Skip to main content

Advertisement

Log in

Regulation of cholesterol homeostasis

  • Review
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Cholesterol homeostasis is among the most intensely regulated processes in biology. Since its isolation from gallstones at the time of the French Revolution, cholesterol has been extensively studied. Insufficient or excessive cellular cholesterol results in pathological processes including atherosclerosis and metabolic syndrome. Mammalian cells obtain cholesterol from the circulation in the form of plasma lipoproteins or intracellularly, through the synthesis of cholesterol from acetyl coenzyme A (acetyl-CoA). This process is tightly regulated at multiple levels. In this review, we provide an overview of the multiple mechanisms by which cellular cholesterol metabolism is regulated. We also discuss the recent advances in the post-transcriptional regulation of cholesterol homeostasis, including the role of small non-coding RNAs (microRNAs). These novel findings may open new avenues for the treatment of dyslipidemias and cardiovascular diseases.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Vance DE, Van den Bosch H (2000) Cholesterol in the year. Biochim Biophys Acta 1529(1–3):1–8

    PubMed  CAS  Google Scholar 

  2. Fernandez C, Lobo Md Mdel V, Gomez-Coronado D, Lasuncion MA (2004) Cholesterol is essential for mitosis progression and its deficiency induces polyploid cell formation. Exp Cell Res 300(1):109–120

    Article  PubMed  CAS  Google Scholar 

  3. Fernandez C, Martin M, Gomez-Coronado D, Lasuncion MA (2005) Effects of distal cholesterol biosynthesis inhibitors on cell proliferation and cell cycle progression. J Lipid Res 46(5):920–929

    Article  PubMed  CAS  Google Scholar 

  4. Ikonen E (2006) Mechanisms for cellular cholesterol transport: defects and human disease. Physiol Rev 86(4):1237–1261

    Article  PubMed  CAS  Google Scholar 

  5. Maxfield FR, Tabas I (2005) Role of cholesterol and lipid organization in disease. Nature 438(7068):612–621

    Article  PubMed  CAS  Google Scholar 

  6. Bloch K (1992) Sterol molecule: structure, biosynthesis, and function. Steroids 57(8):378–383

    Article  PubMed  CAS  Google Scholar 

  7. Ponticorvo L, Rittenberg D, Bloch K (1949) The utilization of acetate for the synthesis of fatty acids, cholesterol, and protoporphyrin. J Biol Chem 179(2):839–842

    PubMed  CAS  Google Scholar 

  8. Brown MS, Goldstein JL (1976) Receptor-mediated control of cholesterol metabolism. Science 191(4223):150–154

    Article  PubMed  CAS  Google Scholar 

  9. Brown MS, Goldstein JL (1986) A receptor-mediated pathway for cholesterol homeostasis. Science 232(4746):34–47

    Article  PubMed  CAS  Google Scholar 

  10. Brown MS, Goldstein JL (1997) The SREBP pathway: regulation of cholesterol metabolism by proteolysis of a membrane-bound transcription factor. Cell 89(3):331–340

    Article  PubMed  CAS  Google Scholar 

  11. Goldstein JL, Brown MS (1990) Regulation of the mevalonate pathway. Nature 343(6257):425–430

    Article  PubMed  CAS  Google Scholar 

  12. Sakakura Y, Shimano H, Sone H, Takahashi A, Inoue N, Toyoshima H, Suzuki S, Yamada N (2001) Sterol regulatory element-binding proteins induce an entire pathway of cholesterol synthesis. Biochem Biophys Res Commun 286(1):176–183

    Article  PubMed  CAS  Google Scholar 

  13. Beaven SW, Tontonoz P (2006) Nuclear receptors in lipid metabolism: targeting the heart of dyslipidemia. Annu Rev Med 57:313–329

    Article  PubMed  CAS  Google Scholar 

  14. Peet DJ, Janowski BA, Mangelsdorf DJ (1998) The LXRs: a new class of oxysterol receptors. Curr Opin Genet Dev 8(5):571–575

    Article  PubMed  CAS  Google Scholar 

  15. Tontonoz P, Mangelsdorf DJ (2003) Liver X receptor signaling pathways in cardiovascular disease. Mol Endocrinol 17(6):985–993

    Article  PubMed  CAS  Google Scholar 

  16. Gerin I, Clerbaux LA, Haumont O, Lanthier N, Das AK, Burant CF, Leclercq IA, Macdougald OA, Bommer GT (2010) Expression of miR-33 from an SREBP2 intron inhibits cholesterol export and fatty acid oxidation. J Biol Chem 285(44):33652–33661

    Article  PubMed  CAS  Google Scholar 

  17. Horie T, Ono K, Horiguchi M, Nishi H, Nakamura T, Nagao K, Kinoshita M, Kuwabara Y, Marusawa H, Iwanaga Y, Hasegawa K, Yokode M, Kimura T, Kita T (2010) MicroRNA-33 encoded by an intron of sterol regulatory element-binding protein 2 (Srebp2) regulates HDL in vivo. Proc Natl Acad Sci USA 107(40):17321–17326

    Article  PubMed  CAS  Google Scholar 

  18. Marquart TJ, Allen RM, Ory DS, Baldan A (2010) miR-33 links SREBP-2 induction to repression of sterol transporters. Proc Natl Acad Sci USA 107(27):12228–12232

    Article  PubMed  CAS  Google Scholar 

  19. Najafi-Shoushtari SH, Kristo F, Li Y, Shioda T, Cohen DE, Gerszten RE, Naar AM (2010) MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 328(5985):1566–1569

    Article  PubMed  CAS  Google Scholar 

  20. Rayner KJ, Suarez Y, Davalos A, Parathath S, Fitzgerald ML, Tamehiro N, Fisher EA, Moore KJ, Fernandez-Hernando C (2010) MiR-33 contributes to the regulation of cholesterol homeostasis. Science 328(5985):1570–1573

    Article  PubMed  CAS  Google Scholar 

  21. Davalos A, Goedeke L, Smibert P, Ramirez CM, Warrier NP, Andreo U, Cirera-Salinas D, Rayner K, Suresh U, Pastor-Pareja JC, Esplugues E, Fisher EA, Penalva LO, Moore KJ, Suarez Y, Lai EC, Fernandez-Hernando C (2011) miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc Natl Acad Sci USA 108 (22):9232–9237

    Google Scholar 

  22. Gerin I, Clerbaux LA, Haumont O, Lanthier N, Das AK, Burant CF, Leclercq IA, MacDougald OA, Bommer GT (2010) Expression of miR-33 from an SREBP2 intron inhibits cholesterol export and fatty acid oxidation. J Biol Chem 285(44):33652–33661

    Article  PubMed  CAS  Google Scholar 

  23. Chen T, Huang Z, Wang L, Wang Y, Wu F, Meng S, Wang C (2009) MicroRNA-125a–5p partly regulates the inflammatory response, lipid uptake, and ORP9 expression in oxLDL-stimulated monocyte/macrophages. Cardiovasc Res 83(1):131–139

    Article  PubMed  CAS  Google Scholar 

  24. Elmen J, Lindow M, Schutz S, Lawrence M, Petri A, Obad S, Lindholm M, Hedtjarn M, Hansen HF, Berger U, Gullans S, Kearney P, Sarnow P, Straarup EM, Kauppinen S (2008) LNA-mediated microRNA silencing in non-human primates. Nature 452(7189):896–899

    Article  PubMed  CAS  Google Scholar 

  25. Esau C, Davis S, Murray SF, Yu XX, Pandey SK, Pear M, Watts L, Booten SL, Graham M, McKay R, Subramaniam A, Propp S, Lollo BA, Freier S, Bennett CF, Bhanot S, Monia BP (2006) miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab 3(2):87–98

    Article  PubMed  CAS  Google Scholar 

  26. Gerin I, Bommer GT, McCoin CS, Sousa KM, Krishnan V, MacDougald OA (2010) Roles for miRNA-378/378* in adipocyte gene expression and lipogenesis. Am J Physiol Endocrinol Metab 299 (2):E198–E206

    Google Scholar 

  27. Iliopoulos D, Drosatos K, Hiyama Y, Goldberg IJ, Zannis VI (2010) MicroRNA-370 controls the expression of microRNA-122 and Cpt1alpha and affects lipid metabolism. J Lipid Res 51 (6):1513–1523

    Google Scholar 

  28. Lin Q, Gao Z, Alarcon RM, Ye J, Yun Z (2009) A role of miR-27 in the regulation of adipogenesis. FEBS J 276(8):2348–2358

    Article  PubMed  CAS  Google Scholar 

  29. Nakanishi N, Nakagawa Y, Tokushige N, Aoki N, Matsuzaka T, Ishii K, Yahagi N, Kobayashi K, Yatoh S, Takahashi A, Suzuki H, Urayama O, Yamada N, Shimano H (2009) The up-regulation of microRNA-335 is associated with lipid metabolism in liver and white adipose tissue of genetically obese mice. Biochem Biophys Res Commun 385(4):492–496

    Article  PubMed  CAS  Google Scholar 

  30. Dietschy JM, Turley SD, Spady DK (1993) Role of liver in the maintenance of cholesterol and low-density lipoprotein homeostasis in different animal species, including humans. J Lipid Res 34(10):1637–1659

    PubMed  CAS  Google Scholar 

  31. Bloch K (1987) Summing up. Annu Rev Biochem 56:1–19

    Article  PubMed  CAS  Google Scholar 

  32. Jasinska M, Owczarek J, Orszulak-Michalak D (2007) Statins: a new insight into their mechanisms of action and consequent pleiotropic effects. Pharmacol Rep 59(5):483–499

    PubMed  CAS  Google Scholar 

  33. Kandutsch AA, Russell AE (1960) Preputial gland tumor sterols. 3. A metabolic pathway from lanosterol to cholesterol. J Biol Chem 235:2256–2261

    PubMed  CAS  Google Scholar 

  34. Baumann NA, Sullivan DP, Ohvo-Rekila H, Simonot C, Pottekat A, Klaassen Z, Beh CT, Menon AK (2005) Transport of newly synthesized sterol to the sterol-enriched plasma membrane occurs via nonvesicular equilibration. Biochemistry 44(15):5816–5826

    Article  PubMed  CAS  Google Scholar 

  35. Chang TY, Chang CC, Cheng D (1997) Acyl-coenzyme A: cholesterol acyltransferase. Annu Rev Biochem 66:613–638

    Article  PubMed  CAS  Google Scholar 

  36. Brown MS, Goldstein JL (1980) Multivalent feedback regulation of HMG CoA reductase, a control mechanism coordinating isoprenoid synthesis and cell growth. J Lipid Res 21(5):505–517

    PubMed  CAS  Google Scholar 

  37. Hooper NM (1999) Detergent-insoluble glycosphingolipid/cholesterol-rich membrane domains, lipid rafts and caveolae (review). Mol Membr Biol 16(2):145–156

    Article  PubMed  CAS  Google Scholar 

  38. Nwokoro NA, Wassif CA, Porter FD (2001) Genetic disorders of cholesterol biosynthesis in mice and humans. Mol Genet Metab 74(1–2):105–119

    Article  PubMed  CAS  Google Scholar 

  39. Smith DW, Lemli L, Opitz JM (1964) A newly recognized syndrome of multiple congenital anomalies. J Pediatr 64:210–217

    Article  PubMed  CAS  Google Scholar 

  40. Brunetti-Pierri N, Corso G, Rossi M, Ferrari P, Balli F, Rivasi F, Annunziata I, Ballabio A, Russo AD, Andria G, Parenti G (2002) Lathosterolosis, a novel multiple-malformation/mental retardation syndrome due to deficiency of 3beta-hydroxysteroid-delta5-desaturase. Am J Hum Genet 71(4):952–958

    Article  PubMed  Google Scholar 

  41. Dietschy JM, Turley SD (2001) Cholesterol metabolism in the brain. Curr Opin Lipidol 12(2):105–112

    Article  PubMed  CAS  Google Scholar 

  42. Jurevics H, Morell P (1995) Cholesterol for synthesis of myelin is made locally, not imported into brain. J Neurochem 64(2):895–901

    Article  PubMed  CAS  Google Scholar 

  43. Snipes GJ, Suter U (1997) Cholesterol and myelin. Subcell Biochem 28:173–204

    Article  PubMed  CAS  Google Scholar 

  44. Fernandez-Hernando C, Suarez Y, Lasuncion MA (2005) Lovastatin-induced PC-12 cell differentiation is associated with RhoA/RhoA kinase pathway inactivation. Mol Cell Neurosci 29(4):591–602

    Article  PubMed  CAS  Google Scholar 

  45. Hayashi H, Campenot RB, Vance DE, Vance JE (2004) Glial lipoproteins stimulate axon growth of central nervous system neurons in compartmented cultures. J Biol Chem 279(14):14009–14015

    Article  PubMed  CAS  Google Scholar 

  46. Holtzman DM, Pitas RE, Kilbridge J, Nathan B, Mahley RW, Bu G, Schwartz AL (1995) Low-density lipoprotein receptor-related protein mediates apolipoprotein E-dependent neurite outgrowth in a central nervous system-derived neuronal cell line. Proc Natl Acad Sci USA 92(21):9480–9484

    Article  PubMed  CAS  Google Scholar 

  47. Block RC, Dorsey ER, Beck CA, Brenna JT, Shoulson I Altered cholesterol and fatty acid metabolism in Huntington disease. J Clin Lipidol 4 (1):17-23

  48. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA (1993) Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer’s disease in late onset families. Science 261(5123):921–923

    Article  PubMed  CAS  Google Scholar 

  49. Strittmatter WJ, Saunders AM, Schmechel D, Pericak-Vance M, Enghild J, Salvesen GS, Roses AD (1993) Apolipoprotein E: high-avidity binding to beta-amyloid and increased frequency of type 4 allele in late-onset familial Alzheimer disease. Proc Natl Acad Sci USA 90(5):1977–1981

    Article  PubMed  CAS  Google Scholar 

  50. Goldstein JL, Brown MS (1982) The LDL receptor defect in familial hypercholesterolemia. Implications for pathogenesis and therapy. Med Clin North Am 66 (2):335-362

    Google Scholar 

  51. Grundy SM (1983) Absorption and metabolism of dietary cholesterol. Annu Rev Nutr 3:71–96

    Article  PubMed  CAS  Google Scholar 

  52. Linder MD, Uronen RL, Holtta-Vuori M, van der Sluijs P, Peranen J, Ikonen E (2007) Rab8-dependent recycling promotes endosomal cholesterol removal in normal and sphingolipidosis cells. Mol Biol Cell 18(1):47–56

    Article  PubMed  CAS  Google Scholar 

  53. Holtta-Vuori M, Tanhuanpaa K, Mobius W, Somerharju P, Ikonen E (2002) Modulation of cellular cholesterol transport and homeostasis by Rab11. Mol Biol Cell 13(9):3107–3122

    Article  PubMed  CAS  Google Scholar 

  54. Ory DS (2004) The Niemann–Pick disease genes; regulators of cellular cholesterol homeostasis. Trends Cardiovasc Med 14(2):66–72

    Article  PubMed  CAS  Google Scholar 

  55. Wang ML, Motamed M, Infante RE, Abi-Mosleh L, Kwon HJ, Brown MS, Goldstein JL (2010) Identification of surface residues on Niemann–Pick C2 essential for hydrophobic handoff of cholesterol to NPC1 in lysosomes. Cell Metab 12(2):166–173

    Article  PubMed  CAS  Google Scholar 

  56. Lebrand C, Corti M, Goodson H, Cosson P, Cavalli V, Mayran N, Faure J, Gruenberg J (2002) Late endosome motility depends on lipids via the small GTPase Rab7. EMBO J 21(6):1289–1300

    Article  PubMed  CAS  Google Scholar 

  57. Ganley IG, Pfeffer SR (2006) Cholesterol accumulation sequesters Rab9 and disrupts late endosome function in NPC1-deficient cells. J Biol Chem 281(26):17890–17899

    Article  PubMed  CAS  Google Scholar 

  58. Maxfield FR, Wustner D (2002) Intracellular cholesterol transport. J Clin Invest 110(7):891–898

    PubMed  CAS  Google Scholar 

  59. Maxfield FR, Mondal M (2006) Sterol and lipid trafficking in mammalian cells. Biochem Soc Trans 34(Pt 3):335–339

    PubMed  CAS  Google Scholar 

  60. Soccio RE, Breslow JL (2004) Intracellular cholesterol transport. Arterioscler Thromb Vasc Biol 24(7):1150–1160

    Article  PubMed  CAS  Google Scholar 

  61. Soccio RE, Breslow JL (2003) StAR-related lipid transfer (START) proteins: mediators of intracellular lipid metabolism. J Biol Chem 278(25):22183–22186

    Article  PubMed  CAS  Google Scholar 

  62. Seedorf U, Ellinghaus P, Roch Nofer J (2000) Sterol carrier protein-2. Biochim Biophys Acta 1486(1):45–54

    PubMed  CAS  Google Scholar 

  63. Gallegos AM, Atshaves BP, Storey SM, Starodub O, Petrescu AD, Huang H, McIntosh AL, Martin GG, Chao H, Kier AB, Schroeder F (2001) Gene structure, intracellular localization, and functional roles of sterol carrier protein-2. Prog Lipid Res 40(6):498–563

    Article  PubMed  CAS  Google Scholar 

  64. Prinz WA (2007) Non-vesicular sterol transport in cells. Prog Lipid Res 46(6):297–314

    Article  PubMed  CAS  Google Scholar 

  65. Martin S, Parton RG (2005) Caveolin, cholesterol, and lipid bodies. Semin Cell Dev Biol 16(2):163–174

    Article  PubMed  CAS  Google Scholar 

  66. Parton RG, Simons K (2007) The multiple faces of caveolae. Nat Rev Mol Cell Biol 8(3):185–194

    Article  PubMed  CAS  Google Scholar 

  67. Uittenbogaard A, Smart EJ (2000) Palmitoylation of caveolin-1 is required for cholesterol binding, chaperone complex formation, and rapid transport of cholesterol to caveolae. J Biol Chem 275(33):25595–25599

    Article  PubMed  CAS  Google Scholar 

  68. Uittenbogaard A, Ying Y, Smart EJ (1998) Characterization of a cytosolic heat-shock protein-caveolin chaperone complex. Involvement in cholesterol trafficking. J Biol Chem 273(11):6525–6532

    Article  PubMed  CAS  Google Scholar 

  69. Heino S, Lusa S, Somerharju P, Ehnholm C, Olkkonen VM, Ikonen E (2000) Dissecting the role of the Golgi complex and lipid rafts in biosynthetic transport of cholesterol to the cell surface. Proc Natl Acad Sci U S A 97(15):8375–8380

    Article  PubMed  CAS  Google Scholar 

  70. Kandutsch AA, Shown EP (1981) Assay of oxysterol-binding protein in a mouse fibroblast, cell-free system. Dissociation constant and other properties of the system. J Biol Chem 256(24):13068–13073

    PubMed  CAS  Google Scholar 

  71. Wang C, JeBailey L, Ridgway ND (2002) Oxysterol-binding-protein (OSBP)-related protein 4 binds 25-hydroxycholesterol and interacts with vimentin intermediate filaments. Biochem J 361(Pt 3):461–472

    Article  PubMed  CAS  Google Scholar 

  72. Ngo M, Ridgway ND (2009) Oxysterol binding protein-related Protein 9 (ORP9) is a cholesterol transfer protein that regulates Golgi structure and function. Mol Biol Cell 20(5):1388–1399

    Article  PubMed  CAS  Google Scholar 

  73. Johansson M, Bocher V, Lehto M, Chinetti G, Kuismanen E, Ehnholm C, Staels B, Olkkonen VM (2003) The two variants of oxysterol binding protein-related protein-1 display different tissue expression patterns, have different intracellular localization, and are functionally distinct. Mol Biol Cell 14(3):903–915

    Article  PubMed  CAS  Google Scholar 

  74. Du X, Kumar J, Ferguson C, Schulz TA, Ong YS, Hong W, Prinz WA, Parton RG, Brown AJ, Yang H (2011) A role for oxysterol-binding protein-related protein 5 in endosomal cholesterol trafficking. J Cell Biol 192(1):121–135

    Article  PubMed  CAS  Google Scholar 

  75. Ponting CP, Aravind L (1999) START: a lipid-binding domain in StAR, HD-ZIP and signalling proteins. Trends Biochem Sci 24(4):130–132

    Article  PubMed  CAS  Google Scholar 

  76. Stocco DM (2001) StAR protein and the regulation of steroid hormone biosynthesis. Annu Rev Physiol 63:193–213

    Article  PubMed  CAS  Google Scholar 

  77. Hanada K, Kumagai K, Yasuda S, Miura Y, Kawano M, Fukasawa M, Nishijima M (2003) Molecular machinery for non-vesicular trafficking of ceramide. Nature 426 (6968):803–809

    Google Scholar 

  78. Rigotti A, Cohen DE, Zanlungo S (2010) STARTing to understand MLN64 function in cholesterol transport. J Lipid Res 51(8):2015–2017

    Article  PubMed  CAS  Google Scholar 

  79. Charman M, Kennedy BE, Osborne N, Karten B (2010) MLN64 mediates egress of cholesterol from endosomes to mitochondria in the absence of functional Niemann–Pick Type C1 protein. J Lipid Res 51(5):1023–1034

    Article  PubMed  CAS  Google Scholar 

  80. Glomset JA, Norum KR (1973) The metabolic role of lecithin: cholesterol acyltransferase: perspectives form pathology. Adv Lipid Res 11:1–65

    CAS  Google Scholar 

  81. Cuchel M, Rader DJ (2006) Macrophage reverse cholesterol transport: key to the regression of atherosclerosis? Circulation 113(21):2548–2555

    Article  PubMed  Google Scholar 

  82. Oram JF, Yokoyama S (1996) Apolipoprotein-mediated removal of cellular cholesterol and phospholipids. J Lipid Res 37(12):2473–2491

    PubMed  CAS  Google Scholar 

  83. Yokoyama S (1998) Apolipoprotein-mediated cellular cholesterol efflux. Biochim Biophys Acta 1392(1):1–15

    PubMed  CAS  Google Scholar 

  84. Yokoyama S (2000) Release of cellular cholesterol: molecular mechanism for cholesterol homeostasis in cells and in the body. Biochim Biophys Acta 1529(1–3):231–244

    PubMed  CAS  Google Scholar 

  85. Jonas A (2000) Lecithin cholesterol acyltransferase. Biochim Biophys Acta 1529(1–3):245–256

    PubMed  CAS  Google Scholar 

  86. Ji Y, Jian B, Wang N, Sun Y, Moya ML, Phillips MC, Rothblat GH, Swaney JB, Tall AR (1997) Scavenger receptor BI promotes high-density lipoprotein-mediated cellular cholesterol efflux. J Biol Chem 272(34):20982–20985

    Article  PubMed  CAS  Google Scholar 

  87. Tanigawa H, Billheimer JT, Tohyama J, Fuki IV, Ng DS, Rothblat GH, Rader DJ (2009) Lecithin: cholesterol acyltransferase expression has minimal effects on macrophage reverse cholesterol transport in vivo. Circulation 120(2):160–169

    Article  PubMed  CAS  Google Scholar 

  88. Calabresi L, Favari E, Moleri E, Adorni MP, Pedrelli M, Costa S, Jessup W, Gelissen IC, Kovanen PT, Bernini F, Franceschini G (2009) Functional LCAT is not required for macrophage cholesterol efflux to human serum. Atherosclerosis 204(1):141–146

    Article  PubMed  CAS  Google Scholar 

  89. Linsel-Nitschke P, Tall AR (2005) HDL as a target in the treatment of atherosclerotic cardiovascular disease. Nat Rev Drug Discov 4(3):193–205

    Article  PubMed  CAS  Google Scholar 

  90. Berge KE, Tian H, Graf GA, Yu L, Grishin NV, Schultz J, Kwiterovich P, Shan B, Barnes R, Hobbs HH (2000) Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science 290(5497):1771–1775

    Article  PubMed  CAS  Google Scholar 

  91. Bhattacharyya AK, Connor WE (1974) Beta-sitosterolemia and xanthomatosis. A newly described lipid storage disease in two sisters. J Clin Invest 53(4):1033–1043

    Article  PubMed  CAS  Google Scholar 

  92. Miettinen TA (1980) Phytosterolaemia, xanthomatosis and premature atherosclerotic arterial disease: a case with high plant sterol absorption, impaired sterol elimination and low cholesterol synthesis. Eur J Clin Invest 10(1):27–35

    Article  PubMed  CAS  Google Scholar 

  93. Yokoyama S (2006) ABCA1 and biogenesis of HDL. J Atheroscler Thromb 13(1):1–15

    Article  PubMed  CAS  Google Scholar 

  94. Tall AR, Yvan-Charvet L, Terasaka N, Pagler T, Wang N (2008) HDL, ABC transporters, and cholesterol efflux: implications for the treatment of atherosclerosis. Cell Metab 7(5):365–375

    Article  PubMed  CAS  Google Scholar 

  95. Brooks-Wilson A, Marcil M, Clee SM, Zhang LH, Roomp K, van Dam M, Yu L, Brewer C, Collins JA, Molhuizen HO, Loubser O, Ouelette BF, Fichter K, Ashbourne-Excoffon KJ, Sensen CW, Scherer S, Mott S, Denis M, Martindale D, Frohlich J, Morgan K, Koop B, Pimstone S, Kastelein JJ, Genest J Jr, Hayden MR (1999) Mutations in ABC1 in Tangier disease and familial high-density lipoprotein deficiency. Nat Genet 22(4):336–345

    Article  PubMed  CAS  Google Scholar 

  96. Neufeld EB, Remaley AT, Demosky SJ, Stonik JA, Cooney AM, Comly M, Dwyer NK, Zhang M, Blanchette-Mackie J, Santamarina-Fojo S, Brewer HB Jr (2001) Cellular localization and trafficking of the human ABCA1 transporter. J Biol Chem 276(29):27584–27590

    Article  PubMed  CAS  Google Scholar 

  97. Neufeld EB, Stonik JA, Demosky SJ Jr, Knapper CL, Combs CA, Cooney A, Comly M, Dwyer N, Blanchette-Mackie J, Remaley AT, Santamarina-Fojo S, Brewer HB Jr (2004) The ABCA1 transporter modulates late endocytic trafficking: insights from the correction of the genetic defect in Tangier disease. J Biol Chem 279(15):15571–15578

    Article  PubMed  CAS  Google Scholar 

  98. Takahashi Y, Smith JD (1999) Cholesterol efflux to apolipoprotein AI involves endocytosis and resecretion in a calcium-dependent pathway. Proc Natl Acad Sci USA 96(20):11358–11363

    Article  PubMed  CAS  Google Scholar 

  99. Choi HY, Karten B, Chan T, Vance JE, Greer WL, Heidenreich RA, Garver WS, Francis GA (2003) Impaired ABCA1-dependent lipid efflux and hypoalphalipoproteinemia in human Niemann–Pick type C disease. J Biol Chem 278(35):32569–32577

    Article  PubMed  CAS  Google Scholar 

  100. Haidar B, Kiss RS, Sarov-Blat L, Brunet R, Harder C, McPherson R, Marcel YL (2006) Cathepsin D, a lysosomal protease, regulates ABCA1-mediated lipid efflux. J Biol Chem 281(52):39971–39981

    Article  PubMed  CAS  Google Scholar 

  101. Nandi S, Ma L, Denis M, Karwatsky J, Li Z, Jiang XC, Zha X (2009) ABCA1-mediated cholesterol efflux generates microparticles in addition to HDL through processes governed by membrane rigidity. J Lipid Res 50(3):456–466

    Article  PubMed  CAS  Google Scholar 

  102. Vaughan AM, Oram JF (2003) ABCA1 redistributes membrane cholesterol independent of apolipoprotein interactions. J Lipid Res 44(7):1373–1380

    Article  PubMed  CAS  Google Scholar 

  103. Vedhachalam C, Duong PT, Nickel M, Nguyen D, Dhanasekaran P, Saito H, Rothblat GH, Lund-Katz S, Phillips MC (2007) Mechanism of ATP-binding cassette transporter A1-mediated cellular lipid efflux to apolipoprotein A-I and formation of high-density lipoprotein particles. J Biol Chem 282(34):25123–25130

    Article  PubMed  CAS  Google Scholar 

  104. Remaley AT, Stonik JA, Demosky SJ, Neufeld EB, Bocharov AV, Vishnyakova TG, Eggerman TL, Patterson AP, Duverger NJ, Santamarina-Fojo S, Brewer HB Jr (2001) Apolipoprotein specificity for lipid efflux by the human ABCAI transporter. Biochem Biophys Res Commun 280(3):818–823

    Article  PubMed  CAS  Google Scholar 

  105. van der Velde AE (2010) Reverse cholesterol transport: from classical view to new insights. World J Gastroenterol 16(47):5908–5915

    PubMed  Google Scholar 

  106. Gelissen IC, Harris M, Rye KA, Quinn C, Brown AJ, Kockx M, Cartland S, Packianathan M, Kritharides L, Jessup W (2006) ABCA1 and ABCG1 synergize to mediate cholesterol export to apoA-I. Arterioscler Thromb Vasc Biol 26(3):534–540

    Article  PubMed  CAS  Google Scholar 

  107. Vaughan AM, Oram JF (2006) ABCA1 and ABCG1 or ABCG4 act sequentially to remove cellular cholesterol and generate cholesterol-rich HDL. J Lipid Res 47(11):2433–2443

    Article  PubMed  CAS  Google Scholar 

  108. Hirsch-Reinshagen V, Zhou S, Burgess BL, Bernier L, McIsaac SA, Chan JY, Tansley GH, Cohn JS, Hayden MR, Wellington CL (2004) Deficiency of ABCA1 impairs apolipoprotein E metabolism in brain. J Biol Chem 279(39):41197–41207

    Article  PubMed  CAS  Google Scholar 

  109. Wahrle SE, Jiang H, Parsadanian M, Hartman RE, Bales KR, Paul SM, Holtzman DM (2005) Deletion of Abca1 increases Abeta deposition in the PDAPP transgenic mouse model of Alzheimer disease. J Biol Chem 280(52):43236–43242

    Article  PubMed  CAS  Google Scholar 

  110. Wahrle SE, Jiang H, Parsadanian M, Kim J, Li A, Knoten A, Jain S, Hirsch-Reinshagen V, Wellington CL, Bales KR, Paul SM, Holtzman DM (2008) Overexpression of ABCA1 reduces amyloid deposition in the PDAPP mouse model of Alzheimer disease. J Clin Invest 118(2):671–682

    PubMed  CAS  Google Scholar 

  111. Horton JD, Goldstein JL, Brown MS (2002) SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 109(9):1125–1131

    PubMed  CAS  Google Scholar 

  112. Horton JD, Shimomura I (1999) Sterol regulatory element-binding proteins: activators of cholesterol and fatty acid biosynthesis. Curr Opin Lipidol 10(2):143–150

    Article  PubMed  CAS  Google Scholar 

  113. Tontonoz P, Kim JB, Graves RA, Spiegelman BM (1993) ADD1: a novel helix-loop-helix transcription factor associated with adipocyte determination and differentiation. Mol Cell Biol 13(8):4753–4759

    PubMed  CAS  Google Scholar 

  114. Duncan EA, Brown MS, Goldstein JL, Sakai J (1997) Cleavage site for sterol-regulated protease localized to a leu-Ser bond in the lumenal loop of sterol regulatory element-binding protein-2. J Biol Chem 272(19):12778–12785

    Article  PubMed  CAS  Google Scholar 

  115. Hua X, Yokoyama C, Wu J, Briggs MR, Brown MS, Goldstein JL, Wang X (1993) SREBP-2, a second basic-helix-loop-helix-leucine zipper protein that stimulates transcription by binding to a sterol regulatory element. Proc Natl Acad Sci USA 90(24):11603–11607

    Article  PubMed  CAS  Google Scholar 

  116. Yang T, Espenshade PJ, Wright ME, Yabe D, Gong Y, Aebersold R, Goldstein JL, Brown MS (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(4):489–500

    Article  PubMed  CAS  Google Scholar 

  117. 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. Proc Natl Acad Sci USA 104(16):6511–6518

    Article  PubMed  CAS  Google Scholar 

  118. Sun LP, Seemann J, Goldstein JL, Brown MS (2007) Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: Insig renders sorting signal in Scap inaccessible to COPII proteins. Proc Natl Acad Sci USA 104(16):6519–6526

    Article  PubMed  CAS  Google Scholar 

  119. Osborne TF (2000) Sterol regulatory element-binding proteins (SREBPs): key regulators of nutritional homeostasis and insulin action. J Biol Chem 275(42):32379–32382

    Article  PubMed  CAS  Google Scholar 

  120. Goldstein JL, DeBose-Boyd RA, Brown MS (2006) Protein sensors for membrane sterols. Cell 124(1):35–46

    Article  PubMed  CAS  Google Scholar 

  121. Espenshade PJ (2006) SREBPs: sterol-regulated transcription factors. J Cell Sci 119(Pt 6):973–976

    Article  PubMed  CAS  Google Scholar 

  122. Brown MS, Goldstein JL (1999) A proteolytic pathway that controls the cholesterol content of membranes, cells, and blood. Proc Natl Acad Sci USA 96(20):11041–11048

    Article  PubMed  CAS  Google Scholar 

  123. Goldstein JL, Rawson RB, Brown MS (2002) Mutant mammalian cells as tools to delineate the sterol regulatory element-binding protein pathway for feedback regulation of lipid synthesis. Arch Biochem Biophys 397(2):139–148

    Article  PubMed  CAS  Google Scholar 

  124. Eberle D, Hegarty B, Bossard P, Ferre P, Foufelle F (2004) SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86(11):839–848

    Article  PubMed  CAS  Google Scholar 

  125. Hegarty BD, Bobard A, Hainault I, Ferre P, Bossard P, Foufelle F (2005) Distinct roles of insulin and liver X receptor in the induction and cleavage of sterol regulatory element-binding protein-1c. Proc Natl Acad Sci USA 102(3):791–796

    Article  PubMed  CAS  Google Scholar 

  126. Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein JL, Mangelsdorf DJ (2000) Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXRalpha and LXRbeta. Genes Dev 14(22):2819–2830

    Article  PubMed  CAS  Google Scholar 

  127. Hirano Y, Yoshida M, Shimizu M, Sato R (2001) Direct demonstration of rapid degradation of nuclear sterol regulatory element-binding proteins by the ubiquitin-proteasome pathway. J Biol Chem 276(39):36431–36437

    Article  PubMed  CAS  Google Scholar 

  128. Hirano Y, Murata S, Tanaka K, Shimizu M, Sato R (2003) Sterol regulatory element-binding proteins are negatively regulated through SUMO-1 modification independent of the ubiquitin/26 S proteasome pathway. J Biol Chem 278(19):16809–16819

    Article  PubMed  CAS  Google Scholar 

  129. Walker AK, Yang F, Jiang K, Ji JY, Watts JL, Purushotham A, Boss O, Hirsch ML, Ribich S, Smith JJ, Israelian K, Westphal CH, Rodgers JT, Shioda T, Elson SL, Mulligan P, Najafi-Shoushtari H, Black JC, Thakur JK, Kadyk LC, Whetstine JR, Mostoslavsky R, Puigserver P, Li X, Dyson NJ, Hart AC, Naar AM (2010) Conserved role of SIRT1 orthologs in fasting-dependent inhibition of the lipid/cholesterol regulator SREBP. Genes Dev 24(13):1403–1417

    Article  PubMed  CAS  Google Scholar 

  130. Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ (1995) LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 9(9):1033–1045

    Article  PubMed  CAS  Google Scholar 

  131. Repa JJ, Mangelsdorf DJ (2000) The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 16:459–481

    Article  PubMed  CAS  Google Scholar 

  132. Attie AD, Kastelein JP, Hayden MR (2001) Pivotal role of ABCA1 in reverse cholesterol transport influencing HDL levels and susceptibility to atherosclerosis. J Lipid Res 42(11):1717–1726

    PubMed  CAS  Google Scholar 

  133. Yu L, Hammer RE, Li-Hawkins J, Von Bergmann K, Lutjohann D, Cohen JC, Hobbs HH (2002) Disruption of Abcg5 and Abcg8 in mice reveals their crucial role in biliary cholesterol secretion. Proc Natl Acad Sci USA 99(25):16237–16242

    Article  PubMed  CAS  Google Scholar 

  134. Bradley MN, Hong C, Chen M, Joseph SB, Wilpitz DC, Wang X, Lusis AJ, Collins A, Hseuh WA, Collins JL, Tangirala RK, Tontonoz P (2007) Ligand activation of LXR beta reverses atherosclerosis and cellular cholesterol overload in mice lacking LXR alpha and apoE. J Clin Invest 117(8):2337–2346

    Article  PubMed  CAS  Google Scholar 

  135. Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B, Heyman RA, Dietschy JM, Mangelsdorf DJ (2000) Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 289(5484):1524–1529

    Article  PubMed  CAS  Google Scholar 

  136. Schultz JR, Tu H, Luk A, Repa JJ, Medina JC, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf DJ, Lustig KD, Shan B (2000) Role of LXRs in control of lipogenesis. Genes Dev 14(22):2831–2838

    Article  PubMed  CAS  Google Scholar 

  137. Song BL, Javitt NB, DeBose-Boyd RA (2005) Insig-mediated degradation of HMG CoA reductase stimulated by lanosterol, an intermediate in the synthesis of cholesterol. Cell Metab 1(3):179–189

    Article  PubMed  CAS  Google Scholar 

  138. Song BL, DeBose-Boyd RA (2004) Ubiquitination of 3-hydroxy-3-methylglutaryl-CoA reductase in permeabilized cells mediated by cytosolic E1 and a putative membrane-bound ubiquitin ligase. J Biol Chem 279(27):28798–28806

    Article  PubMed  CAS  Google Scholar 

  139. Song BL, Sever N, DeBose-Boyd RA (2005) Gp78, a membrane-anchored ubiquitin ligase, associates with Insig-1 and couples sterol-regulated ubiquitination to degradation of HMG CoA reductase. Mol Cell 19(6):829–840

    Article  PubMed  CAS  Google Scholar 

  140. Sever N, Song BL, 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(52):52479–52490

    Article  PubMed  CAS  Google Scholar 

  141. Gill S, Stevenson J, Kristiana I, Brown AJ (2011) Cholesterol-dependent degradation of squalene monooxygenase, a control point in cholesterol synthesis beyond HMG-CoA reductase. Cell Metab 13 (3):260–273

    Google Scholar 

  142. Beg ZH, Allmann DW, Gibson DM (1973) Modulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity with cAMP and wth protein fractions of rat liver cytosol. Biochem Biophys Res Commun 54(4):1362–1369

    Article  PubMed  CAS  Google Scholar 

  143. Clarke PR, Hardie DG (1990) Regulation of HMG-CoA reductase: identification of the site phosphorylated by the AMP-activated protein kinase in vitro and in intact rat liver. EMBO J 9(8):2439–2446

    PubMed  CAS  Google Scholar 

  144. Hardie DG, Pan DA (2002) Regulation of fatty acid synthesis and oxidation by the AMP-activated protein kinase. Biochem Soc Trans 30(Pt 6):1064–1070

    PubMed  CAS  Google Scholar 

  145. Sato R, Goldstein JL, Brown MS (1993) Replacement of serine-871 of hamster 3-hydroxy-3-methylglutaryl-CoA reductase prevents phosphorylation by AMP-activated kinase and blocks inhibition of sterol synthesis induced by ATP depletion. Proc Natl Acad Sci USA 90(20):9261–9265

    Article  PubMed  CAS  Google Scholar 

  146. Omkumar RV, Darnay BG, Rodwell VW (1994) Modulation of Syrian hamster 3-hydroxy-3-methylglutaryl-CoA reductase activity by phosphorylation. Role of serine 871. J Biol Chem 269(9):6810–6814

    PubMed  CAS  Google Scholar 

  147. Benjannet S, Rhainds D, Essalmani R, Mayne J, Wickham L, Jin W, Asselin MC, Hamelin J, Varret M, Allard D, Trillard M, Abifadel M, Tebon A, Attie AD, Rader DJ, Boileau C, Brissette L, Chretien M, Prat A, Seidah NG (2004) NARC-1/PCSK9 and its natural mutants: zymogen cleavage and effects on the low-density lipoprotein (LDL) receptor and LDL cholesterol. J Biol Chem 279(47):48865–48875

    Article  PubMed  CAS  Google Scholar 

  148. Maxwell KN, Breslow JL (2004) Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc Natl Acad Sci USA 101(18):7100–7105

    Article  PubMed  CAS  Google Scholar 

  149. Rashid S, Curtis DE, Garuti R, Anderson NN, Bashmakov Y, Ho YK, Hammer RE, Moon YA, Horton JD (2005) Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9. Proc Natl Acad Sci USA 102(15):5374–5379

    Article  PubMed  CAS  Google Scholar 

  150. Chan JC, Piper DE, Cao Q, Liu D, King C, Wang W, Tang J, Liu Q, Higbee J, Xia Z, Di Y, Shetterly S, Arimura Z, Salomonis H, Romanow WG, Thibault ST, Zhang R, Cao P, Yang XP, Yu T, Lu M, Retter MW, Kwon G, Henne K, Pan O, Tsai MM, Fuchslocher B, Yang E, Zhou L, Lee KJ, Daris M, Sheng J, Wang Y, Shen WD, Yeh WC, Emery M, Walker NP, Shan B, Schwarz M, Jackson SM (2009) A proprotein convertase subtilisin/kexin type 9 neutralizing antibody reduces serum cholesterol in mice and nonhuman primates. Proc Natl Acad Sci USA 106(24):9820–9825

    Article  PubMed  CAS  Google Scholar 

  151. Zelcer N, Hong C, Boyadjian R, Tontonoz P (2009) LXR regulates cholesterol uptake through Idol-dependent ubiquitination of the LDL receptor. Science 325(5936):100–104

    Article  PubMed  CAS  Google Scholar 

  152. Hong C, Duit S, Jalonen P, Out R, Scheer L, Sorrentino V, Boyadjian R, Rodenburg KW, Foley E, Korhonen L, Lindholm D, Nimpf J, van Berkel TJ, Tontonoz P, Zelcer N (2010) The E3 ubiquitin ligase IDOL induces the degradation of the low-density lipoprotein receptor family members VLDLR and ApoER2. J Biol Chem 285 (26):19720–19726

    Google Scholar 

  153. Ambros V (2003) MicroRNA pathways in flies and worms: growth, death, fat, stress, and timing. Cell 113(6):673–676

    Article  PubMed  CAS  Google Scholar 

  154. Ambros V (2004) The functions of animal microRNAs. Nature 431(7006):350–355

    Article  PubMed  CAS  Google Scholar 

  155. Bartel DP (2009) MicroRNAs: target recognition and regulatory functions. Cell 136(2):215–233

    Article  PubMed  CAS  Google Scholar 

  156. Filipowicz W, Bhattacharyya SN, Sonenberg N (2008) Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat Rev Genet 9(2):102–114

    Article  PubMed  CAS  Google Scholar 

  157. Elmen J, Lindow M, Silahtaroglu A, Bak M, Christensen M, Lind-Thomsen A, Hedtjarn M, Hansen JB, Hansen HF, Straarup EM, McCullagh K, Kearney P, Kauppinen S (2008) Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to up-regulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res 36(4):1153–1162

    Article  PubMed  CAS  Google Scholar 

  158. Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W, Tuschl T (2002) Identification of tissue-specific microRNAs from mouse. Curr Biol 12(9):735–739

    Article  PubMed  CAS  Google Scholar 

  159. Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk ME, Kauppinen S, Orum H (2010) Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 327(5962):198–201

    Article  PubMed  CAS  Google Scholar 

  160. Esau C, Kang X, Peralta E, Hanson E, Marcusson EG, Ravichandran LV, Sun Y, Koo S, Perera RJ, Jain R, Dean NM, Freier SM, Bennett CF, Lollo B, Griffey R (2004) MicroRNA-143 regulates adipocyte differentiation. J Biol Chem 279(50):52361–52365

    Article  PubMed  CAS  Google Scholar 

  161. Rayner KJ, Sheedy FJ, Esau CC, Hussain FN, Temel RE, Parathath S, van Gils JM, Rayner AJ, Chang AN, Suarez Y, Fernandez-Hernando C, Fisher EA, Moore KJ (2011) Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J Clin Invest 121 (7):2921–2931

    Google Scholar 

  162. Fish JE, Santoro MM, Morton SU, Yu S, Yeh RF, Wythe JD, Ivey KN, Bruneau BG, Stainier DY, Srivastava D (2008) miR-126 regulates angiogenic signaling and vascular integrity. Dev Cell 15(2):272–284

    Article  PubMed  CAS  Google Scholar 

  163. Wang S, Aurora AB, Johnson BA, Qi X, McAnally J, Hill JA, Richardson JA, Bassel-Duby R, Olson EN (2008) The endothelial-specific microRNA miR-126 governs vascular integrity and angiogenesis. Dev Cell 15(2):261–271

    Article  PubMed  CAS  Google Scholar 

  164. Harris TA, Yamakuchi M, Ferlito M, Mendell JT, Lowenstein CJ (2008) MicroRNA-126 regulates endothelial expression of vascular cell adhesion molecule 1. Proc Natl Acad Sci USA 105(5):1516–1521

    Article  PubMed  CAS  Google Scholar 

  165. Zhang Q, Kandic I, Kutryk MJ (2010) Dysregulation of angiogenesis-related microRNAs in endothelial progenitor cells from patients with coronary artery disease. Biochem Biophys Res Commun 405 (1):42–46

    Google Scholar 

  166. Boettger T, Beetz N, Kostin S, Schneider J, Kruger M, Hein L, Braun T (2009) Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. J Clin Invest 119(9):2634–2647

    Article  PubMed  CAS  Google Scholar 

  167. Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee TH, Miano JM, Ivey KN, Srivastava D (2009) miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 460(7256):705–710

    PubMed  CAS  Google Scholar 

  168. Elia L, Quintavalle M, Zhang J, Contu R, Cossu L, Latronico MV, Peterson KL, Indolfi C, Catalucci D, Chen J, Courtneidge SA, Condorelli G (2009) The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death Differ 16(12):1590–1598

    Article  PubMed  CAS  Google Scholar 

  169. Xin M, Small EM, Sutherland LB, Qi X, McAnally J, Plato CF, Richardson JA, Bassel-Duby R, Olson EN (2009) MicroRNAs miR-143 and miR-145 modulate cytoskeletal dynamics and responsiveness of smooth muscle cells to injury. Genes Dev 23(18):2166–2178

    Article  PubMed  CAS  Google Scholar 

  170. Raitoharju E, Lyytikainen LP, Levula M, Oksala N, Mennander A, Tarkka M, Klopp N, Illig T, Kahonen M, Karhunen PJ, Laaksonen R, Lehtimaki T (2011) miR-21, miR-210, miR-34a, and miR-146a/b are up-regulated in human atherosclerotic plaques in the Tampere Vascular Study. Atherosclerosis

  171. Mitchell PS, Parkin RK, Kroh EM, Fritz BR, Wyman SK, Pogosova-Agadjanyan EL, Peterson A, Noteboom J, O’Briant KC, Allen A, Lin DW, Urban N, Drescher CW, Knudsen BS, Stirewalt DL, Gentleman R, Vessella RL, Nelson PS, Martin DB, Tewari M (2008) Circulating microRNAs as stable blood-based markers for cancer detection. Proc Natl Acad Sci USA 105(30):10513–10518

    Article  PubMed  CAS  Google Scholar 

  172. Zomer A, Vendrig T, Hopmans ES, van Eijndhoven M, Middeldorp JM, Pegtel DM (2010) Exosomes: Fit to deliver small RNA. Commun Integr Biol 3 (5):447–450

    Google Scholar 

  173. Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, Weber M, Hamm CW, Roxe T, Muller-Ardogan M, Bonauer A, Zeiher AM, Dimmeler S (2010) Circulating microRNAs in patients with coronary artery disease. Circ Res 107 (5):677–684

    Google Scholar 

  174. Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M (2010) Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ Res 107 (6):810–817

    Google Scholar 

  175. Vickers KC, Palmisano BT, Shoucri BM, Shamburek RD, Remaley AT (2011) MicroRNAs are transported in plasma and delivered to recipient cells by high-density lipoproteins. Nat Cell Biol 13 (4):423–433

    Google Scholar 

  176. Fazi F, Rosa A, Fatica A, Gelmetti V, De Marchis ML, Nervi C, Bozzoni I (2005) A minicircuitry comprised of microRNA-223 and transcription factors NFI-A and C/EBPalpha regulates human granulopoiesis. Cell 123(5):819–831

    Article  PubMed  CAS  Google Scholar 

  177. Izumi B, Nakasa T, Tanaka N, Nakanishi K, Kamei N, Yamamoto R, Nakamae T, Ohta R, Fujioka Y, Yamasaki K, Ochi M (2011) MicroRNA-223 expression in neutrophils in the early phase of secondary damage after spinal cord injury. Neurosci Lett 492 (2):114–118

    Google Scholar 

  178. Sugatani T, Hruska KA (2007) MicroRNA-223 is a key factor in osteoclast differentiation. J Cell Biochem 101(4):996–999

    Article  PubMed  CAS  Google Scholar 

  179. Krutzfeldt J, Rajewsky N, Braich R, Rajeev KG, Tuschl T, Manoharan M, Stoffel M (2005) Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438(7068):685–689

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to thank Dr. Yajaira Suárez for her helpful comments and stimulating discussions. C.F.-H. laboratory is supported by grants from the National Institute of Health (R01HL106063 and R01HL107953). We apologize to those whose work could not be cited.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carlos Fernández-Hernando.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Goedeke, L., Fernández-Hernando, C. Regulation of cholesterol homeostasis. Cell. Mol. Life Sci. 69, 915–930 (2012). https://doi.org/10.1007/s00018-011-0857-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-011-0857-5

Keywords

Navigation