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.

  • Letter
  • Published:

Kinesin-mediated axonal transport of a membrane compartment containing β-secretase and presenilin-1 requires APP

Abstract

Proteolytic processing of amyloid precursor protein (APP) generates amyloid-β peptide and has been implicated in the pathogenesis of Alzheimer's disease1. However, the normal function of APP, whether this function is related to the proteolytic processing of APP, and where this processing takes place in neurons in vivo remain unknown. We have previously shown that the axonal transport of APP in neurons is mediated by the direct binding of APP to the kinesin light chain subunit of kinesin-I, a microtubule motor protein2. Here we identify an axonal membrane compartment that contains APP, β-secretase and presenilin-1. The fast anterograde axonal transport of this compartment is mediated by APP and kinesin-I. Proteolytic processing of APP can occur in the compartment in vitro and in vivo in axons. This proteolysis generates amyloid-β and a carboxy-terminal fragment of APP, and liberates kinesin-I from the membrane. These results suggest that APP functions as a kinesin-I membrane receptor, mediating the axonal transport of β-secretase and presenilin-1, and that processing of APP to amyloid-β by secretases can occur in an axonal membrane compartment transported by kinesin-I.

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: Axonal transport of Bace, Ps1 and other kinesin-I cargoes is decreased in APP-null mutant mice.
Figure 2: APP, Bace, Ps1 and kinesin-I are present in a specific subset of vesicles in mouse sciatic nerves and corpus callosum.
Figure 3: Time-dependent generation of Aβ and release of kinesin-I and the C terminus of APP.

Similar content being viewed by others

References

  1. Selkoe, D. J. Translating cell biology into therapeutic advances in Alzheimer's disease. Nature 399, A23–A31 (1999).

    Article  ADS  CAS  Google Scholar 

  2. Kamal, A., Stokin, G. B., Yang, Z., Xia, C. & Goldstein, L. S. Axonal transport of amyloid precursor protein is mediated by direct binding to the kinesin light chain subunit of kinesin-I. Neuron 28, 449–459 (2000).

    Article  CAS  Google Scholar 

  3. Vassar et al. β-secretase cleavage of Alzheimer's amyloid precursor protein by the transmembrane aspartic protease BACE. Science 286, 735–741 (1999).

    Article  CAS  Google Scholar 

  4. De Strooper, B. et al. Deficiency of presenilin-1 inhibits the normal cleavage of amyloid precursor protein. Nature 391, 387–390 (1998).

    Article  ADS  CAS  Google Scholar 

  5. Selkoe, D. J. & Wolfe, M. S. In search of γ-secretase: presenilin at the cutting edge. Proc. Natl Acad. Sci. USA 97, 5690–5692 (2000).

    Article  ADS  CAS  Google Scholar 

  6. De Strooper, B. et al. A presenilin-1-dependent γ-secretase-like protease mediates release of Notch intracellular domain. Nature 398, 518–522 (1999).

    Article  ADS  CAS  Google Scholar 

  7. Ray, W. J. et al. Cell surface presenilin-1 participates in the γ-secretase-like proteolysis of Notch. J. Biol. Chem. 274, 36801–36807 (1999).

    Article  CAS  Google Scholar 

  8. Annaert, W. G. et al. Presenilin 1 controls γ -secretase processing of amyloid precursor protein in pre-Golgi compartments of hippocampal neurons. J. Cell Biol. 147, 277–294 (1999).

    Article  CAS  Google Scholar 

  9. Walter, J. et al. The Alzheimer's disease-associated presenilins are differentially phosphorylated proteins located predominantly within the endoplasmic reticulum. Mol. Med. 2, 673–691 (1996).

    Article  CAS  Google Scholar 

  10. Koo, E. H. & Squazzo, S. L. Evidence that production and release of amyloid β-protein involves the endocytic pathway. J. Biol. Chem. 269, 17386–17389 (1994).

    Article  CAS  Google Scholar 

  11. Thinakaran, G., Teplow, D. B., Siman, R., Greenberg, B. & Sisodia, S. S. Metabolism of the “Swedish” amyloid precursor protein variant in neuro2a (N2a) cells. Evidence that cleavage at the “β-secretase” site occurs in the Golgi apparatus. J. Biol. Chem. 271, 9390–9397 (1996).

    Article  CAS  Google Scholar 

  12. Xu, H. et al. Generation of Alzheimer β-amyloid protein in the trans-Golgi network in the apparent absence of vesicle formation. Proc. Natl Acad. Sci. USA 94, 3748–3752 (1997).

    Article  ADS  CAS  Google Scholar 

  13. Chyung, A. S. C., Greenberg, B. D., Cook, D. G., Doms, R. W. & Lee, V. M. Novel β-secretase cleavage of β-amyloid precursor protein in the endoplasmic reticulum/intermediate compartment of NT2N cells. J. Cell Biol. 138, 671–680 (1997).

    Article  CAS  Google Scholar 

  14. Skovronsky, D. M., Doms, R. W. & Lee, V. M. Detection of a novel intraneuronal pool of insoluble amyloid β protein that accumulates with time in culture. J. Cell Biol. 141, 1031–1039 (1998).

    Article  CAS  Google Scholar 

  15. Maltese, W. A. et al. Retention of the Alzheimer's amyloid precursor fragment C99 in the endoplasmic reticulum prevents formation of amyloid β-peptide. J. Biol. Chem. 276, 20267–20279 (2001).

    Article  CAS  Google Scholar 

  16. Cupers, P. et al. The discrepancy between presenilin subcellular localization and γ-secretase processing of amyloid precursor protein. J. Cell Biol. 154, 731–740 (2001).

    Article  CAS  Google Scholar 

  17. Zheng, H. et al. β-amyloid precursor protein-deficient mice show reactive gliosis and decreased locomotor activity. Cell 81, 525–531 (1995).

    Article  CAS  Google Scholar 

  18. Dawson, G. R. et al. Age-related cognitive deficits, impaired long-term potentiation and reduction in synaptic marker density in mice lacking the β-amyloid precursor protein. Neuroscience 90, 1–13 (1999).

    Article  CAS  Google Scholar 

  19. Seabrook, G. R. et al. Mechanisms contributing to the deficits in hippocampal synaptic plasticity in mice lacking amyloid precursor protein. Neuropharmacology 38, 349–359 (1999).

    Article  CAS  Google Scholar 

  20. Magara, F. et al. Genetic background changes the pattern of forebrain commissure defects in transgenic mice underexpressing the β-amyloid-precursor protein. Proc. Natl Acad. Sci. USA 96, 4656–4661 (1999).

    Article  ADS  CAS  Google Scholar 

  21. Ferreira, A., Niclas, J., Vale, R. D., Banker, G. & Kosik, K. S. Suppression of kinesin expression in cultured hippocampal neurons using antisense oligonucleotides. J. Cell Biol. 117, 595–606 (1992).

    Article  CAS  Google Scholar 

  22. Okada, Y., Yamazaki, H., Sekine-Aizawa, Y. & Hirokawa, N. The neuron-specific kinesin superfamily protein KIF1A is a unique monomeric motor for anterograde axonal transport of synaptic vesicle precursors. Cell 81, 769–780 (1995).

    Article  CAS  Google Scholar 

  23. Otsuka, A. J. et al. The C. elegans unc-104 gene encodes a putative kinesin heavy chain-like protein. Neuron 6, 113–122 (1991).

    Article  CAS  Google Scholar 

  24. Potempska, A., Styles, J., Mehta, P., Kim, K. S. & Miller, D. L. Purification and tissue level of the β-amyloid peptide precursor of rat brain. J. Biol. Chem. 266, 8464–8469 (1991).

    Article  CAS  Google Scholar 

  25. Hollenbeck, P. J. The distribution, abundance and subcellular localization of kinesin. J. Cell Biol. 108, 2335–2342 (1989).

    Article  CAS  Google Scholar 

  26. Xia, W. et al. Presenilin complexes with the C-terminal fragments of amyloid precursor protein at the sites of amyloid β-protein generation. Proc. Natl Acad. Sci. USA 97, 9299–9304 (2000).

    Article  ADS  CAS  Google Scholar 

  27. Bowman, A. B. et al. Kinesin-dependent axonal transport is mediated by the Sunday driver (SYD) protein. Cell 103, 583–594 (2000).

    Article  CAS  Google Scholar 

  28. Verhey, K. J. et al. Cargo of kinesin identified as JIP scaffolding proteins and associated signaling molecules. J. Cell Biol. 152, 959–970 (2001).

    Article  CAS  Google Scholar 

  29. Kim, S. H., Lah, J. J., Thinakaran, G., Levey, A. & Sisodia, S. S. Subcellular localization of presenilins: association with a unique membrane pool in cultured cells. Neurobiol Dis. 7, 99–117 (2000).

    Article  CAS  Google Scholar 

  30. Cao, X. & Sudhof, T. C. A transcriptively active complex of APP with Fe65 and histone acetyltransferase Tip60. Science 293, 115–120 (2001).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank G. Stokin for the Kif5b antibody, S. Brady for the 63-90 Klc antibody, S. Sisodia for providing the Ps1, Aplp1 and Aplp2 antibodies, and Merck for the APP-null mice. We thank D. Cleveland for discussions, and S. Emr for use of the deconvolution microscope. This work is supported by a grant from the National Institutes for Health. L.S.B.G. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lawrence S. B. Goldstein.

Supplementary information

Figure 1

(JPG 24.06 KB)

a, APP, BACE and PS1 accumulate proximal to a sciatic ligation. The sciatic nerves of wild-type mice were ligated unilaterally at the midpoint for 6hrs, sections were processed for immunohistochemistry as described in methods, and stained with antibodies to APP, BACE and PS1. Bar is 100 microns. b, APP, BACE, and PS1 co-localize proximal to a sciatic ligation. Wild-type mouse sciatic nerves were ligated unilaterally at the midpoint for 6hrs, sections were processed for immunohistochemistry as described in methods, co-stained with antibodies to BACE and APP or BACE and PS1. Arrows show co-localization whereas arrowheads point to nerve fibers where this is little co-localization. Bar is 2 microns.

Figure 2

(JPG 36.35 KB)

Immunofluoresence localization of kinesin-I, APP, BACE, and PS1 in mouse sciatic nerves. Cross-sections of mouse sciatic nerves were double-stained with antibodies to KLC1 and axonal marker SMI-31, KLC1 and Schwann marker S100, APP and S100, BACE and S100, or PS1 and S100, and then detected with FITC (green) or Texas-red (red) secondary antibodies; merged images in each panel are shown on the right. Arrows indicate axons, and arrowheads indicate Schwann cells. Comparable staining (not shown) of APP null mice with the APP antibody revealed a much reduced intensity of staining, which is presumably due to cross-reactivity with APLP1 and APLP2. Bar is 10 micron.

Figure 3

(JPG 22.82 KB)

APP, BACE, PS1 and kinesin-I are present in a specific subset of vesicles in mouse sciatic nerves. a, Fractionation profile of vesicles. Mouse sciatic nerves were fractionated as described in methods. 50ug each of the various fractions were analyzed by SDS-PAGE and Western blots with the indicated antibodies. b, Immunoisolation of vesicles from P3. Vesicles were immunoisolated with either no antibody (No Ab) or antibodies to KLC (63-90), KIF5B, APP (C-terminal pAb), or synaptotagmin (SYT). Equal amounts of bound (vesicle IP) and unbound vesicles were analyzed by SDS-PAGE and Western blots with the indicated antibodies. c, Immunoisolation of vesicle proteins in the presence of detergent. Vesicles were prepared and immunoisolated in the presence of 1% NP-40 detergent. The unbound supernatant (supe) and detergent vesicle IP’s were analyzed by SDS-PAGE and Western blots using the indicated antibodies.

Figure 4

(JPG 30.32 KB)

Ab is present mouse sciatic nerves. a, Ab40 and Ab42 immunoreactivity is present in mouse sciatic nerve extracts. Mouse sciatic nerve extracts were prepared from wild-type (wt) and mutant (-/-) APP mice, and analyzed on Tris-tricine gels and Western blots using the three Ab antibodies, Ab40 or Ab42 or 4G8. b, Ab40 and Ab42 antibodies recognize synthetic peptides and can co-immunoprecipitate Ab40 and Ab42 from sciatic nerve extracts. Wild-type mouse sciatic nerves were immunoprecipitated with the antibodies Ab40 or Ab42 or 4G8, and the immunoprecipitates (IP) and supernatant (supe) remaining were analyzed by Tris-tricine gels and Western blots using the indicated antibodies; Ab40 and Ab42 synthetic peptides (Calbiochem) were loaded as controls. c, Detection of Ab fragments in mouse sciatic nerves by SELDI mass spectroscopy. Spectra from SELDI analyses of immunoprecipitates of Ab from sciatic nerves of wild-type (wt) or APP null mutant mice using either Ab40 or Ab42 or 4G8 antibodies (see Table I for sequence assignments).

Table 2 Table II

Rights and permissions

Reprints and permissions

About this article

Cite this article

Kamal, A., Almenar-Queralt, A., LeBlanc, J. et al. Kinesin-mediated axonal transport of a membrane compartment containing β-secretase and presenilin-1 requires APP. Nature 414, 643–648 (2001). https://doi.org/10.1038/414643a

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/414643a

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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