1932

Abstract

Scap is a polytopic membrane protein that functions as a molecular machine to control the cholesterol content of membranes in mammalian cells. In the 21 years since our laboratory discovered Scap, we have learned how it binds sterol regulatory element-binding proteins (SREBPs) and transports them from the endoplasmic reticulum (ER) to the Golgi for proteolytic processing. Proteolysis releases the SREBP transcription factor domains, which enter the nucleus to promote cholesterol synthesis and uptake. When cholesterol in ER membranes exceeds a threshold, the sterol binds to Scap, triggering several conformational changes that prevent the Scap–SREBP complex from leaving the ER. As a result, SREBPs are no longer processed, cholesterol synthesis and uptake are repressed, and cholesterol homeostasis is restored. This review focuses on the four domains of Scap that undergo concerted conformational changes in response to cholesterol binding. The data provide a molecular mechanism for the control of lipids in cell membranes.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-biochem-062917-011852
2018-06-20
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/biochem/87/1/annurev-biochem-062917-011852.html?itemId=/content/journals/10.1146/annurev-biochem-062917-011852&mimeType=html&fmt=ahah

Literature Cited

  1. 1.  Hua X, Nohturfft A, Goldstein JL, Brown MS 1996. Sterol resistance in CHO cells traced to point mutation in SREBP cleavage activating protein (SCAP). Cell 87:415–26
    [Google Scholar]
  2. 2.  Brown MS, Goldstein JL 1997. The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89:331–40
    [Google Scholar]
  3. 3.  Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW et al. 2003. Combined analysis of olignucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. PNAS 100:12027–32
    [Google Scholar]
  4. 4.  Radhakrishnan A, Goldstein JL, McDonald JG, Brown MS 2008. Switch-like control of SREBP-2 transport triggered by small changes in ER cholesterol: a delicate balance. Cell Metab 8:512–21
    [Google Scholar]
  5. 5.  Brown MS, Dana SE, Goldstein JL 1973. Regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in human fibroblasts by lipoproteins. PNAS 70:2162–66
    [Google Scholar]
  6. 6.  Goldstein JL, Brown MS 1973. Familial hypercholesterolemia: identification of a defect in the regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity associated with overproduction of cholesterol. PNAS 70:2804–8
    [Google Scholar]
  7. 7.  Brown MS, Goldstein JL 1974. Familial hypercholesterolemia: defective binding of lipoproteins to cultured fibroblasts associated with impaired regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity. PNAS 71:788–92
    [Google Scholar]
  8. 8.  Goldstein JL, Brown MS 2009. History of discovery: the LDL receptor. Arterioscler. Thromb. Vasc. Biol. 29:431–38
    [Google Scholar]
  9. 9.  Anderson RGW, Brown MS, Goldstein JL 1977. Role of the coated endocytic vesicle in the uptake of receptor-bound low density lipoprotein in human fibroblasts. Cell 10:351–64
    [Google Scholar]
  10. 10.  Brown MS, Goldstein JL 1986. A receptor-mediated pathway for cholesterol homeostasis. Science 232:34–47
    [Google Scholar]
  11. 11.  Goldstein JL, Hobbs HH, Brown MS 1995. Familial hypercholesterolemia. The Metabolic and Molecular Bases of Inherited Disease CR Scriver, AL Beaudet, WS Sly, D Valle 1981–2030 New York: McGraw-Hill
    [Google Scholar]
  12. 12.  Brown MS, Goldstein JL 1975. Regulation of the activity of the low density lipoprotein receptor in human fibroblasts. Cell 6:307–16
    [Google Scholar]
  13. 13.  Brown MS, Goldstein JL 2009. Cholesterol feedback: from Schoenheimer's bottle to Scap's MELADL. J. Lipid Res. 50:S15–27
    [Google Scholar]
  14. 14.  Südhof TC, van der Westhuyzen DR, Goldstein JL, Brown MS, Russell DW 1987. Three direct repeats and a TATA-like sequence are required for regulated expression of the human LDL receptor gene. J. Biol. Chem. 262:10773–79
    [Google Scholar]
  15. 15.  Briggs MR, Yokoyama C, Wang X, Brown MS, Goldstein JL 1993. Nuclear protein that binds sterol regulatory element of low density lipoprotein receptor promoter: I. Identification of the protein and delineation of its target nucleotide sequence. J. Biol. Chem. 268:14490–96
    [Google Scholar]
  16. 16.  Wang X, Briggs MR, Hua X, Yokoyama C, Goldstein JL, Brown MS 1993. Nuclear protein that binds sterol regulatory element of low density lipoprotein receptor promoter: II. Purification and characterization. J. Biol. Chem. 268:14497–504
    [Google Scholar]
  17. 17.  Yokoyama C, Wang X, Briggs MR, Admon A, Wu J et al. 1993. SREBP-1, a basic helix-loop-helix leucine zipper protein that controls transcription of the LDL receptor gene. Cell 75:187–97
    [Google Scholar]
  18. 18.  Wang X, Sato R, Brown MS, Hua X, Goldstein JL 1994. SREBP-1, a membrane-bound transcription factor released by sterol-regulated proteolysis. Cell 77:53–62
    [Google Scholar]
  19. 19.  Hua X, Sakai J, Ho YK, Goldstein JL, Brown MS 1995. Hairpin orientation of sterol regulatory element binding protein-2 in cell membranes as determined by protease protection. J. Biol. Chem. 270:29422–27
    [Google Scholar]
  20. 20.  Hua X, Yokoyama C, Wu J, Briggs MR, Brown MS et al. 1993. SREBP-2, a second basic–helix–loop–helix–leucine zipper protein that stimulates transcription by binding to a sterol regulatory element. PNAS 90:11603–7
    [Google Scholar]
  21. 21.  Goldstein JL, DeBose-Boyd RA, Brown MS 2006. Protein sensors for membrane sterols. Cell 124:35–46
    [Google Scholar]
  22. 22.  Horton JD, Goldstein JL, Brown MS 2002. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Investig. 109:1125–31
    [Google Scholar]
  23. 23.  Sakai J, Duncan EA, Rawson RB, Hua X, Brown MS, Goldstein JL 1996. Sterol-regulated release of SREBP-2 from cell membranes requires two sequential cleavages, one within a transmembrane segment. Cell 85:1037–46
    [Google Scholar]
  24. 24.  Rawson RB, Zelenski NG, Nijhawan D, Ye J, Sakai J et al. 1997. Complementation cloning of S2P, a gene encoding a putative metalloprotease required for intramembrane cleavage of SREBPs. Mol. Cell 1:47–57
    [Google Scholar]
  25. 25.  Sakai J, Rawson RB, Espenshade PJ, Cheng D, Seegmiller AC et al. 1998. Molecular identification of the sterol-regulated luminal protease that cleaves SREBPs and controls lipid composition of animal cells. Mol. Cell 2:505–14
    [Google Scholar]
  26. 26.  Chang TY, Limanek JS 1980. Regulation of cytosolic acetoacetyl coenzyme A thiolase, 3-hydroxy-3-methylglutaryl coenzyme A synthase, 3-hydroxy-3-methylglutaryl coenzyme A reductase, and mevalonate kinase by low density lipoprotein and by 25-hydroxycholesterol in Chinese hamster ovary cells. J. Biol. Chem. 255:7787–95
    [Google Scholar]
  27. 27.  Hasan MT, Chang TY 1994. Somatic cell genetic analysis of two classes of CHO cell mutants expressing opposite phenotypes in sterol-dependent regulation of cholesterol metabolism. Somat. Cell Mol. Genet. 20:481–91
    [Google Scholar]
  28. 28.  Gil G, Faust JR, Chin DJ, Goldstein JL, Brown MS 1985. Membrane-bound domain of HMG CoA reductase is required for sterol-enhanced degradation of the enzyme. Cell 41:249–58
    [Google Scholar]
  29. 29.  Nohturfft A, Brown MS, Goldstein JL 1998. Topology of SREBP cleavage-activating protein, a polytopic membrane protein with a sterol-sensing domain. J. Biol. Chem. 273:17243–50
    [Google Scholar]
  30. 30.  Olender EH, Simoni RD 1992. The intracellular targeting and membrane topology of 3-hydroxy-3-methylglutaryl-CoA reductase. J. Biol. Chem. 267:4223–35
    [Google Scholar]
  31. 31.  Sakai J, Nohturfft A, Cheng D, Ho YK, Brown MS, Goldstein JL 1997. Identification of complexes between the COOH-terminal domains of sterol regulatory element-binding proteins (SREBPs) and SREBP cleavage-activating protein (SCAP). J. Biol. Chem. 272:20213–21
    [Google Scholar]
  32. 32.  Sakai J, Nohturfft A, Goldstein JL, Brown MS 1998. Cleavage of sterol regulatory element binding proteins (SREBPs) at site-1 requires interaction with SREBP cleavage-activating protein. Evidence from in vivo competition studies. J. Biol. Chem. 273:5785–93
    [Google Scholar]
  33. 33.  Nohturfft A, Brown MS, Goldstein JL 1998. Sterols regulate processing of carbohydrate chains of wild-type SREBP cleavage-activating protein (SCAP), but not sterol-resistant mutants Y298C or D443N. PNAS 95:12848–53
    [Google Scholar]
  34. 34.  Yabe D, Xia Z-P, Adams CM, Rawson RB 2002. Three mutations in sterol-sensing domain of SCAP block interaction with Insig and render SREBP cleavage insensitive to sterols. PNAS 99:16672–77
    [Google Scholar]
  35. 35.  Nohturfft A, Yabe D, Goldstein JL, Brown MS, Espenshade PJ 2000. Regulated step in cholesterol feedback localized to budding of SCAP from ER membranes. Cell 102:315–23
    [Google Scholar]
  36. 36.  DeBose-Boyd RA, Brown MS, Li W-P, Nohturfft A, Goldstein JL, Espenshade PJ 1999. Transport-dependent proteolysis of SREBP: Relocation of site-1 protease from Golgi to ER obviates the need for SREBP transport to Golgi. Cell 99:703–12
    [Google Scholar]
  37. 37.  Brown AJ, Sun L, Feramisco JD, Brown MS, Goldstein JL 2002. Cholesterol addition to ER membranes alters conformation of SCAP, the SREBP escort protein that regulates cholesterol metabolism. Mol. Cell 10:237–45
    [Google Scholar]
  38. 38.  Yang T, Goldstein JL, Brown MS 2000. Overexpression of membrane domain of SCAP prevents sterols from inhibiting SCAP/SREBP exit from endoplasmic reticulum. J. Biol. Chem. 275:29881–86
    [Google Scholar]
  39. 39.  Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y et al. 2002. Crucial step in cholesterol homeostasis: sterols promote binding of SCAP to INSIG-1, a membrane protein that facilitates retention of SREBPs in ER. Cell 110:489–500
    [Google Scholar]
  40. 40.  Mohn KL, Laz TM, Hsu J-C, Melby AE, Bravo R, Taub R 1991. The immediate-early growth response in regenerating liver and insulin-stimulated H-35 cells: comparison with serum-stimulated 3T3 cells and identification of 41 novel immediate-early genes. Mol. Cell Biol. 11:381–90
    [Google Scholar]
  41. 41.  Luong A. 2000. Identification of three novel SREBP-activated target genes: acetyl CoA synthetase, 3-β-hydroxysterol dehydrogenase, and CL-6/Insig1 PhD Thesis Univ. Tex. Southwest. Med. Cent. Dallas:
  42. 42.  Yabe D, Brown MS, Goldstein JL 2002. Insig-2, a second endoplasmic reticulum protein that binds SCAP and blocks export of sterol regulatory element-binding proteins. PNAS 99:12753–58
    [Google Scholar]
  43. 43.  Feramisco JD, Goldstein JL, Brown MS 2004. Membrane topology of human Insig-1, a protein regulator of lipid synthesis. J. Biol. Chem. 279:8487–96
    [Google Scholar]
  44. 44.  Gong Y, Lee JN, Lee PCW, Goldstein JL, Brown MS, Ye J 2006. Sterol-regulated ubiquitination and degradation of Insig-1 creates a convergent mechanism for feedback control of cholesterol synthesis and uptake. Cell Metab 3:15–24
    [Google Scholar]
  45. 45.  Kandutsch AA, Chen HW 1974. Inhibition of sterol synthesis in cultured mouse cells by cholesterol derivatives oxygenated in the side chain. J. Biol. Chem. 249:6057–61
    [Google Scholar]
  46. 46.  Brown MS, Goldstein JL 1974. Suppression of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity and inhibition of growth of human fibroblasts by 7-ketocholesterol. J. Biol. Chem. 249:7306–14
    [Google Scholar]
  47. 47.  Adams CM, Reitz J, DeBrabander JK, Feramisco JD, Li L et al. 2004. Cholesterol and 25-hydroxycholesterol inhibit activation of SREBPs by different mechanisms, both involving SCAP and Insigs. J. Biol. Chem. 279:52772–80
    [Google Scholar]
  48. 48.  Radhakrishnan A, Sun L-P, Kwon HJ, Brown MS, Goldstein JL 2004. Direct binding of cholesterol to the purified membrane region of SCAP: mechanism for a sterol-sensing domain. Mol. Cell 15:259–68
    [Google Scholar]
  49. 49.  Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL 2007. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Oxysterols block transport by binding to Insig. PNAS 104:6511–18
    [Google Scholar]
  50. 50.  Dobrosotskaya I, Goldstein JL, Brown MS, Rawson RB 2003. Reconstitution of sterol-regulated endoplasmic reticulum-to-Golgi transport of SREBP-2 in insect cells by co-expression of mammalian SCAP and Insigs. J. Biol. Chem. 278:35837–43
    [Google Scholar]
  51. 51.  Sun L-P, Seemann J, Brown MS, Goldstein JL 2007. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins. PNAS 104:6519–26
    [Google Scholar]
  52. 52.  Zambrano F, Fleischer S, Fleischer B 1975. Lipid composition of the Golgi apparatus of rat kidney and liver in comparison with other subcellular organelles. Biochim. Biophys. Acta 380:357–69
    [Google Scholar]
  53. 53.  Lange Y, Steck TL 1997. Quantitation of the pool of cholesterol associated with acyl-CoA: cholesterol acyltransferase in human fibroblasts. J. Biol. Chem. 272:13103–8
    [Google Scholar]
  54. 54.  Gong X, Qian H, Shao W, Li J, Wu J et al. 2016. Complex structure of the fission yeast SREBP-SCAP binding domains reveals an oligomeric organization. Cell Res 26:1197–211
    [Google Scholar]
  55. 55.  Espenshade PJ, Li W-P, Yabe D 2002. Sterols block binding of COPII proteins to SCAP, thereby controlling SCAP sorting in ER. PNAS 99:11694–99
    [Google Scholar]
  56. 56.  Sun L-P, Li L, Goldstein JL, Brown MS 2005. Insig required for sterol-mediated inhibition of Scap/SREBP binding to COPII proteins in vitro. J. Biol. Chem. 280:26483–90
    [Google Scholar]
  57. 57.  Feramisco JD, Radhakrishnan A, Ikeda Y, Reitz J, Brown MS, Goldstein JL 2005. Intramembrane aspartic acid in SCAP protein governs cholesterol-induced conformational change. PNAS 102:3242–47
    [Google Scholar]
  58. 58.  Sever N, Yang T, Brown MS, Goldstein JL, DeBose-Boyd RA 2003. Accelerated degradation of HMG CoA reductase mediated by binding of Insig-1 to its sterol-sensing domain. Mol. Cell 11:25–33
    [Google Scholar]
  59. 59.  Sever N, Song B-L, Yabe D, Goldstein JL, Brown MS, DeBose-Boyd RA 2003. Insig-dependent ubiquitination and degradation of mammalian 3-hydroxy-3-methylglutaryl-CoA reductase stimulated by sterols and geranylgeraniol. J. Biol. Chem. 278:52479–90
    [Google Scholar]
  60. 60.  Zhang Y, Lee KM, Kinch LN, Clark L, Grishin NV et al. 2016. Direct demonstration that Loop1 of Scap binds to Loop7, a crucial event in cholesterol homeostasis. J. Biol. Chem. 291:12888–96
    [Google Scholar]
  61. 61.  Motamed M, Zhang Y, Wang ML, Seemann J, Kwon HJ et al. 2011. Identification of luminal Loop 1 of Scap protein as the sterol sensor that maintains cholesterol homeostasis. J. Biol. Chem. 286:18002–12
    [Google Scholar]
  62. 62.  Zhang Y, Motamed M, Seemann J, Brown MS, Goldstein JL 2013. Point mutation in luminal Loop 7 of Scap blocks interaction with Loop 1 and abolishes movement to Golgi. J. Biol. Chem. 288:14059–67
    [Google Scholar]
  63. 63.  Gao Y, Zhou Y, Goldstein JL, Brown MS, Radhakrishnan A 2017. Cholesterol-induced conformational changes in the sterol-sensing domain of the Scap protein suggest feedback mechanism to control cholesterol synthesis. J. Biol. Chem. 292:8729–37
    [Google Scholar]
  64. 64.  Lee PCW, Sever N, DeBose-Boyd RA 2005. Isolation of sterol-resistant Chinese hamster ovary cells with genetic deficiencies in both Insig-1 and Insig-2. J. Biol. Chem. 280:25242–49
    [Google Scholar]
  65. 65.  Prabhu AV, Luu W, Li D, Sharpe LJ, Brown AJ 2016. DHCR7: a vital enzyme switch between cholesterol and vitamin D production. Prog. Lipid Res. 64:138–51
    [Google Scholar]
  66. 66.  Carstea ED, Morris JA, Coleman KG, Loftus SK, Zhang D et al. 1997. Niemann-Pick C1 disease gene: homology to mediators of cholesterol homeostasis. Science 277:228–31
    [Google Scholar]
  67. 67.  Altmann SW, Davis HR Jr., Zhu L-J, Yao X, Hoos LM et al. 2004. Niemann-Pick C1 Like 1 protein is critical for intestinal cholesterol absorption. Science 303:1201–4
    [Google Scholar]
  68. 68.  Burke R, Nellen D, Bellotto M, Hafen E, Senti K-A et al. 1999. Dispatched, a novel sterol-sensing domain protein dedicated to the release of cholesterol-modified hedgehog from signaling cells. Cell 99:803–15
    [Google Scholar]
  69. 69.  Jo Y, Lee PCW, Sguigna PV, DeBose-Boyd RA 2011. Sterol-induced degradation of HMG CoA reductase depends on interplay of two Insigs and two ubiquitin ligases, gp78 and Trc8. PNAS 108:20503–8
    [Google Scholar]
  70. 70.  Lin PH, Lan WM, Chau LY 2013. TRC8 suppresses tumorigenesis through targeting heme oxygenase-1 for ubiquitination and degradation. Oncogene 32:2325–34
    [Google Scholar]
  71. 71.  Kuwabara PE, Labouesse M 2002. The sterol-sensing domain: multiple families, a unique role?. Trends Genet 18:193–201
    [Google Scholar]
/content/journals/10.1146/annurev-biochem-062917-011852
Loading
/content/journals/10.1146/annurev-biochem-062917-011852
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error