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:

Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance

An Author Correction to this article was published on 20 November 2023

This article has been updated

Abstract

PICALM is a highly validated genetic risk factor for Alzheimer's disease (AD). We found that reduced expression of PICALM in AD and murine brain endothelium correlated with amyloid-β (Aβ) pathology and cognitive impairment. Moreover, Picalm deficiency diminished Aβ clearance across the murine blood-brain barrier (BBB) and accelerated Aβ pathology in a manner that was reversible by endothelial PICALM re-expression. Using human brain endothelial monolayers, we found that PICALM regulated PICALM/clathrin-dependent internalization of Aβ bound to the low density lipoprotein receptor related protein-1, a key Aβ clearance receptor, and guided Aβ trafficking to Rab5 and Rab11, leading to Aβ endothelial transcytosis and clearance. PICALM levels and Aβ clearance were reduced in AD-derived endothelial monolayers, which was reversible by adenoviral-mediated PICALM transfer. Inducible pluripotent stem cell–derived human endothelial cells carrying the rs3851179 protective allele exhibited higher PICALM levels and enhanced Aβ clearance. Thus, PICALM regulates Aβ BBB transcytosis and clearance, which has implications for Aβ brain homeostasis and clearance therapy.

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: PICALM reductions in brain capillary endothelium in AD.
Figure 2: Diminished Aβ clearance in Picalm +/− mice.
Figure 3: Diminished Aβ clearance and accelerated pathology in APP sw/0 Picalm +/− mice.
Figure 4: Endothelial specific rescue of PICALM deficiency in the hippocampus of APP sw/0; Picalm +/− mice.
Figure 5: PICALM/clathrin-dependent endocytosis of Aβ-LRP1 complex by brain endothelial cells.
Figure 6: PICALM associates with LRP1 during Aβ transcytosis across endothelial monolayer.
Figure 7: PICALM interacts with Rab5 and Rab11 during Aβ transcytosis across endothelial monolayer.
Figure 8: Aβ transcytosis across AD-derived endothelial monolayer and iPSC-derived endothelium carrying the rs3851179 PICALM variants.

Similar content being viewed by others

Change history

References

  1. Dreyling, M.H. et al. The t(10;11)(p13;q14) in the U937 cell line results in the fusion of the AF10 gene and CALM, encoding a new member of the AP–3 clathrin assembly protein family. Proc. Natl. Acad. Sci. USA 93, 4804–4809 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Tebar, F., Bohlander, S.K. & Sorkin, A. Clathrin assembly lymphoid myeloid leukemia (CALM) protein: localization in endocytic–coated pits, interactions with clathrin, and the impact of overexpression on clathrin–mediated traffic. Mol. Biol. Cell 10, 2687–2702 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Ford, M.G. et al. Simultaneous binding of PtdIns(4,5)P2 and clathrin by AP180 in the nucleation of clathrin lattices on membranes. Science 291, 1051–1055 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Marsh, M. & McMahon, H.T. The structural era of endocytosis. Science 285, 215–220 (1999).

    Article  CAS  PubMed  Google Scholar 

  5. Sorkin, A. & von Zastrow, M. Endocytosis and signaling: intertwining molecular networks. Nat. Rev. Mol. Cell Biol. 10, 609–622 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Treusch, S. et al. Functional links between Aβ toxicity, endocytic trafficking, and Alzheimer's disease risk factors in yeast. Science 334, 1241–1245 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Miller, S.E. et al. The molecular basis for the endocytosis of small R–SNAREs by the clathrin adaptor CALM. Cell 147, 1118–1131 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Vecchi, M. et al. Nucleocytoplasmic shuttling of endocytic proteins. J. Cell Biol. 153, 1511–1517 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Harold, D. et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer's disease. Nat. Genet. 41, 1088–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lambert, J.C. et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer's disease. Nat. Genet. 41, 1094–1099 (2009).

    Article  CAS  PubMed  Google Scholar 

  11. Tanzi, R.E. The genetics of Alzheimer disease. Cold Spring Harbor Perspect. Med. 2, a006296 (2012).

    Article  Google Scholar 

  12. Lambert, J.C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Carrasquillo, M.M. et al. Late-onset Alzheimer disease genetic variants in posterior cortical atrophy and posterior AD. Neurology 82, 1455–1462 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Zlokovic, B.V. Neurovascular pathways to neurodegeneration in Alzheimer's disease and other disorders. Nat. Rev. Neurosci. 12, 723–738 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hardy, J. & Selkoe, D.J. The amyloid hypothesis of Alzheimer's disease: progress and problems on the road to therapeutics. Science 297, 353–356 (2002).

    Article  CAS  PubMed  Google Scholar 

  16. Querfurth, H.W. & LaFerla, F.M. Alzheimer's disease. N. Engl. J. Med. 362, 329–344 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Tian, Y., Chang, J.C., Fan, E.Y., Flajolet, M. & Greengard, P. Adaptor complex AP2/PICALM, through interaction with LC3, targets Alzheimer's APP–CTF for terminal degradation via autophagy. Proc. Natl. Acad. Sci. USA 110, 17071–17076 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Xiao, Q. et al. Role of phosphatidylinositol clathrin assembly lymphoid–myeloid leukemia (PICALM) in intracellular amyloid precursor protein (APP) processing and amyloid plaque pathogenesis. J. Biol. Chem. 287, 21279–21289 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kanatsu, K. et al. Decreased CALM expression reduces Aβ42 to total Aβ ratio through clathrin-mediated endocytosis of γ-secretase. Nat. Commun. 5, 3386 (2014).

    Article  PubMed  Google Scholar 

  20. Baig, S. et al. Distribution and expression of picalm in Alzheimer disease. J. Neuropathol. Exp. Neurol. 69, 1071–1077 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Parikh, I., Fardo, D.W. & Estus, S. Genetics of PICALM expression and Alzheimer's disease. PLoS ONE 9, e91242 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Shibata, M. et al. Clearance of Alzheimer's amyloid-ss(1–40) peptide from brain by LDL receptor–related protein-1 at the blood-brain barrier. J. Clin. Invest. 106, 1489–1499 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  24. Wu, Z. et al. Role of the MEOX2 homeobox gene in neurovascular dysfunction in Alzheimer disease. Nat. Med. 11, 959–965 (2005).

    Article  CAS  PubMed  Google Scholar 

  25. Bell, R.D. et al. SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells. Nat. Cell Biol. 11, 143–153 (2009).

    Article  CAS  PubMed  Google Scholar 

  26. Kanekiyo, T., Liu, C.C., Shinohara, M., Li, J. & Bu, G. LRP1 in brain vascular smooth muscle cells mediates local clearance of Alzheimer's amyloid-β. J. Neurosci. 32, 16458–16465 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sagare, A.P. et al. Pericyte loss influences Alzheimer-like neurodegeneration in mice. Nat. Commun. 4, 2932 (2013).

    Article  PubMed  Google Scholar 

  28. Ando, K. et al. Clathrin adaptor CALM/PICALM is associated with neurofibrillary tangles and is cleaved in Alzheimer's brains. Acta Neuropathol. 125, 861–878 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Hsiao, K. et al. Correlative memory deficits, Abeta elevation, and amyloid plaques in transgenic mice. Science 274, 99–102 (1996).

    Article  CAS  PubMed  Google Scholar 

  30. Zhu, D. et al. Protein S controls hypoxic/ischemic blood-brain barrier disruption through the TAM receptor Tyro3 and sphingosine 1–phosphate receptor. Blood 115, 4963–4972 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Li, Y., Lu, W., Marzolo, M.P. & Bu, G. Differential functions of members of the low density lipoprotein receptor family suggested by their distinct endocytosis rates. J. Biol. Chem. 276, 18000–18006 (2001).

    Article  CAS  PubMed  Google Scholar 

  32. Bell, R.D. et al. Apolipoprotein E controls cerebrovascular integrity via cyclophilin A. Nature 485, 512–516 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Armulik, A. et al. Pericytes regulate the blood–brain barrier. Nature 468, 557–561 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Bell, R.D. et al. Pericytes control key neurovascular functions and neuronal phenotype in the adult brain and during brain aging. Neuron 68, 409–427 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Daneman, R., Zhou, L., Kebede, A.A. & Barres, B.A. Pericytes are required for blood–brain barrier integrity during embryogenesis. Nature 468, 562–566 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Winkler, E.A., Bell, R.D. & Zlokovic, B.V. Pericyte-specific expression of PDGF beta receptor in mouse models with normal and deficient PDGF beta receptor signaling. Mol. Neurodegener. 5, 32 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Kanekiyo, T. et al. Neuronal clearance of amyloid–β by endocytic receptor LRP1. J. Neurosci. 33, 19276–19283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Stenmark, H. Rab GTPases as coordinators of vesicle traffic. Nat. Rev. Mol. Cell Biol. 10, 513–525 (2009).

    Article  CAS  PubMed  Google Scholar 

  39. Li, J. et al. Differential regulation of amyloid-β endocytic trafficking and lysosomal degradation by apolipoprotein E isoforms. J. Biol. Chem. 287, 44593–44601 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Xu, S. et al. A Rab11a-enriched subapical membrane compartment regulates a cytoskeleton-dependent transcytotic pathway in secretory epithelial cells of the lacrimal gland. J. Cell Sci. 124, 3503–3514 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Lapierre, L.A. et al. Phosphorylation of Rab11-FIP2 regulates polarity in MDCK cells. Mol. Biol. Cell 23, 2302–2318 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Yui, N. et al. Basolateral targeting and microtubule-dependent transcytosis of the aquaporin-2 water channel. Am. J. Physiol. Cell Physiol. 304, C38–C48 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Takahashi, S. et al. Rab11 regulates exocytosis of recycling vesicles at the plasma membrane. J. Cell Sci. 125, 4049–4057 (2012).

    CAS  PubMed  Google Scholar 

  44. Zeigerer, A. et al. Rab5 is necessary for the biogenesis of the endolysosomal system in vivo. Nature 485, 465–470 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Raj, T. et al. Alzheimer disease susceptibility loci: evidence for a protein network under natural selection. Am. J. Hum. Genet. 90, 720–726 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Okita, K. et al. A more efficient method to generate integration-ree human iPS cells. Nat. Methods 8, 409–412 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Mali, P. et al. RNA–guided human genome engineering via Cas9. Science 339, 823–826 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Adams, W.J. et al. Functional vascular endothelium derived from human induced pluripotent stem cells. Stem Cell Reports 1, 105–113 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Bushlin, I. et al. Clathrin assembly protein AP180 and CALM differentially control axogenesis and dendrite outgrowth in embryonic hippocampal neurons. J. Neurosci. 28, 10257–10271 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Harel, A., Mattson, M.P. & Yao, P.J. CALM, a clathrin assembly protein, influences cell surface GluR2 abundance. Neuromolecular Med. 13, 88–90 (2011).

    Article  CAS  PubMed  Google Scholar 

  51. Zlokovic, B.V., Mackic, J.B., Wang, L., McComb, J.G. & McDonough, A. Differential expression of Na,K-ATPase alpha and beta subunit isoforms at the blood-brain barrier and the choroid plexus. J. Biol. Chem. 268, 8019–8025 (1993).

    Article  CAS  PubMed  Google Scholar 

  52. Wu, Z., Hofman, F.M. & Zlokovic, B.V. A simple method for isolation and characterization of mouse brain microvascular endothelial cells. J. Neurosci. Methods 130, 53–63 (2003).

    Article  CAS  PubMed  Google Scholar 

  53. Ishikawa, Y. et al. Role of the clathrin adaptor PICALM in normal hematopoiesis and polycythemia vera pathophysiology. Haematologica 100, 439–451 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cirrito, J.R. et al. P-glycoprotein deficiency at the blood-brain barrier increases amyloid-beta deposition in an Alzheimer disease mouse model. J. Clin. Invest. 115, 3285–3290 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Grimm, D. et al. In vitro and in vivo gene therapy vector evolution via multispecies interbreeding and retargeting of adeno-associated viruses. J. Virol. 82, 5887–5911 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Mackic, J.B. et al. Human blood-brain barrier receptors for Alzheimer's amyloid-beta 1-40. Asymmetrical binding, endocytosis, and transcytosis at the apical side of brain microvascular endothelial cell monolayer. J. Clin. Invest. 102, 734–743 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Deane, R. et al. RAGE mediates amyloid-beta peptide transport across the blood-brain barrier and accumulation in brain. Nat. Med. 9, 907–913 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institutes of Health grants R37NS34467 (B.V.Z.), R37AG23084 (B.V.Z.), R01AG039452 (B.V.Z.), R01AG035355 (G.B.), R01AG027924 (G.B.) and R00NS07743 (J.K.I.), the Cure for Alzheimer Fund (B.V.Z.), the American Cancer Society (grant RSG-13-379-01-LIB to T.M.), the Rainwater Charitable Foundation (J.K.I.), the Donald E. and Delia B. Baxter Foundation (J.K.I.), and the Daiichi Sankyo Foundation of Life Science (T.S.).

AUTHORS CONTRIBUTIONS

Z.Z., A.P.S. and Q.M. designed and performed the experiments and analyzed the data. M.R.H., P.K., K.K., N.C.O., S.V.R., G.S., A.A. and T.S. performed the experiments. E.A.W. performed the pilot experiments. A.R. performed in vivo microdialysis experiments. T.K. and G.B. contributed critical materials. D.Z. generated pilot data. C.A.M., J.A.S., M.M. and T.M. provided critical materials. J.K.I. designed the iPSC study. B.V.Z. designed all of the experiments, analyzed the data and wrote the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Berislav V Zlokovic.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 PICALM expression in brain capillary endothelium in human brain and in mouse brain.

a, Double immunostaining for PICALM (red, left) and lectin–positive endothelial profiles (blue, right) in the prefrontal cortex (Brodmann 9/10; pink–merged, right) of an age–matched control Braak I and AD individual Braak VI. Bar=20 µm. b, Immunoblotting for PICALM relative to tubulin in brain microvessels and microvessel–depleted brains in 2–3 month old control mice. c–d, Immunoblotting for PICALM relative to β–actin in brain microvessels (c) and microvessel–depleted brains (d) in 15 month old APPsw/0 and age–matched littermate control mice. Mean + s.e.m., n=6 mice per group. e, Immunoblotting for PICALM relative to β–actin in primary mouse brain endothelial cells (BEC) cultured with or without Aβ40 (1 µM) for 72 hr. Mean ± s.e.m. from 3 independent primary isolates in triplicate. P<0.05 by Student’s t–test; NS, non–significant.

Supplementary Figure 2 Generation of Picalm +/– mouse, biochemistry and behavioral characterization.

Generation of Picalm +/– mouse: a, A diagram showing the targeting strategy for generating the Picalm knockout mouse. See additional Methods for more detailed information. Biochemistry: b─m, Picalm +/– mice show no changes in blood glucose, liver and renal tests and serum electrolytes compared to wild type littermate controls. The tests include: (b) Glucose; (c) Serum alkaline phosphatase; (d) Serum alanine aminotransferase (ALT); (e) Serum aspartate aminotransferase (AST); (f) Creatine phosphokinase (CPK); (g) Albumin; (h) Total protein; (i) Total bilirubin; (j) Blood urea nitrogen; (k) Creatinine level; (l) Calcium level; (m) Phosphorous level. Mean + s.e.m., n=5 mice per group. Behavioral tests: n–q, Nest construction (n), burrowing (o), novel object recognition (NOR) (p) and location (NOL) (q) tests showing no cognitive difference in 9 month old Picalm +/– mice compared to age–matched Picalm +/+ littermates. Means + s.e.m. n=12 mice per group. Statistical significance by Student’s t–test.

Supplementary Figure 3 Additional characterization of APP sw/0 Picalm +/– mice.

a–b, Representative western blots and densitometry analysis of PICALM in brain microvessels (a) and microvessel–depleted brain homogenate (b) in 3 month old APP sw/0 Picalm +/– mice and age–matched APP sw/0 Picalm +/+ littermate controls. Mean + s.e.m., n=3–4 mice per group. Statistical significance by Student’s t–test. β–actin was used as a loading control. c, Representative images of brain tissue sections stained with human Aβ–specific antibodies show accelerated Aβ deposition in the hippocampus and cortex and development of early cerebral amyloid angiopathy (CAA) in 6 month–old APP sw/0; Picalm +/– mice compared to age–matched APP sw/0; Picalm +/+ littermate controls. d, Increased Aβ load in the hippocampus and cortex in 6 month–old APP sw/0; Picalm +/– mice compared to age–matched APP sw/0; Picalm +/+ littermate controls. Mean + s.e.m., n=5–6 mice per group. Statistical significance by Student’s t–test.

Supplementary Figure 4 Endothelial-specific expression of Tdtomato after injection of the AAV-Flex-Tdtomato into the hippocampus of APP sw/0; Picalm +/–; Tie2-Cre mice, and Aβ clearance from the hippocampus in APP sw/0; Picalm +/–; Tie2-Cre mice after injection of the AAV-Flex-Picalm into the hippocampus and endothelial-specific rescue of PICALM.

a, Representative confocal images in 5 month old APP sw/0; Picalm +/–; Tie2–Cre mice show Tie2–Cre–dependent expression of Tdtomato (red) in >50% of lectin–positive endothelial vascular profiles after injection of the AAV–Flex–Tdtomato into the hippocampus. Co–injection of AAV–Synapsin–GFP shows that ~3% of hippocampal neurons express Tdtomato indicating minimal leakage. b, Coronal brain section of a 6 month old APP sw/0; Picalm +/–; Tie2–Cre mouse injected into the hippocampus with AAV–Flex (control, left) or AAV–Flex–Picalm (right) show clearance of Aβ from the hippocampus after endothelial–specific re–expression of PICALM (right; see also main Fig. 4b). Aβ immunoststaing – green; Dapi – blue. Data are representative from 3–5 independent experiments.

Supplementary Figure 5 Characterization of Picalm +/– mice and APP sw/0 Picalm +/– mice.

a–d, APP abundance relative to β–actin (a), β–secretase activity (b) sAPPβ levels (c) and γ–secretase activity determined by the production of Notch intracellular domain (NICD) fragment from Notch protein (d) were studied in the forebrain lysates from 3 month old Picalm +/– and age–matched littermate controls. e–j, Picalm +/– mice show no changes in the expression of major Aβ transporters in the brain microvessels, including Pgp (e), LRP1 by immunoblottting of microvessels (f) and double staining for LRP1 and endothelial–specific lectin (g) and RAGE (h), and no change in the levels of Aβ–degrading enzymes in the brain including neprilysin (i) and insulin–degrading enzyme (IDE) (j). k–n, APP abundance relative to β–actin (k), β–secretase activity (l) sAPPβ levels (m) and γ–secretase activity (n) were studied in the forebrain lysates from 3 month old APP sw/0 Picalm +/– and age–matched littermate APP sw/0 Picalm +/+ controls. o–s, APP sw/0 Picalm +/– mice show no changes in the expression of major Aβ transporters in the brain microvessels, including Pgp (o), LRP1 (p) and RAGE (q), and no change in expression of Aβ–degrading enzymes in the brain including neprilysin (r) and IDE (s). Mean + s.e.m., from 3–4 mice per group. Statistical significance was determined by Student’s t–test. NS, non–significant.

Supplementary Figure 6 Binding of Aβ to LRP1 and interactions of Aβ-LRP1 complexes with AP-2 and PICALM.

a, Proximity ligation assay (PLA) showing binding of Aβ40 (1 nM) to LRP1 on the cell surface of brain endothelial cells (BEC) at 4°C for 30 min. b, LRP1–Aβ40 complexes colocalize with AP–2 30 s after treatment of BEC with FAMAβ40 (250 nM). c, LRP1–Aβ42 complexes colocalize with PICALM 30 s after treatment of BEC with FAMAβ42 (250 nM) for 30 s. Dapi, nuclear staining (blue). Insets: high magnification depicting colocalizations. Bar=10 μm. d, Minimal internalization and lack of colocalization of scramble FAM–Aβ42 (green) with LRP1 (red). Bar=10 μm. Representative findings are from 3 independent primary isolates determined in triplicates (i.e., 3 different cell cultures per isolate; >20 cells in each replicate). e, Control assay for Fig. 5g shows no binding between GST and recombinant human PICALM.

Supplementary Figure 7 Apolipoprotein E and activated α2-macroglobulin do not initiate PICALM binding to LRP1.

a, PICALM does not bind to LRP1 in BEC treated with lipidated apoE3 (40 nM), apoE4 (40 nM) or activated α2–Macroglobulin (α2–M*, activated with methylamine, 0.25 nM) (left). Binding of PICALM to LRP1 in BEC treated with Aβ40 (1 nM, positive control), and loss of PICALM binding to LRP1 in BEC treated with pre–formed complexes between Aβ40 and apoE3, apoE4 or α2–M* (right). In these experiments co–immunoprecipitation of PICALM was performed by LRP1–specific antibody (IP: LRP1) within 1 min of BEC exposure to different LRP1 ligands using assay conditions as in main Fig. 5e and described in Supplementary Methods b, Control LRP1 internalization assay with Aβ. BEC were incubated with Aβ40 (230 nM) at 4°C for 15 min and then transferred to 37°C for 1 min for LRP1 internalization assay. The cell surface LRP1 was immunodetected with the N–terminus specific LRP1 antibody (N–20). c, LRP1 internalization is not triggered by apoE3, apoE4 or α2M*. BEC were incubated with Aβ40 (230 nM, control) or pre–formed complexes between Aβ40 and apoE3, apoE4 or α2M* at 4°C for 15 min and then transferred to 37 °C for 1 min for LRP1 internalization assay. Values are means + s.d. from 3 independent BEC isolates from 3 different donors using 3 replicate cultures. The number of cells counted for each culture was 20. The total number of cells in each group was 180. Statistical significance by ANOVA followed by Tukey’s posthoc test.

Supplementary Figure 8 Endothelial cell polarity of LRP1 and RAGE in brain capillaries in human brain in situ and in an in vitro blood-brain barrier model.

a, A representative confocal scanning analysis of lectin–positive endothelium (blue) and LRP1 immunodetection in capillary endothelium (red) in a control human brain. Merged: purple. Bar represents 5 μm. Chart: LRP1 relative signal intensity (red) plotted over the endothelial–specific lectin signal intensity (blue). b, A representative confocal scanning analysis of lectin–positive endothelium (blue) and RAGE immunodetection in capillary endothelium (red) in a control human brain. Merged: purple. Bar represents 5 μm. Chart: RAGE relative signal intensity (red) plotted over the endothelial–specific lectin signal intensity (blue). c, Expression of RAGE and LRP1 in the endothelial monolayer (left) and polarized expression of RAGE (red) and LRP1 (green) to the apical and basolateral side of the monolayer (right), respectively. Representative findings are from 3 independent primary isolates determined triplicates (3 different cultures per isolate; >20 cells in each replicate). d, Inhibition of synthetic human Aβ40 basolateral–to–apical transendothelial transport across an in vitro BBB monolayer by RAP (the receptor associated protein) and anti–LRP1 specific antibody, but not by anti–LRP2, anti–VLDLR and anti–LDLR antibodies, and in monolayers after BEC transfection with si.LRP1 but not control si.Scarmble. Aβ40 transcellular transport was determined by ELISA within 30 min and was corrected for the paracellular diffusion of simultaneously measured 14C–inulin basolateral–to–apical transport as described in detail in the Supplementary Methods. Aβ40 transport in the absence of potential inhibitors or competitors was arbitrarily taken as 100%. Mean + s.e.m., from 3 independent isolates determined in triplicate.

Supplementary Figure 9 PICALM regulates Aβ transcytosis in brain endothelial monolayers.

a, A representative proximity ligation assay (PLA) showing colocalization of Aβ–LRP1 complexes (green) near the apical membrane of the endothelial monolayer within 5 min of Aβ40 (1 nM) application to the basolateral side. Bar=20 µm. b, Aβ40 (1 nM) transendothelial transport across an in vitro BBB monolayer expressed as the percentage of dose (%) of Aβ transported from the basoalateral to apical chamber. Primary human brain endothelial cells have been transfected with scramble si.RNA (si.Scramble), or si.RNA targeting RAB11a or RAB11b, respectively. Mean + s.e.m., from 3 independent primary isolates determined in triplicate (3 different cultures per isolate; >20 cells in each replicate). c, Immunoblotting of different Rab proteins in Madin–Darby canine kidney (MDCK) epithelial cells and human brain endothelial cells (BEC), showing that Rab11b is mainly expressed in BEC, while Rab11a is mainly expressed in MDCK cells. Immunoblot is representative of 3 independent experiments. For each experiment we used at least 3 different samples for each condition. d, Left panel: HPLC elution profile of 125I–Aβ40 in the apical chamber after 30 min transport from the basolateral to apical chamber shows a single peak corresponding to the elution profile of Aβ40 standard indicating no degradation. Right panel: SDS–PAGE analysis showing intact 125I–Aβ40 in the apical chamber after 30 min of transendothelial transport across an in vitro BBB monolayer.

Supplementary Figure 10 PICALM and LRP1-Aβ complexes associate with EEA1, Rab5 and Rab11, but not Rab7 or LAMP1 (a lysosomal marker), in human brain endothelial cells cultured with Aβ.

a, Colocalization between PICALM (red) and EEA1 (green) in BEC cultured with and without FAM–Aβ40 (250 nM) for 2 min. Dapi, nuclear staining (blue). Insets: high magnification depicting colocalization. Bar=10 µm. Graph (on the left) – quantification of colocalized PICALM and EEA1 puncta. b, Colocalization between Aβ–LRP1 complexes visualized by the proximity ligation assay (PLA) (green) and Rab5 (red) in cultures incubated with FAM–Aβ40 (250 nM) for 2 min. c, Lack of association between PICALM (red) and a lysosomal marker LAMP1 (green) in BEC cultured without and with FAM–Aβ40 (250 nM) for 5 min. Dapi, nuclear staining (blue). Insets: high magnification. Bar=10 μm. Graph (on the left) – quantification of colocalized PICALM and LAMP1 puncta. Mean + s.d., from 3 independent primary isolates determined in triplicates (3 different cultures per each isolate; >20 cells in each replicate). Statistical significance by Student’s t-test. NS, non–significant.

Supplementary Figure 11 Association of Aβ42 with Rab5 and Rab11 and low colocalization between PICALM and Rab GTPases in the absence of Aβ in primary human endothelial cell cultures.

a–b, Association of Aβ42 with Rab 5 (a) and Rab 11 (b) in brain endothelial cells cultured with FAM–Aβ42 (250 nM) for 90 s and 5 min, respectively. Dapi, nuclear staining (blue). Insets: high magnification depicting colocalization. c–e, Control experiment showing barely detectable colocalizations between PICALM (red) and Rab5 (green) (c), Rab7 (d) and Rab11 (e) in primary human brain endothelial cells cultured with vehicle (–Aβ) for 2 and 5 min, respectively. Dapi, nuclear staining (blue). Insets: high magnification depicting colocalization. Bar: 10 μm. Mean + s.d., from 3 independent primary isolates determined in triplicate (3 different cultures per isolate; >20 cells analyzed in each replicate).

Supplementary Figure 12 Reduced PICALM expression in brain endothelial monolayers derived from Alzheimer’s disease patients.

a, A primary AD brain endothelial monolayer showing ZO–1 tight junction protein (green) and reduced expression of PICALM (red). A representative image was taken from 6 independent primary isolates determined in triplicate (3 different cultures for each isolate; >20 cells in each replicate). b, Transendothelial electrical resistance (TEER) showing normal TEER values in in vitro BBB monolayers from AD patients compared to controls. Mean + s.e.m. for 8 isolates per group studied in triplicate. c, Permeability of an in vitro BBB model derived from AD cells compared to controls to 40 kDa and 2,000 kDa FITC–Dextran. Mean + s.e.m. from 3 isolates per group in triplicate. d, A representative western blot analysis showing the expression of Flag–PICLAM, LRP1 minigene (mLRP1), and Flag–PICLAM+mLRP1 in the BBB AD monolayers after adenoviral expression of PICALM and mLRP1. Tubulin is used as loading control. Immunoblot is representative of 3 independent experiments. For each experiment we used at least 3 different samples for each condition.

Supplementary Figure 13 Diagram

The diagram showing PICALM regulates PICALM/clathrin–dependent internalization of Aβ–LRP1 complex and guides Aβ trafficking to Rab5 and Rab11 leading to Aβ endothelial transcytosis and clearance across the BBB.

Supplementary Figure 14 Original western blots.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14 and Supplementary Table 1 (PDF 6072 kb)

Supplementary Methods Checklist (PDF 423 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhao, Z., Sagare, A., Ma, Q. et al. Central role for PICALM in amyloid-β blood-brain barrier transcytosis and clearance. Nat Neurosci 18, 978–987 (2015). https://doi.org/10.1038/nn.4025

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.4025

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