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:

Structural mechanism for specific binding of chemical compounds to amyloid fibrils

Abstract

Amyloid fibril is an important pharmaceutical target for diagnostic and therapeutic treatment of neurodegenerative diseases. However, rational design of chemical compounds that interact with amyloid fibrils is unachievable due to the lack of mechanistic understanding of the ligand–fibril interaction. Here we used cryoelectron microscopy to survey the amyloid fibril-binding mechanism of a series of compounds including classic dyes, (pre)clinical imaging tracers and newly identified binders from high-throughput screening. We obtained clear densities of several compounds in complex with an α-synuclein fibril. These structures unveil the basic mechanism of the ligand–fibril interaction, which exhibits remarkable difference from the canonical ligand–protein interaction. In addition, we discovered a druggable pocket that is also conserved in the ex vivo α-synuclein fibrils from multiple system atrophy. Collectively, these findings expand our knowledge of protein–ligand interaction in the amyloid fibril state, which will enable rational design of amyloid binders in a medicinally beneficial way.

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

Fig. 1: Binding of chemical ligands to α-syn fibrils.
Fig. 2: Cryo-EM structure of the CCA–α-syn fibril.
Fig. 3: Cryo-EM structure of the EB–α-syn fibril.
Fig. 4: Cryo-EM structures for the ligands binding to the N-pocket of the α-syn fibril.
Fig. 5: Cryo-EM structures of ThT–α-syn and PiB–α-syn fibrils.
Fig. 6: Interligand π–π interactions in ligand-bound α-syn fibrils.

Similar content being viewed by others

Data availability

Cryo-EM maps have been deposited in the Electron Microscopy Data Bank (EMDB) under accession nos. EMD-33960 for CCA-bound α-syn fibrils, EMD-33961 for CR-bound α-syn fibrils, EMD-33965 for EB-bound α-syn fibrils, EMD-33966 for ThT-bound α-syn fibrils, EMD-33968 for BF227a-bound α-syn fibrils, EMD-33967 for PiB-bound α-syn fibrils, EMD-33969 for C05-03-bound α-syn fibrils, EMD-33970 for SIL5-bound α-syn fibrils and EMD-33971 for pFTAA-bound α-syn fibrils. The corresponding atomic models have been deposited in the PDB under the following accession nos.: 7YNF for CCA-bound α-syn fibrils, 7YNG for CR-bound α-syn fibrils, 7YNL for EB-bound α-syn fibrils, 7YNM for ThT-bound α-syn fibrils (conformation 1), 7YNN for ThT-bound α-syn fibrils (conformation 2), 7YNP for BF227a-bound α-syn fibrils, 7YNO for PiB-bound α-syn fibrils (conformation 1), 7YNQ for PiB-bound α-syn fibrils (conformation 2), 7YNR for C05-03-bound α-syn fibrils, 7YNS for SIL5-bound α-syn fibrils and 7YNT for pFTAA-bound α-syn fibrils, respectively. The structural models used in the present study are available in the PDB database under accession nos. 6A6B (apo-α-syn fibrils), 6XYO (MSA type I), 6XYP (MSA type II1) and 6XYQ (MSA type II2). Source data are provided with this paper.

References

  1. Hardy, J. A. & Higgins, G. A. Alzheimer’s disease: the amyloid cascade hypothesis. Science 256, 184–185 (1992).

    Article  CAS  PubMed  Google Scholar 

  2. Iwatsubo, T. et al. Visualization of Aβ42 (43) and Aβ40 in senile plaques with end-specific Aβ monoclonals: evidence that an initially deposited species is Aβ42 (43). Neuron 13, 45–53 (1994).

    Article  CAS  PubMed  Google Scholar 

  3. Spillantini, M. G. et al. α-Synuclein in Lewy bodies. Nature 388, 839–840 (1997).

    Article  CAS  PubMed  Google Scholar 

  4. Ballatore, C., Lee, V. M.-Y. & Trojanowski, J. Q. Tau-mediated neurodegeneration in Alzheimer’s disease and related disorders. Nat. Rev. Neurosci. 8, 663–672 (2007).

    Article  CAS  PubMed  Google Scholar 

  5. Kam, T.-I. et al. Poly(ADP-ribose) drives pathologic α-synuclein neurodegeneration in Parkinson’s disease. Science 362, eaat8407 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Zhou, X. et al. Mutations linked to neurological disease enhance self-association of low-complexity protein sequences. Science 377, eabn5582 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Bennhold, H. Specific staining of amyloid by Congo red. München. Med. Wochenschr. 69, 1537–1538 (1922).

    Google Scholar 

  8. CF, C. Fluorescent stains, with special reference to amyloid and connective tissues. Arch. Pathol. 68, 487–498 (1959).

    Google Scholar 

  9. Klunk, W. E., Pettegrew, J. & Abraham, D. J. Quantitative evaluation of congo red binding to amyloid-like proteins with a beta-pleated sheet conformation. J. Histochem. Cytochem. 37, 1273–1281 (1989).

    Article  CAS  PubMed  Google Scholar 

  10. Aliyan, A., Cook, N. P. & Martí, A. A. Interrogating amyloid aggregates using fluorescent probes. Chem. Rev. 119, 11819–11856 (2019).

    Article  CAS  PubMed  Google Scholar 

  11. Villemagne, V. L., Doré, V., Burnham, S. C., Masters, C. L. & Rowe, C. C. Imaging tau and amyloid-β proteinopathies in Alzheimer disease and other conditions. Nat. Rev. Neurol. 14, 225–236 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Korat, Š. et al. Alpha-synuclein PET tracer development—an overview about current efforts. Pharmaceuticals 14, 847 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Alzghool, O. M., van Dongen, G., van de Giessen, E., Schoonmade, L. & Beaino, W. α‐Synuclein radiotracer development and in vivo imaging: recent advancements and new perspectives. Mov. Disord. 37, 936–948 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Vandenberghe, R. et al. 18F‐flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann. Neurol. 68, 319–329 (2010).

    Article  PubMed  Google Scholar 

  15. Curtis, C. et al. Phase 3 trial of flutemetamol labeled with radioactive fluorine 18 imaging and neuritic plaque density. JAMA Neurol. 72, 287–294 (2015).

    Article  PubMed  Google Scholar 

  16. Pontecorvo, M. J. et al. Relationships between flortaucipir PET tau binding and amyloid burden, clinical diagnosis, age and cognition. Brain 140, 748–763 (2017).

    PubMed  PubMed Central  Google Scholar 

  17. Ossenkoppele, R. et al. Discriminative accuracy of [18F]flortaucipir positron emission tomography for Alzheimer disease vs other neurodegenerative disorders. Jama 320, 1151–1162 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Fleisher, A. S. et al. Positron emission tomography imaging with [18F]flortaucipir and postmortem assessment of Alzheimer disease neuropathologic changes. JAMA Neurol. 77, 829–839 (2020).

    Article  PubMed  Google Scholar 

  19. Hsu, J.-L. et al. The imaging features and clinical associations of a novel tau PET tracer—18F-APN1607 in Alzheimer disease. Clin. Nucl. Med. 45, 747–756 (2020).

    Article  PubMed  Google Scholar 

  20. Tagai, K. et al. High-contrast in vivo imaging of tau pathologies in Alzheimer’s and non-Alzheimer’s disease tauopathies. Neuron 109, 42–58.e8 (2021).

  21. Ono, M. et al. In vivo visualization of propagating α-synuclein pathologies in mouse and marmoset models by a bimodal imaging probe, C05-05. Preprint at bioRxiv https://doi.org/10.1101/2020.10.23.349860 (2021).

  22. Lu, J.-X. et al. Molecular structure of β-amyloid fibrils in Alzheimer’s disease brain tissue. Cell 154, 1257–1268 (2013).

    Article  CAS  PubMed  Google Scholar 

  23. 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.e616 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Schweighauser, M. et al. Structures of α-synuclein filaments from multiple system atrophy. Nature 585, 464–469 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Li, D. & Liu, C. Conformational strains of pathogenic amyloid proteins in neurodegenerative diseases. Nat. Rev. Neurosci. 23, 523–534 (2022).

  27. Babine, R. E. & Bender, S. L. Molecular recognition of protein−ligand complexes: applications to drug design. Chem. Rev. 97, 1359–1472 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Sawaya, M. R., Hughes, M. P., Rodriguez, J. A., Riek, R. & Eisenberg, D. S. The expanding amyloid family: structure, stability, function, and pathogenesis. Cell 184, 4857–4873 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gohlke, H. & Klebe, G. Approaches to the description and prediction of the binding affinity of small‐molecule ligands to macromolecular receptors. Angew. Chem. Int. Ed. 41, 2644–2676 (2002).

    Article  CAS  Google Scholar 

  30. Moore, P. B., Hendrickson, W. A., Henderson, R. & Brunger, A. T. The protein-folding problem: not yet solved. Science 375, 507–507 (2022).

    Article  PubMed  Google Scholar 

  31. Groenning, M. et al. Binding mode of thioflavin T in insulin amyloid fibrils. J. Struct. Biol. 159, 483–497 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Wu, C., Biancalana, M., Koide, S. & Shea, J.-E. Binding modes of thioflavin-T to the single-layer β-sheet of the peptide self-assembly mimics. J. Mol. Biol. 394, 627–633 (2009).

    Article  CAS  PubMed  Google Scholar 

  33. Biancalana, M., Makabe, K., Koide, A. & Koide, S. Molecular mechanism of thioflavin-T binding to the surface of β-rich peptide self-assemblies. J. Mol. Biol. 385, 1052–1063 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. Amdursky, N., Erez, Y. & Huppert, D. Molecular rotors: what lies behind the high sensitivity of the thioflavin-T fluorescent marker. Acc. Chem. Res. 45, 1548–1557 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Carter, D. & Chou, K.-C. A model for structure-dependent binding of Congo red to Alzheimer β-amyloid fibrils. Neurobiol. Aging 19, 37–40 (1998).

    Article  CAS  PubMed  Google Scholar 

  36. Childers, W. S., Mehta, A. K., Lu, K. & Lynn, D. G. Templating molecular arrays in amyloid’s cross-β grooves. J. Am. Chem. Soc. 131, 10165–10172 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Schütz, A. K. et al. The amyloid–Congo red interface at atomic resolution. Angew. Chem. Int. Ed. 50, 5956–5960 (2011).

    Article  Google Scholar 

  38. Frieg, B., Gremer, L., Heise, H., Willbold, D. & Gohlke, H. Binding modes of thioflavin T and Congo red to the fibril structure of amyloid-β (1–42). Chem. Commun. 56, 7589–7592 (2020).

    Article  CAS  Google Scholar 

  39. Espargaró, A. et al. On the binding of Congo Red to amyloid fibrils. Angew. Chem. 132, 8181–8184 (2020).

    Article  Google Scholar 

  40. Duan, P. et al. Binding sites of a position emission tomography imaging agent in Alzheimer’s β-amyloid fibrils studied using 19F solid-state NMR. J. Am. Chem. Soc. 144, 1416–1430 (2022).

  41. Shi, Y. et al. Cryo-EM structures of tau filaments from Alzheimer’s disease with PET ligand APN-1607. Acta Neuropathol. 141, 697–708 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Holmes, B. B. et al. Heparan sulfate proteoglycans mediate internalization and propagation of specific proteopathic seeds. Proc. Natl Acad. Sci. USA 110, E3138–E3147 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Tao, Y. et al. Heparin induces α-synuclein to form new fibril polymorphs with attenuated neuropathology. Nat. Commun. 13, 4226 (2022).

  44. Tumolo, T. & Lanfer-Marquez, U. M. Copper chlorophyllin: a food colorant with bioactive properties? Food Res. Int. 46, 451–459 (2012).

    Article  CAS  Google Scholar 

  45. Saria, A. & Lundberg, J. M. Evans blue fluorescence: quantitative and morphological evaluation of vascular permeability in animal tissues. J. Neurosci. Methods 8, 41–49 (1983).

    Article  CAS  PubMed  Google Scholar 

  46. Klunk, W. E. et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh Compound‐B. Ann. Neurol. 55, 306–319 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Fodero-Tavoletti, M. T. et al. In vitro characterisation of BF227 binding to α-synuclein/Lewy bodies. Eur. J. Pharmacol. 617, 54–58 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Kikuchi, A. et al. In vivo visualization of α-synuclein deposition by carbon-11-labelled 2-[2-(2-dimethylaminothiazol-5-yl) ethenyl]-6-[2-(fluoro) ethoxy] benzoxazole positron emission tomography in multiple system atrophy. Brain 133, 1772–1778 (2010).

  49. Liu, F. T. et al. 18F‐Florzolotau tau positron emission tomography imaging in patients with multiple system atrophy–parkinsonian subtype. Mov. Disord. 37, 1915–1923 (2022).

    Article  CAS  PubMed  Google Scholar 

  50. Yu, L. et al. Synthesis and in vitro evaluation of α-synuclein ligands. Bioorg. Med. Chem. 20, 4625–4634 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Åslund, A. et al. Novel pentameric thiophene derivatives for in vitro and in vivo optical imaging of a plethora of protein aggregates in cerebral amyloidoses. ACS Chem. Biol. 4, 673–684 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Herrmann, U. S. et al. Structure-based drug design identifies polythiophenes as antiprion compounds. Sci. Transl. Med. 7, 299ra123–299ra123 (2015).

    Article  PubMed  Google Scholar 

  53. Zivanov, J. et al. A Bayesian approach to single-particle electron cryo-tomography in RELION-4.0. eLife 11, e83724 (2022).

  54. 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 

  55. Meyer, E. A., Castellano, R. K. & Diederich, F. Interactions with aromatic rings in chemical and biological recognition. Angew. Chem. Int. Ed. 42, 1210–1250 (2003).

    Article  CAS  Google Scholar 

  56. Martinez, C. R. & Iverson, B. L. Rethinking the term ‘pi-stacking’. Chem. Sci. 3, 2191–2201 (2012).

    Article  CAS  Google Scholar 

  57. Zhao, Y. et al. Conformational preferences of π–π stacking between ligand and protein, analysis derived from crystal structure data geometric preference of π–π interaction. Interdiscip. Sci. 7, 211–220 (2015).

    Article  CAS  PubMed  Google Scholar 

  58. Jiang, L. et al. Structure-based discovery of fiber-binding compounds that reduce the cytotoxicity of amyloid beta. eLife 2, e00857 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Fodero-Tavoletti, M. T. et al. In vitro characterization of Pittsburgh Compound-B binding to Lewy bodies. J. Neurosci. 27, 10365–10371 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Klingstedt, T. et al. Luminescent conjugated oligothiophenes distinguish between α-synuclein assemblies of Parkinson’s disease and multiple system atrophy. Acta Neuropathol. Commun. 7, 193 (2019).

  61. Yang, Y. et al. Structures of α-synuclein filaments from human brains with Lewy pathology. Nature 610, 791–795 (2022).

    Article  CAS  PubMed  Google Scholar 

  62. Mastronarde, D. N. Automated electron microscope tomography using robust prediction of specimen movements. J. Struct. Biol. 152, 36–51 (2005).

    Article  PubMed  Google Scholar 

  63. Zheng, S. Q. et al. MotionCor2: anisotropic correction of beam-induced motion for improved cryo-electron microscopy. Nat. Methods 14, 331–332 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. 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 

  65. Adams, P. D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D 66, 213–221 (2010).

  66. Pettersen, E. F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

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

  68. The PyMOL Molecular Graphics System v.1.8 (Schrodinger LLC, 2015).

Download references

Acknowledgements

We thank the Cryo-Electron Microscopy center at Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry for help with cryo-EM data collection in EB, ThT, BF227a, PiB, C05-03 and SIL5 cases. We thank the Bio-Electron Microscopy Facility of ShanghaiTech University for help with cryo-EM data collection in CCA, CR and pFTAA cases. We thank Y. Sun and Q. Cao for their helpful discussion on cryo-EM data processing. We thank W. Kong from Cytiva and Y. Zhang from the Discovery Technology Platform at the Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University for providing technical support of Biacore 8K. This work was supported by the National Natural Science Foundation of China (grant nos. 82188101 and 32171236 to C.L. and 32170683 to D.L.), the Major State Basic Research Development Program (grant no. 2019YFE0120600 to C.L.), the Science and Technology Commission of Shanghai Municipality (grant nos. 20XD1425000, 2019SHZDZX02 and 22JC1410400 to C.L.) and the Shanghai Pilot Program for Basic Research—Chinese Academy of Science, Shanghai Branch (grant no. CYJ-SHFY-2022-005 to C.L.).

Author information

Authors and Affiliations

Authors

Contributions

Y.T., D.L. and C.L. designed the project. Y.T., W.X., Q.Z. and S.Z. prepared cryo-EM samples of ligand-bound α-syn fibrils, and performed cryo-EM data collection and processing. H.X., C.H., W.G., W.T. and L.T. synthesized the chemical compounds. Y.T., Q.Z. and Y.L. performed the high-throughput screening for binders of α-syn fibrils. Y.T. performed the ligand-α-syn fibril-binding assays. S.Z. performed the SPR experiment. All the authors were involved in analyzing the data and contributed to discussion of the paper and editing. Y.T., D.L. and C.L. wrote the paper.

Corresponding authors

Correspondence to Dan Li or Cong Liu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Chemical Biology thanks Salvador Ventura, Michael Stowell and Myungwoon Lee for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 ELISA-based screening assay for identification of chemical binders of α-syn fibrils.

a, Schematic illustration of the ELISA-based high-throughput screening assay. ELISA plates are coated with heparin. 100 nM of sonicated FLAG-tagged α-syn PFFs are pre−mixed with 20 μM of each compound before added to each well. Bound α-syn PFFs are detected by horse radish peroxidase (HRP) conjugated anti-FLAG antibody (Anti-FLAG-Ab-HRP). b, The binding affinities of heparin to α-syn PFFs. Top, ELISA-based binding saturation assay for heparin-α-syn PFF interaction. ELISA absorbance values are normalized and plotted against FLAG-tagged α-syn PFFs concentrations. Data are shown as mean ± s.d., n = 4 independent samples. Nonlinear regression is used to fit the curve to a binding saturation equation in GraphPad Prism 8.0.2 with R2 0.9966. Bottom, binding affinity (KD) of heparin to α-syn PFFs measured by SPR. Compound concentrations are indicated. c, IC50 measurement of CCA (left), EB (right, top) and CR (right, bottom) against α-syn PFFs-heparin interaction by ELISA. ELISA absorbance values are normalized and plotted against compound concentrations. Data are shown as mean ± s.d., n = 4 independent samples. Nonlinear regression is used to fit each curve to a sigmoidal four parameters [inhibitor]-response (variable slope) equation in GraphPad Prism 8.0.2. with R2 0.9982 for CCA curve, 0.9987 for EB curve, and 0.9995 for CR curve, respectively.

Source data

Extended Data Fig. 2 Binding affinities of ligands to α-syn fibrils measured by fluorescent binding saturation curves.

a, Differential fluorescent spectra of 50 μM ligands in the presence and absence of 50 μM α-syn fibrils. Differential fluorescence signal intensities (Ligandbound - Ligandfree) are shown as mean ± s.d., n = 3 independently prepared samples. The chemical structures of the ligands are shown. b, The fluorescent binding curves of the ligands to α-syn fibrils measured at the excited and maximum emission wavelengths in (a). 1 µM of α-syn fibrils were added. Data are shown as mean ± s.d., n = 3 independent samples. The curves were analyzed by nonlinear regression with a saturation binding equation. c, The fitting results of (b). Binding affinity KD is the concentration of ligand required to reach half-maximal binding, and h is the Hill coefficient. Bmax is the maximal binding signal.

Source data

Extended Data Fig. 3 Binding affinities of ligands to α-syn PFFs measured by surface plasmon resonance (SPR).

For each panel, the chemical structure of ligand is shown on the top. The binding affinities (KD) of ligands to α-syn PFFs were calculated based on the SPR association and dissociation curves.

Extended Data Fig. 4 Cryo-EM 3D reconstruction density maps of the ligand-bound α-syn fibrils.

For each panel, density maps are displayed as a half pitch of side view (left), enlarged top (right, top) and side views (right, bottom). Fibril parameters including the length of half pitch (180° helical turn), twist angle and helical rise are indicated. Extra densities are colored in line with the ligand colors. Chemical structures of the ligands are provided along with the density maps. The thresholds of density maps: CCA, 0.0096; CR, 0.008; EB, 0.013; ThT, 0.0098; BF227a, 0.0045; PiB, 0.0085; C05-03, 0.0045; SIL5, 0.0069; pFTAA, 0.0233.

Extended Data Fig. 5 Ligand densities in the N-pocket, C-pocket, inter-pocket, and on the back-surface around the α-syn fibril.

a, Structure model of the α-syn fibril with all residues and pockets labeled. b, For each ligand-binding pocket/surface around the α-syn fibril, cryo-EM densities and structures are shown and colored in gray. For each ligand, their densities are shown and colored in line with the ligand color. The protein density is restricted to areas within a 2-Å radius of the α-syn model, and combined with the ligand densities displayed with the same threshold: CCA, 0.0096; CR, 0.008; EB, 0.013; ThT, 0.0098; BF227a, 0.0045; PiB, 0.0085; C05-03, 0.0045; SIL5, 0.0069; pFTAA, 0.0233. Chemical structures of the ligands are provided under the ligand names.

Extended Data Fig. 6 Cryo-EM densities and structure models of CCA molecules at different ligand-binding sites of the α-syn fibril.

CCA molecules are shown as sticks. The densities are displayed with a threshold of 0.0096. Alternative orientations of CCA molecules in inter-pocket and back-surfaces are also shown.

Extended Data Fig. 7 Y39 shapes the N-pocket of α-syn fibril.

a, Structure of one layer of apo-α-syn fibril (PBD ID: 6A6B). N-pocket composed of residues 39–46 and 80–83 is highlighted in yellow. b, Two conformations of Y39 observed in the ligand-bound α-syn fibrils. When the side chain of Y39 points outwards (open state), the pocket is in the shape of a narrow deep groove preferable for specific ligand binding. When the side chain of Y39 points inwards (closed state), the pocket is in the shape of a wide shallow groove. Top and side views of N-pocket are shown. Y39 is highlighted in red.

Extended Data Fig. 8 Conserved ligand-binding N-pocket in in vitro and ex vivo α-syn fibrils from MSA.

a, Overlay of the α-syn monomeric structures in the fibrils including BF227a-α-syn fibril (yellow), type I filaments from MSA (red, PDB 6XYO), type II1 filaments from MSA (blue, PDB 6XYP), and type II2 filaments from MSA (green, PDB 6XYQ). α-Syn is shown in ribbons; BF227a is shown in sticks and colored in orange. RMSDs of BF227a-α-syn fibril versus MSA type I is 2.285 Å over 53 C-α atoms (global alignment); BF227a-α-syn fibril versus MSA type II1 is 4.577 Å over 58 C-α atoms; BF227a-α-syn fibril versus MSA type II2 is 4.568 Å over 58 C-α atoms. The N- and C-pockets are indicated by dashed box. The N-pocket is zoomed in in (b) with residue side chains shown in sticks. c, The surface of α-syn fibrils is shown and colored in line with (a). N-pocket of each fibril is framed and enlarged. Y39 is highlighted in black.

Extended Data Fig. 9 Interactions between ThT and the N-pocket of α-syn fibril.

a, π-π interactions between the benzothiazole rings of ThT (top, conformation 1, colored in yellow; bottom, conformation 2, colored in magenta) and Y39 residues (colored in wheat) in three consecutive α-syn molecules (i, i + 1, i + 2) along the fibril. Detailed interaction distances of π planes are labeled. b, Hydrophobic interactions between the methyl groups of ThT (conformation 1, colored in yellow) and the side chains of K80 and V82. The benzene ring of ThT forms cation-π interactions with the side chains of K80. Detailed interaction distances are labeled. c, Summary of the molecular interactions between ThT (conformation 1, left, colored in yellow; conformation 2, right, colored in magenta) and α-syn fibril (black). Interactions are depicted as blue dashed lines.

Extended Data Fig. 10 Interactions between PiB and the N-pocket of α-syn fibril.

a, Interactions of two alternative conformations of PiB (colored in yellow and pink, respectively) with Y39 (colored in wheat) and V82 (colored in light purple) of α-syn. Three layers of α-syn molecules (i, i + 1, i + 2) are shown. Detailed interaction distances are labeled. b, Summary of the molecular interactions between PiB (conformation 1, left; conformation 2, right) and α-syn fibril (black). Interactions are depicted as blue dashed lines. c, π-π stacking of PiB conformations along the fibril axis. Detailed distances and angles are labeled.

Supplementary information

Supplementary Information

Supplementary Figs. 1–12, Tables 1–2 and Note.

Reporting Summary

Supplementary Data 1

Statistical source data for Supplementary Fig. 4.

Source data

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Extended Data Fig. 1

Statistical source data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical source data for Extended Data Fig. 2.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tao, Y., Xia, W., Zhao, Q. et al. Structural mechanism for specific binding of chemical compounds to amyloid fibrils. Nat Chem Biol 19, 1235–1245 (2023). https://doi.org/10.1038/s41589-023-01370-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-023-01370-x

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