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:

Proteome-wide mapping of cholesterol-interacting proteins in mammalian cells

Abstract

Cholesterol is an essential structural component of cellular membranes and serves as a precursor for several classes of signaling molecules. Cholesterol exerts its effects and is, itself, regulated in large part by engagement in specific interactions with proteins. The full complement of sterol-binding proteins that exist in mammalian cells, however, remains unknown. Here we describe a chemoproteomic strategy that uses clickable, photoreactive sterol probes in combination with quantitative mass spectrometry to globally map cholesterol-protein interactions directly in living cells. We identified over 250 cholesterol-binding proteins, including receptors, channels and enzymes involved in many established and previously unreported interactions. Prominent among the newly identified interacting proteins were enzymes that regulate sugars, glycerolipids and cholesterol itself as well as proteins involved in vesicular transport and protein glycosylation and degradation, pointing to key nodes in biochemical pathways that may couple sterol concentrations to the control of other metabolites and protein localization and modification.

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: Clickable photoreactive sterol probes.
Figure 2: Gel-based profiling of sterol-binding proteins in HeLa cells.
Figure 3: Mass spectrometry–based profiling of sterol-binding proteins in HeLa cells.
Figure 4: Analysis of group I proteins.

Similar content being viewed by others

References

  1. Schroeder, F. et al. Caveolin, sterol carrier protein-2, membrane cholesterol-rich microdomains and intracellular cholesterol trafficking. Subcell. Biochem. 51, 279–318 (2010).

    Article  CAS  Google Scholar 

  2. Russell, D.W. Oxysterol biosynthetic enzymes. Biochim. Biophys. Acta 1529, 126–135 (2000).

    Article  CAS  Google Scholar 

  3. McLean, K.J., Hans, M. & Munro, A.W. Cholesterol, an essential molecule: diverse roles involving cytochrome P450 enzymes. Biochem. Soc. Trans. 40, 587–593 (2012).

    Article  CAS  Google Scholar 

  4. Russell, D.W. Fifty years of advances in bile acid synthesis and metabolism. J. Lipid Res. 50 (suppl.), S120–S125 (2009).

    Article  Google Scholar 

  5. Badimon, L. & Vilahur, G. LDL-cholesterol versus HDL-cholesterol in the atherosclerotic plaque: inflammatory resolution versus thrombotic chaos. Ann. NY Acad. Sci. 1254, 18–32 (2012).

    Article  CAS  Google Scholar 

  6. Mirza, R. et al. DHCR24 gene knockout mice demonstrate lethal dermopathy with differentiation and maturation defects in the epidermis. J. Invest. Dermatol. 126, 638–647 (2006).

    Article  CAS  Google Scholar 

  7. Porter, F.D. Human malformation syndromes due to inborn errors of cholesterol synthesis. Curr. Opin. Pediatr. 15, 607–613 (2003).

    Article  Google Scholar 

  8. Herman, G.E. Disorders of cholesterol biosynthesis: prototypic metabolic malformation syndromes. Hum. Mol. Genet. 12 Spec No 1, R75–R88 (2003).

    Article  CAS  Google Scholar 

  9. Cianciola, N.L., Carlin, C.R. & Kelley, T.J. Molecular pathways for intracellular cholesterol accumulation: common pathogenic mechanisms in Niemann-Pick disease Type C and cystic fibrosis. Arch. Biochem. Biophys. 515, 54–63 (2011).

    Article  CAS  Google Scholar 

  10. Rosenbaum, A.I. & Maxfield, F.R. Niemann-Pick type C disease: molecular mechanisms and potential therapeutic approaches. J. Neurochem. 116, 789–795 (2011).

    Article  CAS  Google Scholar 

  11. Weinhofer, I., Forss-Petter, S., Kunze, M., Zigman, M. & Berger, J. X-linked adrenoleukodystrophy mice demonstrate abnormalities in cholesterol metabolism. FEBS Lett. 579, 5512–5516 (2005).

    Article  CAS  Google Scholar 

  12. Brown, M.S. & Goldstein, J.L. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89, 331–340 (1997).

    Article  CAS  Google Scholar 

  13. Gill, S., Chow, R. & Brown, A.J. Sterol regulators of cholesterol homeostasis and beyond: the oxysterol hypothesis revisited and revised. Prog. Lipid Res. 47, 391–404 (2008).

    Article  CAS  Google Scholar 

  14. Jeon, T.I. & Osborne, T.F. SREBPs: metabolic integrators in physiology and metabolism. Trends Endocrinol. Metab. 23, 65–72 (2012).

    Article  CAS  Google Scholar 

  15. Heal, W.P., Jovanovic, B., Bessin, S., Wright, M.H., Magee, A.I. & Tate, E.W. Bioorthogonal chemical tagging of protein cholesterylation in living cells. Chem. Commun. (Camb.) 47, 4081–4083 (2011).

    Article  CAS  Google Scholar 

  16. Hoop, C.L., Sivanandam, V.N., Kodali, R., Srnec, M.N. & van der Wel, P.C. Structural characterization of the caveolin scaffolding domain in association with cholesterol-rich membranes. Biochemistry 51, 90–99 (2012).

    Article  CAS  Google Scholar 

  17. Theesfeld, C.L., Pourmand, D., Davis, T., Garza, R.M. & Hampton, R.Y. The sterol-sensing domain (SSD) directly mediates signal-regulated endoplasmic reticulum-associated degradation (ERAD) of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase isozyme Hmg2. J. Biol. Chem. 286, 26298–26307 (2011).

    Article  CAS  Google Scholar 

  18. Motamed, M. et al. Identification of luminal Loop 1 of Scap protein as the sterol sensor that maintains cholesterol homeostasis. J. Biol. Chem. 286, 18002–18012 (2011).

    Article  CAS  Google Scholar 

  19. Thiele, C., Hannah, M.J., Fahrenholz, F. & Huttner, W.B. Cholesterol binds to synaptophysin and is required for biogenesis of synaptic vesicles. Nat. Cell Biol. 2, 42–49 (2000).

    Article  CAS  Google Scholar 

  20. Charrin, S. et al. A physical and functional link between cholesterol and tetraspanins. Eur. J. Immunol. 33, 2479–2489 (2003).

    Article  CAS  Google Scholar 

  21. Rostovtsev, V.V., Green, J.G., Fokin, V.V. & Sharpless, K.B. A stepwise Huisgen cycloaddition process: copper(I)-catalyzed regioselective “ligation” of azides and terminal alkynes. Angew. Chem. Int. Edn. Engl. 41, 2596–2599 (2002).

    Article  CAS  Google Scholar 

  22. Speers, A.E. & Cravatt, B.F. Profiling enzyme activities in vivo using click chemistry methods. Chem. Biol. 11, 535–546 (2004).

    Article  CAS  Google Scholar 

  23. Mintzer, E.A., Waarts, B.L., Wilschut, J. & Bittman, R. Behavior of a photoactivatable analog of cholesterol, 6-photocholesterol, in model membranes. FEBS Lett. 510, 181–184 (2002).

    Article  CAS  Google Scholar 

  24. 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 

  25. Roberts, J.D. & Caserio, M. Basic Principles of Organic Chemistry 445–487 (Addison-Wesley, 1977).

  26. Ong, S.E., Foster, L.J. & Mann, M. Mass spectrometric-based approaches in quantitative proteomics. Methods 29, 124–130 (2003).

    Article  CAS  Google Scholar 

  27. Tong, F. et al. Decreased expression of ARV1 results in cholesterol retention in the endoplasmic reticulum and abnormal bile acid metabolism. J. Biol. Chem. 285, 33632–33641 (2010).

    Article  CAS  Google Scholar 

  28. Thompson, A.A. et al. GPCR stabilization using the bicelle-like architecture of mixed sterol-detergent micelles. Methods 55, 310–317 (2011).

    Article  CAS  Google Scholar 

  29. Huang da, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  Google Scholar 

  30. Huang da, W., Sherman, B.T. & Lempicki, R.A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  Google Scholar 

  31. Pastorino, J.G. & Hoek, J.B. Regulation of hexokinase binding to VDAC. J. Bioenerg. Biomembr. 40, 171–182 (2008).

    Article  CAS  Google Scholar 

  32. Christian, A.E., Haynes, M.P., Phillips, M.C. & Rothblat, G.H. Use of cyclodextrins for manipulating cellular cholesterol content. J. Lipid Res. 38, 2264–2272 (1997).

    CAS  PubMed  Google Scholar 

  33. Martin, B.R., Wang, C., Adibekian, A., Tully, S.E. & Cravatt, B.F. Global profiling of dynamic protein palmitoylation. Nat. Methods 9, 84–89 (2012).

    Article  CAS  Google Scholar 

  34. Washburn, M.P., Wolters, D. & Yates, J.R. III. Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat. Biotechnol. 19, 242–247 (2001).

    Article  CAS  Google Scholar 

  35. Eng, J., McCormack, A.L. & Yates, J.R. An approach to correlate MS/MS data to amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 5, 976–989 (1994).

    Article  CAS  Google Scholar 

  36. Weerapana, E. et al. Quantitative reactivity profiling predicts functional cysteines in proteomes. Nature 468, 790–795 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank P. Baran for helpful advice on the synthesis of sterol probes, and A. Rheingold and C. Moore for X-ray crystallographic analysis. This work was supported by US National Institutes of Health (CA132630) and the Skaggs Institute for Chemical Biology.

Author information

Authors and Affiliations

Authors

Contributions

J.J.H., S.E.T., M.J.N. and B.F.C. designed experiments; J.J.H., A.B.C. and S.E.T. performed experiments; J.J.H. and B.F.C. analyzed data; J.J.H. and B.F.C. wrote the manuscript.

Corresponding author

Correspondence to Benjamin F Cravatt.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7, Supplementary Table 5, Supplementary Note (PDF 2876 kb)

Supplementary Table 1

List of group I–IV proteins with experimental SILAC ratios. Groups I, II, III, and IV are presented on separate sheets, with the mean SILAC ratio obtained in the trans versus no–UV light, trans versus PEA-DEA, and trans versus 10 × cholesterol competition (Ch) shown. For proteins that were not detected in the no–UV light experiments but were found in the vehicle control experiments, the vehicle ratios are shown instead (colored in blue). For group I proteins, gene expression data are presented as 1 = yes; 0 = no, where ‘yes’ means that the gene expression change was > twofold at 30 min cholesterol treatment or > twofold at 12 h cholesterol treatment. All quantifications were manually filtered here. ND, not detected. See Supplementary Table 2 for a list of individual peptide sequences and ratios detected for each protein. (XLSX 152 kb)

Supplementary Table 2

Complete proteomic data for trans versus no UV light and versus vehicle. Data sets include the individual peptides detected for each protein, their SILAC ratios and charge states. SILAC ratios are presented as the mean of duplicates but are not manually filtered. (XLSX 1574 kb)

Supplementary Table 3

Data sets for each set of SILAC experiments. Mean ratios are presented on separate sheets for each set of SILAC experiments. Control experiments (no UV light and vehicle comparisons) are shown together, are unfiltered and were each run in duplicate. PEA-DA comparisons and cholesterol competition, shown together, were run in duplicate and quadruplicate, respectively, and background proteins from control runs are removed. Probe comparisons (trans versus epi, cis and trans), shown together, were run in duplicate, and background proteins are removed. All quantifications are raw and were not manually filtered. (XLSX 211 kb)

Supplementary Table 4

Complete gene expression profiles for cholesterol and probe treatments. The signal intensity for each gene in each microarray experiment is included. The three conditions tested were: (i) control/vehicle: cells were untreated, and were collected and processed in parallel with the other conditions; (ii) 30 min: cells received 20 μM cis probe + 100 μM cholesterol for 30 min before harvest; (iii) 12 h: cells received 20 μM cis probe + 100 μM cholesterol for 12 h before harvest. Total mRNA was extracted for each condition, and gene expression was analyzed by HU133 Set GeneChip from Affymetrix. (XLSX 6362 kb)

Supplementary Table 6

Unenriched versus trans-enrichment membrane proteome comparison by spectral counting. Raw data set presenting proteins in groups I–IV that were detected with >10 average spectral counts over quadruplicate trans probe–mediated enrichments (columns t1–t4) or unenriched membrane proteomes (columns u1–u4). (XLSX 108 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Hulce, J., Cognetta, A., Niphakis, M. et al. Proteome-wide mapping of cholesterol-interacting proteins in mammalian cells. Nat Methods 10, 259–264 (2013). https://doi.org/10.1038/nmeth.2368

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nmeth.2368

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