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:

ER–lysosome contacts enable cholesterol sensing by mTORC1 and drive aberrant growth signalling in Niemann–Pick type C

Abstract

Cholesterol activates the master growth regulator, mTORC1 kinase, by promoting its recruitment to the surface of lysosomes by the Rag guanosine triphosphatases (GTPases). The mechanisms that regulate lysosomal cholesterol content to enable mTORC1 signalling are unknown. Here, we show that oxysterol binding protein (OSBP) and its anchors at the endoplasmic reticulum (ER), VAPA and VAPB, deliver cholesterol across ER–lysosome contacts to activate mTORC1. In cells lacking OSBP, but not other VAP-interacting cholesterol carriers, the recruitment of mTORC1 by the Rag GTPases is inhibited owing to impaired transport of cholesterol to lysosomes. By contrast, OSBP-mediated cholesterol trafficking drives constitutive mTORC1 activation in a disease model caused by the loss of the lysosomal cholesterol transporter, Niemann–Pick C1 (NPC1). Chemical and genetic inactivation of OSBP suppresses aberrant mTORC1 signalling and restores autophagic function in cellular models of Niemann–Pick type C (NPC). Thus, ER–lysosome contacts are signalling hubs that enable cholesterol sensing by mTORC1, and targeting the sterol-transfer activity of these signalling hubs could be beneficial in patients with NPC.

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

Fig. 1: Cholesterol accumulates at the limiting membrane of NPC1-deficient lysosomes in an OSBP-dependent manner.
Fig. 2: OSBP is required for cholesterol-dependent lysosomal recruitment and activation of mTORC1.
Fig. 3: OSBP regulates mTORC1 upstream of the Rag GTPases.
Fig. 4: Both the lipid-transporting and lysosome-targeting functions of OSBP are required for mTORC1 activation by cholesterol.
Fig. 5: Excess lysosomal PtdIns4P after OSBP inhibition does not cause mTORC1 inhibition.
Fig. 6: VAPA and VAPB, the ER anchors for OSBP, are essential for cholesterol-dependent mTORC1 activation.
Fig. 7: OSBP inhibition abrogates aberrant mTORC1 signalling in NPC1-deficient cells.
Fig. 8: OSBP inhibition restores autophagic function in NPC1-deficient cells.

Similar content being viewed by others

Data availability

Mass spectrometry data that support the findings of this study are provided in Supplementary Table 1 and have been deposited in ProteomeXchange with accession number PXD014733. Statistics source data for Figs. 14 and 68 and Supplementary Figs. 16 are provided in Supplementary Table 2. All data supporting the findings of this study are available from the corresponding author on reasonable request.

References

  1. Gatta, A. T. & Levine, T. P. Piecing together the patchwork of contact sites. Trends Cell Biol. 27, 214–229 (2017).

    Article  CAS  Google Scholar 

  2. Phillips, M. J. & Voeltz, G. K. Structure and function of ER membrane contact sites with other organelles. Nat. Rev. Mol. Cell Biol. 17, 69–82 (2016).

    Article  CAS  Google Scholar 

  3. Thelen, A. M. & Zoncu, R. Emerging roles for the lysosome in lipid metabolism. Trends Cell Biol. 27, 833–850 (2017).

    Article  CAS  Google Scholar 

  4. Kim, E., Goraksha-Hicks, P., Li, L., Neufeld, T. P. & Guan, K. L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat. Cell Biol. 10, 935–945 (2008).

    Article  CAS  Google Scholar 

  5. Sancak, Y. et al. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141, 290–303 (2010).

    Article  CAS  Google Scholar 

  6. Sancak, Y. et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320, 1496–1501 (2008).

    Article  CAS  Google Scholar 

  7. Castellano, B. M. et al. Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science 355, 1306–1311 (2017).

    Article  CAS  Google Scholar 

  8. Jung, J., Genau, H. M. & Behrends, C. Amino acid-dependent mTORC1 regulation by the lysosomal membrane protein SLC38A9. Mol. Cell. Biol. 35, 2479–2494 (2015).

    Article  CAS  Google Scholar 

  9. Rebsamen, M. et al. SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1. Nature 519, 477–481 (2015).

    Article  CAS  Google Scholar 

  10. Wang, S. et al. Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1. Science 347, 188–194 (2015).

    Article  CAS  Google Scholar 

  11. Gong, X. et al. Structural insights into the Niemann-Pick C1 (NPC1)-mediated cholesterol transfer and ebola infection. Cell 165, 1467–1478 (2016).

    Article  CAS  Google Scholar 

  12. Kwon, H. J. et al. Structure of N-terminal domain of NPC1 reveals distinct subdomains for binding and transfer of cholesterol. Cell 137, 1213–1224 (2009).

    Article  Google Scholar 

  13. Li, X., Saha, P., Li, J., Blobel, G. & Pfeffer, S. R. Clues to the mechanism of cholesterol transfer from the structure of NPC1 middle lumenal domain bound to NPC2. Proc. Natl Acad. Sci. USA 113, 10079–10084 (2016).

    Article  CAS  Google Scholar 

  14. Maxfield, F. R. Role of endosomes and lysosomes in human disease. Cold Spring Harb. Perspect. Biol. 6, a016931 (2014).

    Article  Google Scholar 

  15. Radhakrishnan, A., Goldstein, J. L., McDonald, J. G. & Brown, M. S. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: a delicate balance. Cell Metab. 8, 512–521 (2008).

    Article  CAS  Google Scholar 

  16. Wilhelm, L. P. et al. STARD3 mediates endoplasmic reticulum-to-endosome cholesterol transport at membrane contact sites. EMBO J. 36, 1412–1433 (2017).

    Article  CAS  Google Scholar 

  17. Im, Y. J., Raychaudhuri, S., Prinz, W. A. & Hurley, J. H. Structural mechanism for sterol sensing and transport by OSBP-related proteins. Nature 437, 154–158 (2005).

    Article  CAS  Google Scholar 

  18. Tong, J., Yang, H., Yang, H., Eom, S. H. & Im, Y. J. Structure of Osh3 reveals a conserved mode of phosphoinositide binding in oxysterol-binding proteins. Structure 21, 1203–1213 (2013).

    Article  CAS  Google Scholar 

  19. Mesmin, B., Antonny, B. & Drin, G. Insights into the mechanisms of sterol transport between organelles. Cell. Mol. Life Sci. 70, 3405–3421 (2013).

    Article  CAS  Google Scholar 

  20. Mesmin, B. et al. Sterol transfer, PI4P consumption, and control of membrane lipid order by endogenous OSBP. EMBO J. 36, 3156–3174 (2017).

    Article  CAS  Google Scholar 

  21. Goto, A., Charman, M. & Ridgway, N. D. Oxysterol-binding protein activation at endoplasmic reticulum-Golgi contact sites reorganizes phosphatidylinositol 4-phosphate pools. J. Biol. Chem. 291, 1336–1347 (2016).

    Article  CAS  Google Scholar 

  22. Dong, R. et al. Endosome-ER contacts control actin nucleation and retromer function through VAP-dependent regulation of PI4P. Cell 166, 408–423 (2016).

    Article  CAS  Google Scholar 

  23. Rowland, A. A., Chitwood, P. J., Phillips, M. J. & Voeltz, G. K. ER contact sites define the position and timing of endosome fission. Cell 159, 1027–1041 (2014).

    Article  CAS  Google Scholar 

  24. Rocha, N. et al. Cholesterol sensor ORP1L contacts the ER protein VAP to control Rab7-RILP-p150 Glued and late endosome positioning. J. Cell Biol. 185, 1209–1225 (2009).

    Article  CAS  Google Scholar 

  25. Willett, R. et al. TFEB regulates lysosomal positioning by modulating TMEM55B expression and JIP4 recruitment to lysosomes. Nat. Commun. 8, 1580 (2017).

    Article  Google Scholar 

  26. Lebrand, C. et al. Late endosome motility depends on lipids via the small GTPase Rab7. EMBO J. 21, 1289–1300 (2002).

    Article  CAS  Google Scholar 

  27. Maekawa, M. & Fairn, G. D. Complementary probes reveal that phosphatidylserine is required for the proper transbilayer distribution of cholesterol. J. Cell. Sci. 128, 1422–1433 (2015).

    Article  CAS  Google Scholar 

  28. Loftus, S. K. et al. Murine model of Niemann-Pick C disease: mutation in a cholesterol homeostasis gene. Science 277, 232–235 (1997).

    Article  CAS  Google Scholar 

  29. Sarkar, S. et al. Impaired autophagy in the lipid-storage disorder Niemann-Pick type C1 disease. Cell Rep. 5, 1302–1315 (2013).

    Article  CAS  Google Scholar 

  30. Lu, F. et al. Identification of NPC1 as the target of U18666A, an inhibitor of lysosomal cholesterol export and Ebola infection. eLife 4, e12177 (2015).

  31. Zoncu, R. et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H+-ATPase. Science 334, 678–683 (2011).

    Article  CAS  Google Scholar 

  32. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    Article  CAS  Google Scholar 

  33. Burgett, A. W. et al. Natural products reveal cancer cell dependence on oxysterol-binding proteins. Nat. Chem. Biol. 7, 639–647 (2011).

    Article  CAS  Google Scholar 

  34. Levine, T. P. & Munro, S. The pleckstrin homology domain of oxysterol-binding protein recognises a determinant specific to Golgi membranes. Curr. Biol. 8, 729–739 (1998).

    Article  CAS  Google Scholar 

  35. Ridgway, N. D., Dawson, P. A., Ho, Y. K., Brown, M. S. & Goldstein, J. L. Translocation of oxysterol binding protein to Golgi apparatus triggered by ligand binding. J. Cell Biol. 116, 307–319 (1992).

    Article  CAS  Google Scholar 

  36. Su, M. Y. et al. Hybrid structure of the RagA/C-Ragulator mTORC1 activation complex. Mol. Cell 68, 835–846 (2017).

    Article  CAS  Google Scholar 

  37. Levin, R. et al. Multiphasic dynamics of phosphatidylinositol 4-phosphate during phagocytosis. Mol. Biol. Cell 28, 128–140 (2017).

    Article  CAS  Google Scholar 

  38. Mesmin, B. et al. A four-step cycle driven by PI(4)P hydrolysis directs sterol/PI(4)P exchange by the ER-Golgi tether OSBP. Cell 155, 830–843 (2013).

    Article  CAS  Google Scholar 

  39. Hammond, G. R., Machner, M. P. & Balla, T. A novel probe for phosphatidylinositol 4-phosphate reveals multiple pools beyond the Golgi. J. Cell Biol. 205, 113–126 (2014).

    Article  CAS  Google Scholar 

  40. Perry, R. J. & Ridgway, N. D. Oxysterol-binding protein and vesicle-associated membrane protein-associated protein are required for sterol-dependent activation of the ceramide transport protein. Mol. Biol. Cell 17, 2604–2616 (2006).

    Article  CAS  Google Scholar 

  41. Bar-Peled, L. et al. A tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1. Science 340, 1100–1106 (2013).

    Article  CAS  Google Scholar 

  42. Panchaud, N., Peli-Gulli, M. P. & De Virgilio, C. Amino acid deprivation inhibits TORC1 through a GTPase-activating protein complex for the Rag family GTPase Gtr1. Sci. Signal. 6, ra42 (2013).

    Article  Google Scholar 

  43. Wolfson, R. L. et al. KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1. Nature 543, 438–442 (2017).

    Article  CAS  Google Scholar 

  44. Menon, S. et al. Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome. Cell 156, 771–785 (2014).

    Article  CAS  Google Scholar 

  45. Zhao, K. & Ridgway, N. D. Oxysterol-binding protein-related protein 1L regulates cholesterol egress from the endo-lysosomal system. Cell Rep. 19, 1807–1818 (2017).

    Article  CAS  Google Scholar 

  46. Hoyer, M. J. et al. A novel class of ER membrane proteins regulates er-associated endosome fission. Cell 175, 254–265 (2018).

    Article  CAS  Google Scholar 

  47. Ordonez, M. P. et al. Disruption and therapeutic rescue of autophagy in a human neuronal model of Niemann Pick type C1. Hum. Mol. Genet. 21, 2651–2662 (2012).

    Article  CAS  Google Scholar 

  48. Pacheco, C. D., Kunkel, R. & Lieberman, A. P. Autophagy in Niemann-Pick C disease is dependent upon Beclin-1 and responsive to lipid trafficking defects. Hum. Mol. Genet. 16, 1495–1503 (2007).

    Article  CAS  Google Scholar 

  49. Kaizuka, T. et al. An autophagic flux probe that releases an internal control. Mol. Cell 64, 835–849 (2016).

    Article  CAS  Google Scholar 

  50. Bjorkoy, G. et al. Monitoring autophagic degradation of p62/SQSTM1. Methods Enzymol. 452, 181–197 (2009).

    Article  Google Scholar 

  51. Chung, J. et al. PI4P/phosphatidylserine countertransport at ORP5- and ORP8-mediated ER–plasma membrane contacts. Science 349, 428–432 (2015).

    Article  CAS  Google Scholar 

  52. Du, X. et al. A role for oxysterol-binding protein-related protein 5 in endosomal cholesterol trafficking. J. Cell Biol. 192, 121–135 (2011).

    Article  CAS  Google Scholar 

  53. Du, X. et al. Oxysterol-binding protein-related protein 5 (ORP5) promotes cell proliferation by activation of mTORC1 signaling. J. Biol. Chem. 293, 3806–3818 (2018).

    Article  CAS  Google Scholar 

  54. Mousley, C. J. et al. A sterol-binding protein integrates endosomal lipid metabolism with TOR signaling and nitrogen sensing. Cell 148, 702–715 (2012).

    Article  CAS  Google Scholar 

  55. Murley, A. et al. Sterol transporters at membrane contact sites regulate TORC1 and TORC2 signaling. J. Cell Biol. 216, 2679–2689 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Ballabio, A. & Gieselmann, V. Lysosomal disorders: from storage to cellular damage. Biochim. Biophys. Acta 1793, 684–696 (2009).

    Article  CAS  Google Scholar 

  57. Infante, R. E. et al. NPC2 facilitates bidirectional transfer of cholesterol between NPC1 and lipid bilayers, a step in cholesterol egress from lysosomes. Proc. Natl Acad. Sci. USA 105, 15287–15292 (2008).

    Article  CAS  Google Scholar 

  58. Infante, R. E. & Radhakrishnan, A. Continuous transport of a small fraction of plasma membrane cholesterol to endoplasmic reticulum regulates total cellular cholesterol. eLife 6, e25466 (2017).

  59. Toulmay, A. & Prinz, W. A. Direct imaging reveals stable, micrometer-scale lipid domains that segregate proteins in live cells. J. Cell Biol. 202, 35–44 (2013).

    Article  CAS  Google Scholar 

  60. Tsuji, T. et al. Niemann-Pick type C proteins promote microautophagy by expanding raft-like membrane domains in the yeast vacuole. eLife 6, e25960 (2017).

  61. Maetzel, D. et al. Genetic and chemical correction of cholesterol accumulation and impaired autophagy in hepatic and neural cells derived from Niemann-Pick Type C patient-specific iPS cells. Stem Cell Rep. 2, 866–880 (2014).

    Article  CAS  Google Scholar 

  62. Fraldi, A. et al. Lysosomal fusion and SNARE function are impaired by cholesterol accumulation in lysosomal storage disorders. EMBO J. 29, 3607–3620 (2010).

    Article  CAS  Google Scholar 

  63. Settembre, C. et al. A block of autophagy in lysosomal storage disorders. Hum. Mol. Genet. 17, 119–129 (2008).

    Article  CAS  Google Scholar 

  64. Goldstein, J. L., Basu, S. K. & Brown, M. S. Receptor-mediated endocytosis of low-density lipoprotein in cultured cells. Methods Enzymol. 98, 241–260 (1983).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank all of the members of the Zoncu laboratory for their insights; R. Perera and M. Rape for reading the manuscript; M. D. Shair (Harvard University) for providing us with the OSW-1 compound; G. Fairn (University of Toronto) for the D4H cholesterol probe cDNA; G. Hammond (University of Pittsburgh) and S. Grinstein (Hospital for Sick Children) for the P4M reporter and the scSac1 catalytic domain cDNAs; H. Arai and N. Kono (University of Tokyo) for the anti-OSBP rabbit polyclonal antibody. C.-Y.L. is the recipient of an American Heart Association postdoctoral fellowship (18POST34070059). This work was supported by the NIH Director’s New Innovator Award (1DP2CA195761-01), the Pew–Stewart Scholarship for Cancer Research, the Damon Runyon-Rachleff Innovation Award, the Edward Mallinckrodt Jr. Foundation Grant and the Packer Wentz Endowment (to R.Z.), a National Institutes of Health grant (R01 HL067773; to D.S.O).

Author information

Authors and Affiliations

Authors

Contributions

C.-Y.L. and R.Z. conceived the study. C.-Y.L., O.B.D., H.R.S. and R.Z. designed experiments. C.-Y.L., H.R.S., O.B.D. and J.Z. performed experiments. X.J., C.A.B. and J.L.C. performed mass spectrometry measurements. D.K.N. and D.S.O. provided advice on experimental design and data analysis. C.-Y.L. and R.Z. wrote the manuscript. All of the authors reviewed and edited the manuscript.

Corresponding author

Correspondence to Roberto Zoncu.

Ethics declarations

Competing interests

R.Z. is a co-founder, consultant and stockholder of Frontier Medicines Corporation. D.K.N. is a co-founder, stock-holder and scientific adviser for Artris Therapeutics and Frontier Medicines Corporation.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 OSBP-delivered cholesterol accumulates on the surface of NPC1-null lysosomes.

a, Coomassie blue staining of recombinant GST-tagged D4H*-mCherry. b, NPC1-deleted HEK-293T cells were treated with 1 μM LysoSensor Green DND-189 for 30 min, fixed, permeabilized with liquid nitrogen (LN2) where indicated, and subjected to cholesterol labeling by GST-D4H*-mCherry and filipin. Scale bar, 10 μm. c, Accumulation of D4H*-mCherry on the plasma membrane of non-permeabilized cells. Control HEK-293T cells were subjected to cholesterol labeling by GST-D4H*-mCherry and filipin, and stained for endogenous LAMP2. Scale bar, 10 μm. d, NPC1 knockout MEFs display cholesterol accumulation at the lysosomal membrane, in addition to the lysosomal lumen. Wild type and NPC1 knockout MEFs were fixed, breached with LN2, subjected to cholesterol labeling by GST-D4H*-mCherry and filipin, and stained for endogenous LAMP1. Scale bar, 10 µm. e, NPC1 inhibition via U18666A treatment induces cholesterol deposition at the limiting membrane as well as in the lumen of lysosomes. Control human fibroblasts were treated with U18666A (5 µg/ml) for 6h and subjected to cholesterol labeling and LAMP2 staining. Scale bar, 10 µm. f, Quantitation of co-localization of D4H*-mCherry with filipin-labeled cholesterol deposits in control human fibroblasts treated with either DMSO or U18666A (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 10 fields of view per treatment; n represents cell number: DMSO (n = 11), U18666A (n = 10), two-tailed, unpaired t-test. ****P = 5.35648 x 10-13 vs. U18666A). g, Depletion of OSBP significantly suppresses the accumulation of lysosomal membrane cholesterol in human NPC1 patient-derived fibroblasts. Human NPC1 patient-derived fibroblasts depleted of ORPs via shRNA were subjected to cholesterol labeling by GST-D4H*-mCherry and filipin, then stained for endogenous LAMP2. Scale bar, 10 µm. See quantitation in Fig. 1f. Experiments in ad, and g performed independently two times. Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 2 OSBP inactivation via OSW-1 treatment abrogates the accumulation of lysosomal membrane cholesterol in NPC1-deficient cells.

a, OSW-1 treatment in human NPC1 patient-derived fibroblasts abolishes cholesterol deposition on the lysosomal limiting membrane. Human patient cells were treated with either DMSO or 20 nM OSW-1 for 8h, fixed, permeabilized with LN2, subjected to cholesterol labeling by GST-D4H*-mCherry and filipin, and stained for endogenous LAMP2. Scale bar, 10 μm. b, Quantitation of co-localization of D4H*-mCherry with filipin-labeled cholesterol deposits in human NPC1 patient-derived cells treated with either DMSO or OSW-1 (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 10 fields of view per treatment; n represents cell number: DMSO (n = 11), OSW-1 (n = 10), two-tailed, unpaired t-test. ****P = 1.07121 x 10-12 vs. DMSO). c, OSW-1 treatment in NPC1 knockout MEFs significantly reduces cholesterol levels on the lysosomal limiting membrane. NPC1 knockout MEFs were treated with either DMSO or 20 nM OSW-1 for 8h, fixed, permeabilized with LN2, subjected to cholesterol labeling by GST-D4H*-mCherry and filipin, and stained for endogenous LAMP1. Scale bar, 10 μm. d, Quantitation of co-localization of D4H*-mCherry with filipin-labeled cholesterol deposits in NPC1 knockout MEFs treated with either DMSO or OSW-1 (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 10 fields of view per treatment; n represents cell number: DMSO (n = 30), OSW-1 (n = 34), two-tailed, unpaired t-test. ****P = 8.39052 x 10-15 vs. DMSO). e, OSW-1 treatment in NPC1-null cells significantly removes cholesterol levels on the lysosomal limiting membrane. NPC1-deleted HEK-293T cells expressing FLAG-GFP-Tmem192 were treated with either DMSO or 20 nM OSW-1 for 8h, fixed, permeabilized with LN2, and subjected to cholesterol labeling by GST-D4H*-mCherry and filipin. Scale bar, 10 μm. f, Quantitation of co-localization of D4H*-mCherry with filipin-labeled cholesterol deposits in NPC1-null cells treated with either DMSO or OSW-1 (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 10 fields of view per treatment; n represents cell number: DMSO (n = 66), OSW-1 (n = 73), two-tailed, unpaired t-test. ****P = 8.69018 x 10-11 vs. DMSO). Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 3 Lysosomal localization of a subset of OSBP in cells.

a, A pool of OSBP is localized to LAMP2-positive lysosomes. (left) HEK-293A cells stably expressing wild type or the indicated variants of FLAG-GFP-OSBP were subjected to immunofluorescence for endogenous LAMP2. Scale bar, 10 µm. (middle) Quantitation of the fraction of LAMP2-positive lysosomes that are positive for OSBP (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 7 fields of view per genotype; n represents cell number: WT (n = 13), ΔELSK (n = 11), ΔPH (n =13), ANOVA with Dunnett’s multiple comparison test. ****Adjusted P = 0.0001 vs. WT cells). (right) Expression levels of the wild type FLAG-GFP-OSBP in relative to those of the endogenous OSBP in HEK-293A cells. Cells were harvested and subjected to immunoblot analysis. Experiment repeated two times. b, A pool of OSBP is localized to LAMP2-positive lysosomes where mTOR and p18 are present. (left) HEK-293A cells stably expressing either wild type or PH domain-deleted FLAG-GFP-OSBP were subjected to immunofluorescence for endogenous LAMP2, mTOR and p18, respectively. Scale bar, 10 µm. (right) Quantitation of the fraction of LAMP2-, mTOR-, p18-labeled vesicles that are positive for OSBP (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 7 fields of view per genotype; n represents cell number: LAMP2 for WT (n = 9), ΔPH (n = 12), two-tailed, unpaired t-test. ****P = 1.07095 x 10-6 vs. WT; mTOR for WT (n = 10), ΔPH (n = 12), two-tailed, unpaired t-test. ****P = 3.89303 x 10-7 vs. WT; p18 for WT (n = 11), ΔPH (n = 9), two-tailed, unpaired t-test. ****P = 1.54976 x 10-7 vs. WT). c, OSW-1 treatment triggers clustering of OSBP on lysosomes. (left) HEK-293A cells stably expressing FLAG-GFP-OSBP were treated with either DMSO or with the OSBP inhibitor OSW-1 (10 nM) for 2h and subjected to immunofluorescence for endogenous LAMP2. Scale bar, 10 µm. (right) Quantitation of the fraction of LAMP2-positive lysosomes that are positive for OSBP (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 7 fields of view per treatment; n represents cell number: DMSO (n = 11), OSW-1 (n = 10), two-tailed, unpaired t-test. ****P = 3.47655 x 10-8 vs. DMSO). d, Depletion of VAPA and VAPB increases the lysosomal localization of OSBP. (left) Cells expressing the indicated shRNAs were subjected to immunofluorescence for endogenous OSBP and LAMP2. Scale bar, 10 µm. (right) Quantitation of the fraction of LAMP2-positive lysosomes that are positive for OSBP (box plots showing the min, 1st quartile, median, 3rd quartile, and max, 10 fields of view per genotype; n represents cell number: shLuc (n = 18), shVAPA/B (n = 17), two-tailed, unpaired t-test. ****P = 9.24193 x 10-11 vs. shLuc). (bottom) knockdown of VAPA/B was validated by immunoblotting. Experiments repeated two times. Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 4 OSBP inhibition causes cholesterol sequestration in the ER and its depletion from lysosomes.

a, Western blot analysis of immuno-isolated lysosomes used for mass spectrometry-based measurement of lysosomal cholesterol content. Lysosomes were purified via anti-HA magnetic beads from cells expressing Tmem192-mRFP-3xHA that have been previously treated with either DMSO or 20 nM OSW-1 for 8h. Samples for lysosomes and post-nuclear supernatant (PNS) were immunoblotted for the indicated proteins. See lysosomal cholesterol measurement in Fig. 2a. Experiments repeated two times. b, (left) Depletion of OSBP induces a significant increase in cholesteryl esters but not total cholesterol. HEK-293T cells were depleted of OSBP by doxycycline-induced shRNA, and total lipid extracts were subjected to mass spectrometry analysis. ‘– Dox’ ‘+ Dox’ indicate control and OSBP-depleted cells (mean ± s.d., n = 5 biologically independent samples per group, two-tailed, unpaired t-test; total cholesterol, ****P = 0.996460171; cholesteryl esters, ****P = 3.69304 x 10-5; cholesteryl oleate, ****P = 0.000215477). (right) OSBP knockdown efficiency was validated by immunoblotting. Experiments repeated two times. c, Lipidomic profiling of HEK-293T cells presented in heat map indicating the relative levels of all the measured lipids normalized to their respective controls (– Dox group as control, n = 5 biologically independent samples per group). d and e, OSBP inactivation induces cholesterol accumulation in the ER. Cells were depleted of OSBP via shRNA (d) or treated with either DMSO or 20 nM OSW-1 for 8h (e), fixed and stained with filipin to visualize intracellular cholesterol distribution. Three independent fields of cells per condition are shown. Scale bar, 10 μm. Experiments repeated two times. Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 5 Exogenously supplied cholesterol is transported to the lysosome via an OSBP-dependent route.

a, HEK-293T cells stably expressing Tmem192-mRFP-3xHA (T192-mRFP) were treated with DMSO or 20 nM OSW-1 for 2h, labeled with TopFluor-cholesterol (TF-Chol) complexed with MCD, chased at the indicated times, fixed, and imaged on a spinning disk confocal microscope. Scale bar, 10 µm. b, Quantitation of co-localization of TF-Chol with T192-mRFP-labeled lysosomes in cells treated with DMSO or OSW-1 (box plots showing the min, 1st quartile, median, 3rd quartile, and max, data expressed as the fraction of T192-mRFP-labeled lysosomes that are positive for TF-Chol, 10 fields of view for each treatment/time point; n represents cell number: DMSO_18 min (n = 106), DMSO_30 min (n = 85), DMSO_60 min (n = 78), DMSO_90 min (n = 77), DMSO_180 min (n = 87), OSW-1_18 min (n = 100), OSW-1_30 min (n = 82), OSW-1_60 min (n = 85), OSW-1_90 min (n = 83), and OSW-1_180 min (n = 107). Total peak area derived from the Area Under Curve (AUC) analysis: DMSO, 60.94 ± 3.111, OSW-1, 14.86 ± 1.849). Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 6 OSBP regulates cholesterol-induced lysosomal recruitment and activation of mTORC1.

a, OSBP drives sterol-dependent lysosomal recruitment of mTORC1. HEK-293T cells expressing the indicated shRNAs were subjected to cholesterol depletion, followed by restimulation with MCD:cholesterol where indicated, and subjected to immunofluorescence for endogenous mTOR and LAMP2. Representative confocal microscopic images are shown. Scale bar, 10 μm. See quantitation in Fig. 2c. Experiments repeated two times. b, OSW-1 abolishes sterol-induced lysosomal recruitment of mTORC1. HEK-293T cells were subjected to cholesterol starvation and restimulation in the presence of DMSO or OSW-1 at the indicated concentrations and subjected to immunofluorescence for endogenous mTOR and LAMP2. Scale bar, 10 µm. c, Quantitation of co-localization between mTOR and LAMP2-positive lysosomes in cells subjected to the indicated treatments (mean ± s.d., 10 fields of view per treatment; n represents cell number: – chol (n = 150), + chol (n = 123), 1 nM OSW-1 + chol (n = 155), 10 nM OSW-1 + chol (n = 163), 20 nM OSW-1 + chol (n = 172), 50 nM OSW-1 + chol (n = 180), ANOVA with Dunnett’s multiple comparison test. **** Adjusted P = 0.0001 vs. + chol). d, Depletion of OSBP inhibits mTORC1 lysosomal localization in cells under full media conditions. HEK-293T cells depleted of OSBP were subjected to immunofluorescence for endogenous mTOR and LAMP2. Representative confocal microscopic images are shown. Scale bar, 10 μm. e, Quantitation of co-localization between mTOR and LAMP2-positive lysosomes in cells cultured in full media (mean ± s.d., 10 fields of view per genotype; n represents cell number: shLuc (n = 103), shOSBP (n = 118), two-tailed, unpaired t-test. ****P = 5.68202 x 10-15 vs. shLuc). f, knockdown of OSBP impairs basal state mTORC1 signaling. Cells expressing the indicated shRNAs were harvested and analyzed by immunoblotting for the indicated proteins and phospho-proteins. Experiments repeated two times. g, Depletion of OSBP has a modest effect on mTORC1 activation by amino acids. HEK-293T cells expressing the indicated shRNAs were either starved of amino acids for 50 min, or starved and restimulated with amino acids for 10 min. Cell lysates were immunoblotted for the indicated proteins and phospho-proteins. Fold changes of the protein phosphorylation are indicated. Experiments repeated two times. h, Loss of OSBP does not affect lysosomal localization of the Ragulator-Rag GTPase complex. HEK-293T cells depleted of OSBP were subjected to cholesterol starvation and restimulation, followed by immunofluorescence for endogenous p18, RagC and LAMP2, respectively. Scale bar, 10 μm. See quantitation in Fig. 3a. Experiment performed once. Statistics source data are provided in Supplementary Table 2.

Supplementary Figure 7 OSBP-mediated mTORC1 regulation requires its membrane tethering and lipid transfer activities.

a, The PH domain and FFAT motif are essential for proper intracellular targeting of OSBP. (left) Domain organization for OSBP full length and the truncations employed in this study. (right) HEK293A cells stably expressing the full length and the indicated truncations of FLAG-GFP-OSBP were subjected to immunofluorescence for endogenous LAMP2. Scale bar, 10 μm. b, HEK-293T cells were depleted of OSBP via shRNA or depleted of OSBP followed by lentivirus-mediated re-expression of FLAG-GFP-OSBP, either wild type or the indicated truncations. Cells were starved of cholesterol for 2h, or starved and restimulated with cholesterol for 2h. Cell lysates were immunoblotted for the indicated proteins and phospho-proteins. c, (upper) OSBP mutants and their corresponding loss of function are shown in the table. (bottom) HEK-293T cells were depleted of OSBP, reconstituted with OSBP wild type and mutants as indicated, and evaluated for cholesterol-dependent mTORC1 signaling. Cell lysates were immunoblotted for the indicated proteins and phospho-proteins. d, HEK-293A cells stably expressing wild type, PH domain-deleted and organelle-targeted FLAG-GFP-OSBP isoforms were subjected to immunofluorescence for endogenous LAMP2. Scale bar, 10 μm. Experiments in a (right), b, c (bottom) and d repeated two times with similar results.

Supplementary Figure 8 OSBP inhibition abolishes mTORC1 signaling and promotes clearance of p62 aggregates in NPC1-deficient cells.

a, Increased ER-lysosome contacts in NPC1-deficient cells. Human fibroblasts co-expressing the lysosomal and ER markers FLAG-GFP-TMEM192 and mCherry-Sec61b, respectively, were seeded on 35mm-glass-bottom dishes and subjected to time-lapse imaging on a spinning disk confocal microscope. The two channels were sequentially acquired at 1 sec intervals. Scale bar, 10 μm; Insets show magnified time-lapse images of the ER-lysosome contacts. Scale bar, 2 μm. b, Electron microscopy (EM) images of lysosomes/autophagic vacuoles in control and NPC1 patient-derived fibroblasts. Scale bar, 0.5 μm; scale bar in insets, 0.2 μm. Arrows indicates the close apposition between lysosomal vesicles and ER tubules. c, OSW-1 treatment eliminates p62/SQSTM1 accumulated in human NPC1 patient-derived fibroblasts in a dose-dependent manner. Cells were treated with OSW-1 at the indicated concentrations for 8h and subjected to immunoblotting for the indicated proteins. d, OSW-1 treatment eliminates p62/SQSTM1 accumulated in Npc1-/- MEFs in a dose-dependent manner. Cells were treated with OSW-1 at the indicated concentrations for 24h and subjected to immunoblotting for the indicated proteins, including p62 and LC3B-II. e, Bafilomycin A1 blocks OSW-1-induced p62 clearance. Npc1-/- MEFs were treated with either 20 nM OSW-1 for 24h, 400 nM Bafilomycin A1 for 4h, or in combination as indicated. f, Combined treatment of Torin1 and OSW-1 has no additional effects on p62 clearance in Npc1-/- MEFs. Cells were treated with either 20 nM OSW-1, 250 nM Torin1, or in combination as indicated for 24h. Cell lysates were immunoblotted for the indicated proteins. Experiments in af repeated two times with similar results.

Supplementary Figure 9 Unprocessed images of all gels and blots.

Lyso-IP (upper, Fig. 1e). OSBP depletion (lower left, Fig. 2d) and OSW-1 (lower right, Fig. 2e) inhibits sterol-induced mTORC1 signaling. OSBP acts upstream of the Rag GTPases (upper, Fig. 3d), GATOR1 (lower left, Fig. 3e) and KICSTOR (lower right, Fig. 3f). OSBP activates mTORC1 via lysosomal contact (upper, Fig. 4d) and lipid transport (lower, Fig. 4e). Excess PI4P does not inhibit mTORC1 in OSBP-depleted cells (Fig. 5d). VAPA/B (upper, Fig. 6c), but not STARD3 (lower left, Fig. 6d) and ORP1L (lower right, Fig. 6e), regulate sterol-dependent mTORC1 signaling. NPC1 regulates sterol-induced OSBP-VAP binding (left, Fig. 7b). OSBP depletion abolishes hyperactive mTORC1 in NPC1-null cells (right, Fig. 7c). OSBP inhibition by OSW-1 (left, Fig. 8b) and shRNA (right, Fig. 8c) restores autophagy in NPC1 fibroblasts. GFP-OSBP expression in HEK-293A cells (left, Supplementary Fig. 3a). Validation of VAPA/B knockdown (right, Supplementary Fig. 3d). Western blots of Lyso-IP (left, Supplementary Fig. 4a). Validation of OSBP knockdown (right, Supplementary Fig. 4b). Basal state (left, Supplementary Fig. 6f) and amino acid-induced (right, Supplementary Fig. 6g) mTORC1 signaling in OSBP-depleted cells. Truncation (left, Supplementary Fig. 7b) and mutation (right, Supplementary Fig. 7c) analysis of OSBP on mTORC1 activation by sterol. OSW-1 activates autophagy in NPC1 fibroblasts (left, Supplementary Fig. 8c; middle, Supplementary Fig. 8d) in a BafA1-sensitive manner (upper right, Supplementary Fig. 8e) via mTORC1 pathway (lower right, Supplementary Fig. 8f).

Supplementary information

Supplementary Information

Supplementary Figures 1–9, titles and legends for Supplementary Tables 1–3.

Reporting Summary

Supplementary Table 1

Full lysosomal proteomics data.

Supplementary Table 2

Statistics source data.

Supplementary Table 3

List of antibodies.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lim, CY., Davis, O.B., Shin, H.R. et al. ER–lysosome contacts enable cholesterol sensing by mTORC1 and drive aberrant growth signalling in Niemann–Pick type C. Nat Cell Biol 21, 1206–1218 (2019). https://doi.org/10.1038/s41556-019-0391-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0391-5

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