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:

Pharmacological chaperones stabilize retromer to limit APP processing

Abstract

Retromer is a multiprotein complex that trafficks cargo out of endosomes. The neuronal retromer traffics the amyloid-precursor protein (APP) away from endosomes, a site where APP is cleaved into pathogenic fragments in Alzheimer's disease. Here we determined whether pharmacological chaperones can enhance retromer stability and function. First, we relied on the crystal structures of retromer proteins to help identify the 'weak link' of the complex and to complete an in silico screen of small molecules predicted to enhance retromer stability. Among the hits, an in vitro assay identified one molecule that stabilized retromer against thermal denaturation. Second, we turned to cultured hippocampal neurons, showing that this small molecule increases the levels of retromer proteins, shifts APP away from the endosome, and decreases the pathogenic processing of APP. These findings show that pharmacological chaperones can enhance the function of a multiprotein complex and may have potential therapeutic implications for neurodegenerative diseases.

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: Characterization of the weak link in retromer complex stability.
Figure 2: Identification of a retromer stabilizing pharmacological chaperone.
Figure 3: The pharmacological chaperone increases retromer levels in neurons.
Figure 4: The pharmacological chaperone decreases Aβ peptide accumulation and reduces the pathogenic pathway of APP.
Figure 5: The pharmacological chaperone shifts the endosomal localization of APP and SorL1.
Figure 6: The pharmacological chaperone specifically modulates Aβ production through the retromer pathway.

Similar content being viewed by others

Accession codes

Accessions

Protein Data Bank

References

  1. Seaman, M.N. The retromer complex—endosomal protein recycling and beyond. J. Cell Sci. 125, 4693–4702 (2012).

    Article  CAS  Google Scholar 

  2. Cullen, P.J. & Korswagen, H.C. Sorting nexins provide diversity for retromer-dependent trafficking events. Nat. Cell Biol. 14, 29–37 (2012).

    Article  CAS  Google Scholar 

  3. Seaman, M.N. Recycle your receptors with retromer. Trends Cell Biol. 15, 68–75 (2005).

    Article  CAS  Google Scholar 

  4. Burd, C. & Cullen, P.J. Retromer: a master conductor of endosome sorting. Cold Spring Harb. Perspect. Biol. 6, a016774 (2014).

    Article  Google Scholar 

  5. Small, S.A. et al. Model-guided microarray implicates the retromer complex in Alzheimer's disease. Ann. Neurol. 58, 909–919 (2005).

    Article  CAS  Google Scholar 

  6. Luo, Y. et al. Mice deficient in BACE1, the Alzheimer's β-secretase, have normal phenotype and abolished β-amyloid generation. Nat. Neurosci. 4, 231–232 (2001).

    Article  CAS  Google Scholar 

  7. Jiang, Y. et al. Alzheimer's-related endosome dysfunction in Down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc. Natl. Acad. Sci. USA 107, 1630–1635 (2010).

    Article  CAS  Google Scholar 

  8. Nikolaev, A., McLaughlin, T., O'Leary, D.D. & Tessier-Lavigne, M. APP binds DR6 to trigger axon pruning and neuron death via distinct caspases. Nature 457, 981–989 (2009).

    Article  CAS  Google Scholar 

  9. Jonsson, T. et al. A mutation in APP protects against Alzheimer's disease and age-related cognitive decline. Nature 488, 96–99 (2012).

    Article  CAS  Google Scholar 

  10. Nishitomi, K. et al. BACE1 inhibition reduces endogenous Aβ and alters APP processing in wild-type mice. J. Neurochem. 99, 1555–1563 (2006).

    Article  CAS  Google Scholar 

  11. Small, S.A. & Gandy, S. Sorting through the cell biology of Alzheimer's disease: intracellular pathways to pathogenesis. Neuron 52, 15–31 (2006).

    Article  CAS  Google Scholar 

  12. Bhalla, A. et al. The location and trafficking routes of the neuronal retromer and its role in amyloid precursor protein transport. Neurobiol. Dis. 47, 126–134 (2012).

    Article  CAS  Google Scholar 

  13. Vieira, S.I. et al. Retrieval of the Alzheimer's amyloid precursor protein from the endosome to the TGN is S655 phosphorylation state-dependent and retromer-mediated. Mol. Neurodegener. 5, 40 (2010).

    Article  Google Scholar 

  14. Muhammad, A. et al. Retromer deficiency observed in Alzheimer's disease causes hippocampal dysfunction, neurodegeneration, and Aβ accumulation. Proc. Natl. Acad. Sci. USA 105, 7327–7332 (2008).

    Article  CAS  Google Scholar 

  15. Lane, R.F. et al. Diabetes-associated SorCS1 regulates Alzheimer's amyloid-β metabolism: evidence for involvement of SorL1 and the retromer complex. J. Neurosci. 30, 13110–13115 (2010).

    Article  CAS  Google Scholar 

  16. Dodson, S.E. et al. LR11/SorLA expression is reduced in sporadic Alzheimer disease but not in familial Alzheimer disease. J. Neuropathol. Exp. Neurol. 65, 866–872 (2006).

    Article  CAS  Google Scholar 

  17. 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 

  18. Fjorback, A.W. et al. Retromer binds the FANSHY sorting motif in SorLA to regulate amyloid precursor protein sorting and processing. J. Neurosci. 32, 1467–1480 (2012).

    Article  CAS  Google Scholar 

  19. 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 

  20. Vardarajan, B.N. et al. Identification of Alzheimer disease-associated variants in genes that regulate retromer function. Neurobiol. Aging 33, 2231.e15–2231.e30 (2012).

    Article  CAS  Google Scholar 

  21. Arighi, C.N., Hartnell, L.M., Aguilar, R.C., Haft, C.R. & Bonifacino, J.S. Role of the mammalian retromer in sorting of the cation-independent mannose 6-phosphate receptor. J. Cell Biol. 165, 123–133 (2004).

    Article  CAS  Google Scholar 

  22. Seaman, M.N. Cargo-selective endosomal sorting for retrieval to the Golgi requires retromer. J. Cell Biol. 165, 111–122 (2004).

    Article  CAS  Google Scholar 

  23. Vergés, M. et al. The mammalian retromer regulates transcytosis of the polymeric immunoglobulin receptor. Nat. Cell Biol. 6, 763–769 (2004).

    Article  Google Scholar 

  24. Norwood, S.J. et al. Assembly and solution structure of the core retromer protein complex. Traffic 12, 56–71 (2011).

    Article  CAS  Google Scholar 

  25. MacLeod, D.A. et al. RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson's disease risk. Neuron 77, 425–439 (2013); erratum 77, 994 (2013).

    Article  CAS  Google Scholar 

  26. Ringe, D. & Petsko, G.A. What are pharmacological chaperones and why are they interesting? J. Biol. 8, 80 (2009).

    Article  Google Scholar 

  27. Lieberman, R.L. et al. Structure of acid β-glucosidase with pharmacological chaperone provides insight into Gaucher disease. Nat. Chem. Biol. 3, 101–107 (2007).

    Article  CAS  Google Scholar 

  28. Hierro, A. et al. Functional architecture of the retromer cargo-recognition complex. Nature 449, 1063–1067 (2007).

    Article  CAS  Google Scholar 

  29. Mattos, C. & Ringe, D. Locating and characterizing binding sites on proteins. Nat. Biotechnol. 14, 595–599 (1996).

    Article  CAS  Google Scholar 

  30. Collins, B.M., Skinner, C.F., Watson, P.J., Seaman, M.N. & Owen, D.J. Vps29 has a phosphoesterase fold that acts as a protein interaction scaffold for retromer assembly. Nat. Struct. Mol. Biol. 12, 594–602 (2005).

    Article  CAS  Google Scholar 

  31. Swarbrick, J.D. et al. VPS29 is not an active metallo-phosphatase but is a rigid scaffold required for retromer interaction with accessory proteins. PLoS ONE 6, e20420 (2011).

    Article  CAS  Google Scholar 

  32. Laurie, A.T. & Jackson, R.M. Q-SiteFinder: an energy-based method for the prediction of protein-ligand binding sites. Bioinformatics 21, 1908–1916 (2005).

    Article  CAS  Google Scholar 

  33. Collins, B.M. et al. Structure of Vps26B and mapping of its interaction with the retromer protein complex. Traffic 9, 366–379 (2008).

    Article  CAS  Google Scholar 

  34. Shi, H., Rojas, R., Bonifacino, J.S. & Hurley, J.H. The retromer subunit Vps26 has an arrestin fold and binds Vps35 through its C-terminal domain. Nat. Struct. Mol. Biol. 13, 540–548 (2006).

    Article  CAS  Google Scholar 

  35. Landon, M.R., Lancia, D.R. Jr., Yu, J., Thiel, S.C. & Vajda, S. Identification of hot spots within druggable binding regions by computational solvent mapping of proteins. J. Med. Chem. 50, 1231–1240 (2007).

    Article  CAS  Google Scholar 

  36. Mucke, L. et al. High-level neuronal expression of Aβ 1–42 in wild-type human amyloid protein precursor transgenic mice: synaptotoxicity without plaque formation. J. Neurosci. 20, 4050–4058 (2000).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the US National Institute on Aging/US National Institutes of Health (NIH) grants AG025161 and AG08702, The Alzheimer's Association, Developmental Therapeutics Program of the National Cancer Institute, Medkoo Biosciences, The Fidelity Biosciences Research Initiative and give special thanks to S. Weninger for advice and encouragement. We also thank The McKnight Endowment for Neuroscience, the Ellison Medical Foundation, and the Gottlieb Family Foundation.

Author information

Authors and Affiliations

Authors

Contributions

V.J.M. identified the retromer stabilizing site and carried out the in silico screens and designed and performed the in vitro retromer chaperone characterization; D.E.B. designed, coordinated and performed the in vivo retromer chaperone characterization; S.S. assisted with the immunohistochemical staining and genetic recombination in neurons; C.V. assisted with neuronal cultures; M.R.A. assisted with the in vitro thermal studies; V.M.P. assisted with the APP fragment analysis; R.T.S. helped with Aβ measurements; and G.A.P., D.R. and S.A.S. conceived of and supervised the studies and wrote the manuscript.

Corresponding authors

Correspondence to Gregory A Petsko, Dagmar Ringe or Scott A Small.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Results, Supplementary Table 1, Supplementary Figures 1–7 and MedKoo certificate. (PDF 1821 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mecozzi, V., Berman, D., Simoes, S. et al. Pharmacological chaperones stabilize retromer to limit APP processing. Nat Chem Biol 10, 443–449 (2014). https://doi.org/10.1038/nchembio.1508

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nchembio.1508

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research