from Fundação para a Ciência e Tecnologia (FCT) (SFRH/BD/52228/2013; SFRH/BPD/109347/2015; SFRH/BPD/87647/2012); L.V.L. and T.F.O. were supported by a grant from the Fritz Thyssen Stiftung (Az. 10.12.2.165), Germany. L.V.L. received an iMM Lisboa internal fund (BIG – Breakthrough Idea Grant) for part of the project. L.V.L. is an Investigator FCT, Portugal. T.F.O. is supported by the DFG Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Germany. LISBOA-01-0145-FEDER-007391, project co-financed by FEDER, POR Lisboa 2020 - Programa Operacional Regional de Lisboa, from PORTUGAL 2020 and by Fundação para a Ciência e a Tecnologia.

Author information

Authors and Affiliations

Authors

Contributions

D.G.F. performed most of the experimental work, analyzed data and wrote the manuscript. M.T.-F., J.E.C., V.L.B. and S.H.V. assisted with behavior and calcium experiments. E.M.S. assisted with animal experiments. I.M.-M. performed the immunohistochemistry experiments. M.S., J.S.R. and I.Z. provided the Prnp−/− mice and experimental support. H.V.M. produced and characterized aSyn species. L.V.L. and T.F.O. coordinated the study, designed the experiments and wrote the manuscript. All of the authors approved the manuscript.

Corresponding authors

Correspondence to Luísa V Lopes or Tiago F Outeiro.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of the aSyn species and biological effects.

(a) SDS–PAGE separation of the different aSyn species. Monomers (aSyn mon) migrate with monomeric molecular weight (15 kDa) whereas aSyn oligomers (aSyn olig), and fibrils (aSyn fib) display SDS-resistant high-molecular weight species. (b) Changes in fEPSP slope induced by theta-burst stimulation recorded from WT rat hippocampal slices pre-incubated with extracellular aSyn monomers (aSyn mon, 90 min, 500 nM, n = 4), fibrils (aSyn fib, 90 min, 500 nM, n = 3) or in control conditions (CTR, n = 4). (c) Plot of the LTP magnitude represented in b (change in fEPSP slope at 50–60 min after theta-burst stimulation, compared to baseline) (means ± s.e.m., P < 0.01, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (d) Plot of the LTP magnitude obtained from WT and Prnp-/- hippocampal slices pre-incubated with extracellular aSyn oligomers (aSyn olig, 90 min, 500 nM, n = 10, 6) or in control conditions (CTR, n = 7, 6) (means ± s.e.m., P < 0.001, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (e) Input/Output (I/O) curves corresponding to fEPSP slope evoked by various stimulation intensities (10 – 120 μA) from WT hippocampal slices pre-incubated with or without aSyn oligomers (n = 5, 7; means ± s.e.m., P < 0.001, F-test). (f) I/O curves from Prnp-/- hippocampal slices pre-incubated with (n = 4) or without (n = 4) aSyn oligomers obtained by the same method as in e (means ± s.e.m., P > 0.05, F-test). (g) I/O curves obtained by the same method as in e, from hippocampal slices CTR (n = 6), pre-incubated with aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 7) or in the presence of the anti-PrP 6D11 antibody (6D11 + aSyn olig, 110 min, 100 nM, n = 4) (means ± s.e.m., P < 0.01, F-test). (h) Changes in fEPSP slope, obtained by the same methods as in b, from WT hippocampal slices in control conditions (CTR, n = 4) and in the presence of aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 6) or together with the anti-PrP antibodies, 8B4 (8B4 + aSyn, 110 min, 10 μg, n = 4) or C-20 (C-20 + aSyn olig, 110 min, 10 μg, n = 4) (means ± s.e.m., P < 0.001, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (i) Paired Pulse Facilitation (PPF) plotted against 200 ms interpulse intervals in WT slices submitted to the same conditions as in g (n = 3-5; means ± s.e.m., P > 0.05, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test).

Supplementary Figure 2 Src pharmacological blockade prevents aSyn oligomer-induced synaptic impairment.

(a) Plot of the LTP magnitude (change in fEPSP slope at 50–60 min after theta-burst stimulation, compared to baseline) from control WT hippocampal slices (CTR, n = 4), slices pre-incubated with the Src antagonist 1-naphthyl-PP1 (PP1, 110 min, 30 μM, n = 3) and slices pre-incubated with extracellular aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 6) or in the presence of PP1 (PP1 + aSyn olig, n = 3) (means ± s.e.m., P < 0.01, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (b) Input/Output (I/O) curves corresponding to fEPSP slope evoked by various stimulation intensities (10 – 120 μA) from control WT hippocampal slices (CTR, n = 5) and slices pre-incubated with extracellular aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 5) or in the presence of PP1 (PP1 + aSyn olig, n = 4) (means ± s.e.m., P < 0.001, F-test). (c) Quantification of the effects of the NMDA receptor antagonist APV (50 μM, 30 min) perfusion on basal fEPSP slope from control WT hippocampal slices (CTR, n = 5), and slices pre-incubated with extracellular aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 4) or in the presence of PP1 (PP1 + aSyn olig, n = 4) (change in slope between baseline and the last 10 min of APV application) (means ± s.e.m., P < 0.01, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (d) Effect of NMDA receptor antagonist APV (50 μM, 30 min) perfusion on basal fEPSP slope in WT hippocampal slices in control conditions (n = 3) or in the presence of aSyn oligomers (n = 4). (e) Quantification of the APV perfusion (50 μM, 30 min) effects on basal fEPSP slope (change in slope between baseline and the last 10 min of APV application) from WT and Prnp-/-hippocampal control slices (CTR) or slices pre-incubated with aSyn oligomers (n = 3-4; means ± s.e.m., P < 0.001). (f) Representative immunoblot and quantitation of NMDA receptor subunit 2B (NMDAR2B) and NMDA receptor subunit 1 (NMDAR1) levels in hippocampal slices from WT and Prnp-/- mice in the same conditions as in d (n = 4; means ± s.e.m., P < 0.001, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). GAPDH was used as a loading control.

Supplementary Figure 3 Extracellular aSyn oligomers induce phosphorylation of SFK kinases and NR2B subunit of NMDA receptors.

(a) Representative image of primary cultures from WT animals at 12 DIV. Mature neurons are labeled with green fluorescence with MAP-2 antibody, astrocytes, in red, are probed with anti-GFAP antibody, and cell nucleus are labeled with Hoechst 33342, in blue fluorescence. Z-stack images were acquired using a confocal microscope at 40x magnification and converted into maximum intensity projections. At the bottom a schematic representation of the aSyn incubation protocol used. (b) Representative immunoblots and quantitation of the aSyn levels in neuronal cultures incubated with extracellular aSyn oligomers over time (n = 4-5; P < 0.01, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). GAPDH was used as a loading control. (c) Representative immunoblots and quantitation of the Fyn levels in neuronal cultures incubated with extracellular aSyn oligomers over time (n = 4; P > 0.05, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). GAPDH was used as a loading control. (d) Representative immunoblots and quantitation of the SFK kinases phosphorylation levels, normalized to Fyn immunoreactivity, in primary neuronal cultures incubated with extracellular aSyn oligomers over time (n = 3-10; P < 0.01, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). (e) Representative immunoblots and quantitation of the PrPC levels in neuronal cultures incubated with extracellular aSyn oligomers over time (n = 6; P > 0.05, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). GAPDH was used as a loading control. (f) Representative immunoblots and quantitation of the NMDAR2B levels in neuronal cultures incubated with extracellular aSyn oligomers over time (n = 4; P > 0.05, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). GAPDH was used as a loading control. (g) Representative immunoblots and quantification of NMDA receptors subunit NR2B phosphorylation levels, normalized to NMDA receptor immunoreactivity, in neuronal cultures incubated with extracellular aSyn oligomers over time. (n = 3-7; P < 0.05, one-way ANOVA followed by a Dunnett’s Multiple Comparison Test). (h) Quantitative analysis of IP:aSyn and IP:PrPC (n = 3; means ± s.e.m., P < 0.05, two-sided unpaired t test). (i) Immunohistochemistry in 1.5 μm hippocampal sections from WT and aSyn Tg mice. aSyn is labelled in red and PrPC is labelled in green (scale bar: 25 μm). At the bottom, details from the 63x magnification images are presented (scale bar: 5 μm).

Supplementary Figure 4 mGluR5 mediated aSyn/PrPC long term potentiation impairment.

(a) Plot of the LTP magnitude (change in fEPSP slope at 50–60 min after theta-burst stimulation, compared to baseline) from control WT hippocampal slices (CTR, n = 4), slices pre-incubated with the mGluR5 antagonist MPEP (110 min, 5 μM, n = 3) and slices pre-incubated with extracellular aSyn oligomers alone (aSyn olig, 90 min, 500 nM, n = 6) or in the presence of MPEP (MPEP + aSyn olig, n = 4) (means ± s.e.m., P < 0.05, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test). (b) Changes in fEPSP slope induced by theta-burst stimulation recorded from WT rat hippocampal slices pre-incubated with extracellular aSyn oligomers alone (aSyn mon, 90 min, 500 nM, n = 6), in the presence of the selective A2AR antagonist SCH-58261 (110 min, 50 nM, SCH + aSyn olig, n = 3) and in the presence of SCH-58261 together with the mGluR5 agonist DHPG (110 min, 10 μM; SCH + DHPG + aSyn olig, n = 4). (c) Plot of the LTP magnitude represented in b and in Fig. 4d (change in fEPSP slope at 50–60 min after theta-burst stimulation, compared to baseline) (means ± s.e.m., P < 0.01, one-way ANOVA followed by a Bonferroni’s Multiple Comparison Test).

Supplementary Figure 5 Characterization of Thy1-aSyn (aSyn Tg) overexpressing mice.

(a) Representative western blot of independent experiments to evaluate aSyn levels in the hippocampus (HIP) and striatum (STR) of WT and aSyn Tg mice. GAPDH was used as a loading control. At the bottom quantification of aSyn immunoreactivity in relation to WT (n = 2-5; means ± s.e.m., P < 0.05, two-sided unpaired t test). (b) Top panels: compositional images of fluorescence immunohistochemistry of WT and aSyn Tg mice hippocampus (scale bar: 500 μm). aSyn is identified in red fluorescence and cell nuclei are stained with Hoechst in blue fluorescence. Bottom panels: maximum intensity projection images of z-stack taken at 63x magnification in the CA1 area of hippocampus (scale bar: 25 μm). At the right, details from the 63x magnification images are presented (scale bar: 5μm). aSyn is identified in red fluorescence and SNAP25 is labeled in green. (c) Representative western blot of independent experiments to evaluate TH levels in the striatum of WT and aSyn Tg mice. GAPDH was used as a loading control. At the bottom the respective quantification of TH immunoreactivity in relation to WT (n = 4; means ± s.e.m., P > 0.05, two-sided unpaired t test). (d) Representative images of TH immunohistochemistry of WT and aSyn Tg mice coronal brain sections. At the bottom the respective quantitation of TH immunoreactivity in relation to WT (n = 3-4; means ± s.e.m., P > 0.05, two-sided unpaired t test). (e) Representative western blot of 3 independent experiments to evaluate PrP levels in the hippocampus of WT, aSyn Tg, and Prnp-/- mice. GAPDH was used as a loading control. At the bottom the respective quantification of PrP immunoreactivity in relation to WT (n = 5-6; means ± s.e.m., P < 0.05, two-sided unpaired t test).

Supplementary Figure 6 Summary diagram of the mechanism by each aSyn oligomers induce an aberrant PrPC-mGluR5-NMDAR2B signaling at the post-synaptic density.

aSyn oligomers physically interact with PrPC (1) to activate mGluR5 (2)50. This leads to the phosphorylation and activation of the Src kinase Fyn (3) followed by the phosphorylation of NMDAR2B (Y1472) (4)27 and robust increase in intracellular Ca2+ (5). Adenosine A2AR are required for the mGluR5-induced NMDAR2B phosphorylation (6)37.

Supplementary Figure 7 Uncropped gels and blots with molecular weight standards complete images

Uncropped versions of representative western blot images from Fig. 2e, 2f, 2g, 2h, 5g, 6g, and 6h and pre-IP lysates corresponding to IPs shown in Fig. 2g and 2h. Membranes were cut prior to antibody staining to allow for simultaneous detection of proteins running at different sizes on the same membrane without reprobing. Representative membranes with the molecular weight (MW) markers used are shown at the bottom right panels.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 1835 kb)

Life Sciences Reporting Summary (PDF 173 kb)

Supplementary Table 1

Complete statistical details (XLSX 14 kb)

aSyn oligomers increase intracellular Ca2+ in neurons

Initial calcium response of FURA-2 AM loaded wildtype neurons to aSyn oligomers (aSyn mon, 500 nM) (AVI 28473 kb)

aSyn oligomers do not increase intracellular Ca2+ in Prnp (-/-) neurons

Initial calcium response of FURA-2 AM loaded Prnp (-/-) neurons to aSyn oligomers (aSyn mon, 500 nM) (AVI 31418 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ferreira, D., Temido-Ferreira, M., Vicente Miranda, H. et al. α-synuclein interacts with PrPC to induce cognitive impairment through mGluR5 and NMDAR2B. Nat Neurosci 20, 1569–1579 (2017). https://doi.org/10.1038/nn.4648

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4648

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