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.

  • Perspective
  • Published:

Cholesterol access in cellular membranes controls Hedgehog signaling

Abstract

The Hedgehog (Hh) signaling pathway coordinates cell–cell communication in development and regeneration. Defects in this pathway underlie diseases ranging from birth defects to cancer. Hh signals are transmitted across the plasma membrane by two proteins, Patched 1 (PTCH1) and Smoothened (SMO). PTCH1, a transporter-like tumor-suppressor protein, binds to Hh ligands, but SMO, a G-protein-coupled-receptor family oncoprotein, transmits the Hh signal across the membrane. Recent structural, biochemical and cell-biological studies have converged at the surprising model that a specific pool of plasma membrane cholesterol, termed accessible cholesterol, functions as a second messenger that conveys the signal between PTCH1 and SMO. Beyond solving a central puzzle in Hh signaling, these studies are revealing new principles in membrane biology: how proteins respond to and remodel cholesterol accessibility in membranes and how the cholesterol composition of organelle membranes is used to regulate protein function.

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: Hedgehog signal transmission across the plasma membrane.
Fig. 2: Multiple sterol binding sites in SMO.
Fig. 3: Structural similarities between PTCH1 and the cholesterol transporter NPC1.
Fig. 4: Cholesterol accessibility in cellular membranes.
Fig. 5: Changes in accessible cholesterol influence Hh signaling in target cells.
Fig. 6: Hh signal transmission by ligand-controlled changes in cholesterol accessibility of the ciliary membrane.
Fig. 7: Two models for the regulation of SMO by PTCH1 at primary cilia.

Similar content being viewed by others

References

  1. Kong, J. H., Siebold, C. & Rohatgi, R. Biochemical mechanisms of vertebrate hedgehog signaling. Development 146, dev166892 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Huangfu, D. et al. Hedgehog signalling in the mouse requires intraflagellar transport proteins. Nature 426, 83–87 (2003).

    Article  CAS  PubMed  Google Scholar 

  3. Goetz, S. C., Ocbina, P. J. R. & Anderson, K. V. The primary cilium as a Hedgehog signal transduction machine. Methods Cell Biol. 94, 199–222 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Wang, C. et al. Structure of the human smoothened receptor bound to an antitumour agent. Nature 497, 338–343 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Porter, J. A., Young, K. E. & Beachy, P. A. Cholesterol modification of Hedgehog signaling proteins in animal development. Science 274, 255–259 (1996).

    Article  CAS  PubMed  Google Scholar 

  6. Pepinsky, R. B. et al. Identification of a palmitic acid-modified form of human Sonic hedgehog. J. Biol. Chem. 273, 14037–14045 (1998).

    Article  CAS  PubMed  Google Scholar 

  7. Chiang, C. et al. Cyclopia and defective axial patterning in mice lacking Sonic Hedgehog gene function. Nature 383, 407–413 (1996).

    Article  CAS  PubMed  Google Scholar 

  8. Cooper, M. K., Porter, J. A., Young, K. E. & Beachy, P. A. Teratogen-mediated inhibition of target tissue response to Shh signaling. Science 280, 1603–1607 (1998).

    Article  CAS  PubMed  Google Scholar 

  9. Cooper, M. K. et al. A defective response to Hedgehog signaling in disorders of cholesterol biosynthesis. Nat. Genet. 33, 508–513 (2003). Cholesterol depletion using MβCD or mutations in genes encoding terminal cholesterol biosynthetic enzymes impair Hh signaling in target cells.

    Article  CAS  PubMed  Google Scholar 

  10. Blassberg, R., Macrae, J. I., Briscoe, J. & Jacob, J. Reduced cholesterol levels impair Smoothened activation in Smith-Lemli-Opitz syndrome. Hum. Mol. Genet. 25, 693–705 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Sharpe, H. J., Wang, W., Hannoush, R. N. & de Sauvage, F. J. Regulation of the oncoprotein Smoothened by small molecules. Nat. Chem. Biol. 11, 246–255 (2015).

    Article  CAS  PubMed  Google Scholar 

  12. Wang, C. et al. Structural basis for Smoothened receptor modulation and chemoresistance to anticancer drugs. Nat. Commun. 5, 4355 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Corcoran, R. B. & Scott, M. P. Oxysterols stimulate Sonic hedgehog signal transduction and proliferation of medulloblastoma cells. Proc. Natl Acad. Sci. USA 103, 8408–8413 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Rohatgi, R., Milenkovic, L. & Scott, M. P. Patched1 regulates hedgehog signaling at the primary cilium. Science 317, 372–376 (2007). PTCH1 is localized in and around primary cilia and inhibits SMO activity and accumulation in the ciliary membrane.

    Article  CAS  PubMed  Google Scholar 

  15. Dwyer, J. R. et al. Oxysterols are novel activators of the hedgehog signaling pathway in pluripotent mesenchymal cells. J. Biol. Chem. 282, 8959–8968 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Nachtergaele, S. et al. Oxysterols are allosteric activators of the oncoprotein Smoothened. Nat. Chem. Biol. 8, 211–220 (2012). Side chain oxysterols induce Hh responses in cultured cells by binding and activating SMO.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Raleigh, D. R. et al. Cilia-associated oxysterols activate Smoothened. Mol. Cell 72, 316–327.e5 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chen, W., Chen, G., Head, D. L., Mangelsdorf, D. J. & Russell, D. W. Enzymatic reduction of oxysterols impairs LXR signaling in cultured cells and the livers of mice. Cell Metab. 5, 73–79 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kinnebrew, M. et al. Cholesterol accessibility at the ciliary membrane controls hedgehog signaling. eLife 8, e50051 (2019). A CRISPR screen and sphingomyelin (SM) depletion showed that Hh signaling is activated selectively by the accessible pool of membrane cholesterol. Fluorescent probes described in ref. 23 showed that Hh ligands trigger an increase in accessible cholesterol in the ciliary membrane by inactivating PTCH1, allowing SMO activation.

    Article  PubMed  PubMed Central  Google Scholar 

  20. McConnell, H. M. & Radhakrishnan, A. Condensed complexes of cholesterol and phospholipids. Biochim. Biophys. Acta 1610, 159–173 (2003).

    Article  CAS  PubMed  Google Scholar 

  21. Demel, R. A., Jansen, J. W., van Dijck, P. W. & van Deenen, L. L. The preferential interaction of cholesterol with different classes of phospholipids. Biochim. Biophys. Acta 465, 1–10 (1977).

    Article  CAS  PubMed  Google Scholar 

  22. Finean, J. B. Phospholipid-cholesterol complex in the structure of myelin. Experientia 9, 17–19 (1953).

    Article  CAS  PubMed  Google Scholar 

  23. Endapally, S. et al. Molecular discrimination between two conformations of sphingomyelin in plasma membranes. Cell 176, 1040–1053.e17 (2019). Fluorescently labeled probes derived from microbial and fungal toxins that can be used to distinguish between the accessible and sequestered pools of cholesterol were developed.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Klein, U., Gimpl, G. & Fahrenholz, F. Alteration of the myometrial plasma membrane cholesterol content with beta-cyclodextrin modulates the binding affinity of the oxytocin receptor. Biochemistry 34, 13784–13793 (1995).

    Article  CAS  PubMed  Google Scholar 

  25. Byrne, E. F. X. et al. Structural basis of Smoothened regulation by its extracellular domains. Nature 535, 517–522 (2016). A 3.2 Å multi-domain crystal structure of SMO unexpectedly revealed a cholesterol molecule bound in the cysteine-rich domain (CRD). Clinically used SMO inhibitors induce a conformational change that prevents cholesterol access to the CRD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Luchetti, G. et al. Cholesterol activates the G-protein coupled receptor Smoothened to promote Hedgehog signaling. eLife 5, e20304 (2016). Refs. 26 and 27 showed that cholesterol can function as an instructive agonist for SMO and is both necessary and sufficient to activate Hh signaling.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Huang, P. et al. Cellular cholesterol directly activates Smoothened in Hedgehog signaling. Cell 166, 1176–1187.e14 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Nedelcu, D., Liu, J., Xu, Y., Jao, C. & Salic, A. Oxysterol binding to the extracellular domain of Smoothened in Hedgehog signaling. Nat. Chem. Biol. 9, 557–564 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Myers, B. R. et al. Hedgehog pathway modulation by multiple lipid binding sites on the smoothened effector of signal response. Dev. Cell 26, 346–357 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Nachtergaele, S. et al. Structure and function of the Smoothened extracellular domain in vertebrate Hedgehog signaling. eLife 2, e01340 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Xiao, X. et al. Cholesterol modification of Smoothened is required for Hedgehog signaling. Mol. Cell 66, 154–162.e10 (2017). Mutations in the CRD cholesterol-binding site abolished SMO function in mouse embryos, and PTCH1 can attenuate cholesterol access to the SMO CRD.

    Article  CAS  PubMed  Google Scholar 

  32. Zhang, X. et al. Crystal structure of a multi-domain human smoothened receptor in complex with a super stabilizing ligand. Nat. Commun. 8, 15383 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Deshpande, I. et al. Smoothened stimulation by membrane sterols drives Hedgehog pathway activity. Nature 571, 284–288 (2019). An active-state structure of SMO bound to two synthetic agonists revealed a second cholesterol-binding site in the middle of the TMD, in addition to the previously identified site in the CRD.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Huang, P. et al. Structural basis of Smoothened activation in Hedgehog signaling. Cell 175, 295–297 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Qi, X. et al. Cryo-EM structure of oxysterol-bound human Smoothened coupled to a heterotrimeric Gi. Nature 571, 279–283 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Yauch, R. L. et al. Smoothened mutation confers resistance to a Hedgehog pathway inhibitor in medulloblastoma. Science 326, 572–574 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Myers, B. R., Neahring, L., Zhang, Y., Roberts, K. J. & Beachy, P. A. Rapid, direct activity assays for Smoothened reveal Hedgehog pathway regulation by membrane cholesterol and extracellular sodium. Proc. Natl Acad. Sci. USA 114, E11141–E11150 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. 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  PubMed  Google Scholar 

  39. Carstea, E. D. et al. Niemann-Pick C1 disease gene: homology to mediators of cholesterol homeostasis. Science 277, 228–231 (1997).

    Article  CAS  PubMed  Google Scholar 

  40. Davies, J. P. & Ioannou, Y. A. Topological analysis of Niemann-Pick C1 protein reveals that the membrane orientation of the putative sterol-sensing domain is identical to those of 3-hydroxy-3-methylglutaryl-CoA reductase and sterol regulatory element binding protein cleavage-activating protein. J. Biol. Chem. 275, 24367–24374 (2000).

    Article  CAS  PubMed  Google Scholar 

  41. Tseng, T. T. et al. The RND permease superfamily: an ancient, ubiquitous and diverse family that includes human disease and development proteins. J. Mol. Microbiol. Biotechnol. 1, 107–125 (1999).

    CAS  PubMed  Google Scholar 

  42. Gong, X. et al. Structural basis for the recognition of Sonic Hedgehog by human Patched1. Science 361, eaas8935 (2018). Refs. 42-45 report liganded and unliganded cryo-EM structures of PTCH1, suggesting a tunnel through the protein that may function in sterol transport. Ref. 45 demonstrated that overexpression of PTCH1 reduces recruitment of a cholesterol-binding probe to the inner leaflet of the plasma membrane.

    Article  PubMed  Google Scholar 

  43. Qi, X., Schmiege, P., Coutavas, E. & Li, X. Two Patched molecules engage distinct sites on Hedgehog yielding a signaling-competent complex. Science 362, aas8843 (2018).

    Article  Google Scholar 

  44. Qi, X., Schmiege, P., Coutavas, E., Wang, J. & Li, X. Structures of human Patched and its complex with native palmitoylated sonic hedgehog. Nature 560, 128–132 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Zhang, Y. et al. Structural basis for cholesterol transport-like activity of the Hedgehog receptor patched. Cell 175, 1352–1364.e14 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Li, X. et al. Structure of human Niemann-Pick C1 protein. Proc. Natl Acad. Sci. USA 113, 8212–8217 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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  PubMed  PubMed Central  Google Scholar 

  48. Qian, H. et al. Structural basis of low-pH-dependent lysosomal cholesterol egress by NPC1 and NPC2. Cell 182, 98–111.e18 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Winkler, M. B. L. et al. Structural insight into eukaryotic sterol transport through Niemann-Pick type C proteins. Cell 179, 485–497.e18 (2019).

    Article  CAS  PubMed  Google Scholar 

  50. Qi, C., Di Minin, G., Vercellino, I., Wutz, A. & Korkhov, V. M. Structural basis of sterol recognition by human hedgehog receptor PTCH1. Sci. Adv. 5, eaaw6490 (2018).

    Article  Google Scholar 

  51. Rudolf, A. F. et al. The morphogen Sonic hedgehog inhibits its receptor Patched by a pincer grasp mechanism. Nat. Chem. Biol. 15, 975–982 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Qian, H. et al. Inhibition of tetrameric Patched1 by Sonic Hedgehog through an asymmetric paradigm. Nat. Commun. 10, 2320 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Long, T. et al. Structural basis for itraconazole-mediated NPC1 inhibition. Nat. Commun. 11, 152 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Infante, R. E. et al. Purified NPC1 protein. I. Binding of cholesterol and oxysterols to a 1278-amino acid membrane protein. J. Biol. Chem. 283, 1052–1063 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Infante, R. E. et al. Purified NPC1 protein: II. Localization of sterol binding to a 240-amino acid soluble luminal loop. J. Biol. Chem. 283, 1064–1075 (2008).

    Article  CAS  PubMed  Google Scholar 

  56. 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  PubMed  PubMed Central  Google Scholar 

  57. 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  PubMed  PubMed Central  Google Scholar 

  58. Bidet, M. et al. The hedgehog receptor patched is involved in cholesterol transport. PLoS One 6, e23834 (2011). PTCH1 can promote efflux of a fluorescent cholesterol analog (BODIPY-cholesterol) from cells.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Litz, J. P., Thakkar, N., Portet, T. & Keller, S. L. Depletion with cyclodextrin reveals two populations of cholesterol in model lipid membranes. Biophys. J. 110, 635–645 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Watts, A., Volotovski, I. D. & Marsh, D. Rhodopsin-lipid associations in bovine rod outer segment membranes. Identification of immobilized lipid by spin-labels. Biochemistry 18, 5006–5013 (1979).

    Article  CAS  PubMed  Google Scholar 

  61. Leathes, J. B. Condensing effect of cholesterol on monolayers. Lancet 208, 853–856 (1925).

    Article  Google Scholar 

  62. Finegold, L.X. Cholesterol in Membrane Models. (CRC Press, 1992).

  63. McConnell, H. & Radhakrishnan, A. Theory of the deuterium NMR of sterol-phospholipid membranes. Proc. Natl Acad. Sci. USA 103, 1184–1189 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Radhakrishnan, A. & McConnell, H. Condensed complexes in vesicles containing cholesterol and phospholipids. Proc. Natl Acad. Sci. USA 102, 12662–12666 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Lange, Y., Tabei, S. M. A., Ye, J. & Steck, T. L. Stability and stoichiometry of bilayer phospholipid-cholesterol complexes: relationship to cellular sterol distribution and homeostasis. Biochemistry 52, 6950–6959 (2013).

    Article  CAS  PubMed  Google Scholar 

  66. Keller, S. L., Radhakrishnan, A. & McConnell, H. M. Saturated phospholipids with high melting temperatures form complexes with cholesterol in monolayers. J. Phys. Chem. B 104, 7522–7527 (2000).

    Article  CAS  Google Scholar 

  67. Lönnfors, M., Doux, J. P. F., Killian, J. A., Nyholm, T. K. M. & Slotte, J. P. Sterols have higher affinity for sphingomyelin than for phosphatidylcholine bilayers even at equal acyl-chain order. Biophys. J. 100, 2633–2641 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Radhakrishnan, A. & McConnell, H. M. Chemical activity of cholesterol in membranes. Biochemistry 39, 8119–8124 (2000).

    Article  CAS  PubMed  Google Scholar 

  69. Ahn, K.-W. & Sampson, N. S. Cholesterol oxidase senses subtle changes in lipid bilayer structure. Biochemistry 43, 827–836 (2004).

    Article  CAS  PubMed  Google Scholar 

  70. Flanagan, J. J., Tweten, R. K., Johnson, A. E. & Heuck, A. P. Cholesterol exposure at the membrane surface is necessary and sufficient to trigger perfringolysin O binding. Biochemistry 48, 3977–3987 (2009).

    Article  CAS  PubMed  Google Scholar 

  71. Gay, A., Rye, D. & Radhakrishnan, A. Switch-like responses of two cholesterol sensors do not require protein oligomerization in membranes. Biophys. J. 108, 1459–1469 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Haynes, M. P., Phillips, M. C. & Rothblat, G. H. Efflux of cholesterol from different cellular pools. Biochemistry 39, 4508–4517 (2000).

    Article  CAS  PubMed  Google Scholar 

  73. Lange, Y., Ye, J. & Steck, T. L. How cholesterol homeostasis is regulated by plasma membrane cholesterol in excess of phospholipids. Proc. Natl Acad. Sci. USA 101, 11664–11667 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Das, A., Goldstein, J. L., Anderson, D. D., Brown, M. S. & Radhakrishnan, A. Use of mutant 125I-perfringolysin O to probe transport and organization of cholesterol in membranes of animal cells. Proc. Natl Acad. Sci. USA 110, 10580–10585 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Das, A., Brown, M. S., Anderson, D. D., Goldstein, J. L. & Radhakrishnan, A. Three pools of plasma membrane cholesterol and their relation to cholesterol homeostasis. eLife 3, e02882 (2014). Sphingomyelin regulates the partitioning of cholesterol between the accessible and sequestered pools.

    Article  PubMed  PubMed Central  Google Scholar 

  76. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  77. Skočaj, M. et al. Tracking cholesterol/sphingomyelin-rich membrane domains with the ostreolysin A-mCherry protein. PLoS One 9, e92783 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Goldstein, J. L. & Brown, M. S. A century of cholesterol and coronaries: from plaques to genes to statins. Cell 161, 161–172 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Slotte, J. P. & Bierman, E. L. Depletion of plasma-membrane sphingomyelin rapidly alters the distribution of cholesterol between plasma membranes and intracellular cholesterol pools in cultured fibroblasts. Biochem. J. 250, 653–658 (1988).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Scheek, S., Brown, M. S. & Goldstein, J. L. Sphingomyelin depletion in cultured cells blocks proteolysis of sterol regulatory element binding proteins at site 1. Proc. Natl Acad. Sci. USA 94, 11179–11183 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Corbit, K. C. et al. Vertebrate Smoothened functions at the primary cilium. Nature 437, 1018–1021 (2005). Hh ligands promote the accumulation of SMO in primary cilia, the likely subcellular location from which it signals to the cytoplasm.

    Article  CAS  PubMed  Google Scholar 

  82. Nachury, M. V. & Mick, D. U. Establishing and regulating the composition of cilia for signal transduction. Nat. Rev. Mol. Cell Biol. 20, 389–405 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Weiss, L. E., Milenkovic, L., Yoon, J., Stearns, T. & Moerner, W. E. Motional dynamics of single Patched1 molecules in cilia are controlled by Hedgehog and cholesterol. Proc. Natl Acad. Sci. USA 116, 5550–5557 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Kaiser, F., Huebecker, M. & Wachten, D. Sphingolipids controlling ciliary and microvillar function. FEBS Lett. https://doi.org/10.1002/1873-3468.13816 (2020).

  85. Serricchio, M. et al. Flagellar membranes are rich in raft-forming phospholipids. Biol. Open 4, 1143–1153 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Tyler, K. M. et al. Flagellar membrane localization via association with lipid rafts. J. Cell Sci. 122, 859–866 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Kaneshiro, E. S., Matesic, D. F. & Jayasimhulu, K. Characterizations of six ethanolamine sphingophospholipids from Paramecium cells and cilia. J. Lipid Res. 25, 369–377 (1984).

    Article  CAS  PubMed  Google Scholar 

  88. Breslow, D. K., Koslover, E. F., Seydel, F., Spakowitz, A. J. & Nachury, M. V. An in vitro assay for entry into cilia reveals unique properties of the soluble diffusion barrier. J. Cell Biol. 203, 129–147 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  89. Kowatsch, C., Woolley, R. E., Kinnebrew, M., Rohatgi, R. & Siebold, C. Structures of vertebrate Patched and Smoothened reveal intimate links between cholesterol and Hedgehog signalling. Curr. Opin. Struct. Biol. 57, 204–214 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Nager, A. R. et al. An actin network dispatches ciliary GPCRs into extracellular vesicles to modulate signaling. Cell 168, 252–263.e14 (2017).

    Article  CAS  PubMed  Google Scholar 

  91. Phua, S. C. et al. Dynamic remodeling of membrane composition drives cell cycle through primary cilia excision. Cell 168, 264–279.e15 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. He, C. et al. Macrophages release plasma membrane-derived particles rich in accessible cholesterol. Proc. Natl Acad. Sci. USA 115, E8499–E8508 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Mondal, M., Mesmin, B., Mukherjee, S. & Maxfield, F. R. Sterols are mainly in the cytoplasmic leaflet of the plasma membrane and the endocytic recycling compartment in CHO cells. Mol. Biol. Cell 20, 581–588 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Courtney, K. C. et al. C24 sphingolipids govern the transbilayer asymmetry of cholesterol and lateral organization of model and live-cell plasma membranes. Cell Reports 24, 1037–1049 (2018).

    Article  CAS  PubMed  Google Scholar 

  95. Liu, S.-L. et al. Orthogonal lipid sensors identify transbilayer asymmetry of plasma membrane cholesterol. Nat. Chem. Biol. 13, 268–274 (2017).

    Article  CAS  PubMed  Google Scholar 

  96. Steck, T. L. & Lange, Y. Transverse distribution of plasma membrane bilayer cholesterol: Picking sides. Traffic 19, 750–760 (2018).

    Article  CAS  PubMed  Google Scholar 

  97. Hausmann, G., von Mering, C. & Basler, K. The hedgehog signaling pathway: where did it come from? PLoS Biol. 7, e1000146 (2009). A hypothesis that Hh signaling may have evolved from an ancient pathway for sensing and transporting hopanoids.

    Article  PubMed  PubMed Central  Google Scholar 

  98. Bazan, J. F. & de Sauvage, F. J. Structural ties between cholesterol transport and morphogen signaling. Cell 138, 1055–1056 (2009).

    Article  CAS  PubMed  Google Scholar 

  99. Chovancova, E. et al. CAVER 3.0: a tool for the analysis of transport pathways in dynamic protein structures. PLOS Comput. Biol. 8, e1002708 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Sokolov, A. & Radhakrishnan, A. Accessibility of cholesterol in endoplasmic reticulum membranes and activation of SREBP-2 switch abruptly at a common cholesterol threshold. J. Biol. Chem. 285, 29480–29490 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge G. Pusapati for help with the figures and comments on the manuscript. C.S. was supported by grants from Cancer Research UK (C20724/A26752) and the European Research Council (647278), R.R. by grants from the National Institutes of Health (GM118082 and GM106078), and A.R. by grants from the NIH (HL20948), Welch Foundation (I-1793) and Leducq Foundation (19CVD04).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Arun Radhakrishnan, Rajat Rohatgi or Christian Siebold.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Radhakrishnan, A., Rohatgi, R. & Siebold, C. Cholesterol access in cellular membranes controls Hedgehog signaling. Nat Chem Biol 16, 1303–1313 (2020). https://doi.org/10.1038/s41589-020-00678-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-020-00678-2

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