Skip to main content

Advertisement

Log in

Structures of fibrils formed by α-synuclein hereditary disease mutant H50Q reveal new polymorphs

  • Article
  • Published:

From Nature Structural & Molecular Biology

View current issue Submit your manuscript

Abstract

Deposits of amyloid fibrils of α-synuclein are the histological hallmarks of Parkinson’s disease, dementia with Lewy bodies and multiple system atrophy, with hereditary mutations in α-synuclein linked to the first two of these conditions. Seeing the changes to the structures of amyloid fibrils bearing these mutations may help to understand these diseases. To this end, we determined the cryo-EM structures of α-synuclein fibrils containing the H50Q hereditary mutation. We find that the H50Q mutation results in two previously unobserved polymorphs of α-synuclein: narrow and wide fibrils, formed from either one or two protofilaments, respectively. These structures recapitulate conserved features of the wild-type fold but reveal new structural elements, including a previously unobserved hydrogen-bond network and surprising new protofilament arrangements. The structures of the H50Q polymorphs help to rationalize the faster aggregation kinetics, higher seeding capacity in biosensor cells and greater cytotoxicity that we observe for H50Q compared to wild-type α-synuclein.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Comparison of wild-type and H50Q polymorphs.
Fig. 2: Cryo-EM structures of H50Q polymorphs.
Fig. 3: Comparison of protofilaments A and B.
Fig. 4: Solvation energy maps and biochemical characterization of H50Q and wild-type α-syn.

Similar content being viewed by others

Data availability

All structural data have been deposited into the Protein Database (PDB) and the Electron Microscopy Data Bank (EMDB) with the following accession codes: H50Q narrow fibril (PDB 6PEO, EMD-20328) and H50Q wide fibril (PDB 6PES, EMD-20331). All other data are available from the authors upon reasonable request.

References

  1. Spillantini, M. G., Crowther, R. A., Jakes, R., Hasegawa, M. & Goedert, M. α-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with Lewy bodies. Proc. Natl Acad. Sci. USA 95, 6469–6473 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Grazia Spillantini, M. et al. Filamentous α-synuclein inclusions link multiple system atrophy with Parkinson’s disease and dementia with Lewy bodies. Neurosci. Lett. 251, 205–208 (1998).

    Article  CAS  Google Scholar 

  3. Spillantini, M. G. & Goedert, M. Neurodegeneration and the ordered assembly of α-synuclein. Cell Tissue Res. 373, 137–148 (2018).

    Article  CAS  PubMed  Google Scholar 

  4. Singleton, A. B. et al. α-Synuclein locus triplication causes parkinson’s disease. Science 302, 841–841 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Chartier-Harlin, M.-C. et al. α-synuclein locus duplication as a cause of familial Parkinson’s disease. Lancet 364, 1167–1169 (2004).

    Article  CAS  Google Scholar 

  6. Ibáñez, P. et al. Causal relation between α-synuclein gene duplication and familial Parkinson’s disease. Lancet 364, 1169–1171 (2004).

  7. Serpell, L. C., Berriman, J., Jakes, R., Goedert, M. & Crowther, R. A. Fiber diffraction of synthetic α-synuclein filaments shows amyloid-like cross-β conformation. Proc. Natl Acad. Sci. USA 97, 4897–4902 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Luk, K. C. et al. Pathological α-synuclein transmission initiates Parkinson-like neurodegeneration in nontransgenic mice. Science 338, 949–953 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Tycko, R. Solid-state NMR studies of amyloid fibril structure. Annu. Rev. Phys. Chem. 62, 279–299 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Bai, X., McMullan, G. & Scheres, S. H. W. How cryo-EM is revolutionizing structural biology. Trends Biochem. Sci. 40, 49–57 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Sawaya, M. R. et al. Atomic structures of amyloid cross-β spines reveal varied steric zippers. Nature 447, 453–457 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Rodriguez, J. A. et al. Structure of the toxic core of α-synuclein from invisible crystals. Nature 525, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Colvin, M. T. et al. Atomic resolution structure of monomorphic Aβ42 amyloid fibrils. J. Am. Chem. Soc. 138, 9663–9674 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Li, B. et al. Cryo-EM of full-length α-synuclein reveals fibril polymorphs with a common structural kernel. Nat. Commun. 9, 3609 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Proukakis, C. et al. A novel α-synuclein missense mutation in Parkinson disease. Neurology 80, 1062–1064 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Appel-Cresswell, S. et al. Alpha-synuclein p.H50Q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 28, 811–813 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Rutherford, N. J., Moore, B. D., Golde, T. E. & Giasson, B. I. Divergent effects of the H50Q and G51D SNCA mutations on the aggregation of α-synuclein. J. Neurochem. 131, 859–867 (2014).

    Article  CAS  PubMed  Google Scholar 

  18. Porcari, R. et al. The H50Q mutation induces a 10-fold decrease in the solubility of α-synuclein. J. Biol. Chem. 290, 2395–2404 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Khalaf, O. et al. The H50Q mutation enhances α-synuclein aggregation, secretion, and toxicity. J. Biol. Chem. 289, 21856–21876 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dearborn, A. D. et al. α-Synuclein amyloid fibrils with two entwined, asymmetrically associated protofibrils. J. Biol. Chem. 291, 2310–2318 (2016).

    Article  CAS  PubMed  Google Scholar 

  21. Fitzpatrick, A. W. P. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease. Nature 547, 185–190 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Liberta, F. et al. Cryo-EM fibril structures from systemic AA amyloidosis reveal the species complementarity of pathological amyloids. Nat. Commun. 10, 1104 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Cao, Q., Boyer, D. R., Sawaya, M. R., Ge, P. & Eisenberg, D. S. Cryo-EM structures of four polymorphic TDP-43 amyloid cores. Nat. Struct. Mol. Biol. 26, 619–627 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Murray, D. T. et al. Structure of FUS protein fibrils and its relevance to self-assembly and phase separation of low-complexity domains. Cell 171, 615–627.e16 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Ghosh, D. et al. The Parkinson’s disease-associated H50Q mutation accelerates α-synuclein aggregation in vitro. Biochemistry 52, 6925–6927 (2013).

    Article  CAS  PubMed  Google Scholar 

  26. Prusiner, S. B. et al. Evidence for α-synuclein prions causing multiple system atrophy in humans with parkinsonism. Proc. Natl Acad. Sci. USA 112, E5308–E5317 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, Y. et al. Amyloid fibril structure of α-synuclein determined by cryo-electron microscopy. Cell Res. 28, 897–903 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Guerrero-Ferreira, R. et al. Cryo-EM structure of α-synuclein fibrils. eLife 7, e36402 (2018).

  29. Tuttle, M. D. et al. Solid-state NMR structure of a pathogenic fibril of full-length human α-synuclein. Nat. Struct. Mol. Biol. 23, 409–415 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Radamaker, L. et al. Cryo-EM structure of a light chain-derived amyloid fibril from a patient with systemic AL amyloidosis. Nat. Commun. 10, 1103 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Swuec, P. et al. Cryo-EM structure of cardiac amyloid fibrils from an immunoglobulin light chain AL amyloidosis patient. Nat. Commun. 10, 1269 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Nelson, R. et al. Structure of the cross-β spine of amyloid-like fibrils. Nature 435, 773–778 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Falcon, B. et al. Novel tau filament fold in chronic traumatic encephalopathy encloses hydrophobic molecules. Nature 568, 420–423 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Liu, C. et al. Out-of-register β-sheets suggest a pathway to toxic amyloid aggregates. Proc. Natl Acad. Sci. USA 109, 20913–20918 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Cohen, S. I. A. et al. Proliferation of amyloid-β42 aggregates occurs through a secondary nucleation mechanism. Proc. Natl Acad. Sci. USA 110, 9758–9763 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Hosp, F. et al. Spatiotemporal proteomic profiling of Huntington’s disease inclusions reveals widespread loss of protein function. Cell Rep. 21, 2291–2303 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bousset, L. et al. Structural and functional characterization of two alpha-synuclein strains. Nat. Commun. 4, 2575 (2013).

    Article  PubMed  CAS  Google Scholar 

  38. Lemkau, L. R. et al. Site-specific perturbations of alpha-synuclein fibril structure by the Parkinson’s disease associated mutations A53T and E46K. PLoS ONE 8, e49750 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lemkau, L. R. et al. Mutant protein A30P α-synuclein adopts wild-type fibril structure, despite slower fibrillation kinetics. J. Biol. Chem. 287, 11526–11532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wakabayashi, M. & Matsuzaki, K. Formation of amyloids by Aβ-(1–42) on NGF-differentiated PC12 cells: roles of gangliosides and cholesterol. J. Mol. Biol. 371, 924–933 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Ono, K., Condron, M. M. & Teplow, D. B. Structure–neurotoxicity relationships of amyloid β-protein oligomers. Proc. Natl Acad. Sci. USA 106, 14745–14750 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. van Meerloo, J., Kaspers, G. J. L. & Cloos, J. in Cancer Cell Culture: Methods and Protocols (ed. Cree, I. A.) 237–245 (Humana Press, 2011).

  43. Suloway, C. et al. Automated molecular microscopy: the new leginon system. J. Struct. Biol. 151, 41–60 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Grant, T. & Grigorieff, N. Automatic estimation and correction of anisotropic magnification distortion in electron microscopes. J. Struct. Biol. 192, 204–208 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Rohou, A. & Grigorieff, N. CTFFIND4: fast and accurate defocus estimation from electron micrographs. J. Struct. Biol. 192, 216–221 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Grant, T. & Grigorieff, N. Measuring the optimal exposure for single particle cryo-EM using a 2.6 Å reconstruction of rotavirus VP6. eLife 4, e06980 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Tang, G. et al. EMAN2: an extensible image processing suite for electron microscopy. J. Struct. Biol. 157, 38–46 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Terwilliger, T. C., Sobolev, O. V., Afonine, P. V. & Adams, P. D. Automated map sharpening by maximization of detail and connectivity. Acta Crystallogr. D 74, 545–559 (2018).

    Article  CAS  Google Scholar 

  49. Emsley, P., Lohkamp, B., Scott, W. G. & Cowtan, K. Features and development of coot. Acta Crystallogr. D 66, 486–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Afonine, P. V. et al. Real-space refinement in PHENIX for cryo-EM and crystallography. Acta Crystallogr. D 74, 531–544 (2018).

    Article  CAS  Google Scholar 

  51. Eisenberg, D. S., Wesson, M. & Yamashita, M. Interpretation of protein folding and binding with atomic solvation parameters. Chem. Scr. 29A, 217–222 (1989).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Zhou for use of Electron Imaging Center for Nanomachines (EICN) resources and P. Ge for assistance in cryo-EM data collection. We acknowledge the use of instruments at the EICN supported by NIH (grant nos. 1S10RR23057 and 1S10OD018111), NSF (grant no. DBI-1338135) and CNSI at UCLA. The authors acknowledge grant nos. NIH AG 060149, NIH AG 054022, NIH AG061847 and DOE DE-FC02-02ER63421 for support. D.R.B. was supported by the National Science Foundation Graduate Research Fellowship Program.

Author information

Authors and Affiliations

Authors

Contributions

D.R.B. and B.L. designed experiments and performed data analysis. B.L. and C.S. expressed and purified the α-syn protein. B.L. grew fibrils of α-syn and performed biochemical experiments. D.R.B. and B.L prepared cryo-EM samples and performed cryo-EM data collection. B.L. and W.F. selected filaments from cryo-EM images. D.R.B. performed cryo-EM data processing and built the atomic models. M.R.S. wrote the software for and D.R.B. carried out solvation energy calculations. All authors analyzed the results and D.R.B wrote the manuscript with input from all authors. L.J. and D.S.E. supervised and guided the project.

Corresponding authors

Correspondence to Lin Jiang or David S. Eisenberg.

Ethics declarations

Competing interests

D.S.E. is an advisor and equity shareholder in ADRx, Inc.

Additional information

Peer review information Inês Chen was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Fourier Shell Analysis.

a) Helical reconstructions of Narrow and Wide Fibrils with minimum and maximum widths labeled. b) Gold-standard half map FSC curves for Narrow (top, left) and Wide (top, right) Fibrils. Map-model FSC curve for Narrow and Wide Fibrils (bottom).

Extended Data Fig. 2 Cryo-EM images and processing.

a) Cryo-EM micrographs and 2D class averages of Narrow (left) and Wide (right) Fibrils. Scale bar = 50 nm. b) 1024 and 288 pixel box size class averages of the Narrow Fibril used to determine crossover distance. 288 pixel box map projections match 2D class averages. c) 686 pixel box size class averages used to determine crossover distance. 686 pixel box map projections match 2D class averages. d) Wide Fibril class averages with a 320 pixel box demonstrate a lack of two-fold symmetry across the fibril axis.

Extended Data Fig. 3 Speculative Atomic Models for Islands 1 and 2.

a) Schematic illustrating possible sequences occupying Islands 1 and 2. 8mers from residues 1–19 and 112–140 were considered as possibilities to occupy Island 1. Island 2 is considered to consist of residues 26–31 followed by a disordered linker formed by residues 32KTKE35. b) Illustration of possible regions from either Protofilament A (left) or Protofilament B (right) that could occupy Island 1. Note that residues from the N-terminus of Protofilament A could account for Island 1 in both the Narrow and Wide Fibril; however, only the Narrow Fibril model is shown here. Island 2 is thought to be formed by the N-terminus of Protofilament A in both Narrow and Wide Fibrils. c) Speculative models for Islands 1 and 2. Check marks indicate plausible models while X’s indicate implausible models. Island 1 models are from either the N-terminus of Protofilament A (blue panels) or the C-terminus of Protofilament B (green panels). Examples of sequences that were found to not be allowed to occupy Island 1 are shown with red dashed circles highlighting steric clashes or β-strand breaking proline residues.

Extended Data Fig. 4 Alternate conformations of K58 and T59 and potential solvent molecules in the α-syn β-arch cavity.

a) Wild-type and H50Q fibrils display alternate conformations of K58 and T59. We note that in order for the Wide Fibril to form, T59 needs to be facing away from the fibril core. Therefore the formation of the Wide Fibril is mutually exclusive with our wild-type rod polymorph. b) Environmental distances of putative water molecule for Protofilament A and B in Wide Fibril and b) Protofilament A in Narrow Fibril.

Extended Data Fig. 5 PreNAC homozipper Island 1 model and additional solvation energy maps.

a) Speculative model of preNAC residues 50QGVATVA56 occupying Island 1 in Protofilament A. b) Atomic solvation map and energetic calculations for Protofilament A with Island 1 as 50QGVATVA56 and Island 2 as 26VAEAAG31. c) Atomic energy solvation map for Wild-type rod polymorph (6cu7). Notice that K58 and T59 can have favorable stabilization energies whether they are facing the solvent or facing the cavity in the β-arch.

Extended Data Fig. 6 Comparison of α-syn protofilament interfaces.

a) Wide Fibril overview (left). 56AEKTKEQV63 homointerface with Wide Fibril electron density (middle). 56AEKTKEQV63 homointerface showing a 2.4 Å rise between mated strands from Protofilament A and Protofilament B and a distance of 7.8 Å between mated sheets of Protofilament A and B (right). b) Van der Waal’s surface, buried surface area, and shape complementarity of 58KTKE61 homointerface, preNAC interface, and NACore interface.

Extended Data Fig. 7 H50Q disrupts the wild-type rod polymorph preNAC protofilament interface.

a) Conformation of H50Q Protofilament A K45 and H50Q. b) Interaction of K45-H50-E57 in the wild-type rod polymorph protofilament interface. c) Hypothetical H50Q double protofilament using the preNAC of Protofilament A as a steric zipper interface. Notice that the H50Q mutation disfavors the protofilament interface due to steric clashes with E57. d) Hypothetical H50Q protofilament interface using preNAC of Protofilament B. Notice the steric clashes between H50Q and E57 at the hypothetical protofilament interface as well as clashes of other parts of the protofilament with Protofilament A.

Extended Data Fig. 8 H50Q fibrils disrupt PC12 cell membranes more than WT fibrils.

Differentiated PC12 cells were treated with sonicated WT and H50Q fibrils and cell permeability was measured via LDH activity in the media (see Methods). H50Q leads to significantly higher cell permeabilization at 1000 and 2000 nM than WT a-syn. Error bars represent standard deviation of four independent experiments. **** = p-value ≤ 0.0001. *** = p-value ≤ 0.001. ns = p-value > 0.05. P-values were calculated using an unpaired, two-tailed t-test with a 95% CI.

Extended Data Fig. 9 Structural alignment of different wild-type and mutant α-syn polymorphs.

a) Structural alignment of H50Q Protofilament A with all wild-type structures determined thus far. b) Structural alignment of residues 50–57 in wild-type and mutant α-syn polymorphs reveals the kernel region is largely conserved while tail regions, especially the N-terminus, adopt variable conformations.

Extended Data Fig. 10

Schematic illustrating possible secondary nucleation of Protofilament B by Narrow Fibrils.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Boyer, D.R., Li, B., Sun, C. et al. Structures of fibrils formed by α-synuclein hereditary disease mutant H50Q reveal new polymorphs. Nat Struct Mol Biol 26, 1044–1052 (2019). https://doi.org/10.1038/s41594-019-0322-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0322-y

  • Springer Nature America, Inc.

This article is cited by

Navigation