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:

MicroRNA-148a regulates LDL receptor and ABCA1 expression to control circulating lipoprotein levels

Abstract

The hepatic low-density lipoprotein receptor (LDLR) pathway is essential for clearing circulating LDL cholesterol (LDL-C). Whereas the transcriptional regulation of LDLR is well characterized, the post-transcriptional mechanisms that govern LDLR expression are just beginning to emerge. Here we develop a high-throughput genome-wide screening assay to systematically identify microRNAs (miRNAs) that regulate LDLR activity in human hepatic cells. From this screen we identified and characterized miR-148a as a negative regulator of LDLR expression and activity and defined a sterol regulatory element–binding protein 1 (SREBP1)-mediated pathway through which miR-148a regulates LDL-C uptake. In mice, inhibition of miR-148a increased hepatic LDLR expression and decreased plasma LDL-C. Moreover, we found that miR-148a regulates hepatic expression of ATP-binding cassette, subfamily A, member 1 (ABCA1) and circulating high-density lipoprotein cholesterol (HDL-C) levels in vivo. These studies uncover a role for miR-148a as a key regulator of hepatic LDL-C clearance through direct modulation of LDLR expression and demonstrate the therapeutic potential of inhibiting miR-148a to ameliorate an elevated LDL-C/HDL-C ratio, a prominent risk factor for cardiovascular disease.

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: Genome-wide miRNA screen identifies novel regulators of LDLR activity.
Figure 2: Post-transcriptional regulation of LDLR expression and activity by miR-148a in human hepatic cells.
Figure 3: LDLR-GFP overexpression rescues LDLR activity in miR-148a–transfected cells.
Figure 4: SREBP1c modulates miR-148a expression in vitro and in vivo.
Figure 5: Inhibition of miR-148a alters plasma cholesterol levels in vivo.
Figure 6: miR-148a post-transcriptionally regulates ABCA1 expression.

Similar content being viewed by others

References

  1. Glass, C.K. & Witztum, J.L. Atherosclerosis. the road ahead. Cell 104, 503–516 (2001).

    Article  CAS  PubMed  Google Scholar 

  2. Lusis, A.J. Atherosclerosis. Nature 407, 233–241 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Hua, X. et al. 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, 11603–11607 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tontonoz, P., Kim, J.B., Graves, R.A. & Spiegelman, B.M. ADD1: a novel helix-loop-helix transcription factor associated with adipocyte determination and differentiation. Mol. Cell. Biol. 13, 4753–4759 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Yokoyama, C. et al. SREBP-1, a basic-helix-loop-helix–leucine zipper protein that controls transcription of the low density lipoprotein receptor gene. Cell 75, 187–197 (1993).

    Article  CAS  PubMed  Google Scholar 

  8. Goldstein, J.L. & Brown, M.S. Regulation of the mevalonate pathway. Nature 343, 425–430 (1990).

    Article  CAS  PubMed  Google Scholar 

  9. Walker, A.K. et al. A conserved SREBP-1–phosphatidylcholine feedback circuit regulates lipogenesis in metazoans. Cell 147, 840–852 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Horton, J.D. et al. Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc. Natl. Acad. Sci. USA 100, 12027–12032 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Maxwell, K.N., Soccio, R.E., Duncan, E.M., Sehayek, E. & Breslow, J.L. Novel putative SREBP and LXR target genes identified by microarray analysis in liver of cholesterol-fed mice. J. Lipid Res. 44, 2109–2119 (2003).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Ambros, V. The functions of animal microRNAs. Nature 431, 350–355 (2004).

    CAS  PubMed  Google Scholar 

  14. Bartel, D.P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. Rayner, K.J. et al. MiR-33 contributes to the regulation of cholesterol homeostasis. Science 328, 1570–1573 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Najafi-Shoushtari, S.H. et al. MicroRNA-33 and the SREBP host genes cooperate to control cholesterol homeostasis. Science 328, 1566–1569 (2010).

    Article  CAS  PubMed  Google Scholar 

  18. Jeon, T.I. et al. An SREBP-responsive microRNA operon contributes to a regulatory loop for intracellular lipid homeostasis. Cell Metab. 18, 51–61 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Rayner, K.J. et al. Antagonism of miR-33 in mice promotes reverse cholesterol transport and regression of atherosclerosis. J. Clin. Invest. 121, 2921–2931 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Rayner, K.J. et al. Inhibition of miR-33a/b in nonhuman primates raises plasma HDL and lowers VLDL triglycerides. Nature 478, 404–407 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rottiers, V. et al. Pharmacological inhibition of a microRNA family in nonhuman primates by a seed-targeting 8-mer antimiR. Sci. Transl. Med. 5, 212ra162 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. de Aguiar Vallim, T.Q. et al. MicroRNA-144 regulates hepatic ABCA1 and plasma HDL following activation of the nuclear receptor FXR. Circ. Res. 112, 1602–1612 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Ramírez, C.M. et al. Control of cholesterol metabolism and plasma HDL levels by miRNA-144. Circ. Res. 112, 1592–1601 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Vickers, K.C. et al. MicroRNA-223 coordinates cholesterol homeostasis. Proc. Natl. Acad. Sci. USA 111, 14518–14523 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Elmén, J. et al. LNA-mediated microRNA silencing in nonhuman primates. Nature 452, 896–899 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Elmén, J. et al. Antagonism of microRNA-122 in mice by systemically administered LNA-antimiR leads to upregulation of a large set of predicted target mRNAs in the liver. Nucleic Acids Res. 36, 1153–1162 (2008).

    Article  CAS  PubMed  Google Scholar 

  27. Esau, C. et al. miR-122 regulation of lipid metabolism revealed by in vivo antisense targeting. Cell Metab. 3, 87–98 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Soh, J., Iqbal, J., Queiroz, J., Fernandez-Hernando, C. & Hussain, M.M. MicroRNA-30c reduces hyperlipidemia and atherosclerosis in mice by decreasing lipid synthesis and lipoprotein secretion. Nat. Med. 19, 892–900 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Brown, M.S., Dana, S.E. & Goldstein, J.L. Regulation of 3-hydroxy-3-methylglutaryl coenzyme A reductase activity in human fibroblasts by lipoproteins. Proc. Natl. Acad. Sci. USA 70, 2162–2166 (1973).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Goldstein, J.L., Basu, S.K., Brunschede, G.Y. & Brown, M.S. Release of low-density lipoprotein from its cell surface receptor by sulfated glycosaminoglycans. Cell 7, 85–95 (1976).

    Article  CAS  PubMed  Google Scholar 

  31. Zhang, J.H., Chung, T.D. & Oldenburg, K.R. A simple statistical parameter for use in evaluation and validation of high-throughput screening assays. J. Biomol. Screen. 4, 67–73 (1999).

    Article  CAS  PubMed  Google Scholar 

  32. Birmingham, A. et al. Statistical methods for analysis of high-throughput RNA interference screens. Nat. Methods 6, 569–575 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Barad, O. et al. MicroRNA expression detected by oligonucleotide microarrays: system establishment and expression profiling in human tissues. Genome Res. 14, 2486–2494 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Landgraf, P. et al. A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 129, 1401–1414 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Vickers, K.C. et al. MicroRNA-27b is a regulatory hub in lipid metabolism and is altered in dyslipidemia. Hepatology 57, 533–542 (2013).

    Article  CAS  PubMed  Google Scholar 

  36. Arora, A. & Simpson, D.A. Individual mRNA expression profiles reveal the effects of specific microRNAs. Genome Biol. 9, R82 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Do, R. et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat. Genet. 45, 1345–1352 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Global Lipids Genetics Consortium. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

  39. Huan, T. et al. Genome-wide identification of microRNA expression quantitative trait loci. Nat. Commun. 6, 6601 (2015).

    Article  CAS  PubMed  Google Scholar 

  40. Trajkovski, M. et al. MicroRNAs 103 and 107 regulate insulin sensitivity. Nature 474, 649–653 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Mercer, J. et al. RNAi screening reveals proteasome- and Cullin3-dependent stages in vaccinia virus infection. Cell Rep. 2, 1036–1047 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. John, B. et al. Human MicroRNA targets. PLoS Biol. 2, e363 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Lewis, B.P., Burge, C.B. & Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120, 15–20 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Dweep, H., Gretz, N. & Sticht, C. miRWalk database for miRNA-target interactions. Methods Mol. Biol. 1182, 289–305 (2014).

    Article  CAS  PubMed  Google Scholar 

  45. Huang, D.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  CAS  Google Scholar 

  46. Szklarczyk, D. et al. The STRING database in 2011: functional interaction networks of proteins, globally integrated and scored. Nucleic Acids Res. 39, D561–D568 (2011).

    Article  CAS  PubMed  Google Scholar 

  47. Thomas, P.D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Down, T.A. & Hubbard, T.J. Computational detection and location of transcription start sites in mammalian genomic DNA. Genome Res. 12, 458–461 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Saini, H.K., Griffiths-Jones, S. & Enright, A.J. Genomic analysis of human microRNA transcripts. Proc. Natl. Acad. Sci. USA 104, 17719–17724 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Ernst, J. & Kellis, M. Discovery and characterization of chromatin states for systematic annotation of the human genome. Nat. Biotechnol. 28, 817–825 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Kent, W.J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Shimomura, I., Bashmakov, Y. & Horton, J.D. Increased levels of nuclear SREBP-1c associated with fatty livers in two mouse models of diabetes mellitus. J. Biol. Chem. 274, 30028–30032 (1999).

    Article  CAS  PubMed  Google Scholar 

  54. Shimomura, I., Shimano, H., Horton, J.D., Goldstein, J.L. & Brown, M.S. Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J. Clin. Invest. 99, 838–845 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Horton, J.D., Bashmakov, Y., Shimomura, I. & Shimano, H. Regulation of sterol regulatory element binding proteins in livers of fasted and refed mice. Proc. Natl. Acad. Sci. USA 95, 5987–5992 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Peet, D.J. et al. Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR alpha. Cell 93, 693–704 (1998).

    Article  CAS  PubMed  Google Scholar 

  57. Yoshikawa, T. et al. Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter. Mol. Cell. Biol. 21, 2991–3000 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Dietschy, J.M., Turley, S.D. & Spady, D.K. Role of liver in the maintenance of cholesterol and low density lipoprotein homeostasis in different animal species, including humans. J. Lipid Res. 34, 1637–1659 (1993).

    CAS  PubMed  Google Scholar 

  59. Oram, J.F. & Vaughan, A.M. ABCA1-mediated transport of cellular cholesterol and phospholipids to HDL apolipoproteins. Curr. Opin. Lipidol. 11, 253–260 (2000).

    Article  CAS  PubMed  Google Scholar 

  60. Yang, M. et al. Identification of miR-185 as a regulator of de novo cholesterol biosynthesis and low density lipoprotein uptake. J. Lipid Res. 55, 226–238 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ramírez, C.M. et al. MicroRNA 33 regulates glucose metabolism. Mol. Cell. Biol. 33, 2891–2902 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Dávalos, A. et al. miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc. Natl. Acad. Sci. USA 108, 9232–9237 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  63. Suárez, Y., Fernandez-Hernando, C., Pober, J.S. & Sessa, W.C. Dicer-dependent microRNAs regulate gene expression and functions in human endothelial cells. Circ. Res. 100, 1164–1173 (2007).

    Article  CAS  PubMed  Google Scholar 

  64. Goedeke, L. et al. A regulatory role for microRNA 33* in controlling lipid metabolism gene expression. Mol. Cell. Biol. 33, 2339–2352 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Allen, R.M., Marquart, T.J., Jesse, J.J. & Baldan, A. Control of very low–density lipoprotein secretion by N-ethylmaleimide–sensitive factor and miR-33. Circ. Res. 115, 10–22 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Calvo, D., Gomez-Coronado, D., Suarez, Y., Lasuncion, M.A. & Vega, M.A. Human CD36 is a high-affinity receptor for the native lipoproteins HDL, LDL and VLDL. J. Lipid Res. 39, 777–788 (1998).

    CAS  PubMed  Google Scholar 

  67. Suárez, Y. et al. Synergistic upregulation of low-density lipoprotein receptor activity by tamoxifen and lovastatin. Cardiovasc. Res. 64, 346–355 (2004).

    Article  CAS  PubMed  Google Scholar 

  68. Chamorro-Jorganes, A., Araldi, E., Rotllan, N., Cirera-Salinas, D. & Suarez, Y. Autoregulation of glypican-1 by intronic microRNA-149 fine tunes the angiogenic response to FGF2 in human endothelial cells. J. Cell Sci. 127, 1169–1178 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Ramirez, C.M. et al. MicroRNA-758 regulates cholesterol efflux through post-transcriptional repression of ATP-binding cassette transporter A1. Arterioscler. Thromb. Vasc. Biol. 31, 2707–2714 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Wang, W. et al. Radixin is required to maintain apical canalicular membrane structure and function in rat hepatocytes. Gastroenterology 131, 878–884 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Mattison, J.A. et al. Resveratrol prevents high fat/sucrose diet-induced central arterial wall inflammation and stiffening in nonhuman primates. Cell Metab. 20, 183–190 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Goedeke, L. et al. Long-term therapeutic silencing of miR-33 increases circulating triglyceride levels and hepatic lipid accumulation in mice. EMBO Mol. Med. 6, 1133–1141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Miller, A.M. et al. MiR-155 has a protective role in the development of non-alcoholic hepatosteatosis in mice. PLoS ONE 8, e72324 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank C. Yun, J. Recio, S. Katz and R. DasGupta at the New York University (NYU) RNAi Core for their advice and assistance with the miRNA screen, K. Harry and members of the Yale University Liver Center for primary mouse hepatocyte isolation, members of the Iwakiri laboratory for reagents and advice on primary hepatocyte culture, the Yale University School of Medicine Mouse Metabolic Phenotyping Center (MMPC) for liver toxicity measurements, P. Tontonoz (UCLA) for generously providing the LDLR-GFP plasmid, the Nonhuman Primate Core of the National Institute on Aging for providing liver samples and E. Fisher (NYU School of Medicine) for kindly providing the human hepatocellular carcinoma cell line (Huh7) and mouse hepatic cell line (Hepa1–6). This work was supported through grants by the US National Institutes of Health (NIH; grants R01HL107953, R01HL107953-04S1 and R01HL106063 (C.F.-H.); grant R01HL105945 (Y.S.); grant 1F31AG043318 (L.G.); grant P30KD034989 (Yale University Liver Center), the American Heart Association (grant 15SDG23000025 (C.M.R.)), the Howard Hughes Medical Institute International Student Research Fellowship (E.A.), the Foundation Leducq Transatlantic Network of Excellence in Cardiovascular Research (C.F.-H.) and the Ministerio de Industria y Comercio, Spain (grant SAF2011-29951 (M.A.L.)). Centro de Investigación Biomédica en Red Fisiopatología de Obesidad y Nutrición (CIBERobn) is an initiative of Instituto de Salud Carlos III (ISCIII), Spain. R.d.C. is supported by the Intramural Research Program of the NIH, National Institute of Aging. A.M.N. and A.W. are supported by NIH grant R01DK 094184. The NYU RNAi core is supported by the Laura and Isaac Perlmutter Cancer Center (NIH, National Cancer Institute grant P30CA16087) and the New York State Stem Cell Science (NYSTEM) contract C026719. The Yale University School of Medicine MMPC is supported by NIH grant U24 DK059635.

Author information

Authors and Affiliations

Authors

Contributions

L.G. and C.F.-H. conceived and designed the study. L.G. optimized and performed the miRNA screen. J.F.A. and A.C.-D. performed confocal experiments, assisted with cloning and analyzed data. L.G. and A.C.-D. performed in vitro experiments and analyzed data. L.G., N.R., C.M.R. and A.C.-D. performed mouse experiments and analyzed data. N.R. performed western blotting for plasma lipoproteins and assessed lipoprotein distribution by FPLC. E.A. cloned the MIR148A promoter. C.-S.L. assisted with western blotting. N.N.A. analyzed gene expression in the livers of fed and fasted wild-type mice and ob/ob mice. R.d.C. designed the nonhuman primate experiments and J.D.H. provided the mouse samples. M.A.L. provided DiI-LDL and native LDL. A.W. and A.M.N. assisted with mouse experiments. Y.S. and C.F.-H. assisted with experimental design and data interpretation. L.G. and C.F.-H. wrote the manuscript, which was commented on by all authors.

Corresponding author

Correspondence to Carlos Fernández-Hernando.

Ethics declarations

Competing interests

C.F.-H. and L.G. have filed a patent (United States PCT/US2014/042196) for the therapeutic use of miR-148 inhibitors in treating cardiometabolic diseases.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Table 2 (PDF 18771 kb)

Supplementary Table 1

Raw data from primary miRNA screen. (XLS 347 kb)

Supplementary Table 3

Functional annotation clusters of human miR-148a targets identified by DAVID. (XLS 768 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Goedeke, L., Rotllan, N., Canfrán-Duque, A. et al. MicroRNA-148a regulates LDL receptor and ABCA1 expression to control circulating lipoprotein levels. Nat Med 21, 1280–1289 (2015). https://doi.org/10.1038/nm.3949

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3949

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