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.

Structural basis for amyloidogenic peptide recognition by sorLA

Abstract

SorLA is a neuronal sorting receptor considered to be a major risk factor for Alzheimer's disease. We have recently reported that it directs lysosomal targeting of nascent neurotoxic amyloid-β (Aβ) peptides by directly binding Aβ. Here, we determined the crystal structure of the human sorLA domain responsible for Aβ capture, Vps10p, in an unbound state and in complex with two ligands. Vps10p assumes a ten-bladed β-propeller fold with a large tunnel at the center. An internal ligand derived from the sorLA propeptide bound inside the tunnel to extend the β-sheet of one of the propeller blades. The structure of the sorLA Vps10p–Aβ complex revealed that the same site is used. Peptides are recognized by sorLA Vps10p in redundant modes without strict dependence on a particular amino acid sequence, thus suggesting a broad specificity toward peptides with a propensity for β-sheet formation.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Specific and pH-dependent binding of peptides to the sorLA Vps10p domain, evaluated by fluorescence polarization.
Figure 2: Transient and multipoint interaction between Aβ40 and sorLA Vps10p.
Figure 3: Structure of the human sorLA Vps10p domain.
Figure 4: Peptide-binding site inside the Vps10p propeller ring.
Figure 5: Peptide binding–induced rearrangements in Vps10p.

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

NCBI Reference Sequence

Protein Data Bank

References

  1. Jacobsen, L. et al. Molecular characterization of a novel human hybrid-type receptor that binds the α2-macroglobulin receptor-associated protein. J. Biol. Chem. 271, 31379–31383 (1996).

    Article  CAS  Google Scholar 

  2. Yamazaki, H. et al. Elements of neural adhesion molecules and a yeast vacuolar protein sorting receptor are present in a novel mammalian low density lipoprotein receptor family member. J. Biol. Chem. 271, 24761–24768 (1996).

    Article  CAS  Google Scholar 

  3. Taira, K. et al. LR11, a mosaic LDL receptor family member, mediates the uptake of ApoE-rich lipoproteins in vitro. Arterioscler. Thromb. Vasc. Biol. 21, 1501–1506 (2001).

    Article  CAS  Google Scholar 

  4. Hampe, W. et al. A head-activator binding protein is present in hydra in a soluble and a membrane-anchored form. Development 126, 4077–4086 (1999).

    CAS  PubMed  Google Scholar 

  5. Schaller, H.C., Hermans-Borgmeyer, I. & Hoffmeister, S.A. Neuronal control of development in hydra. Int. J. Dev. Biol. 40, 339–344 (1996).

    CAS  PubMed  Google Scholar 

  6. Rezgaoui, M. et al. The neuropeptide head activator is a high-affinity ligand for the orphan G-protein-coupled receptor GPR37. J. Cell Sci. 119, 542–549 (2006).

    Article  CAS  Google Scholar 

  7. Scherzer, C.R. et al. Loss of apolipoprotein E receptor LR11 in Alzheimer disease. Arch. Neurol. 61, 1200–1205 (2004).

    Article  Google Scholar 

  8. Andersen, O.M. et al. Neuronal sorting protein-related receptor sorLA/LR11 regulates processing of the amyloid precursor protein. Proc. Natl. Acad. Sci. USA 102, 13461–13466 (2005).

    Article  CAS  Google Scholar 

  9. Dodson, S.E. et al. Loss of LR11/SORLA enhances early pathology in a mouse model of amyloidosis: evidence for a proximal role in Alzheimer's disease. J. Neurosci. 28, 12877–12886 (2008).

    Article  CAS  Google Scholar 

  10. Rogaeva, E. et al. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nat. Genet. 39, 168–177 (2007).

    Article  CAS  Google Scholar 

  11. Bettens, K. et al. SORL1 is genetically associated with increased risk for late-onset Alzheimer disease in the Belgian population. Hum. Mutat. 29, 769–770 (2008).

    Article  Google Scholar 

  12. Pottier, C. et al. High frequency of potentially pathogenic SORL1 mutations in autosomal dominant early-onset Alzheimer disease. Mol. Psychiatry 17, 875–879 (2012).

    Article  CAS  Google Scholar 

  13. Caglayan, S. et al. Lysosomal sorting of amyloid-β by the SORLA receptor is impaired by a familial Alzheimer's disease mutation. Sci. Transl. Med. 6, 223ra20 (2014).

    Article  Google Scholar 

  14. Marcusson, E.G., Horazdovsky, B.F., Cereghino, J.L., Gharakhanian, E. & Emr, S.D. The sorting receptor for yeast vacuolar carboxypeptidase Y is encoded by the VPS10 gene. Cell 77, 579–586 (1994).

    Article  CAS  Google Scholar 

  15. Willnow, T.E., Petersen, C.M. & Nykjaer, A. VPS10P-domain receptors: regulators of neuronal viability and function. Nat. Rev. Neurosci. 9, 899–909 (2008).

    Article  CAS  Google Scholar 

  16. Quistgaard, E.M. et al. Ligands bind to Sortilin in the tunnel of a ten-bladed β-propeller domain. Nat. Struct. Mol. Biol. 16, 96–98 (2009).

    Article  CAS  Google Scholar 

  17. Jacobsen, L. et al. Activation and functional characterization of the mosaic receptor SorLA/LR11. J. Biol. Chem. 276, 22788–22796 (2001).

    Article  CAS  Google Scholar 

  18. Rossi, A.M. & Taylor, C.W. Analysis of protein-ligand interactions by fluorescence polarization. Nat. Protoc. 6, 365–387 (2011).

    Article  CAS  Google Scholar 

  19. Deane, R. et al. LRP/amyloid beta-peptide interaction mediates differential brain efflux of Abeta isoforms. Neuron 43, 333–344 (2004).

    Article  CAS  Google Scholar 

  20. Nakata, Z. et al. Crystallization and preliminary crystallographic analysis of human LR11 Vps10p domain. Acta Crystallogr. Sect. F Struct. Biol. Cryst. Commun. 67, 129–132 (2011).

    Article  CAS  Google Scholar 

  21. Chiti, F. & Dobson, C.M. Amyloid formation by globular proteins under native conditions. Nat. Chem. Biol. 5, 15–22 (2009).

    Article  CAS  Google Scholar 

  22. Trovato, A., Seno, F. & Tosatto, S.C.E. The PASTA server for protein aggregation prediction. Protein Eng. Des. Sel. 20, 521–523 (2007).

    Article  CAS  Google Scholar 

  23. Tycko, R. & Wickner, R.B. Molecular structures of amyloid and prion fibrils: consensus versus controversy. Acc. Chem. Res. 46, 1487–1496 (2013).

    Article  CAS  Google Scholar 

  24. Yagi-Utsumi, M. et al. NMR characterization of the interaction of GroEL with amyloid beta as a model ligand. FEBS Lett. 587, 1605–1609 (2013).

    Article  CAS  Google Scholar 

  25. Mittag, T. et al. Dynamic equilibrium engagement of a polyvalent ligand with a single-site receptor. Proc. Natl. Acad. Sci. USA 105, 17772–17777 (2008).

    Article  CAS  Google Scholar 

  26. Libich, D.S., Fawzi, N.L., Ying, J.F. & Clore, G.M. Probing the transient dark state of substrate binding to GroEL by relaxation-based solution NMR. Proc. Natl. Acad. Sci. USA 110, 11361–11366 (2013).

    Article  CAS  Google Scholar 

  27. Beglova, N. & Blacklow, S.C. The LDL receptor: how acid pulls the trigger. Trends Biochem. Sci. 30, 309–317 (2005).

    Article  CAS  Google Scholar 

  28. Ohwaki, K. et al. A secreted soluble form of LR11, specifically expressed in intimal smooth muscle cells, accelerates formation of lipid-laden macrophages. Arterioscler. Thromb. Vasc. Biol. 27, 1050–1056 (2007).

    Article  CAS  Google Scholar 

  29. Mizushima, S. & Nagata, S. Pef-Bos, a powerful mammalian expression vector. Nucleic Acids Res. 18, 5322 (1990).

    Article  CAS  Google Scholar 

  30. Stanley, P. Chinese hamster ovary cell mutants with multiple glycosylation defects for production of glycoproteins with minimal carbohydrate heterogeneity. Mol. Cell. Biol. 9, 377–383 (1989).

    Article  CAS  Google Scholar 

  31. Fujii, Y. et al. PA tag: a versatile protein tagging system using a super high affinity antibody against a dodecapeptide derived from human podoplanin. Protein Expr. Purif. 95, 240–247 (2014).

    Article  CAS  Google Scholar 

  32. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  Google Scholar 

  33. Vagin, A. & Teplyakov, A. Molecular replacement with MOLREP. Acta Crystallogr. D Biol. Crystallogr. 66, 22–25 (2010).

    Article  CAS  Google Scholar 

  34. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  Google Scholar 

  35. Murshudov, G.N. et al. REFMAC5 for the refinement of macromolecular crystal structures. Acta Crystallogr. D Biol. Crystallogr. 67, 355–367 (2011).

    Article  CAS  Google Scholar 

  36. McCoy, A.J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  38. Chen, V.B. et al. MolProbity: all-atom structure validation for macromolecular crystallography. Acta Crystallogr. D Biol. Crystallogr. 66, 12–21 (2010).

    Article  CAS  Google Scholar 

  39. Yagi-Utsumi, M., Matsuo, K., Yanagisawa, K., Gekko, K. & Kato, K. Spectroscopic characterization of intermolecular interaction of amyloid β promoted on GM1 micelles. Int. J. Alzheimers Dis. 2011, 925073 (2010).

    PubMed  PubMed Central  Google Scholar 

  40. Flanagan, J.G. et al. Alkaline phosphatase fusions of ligands or receptors as in situ probes for staining of cells, tissues, and embryos. Methods Enzymol. 327, 19–35 (2000).

    Article  CAS  Google Scholar 

  41. Tabata, S. et al. A rapid screening method for cell lines producing singly-tagged recombinant proteins using the “TARGET tag” system. J. Proteomics 73, 1777–1785 (2010).

    Article  CAS  Google Scholar 

  42. Pronk, S. et al. Gromacs 4.5: a high-throughput and highly parallel open source molecular simulation toolkit. Bioinformatics 29, 845–854 (2013).

    Article  CAS  Google Scholar 

  43. Oostenbrink, C., Villa, A., Mark, A.E. & Van Gunsteren, W.F. A biomolecular force field based on the free enthalpy of hydration and solvation: The gromos force-field parameter sets 53A5 and 53A6. J. Comput. Chem. 25, 1656–1676 (2004).

    Article  CAS  Google Scholar 

  44. Berendsen, H.J.C., Postma, J.P.M., Vangunsteren, W.F., Dinola, A. & Haak, J.R. Molecular-dynamics with coupling to an external bath. J. Chem. Phys. 81, 3684–3690 (1984).

    Article  CAS  Google Scholar 

  45. Hess, B., Kutzner, C., van der Spoel, D. & Lindahl, E. GROMACS 4: algorithms for highly efficient, load-balanced, and scalable molecular simulation. J. Chem. Theory Comput. 4, 435–447 (2008).

    Article  CAS  Google Scholar 

  46. Darden, T., York, D. & Pedersen, L. Particle mesh Ewald: an N•log(N) method for Ewald sums in large systems. J. Chem. Phys. 98, 10089–10092 (1993).

    Article  CAS  Google Scholar 

  47. Parrinello, M. & Rahman, A. Polymorphic transitions in single-crystals: a new molecular-dynamics method. J. Appl. Phys. 52, 7182–7190 (1981).

    Article  CAS  Google Scholar 

  48. Humphrey, W., Dalke, A. & Schulten, K. VMD: visual molecular dynamics. J. Mol. Graph. 14, 33–38 (1996).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We would like to thank the staff of the beamlines at Photon Factory and SPring-8 for their help with X-ray data collection, K. Yamashita for help in the setup of MD simulations and for discussions on data analysis, S. Thompson for critical reading and editing of the manuscript, K. Tamura-Kawakami for excellent technical assistance and M. Sakai for preparation of the manuscript. This work was supported by the Grant-in-Aid for Scientific Research on Innovative Areas 'Analysis and Synthesis of Multidimensional Immune Organ Network' (no. 24111006 to J.T.) and 'Dynamic Ordering of Biomolecular Systems for Creation of Integrated Functions' (no. 25102008 to K.K.) from the Ministry of Education, Culture, Sports, Science and Technology of Japan (MEXT), by the 'Platform for Drug Discovery, Informatics, and Structural Life Science' grant from the MEXT (no. 12736015 to J.T.), by the 'X-ray Free Electron Laser Priority Strategy Program' grant from the MEXT (no. 12004060 to J.T.), by the Nanotechnology Platform Project (no. S-14-MS-1039 to J.T.) from the MEXT, by the Okazaki ORION project (to K.K.) and by the Research Funding for Longevity Sciences from the National Center for Geriatrics and Gerontology of Japan (no. 25-19 to K.K.).

Author information

Authors and Affiliations

Authors

Contributions

Y.K. performed the structure determination of sorLA Vps10p in complex with Aβ peptide, carried out binding experiments and the MD simulations and wrote the manuscript. M.N. and Z.N. performed the structure determination of ligand-free and propeptide-bound sorLA Vps10p. M.Y.-U. and K.K. performed the NMR experiments and analyzed the data. S.T.-N. and E.M. performed binding experiments. T.N. assisted with the structural determination and validation. J.T. conceived the experimental design, analyzed the data and wrote the manuscript. All authors contributed to the preparation of the manuscript.

Corresponding author

Correspondence to Junichi Takagi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Quantitation of the binding of pro53 peptide to sorLA Vps10p measured by the AP reporter assay.

The graph shows tracings of the typical chromogenic AP reaction observed with AP-pro53 (orange) or control AP-MycHis (blue) eluted from the sorLA Vps10p-beads. Note that gradual increase in the absorbance with the AP-MycHis is indistinguishable from that with mock sample (where no AP activity is present in the reaction mixture), indicating that the background nonspecific binding of AP protein to the beads is negligible.

Supplementary Figure 2 Sequence alignment of the Vps10p domains.

Amino acid sequences are from human sorLA (SORLA_hu, NP_003096.1), human sortilin (sort_hu, CCA66904), mouse SorCS1 (sorCS1_mo, Q9JLC4), and yeast Vps10p (Vps10p_sp, O42930). Secondary structure elements are denoted by straight (strands) or wavy (helices) lines below each sequence. Strand designation is shown above the alignment with the same color code as in Fig. 3. The L1 and L2 segments are highlighted in salmon and cyan, respectively. Cysteines are shown with a grey background with lines connecting the disulfide-bonded pair. The Gly511 mutated in a familial AD patient is marked by a red box. Propeptide cleavage sites are indicated by “//”.

Supplementary Figure 3 Close-up view of the propeptide-binding site in the propeller tunnel.

The Vps10p propeller domain (gray) is shown in surface (left) or ribbon (right) presentations, with L1, L2, and the AD-causing mutation residue Gly511 colored in salmon, cyan, and red, respectively. Propeptide ligand bound inside the tunnel is shown in CPK model in both panels.

Supplementary Figure 4 Residue-wise conformational flexibility of the sorLA Vps10p domain.

(a) Structural changes that accompany propeptide binding. The residue number is plotted against distance between mainchain Cα atoms in the ligand-free (at pH 4.5) and the propeptide-bound (at pH 6.5) forms of the sorLA Vps10p domain after structural superposition. Regions disordered in either structure are indicated by horizontal light blue bars. Note that the largest structural differences are found in L1 and the 10CC-b segments. (b) RMSF values for each residue during the MD simulation of the propeptide-bound form of sorLA Vps10p domain (see the legend to Figure 5d) are plotted similarly to the (a).

Supplementary Figure 5 Prediction of β-aggregation tendency of various sorLA Vps10p ligand peptides with the PASTA server.

The amino acid sequences of (a) the sorLA propeptide (53 residues), (b) Aβ40 (40 residues), (c) NT (13 residues), (d) HA (11 residues), and the sortilin propeptide (44 residues) were fed to the PASTA server (http://biocomp.bio.unipd.it/pasta/) and the resultant per-residue aggregation probability scores, h(k), are plotted.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 and Supplementary Tables 1–4 (PDF 2120 kb)

Supplementary Data Set 1

Uncropped gel images (PDF 259 kb)

A 10-ns MD simulation of sorLA Vps10p-propeptide complex

The movie depicts the trajectory from a representative 10-ns MD simulation of sorLA Vps10p-propeptide complex. Structure is color-coded as in Fig. 4a, where L1, L2, and bound peptide is shown in magenta, cyan, and blue, respectively (MOV 1107 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kitago, Y., Nagae, M., Nakata, Z. et al. Structural basis for amyloidogenic peptide recognition by sorLA. Nat Struct Mol Biol 22, 199–206 (2015). https://doi.org/10.1038/nsmb.2954

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2954

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