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:

The long noncoding RNA CHROME regulates cholesterol homeostasis in primates

Abstract

The human genome encodes thousands of long noncoding RNAs (lncRNAs), the majority of which are poorly conserved and uncharacterized. Here we identify a primate-specific lncRNA (CHROME), which is elevated in the plasma and atherosclerotic plaques of individuals with coronary artery disease, and regulates cellular and systemic cholesterol homeostasis. Expression of the lncRNA CHROME is influenced by dietary and cellular cholesterol through the sterol-activated liver X receptor transcription factors, which control genes that mediate responses to cholesterol overload. Using gain- and loss-of-function approaches, we show that CHROME promotes cholesterol efflux and high-density lipoprotein (HDL) biogenesis by curbing the actions of a set of functionally related microRNAs that repress genes in those pathways. CHROME knockdown in human hepatocytes and macrophages increases the levels of miR-27b, miR-33a, miR-33b and miR-128, thereby reducing the expression of their overlapping target gene networks and associated biological functions. In particular, cells that lack CHROME show reduced expression of ABCA1, which regulates cholesterol efflux and nascent HDL particle formation. Collectively, our findings identify CHROME as a central component of the noncoding RNA circuitry that controls cholesterol homeostasis in humans.

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

Fig. 1: Levels of lncRNA CHROME are increased in atherosclerotic cardiovascular disease.
Fig. 2: The lncRNA CHROME is regulated by dietary and cellular cholesterol by LXR transcription factors.
Fig. 3: CHROME depletion in hepatocytes reduces cholesterol efflux and formation of nascent HDL particles.
Fig. 4: CHROME interacts with a set of miRNAs known to repress cholesterol efflux.
Fig. 5: Gain or loss of CHROME in hepatocytes alters levels of its interacting miRNAs and their common target mRNAs involved in cholesterol metabolism.
Fig. 6: CHROME regulates cholesterol efflux in macrophages through its interaction with miRNAs.

Similar content being viewed by others

Data availabilty

The Biobank of Karolinska Endarterectomies (BiKE) microarray dataset has been deposited in the Gene Expression Omnibus (GEO) and is available under accession GSE21545. HepG2 RNA-sequencing datasets have been deposited in GEO under accession GSE97469. Data that support the plots within this paper and other findings of this study are available from the corresponding author upon reasonable request.

References

  1. Goedeke, L. & Fernandez-Hernando, C. Regulation of cholesterol homeostasis. Cell. Mol. Life Sci. 69, 915–930 (2012).

    Article  CAS  Google Scholar 

  2. Siddiqi, H. K., Kiss, D. & Rader, D. HDL-cholesterol and cardiovascular disease: rethinking our approach. Curr. Opin. Cardiol. 30, 536–542 (2015).

    Article  Google Scholar 

  3. Lee, S. D. & Tontonoz, P. Liver X receptors at the intersection of lipid metabolism and atherogenesis. Atherosclerosis 242, 29–36 (2015).

    Article  CAS  Google Scholar 

  4. Scacalossi, K. R., van Solingen, C. & Moore, K. J. Long non-coding RNAs regulating macrophage functions in homeostasis and disease. Vascul. Pharmacol. https://doi.org/10.1016/j.vph.2018.02.011 (2018).

  5. van Solingen, C., Scacalossi, K. R. & Moore, K. J. Long noncoding RNAs in lipid metabolism. Curr. Opin. Lipidol. 29, 224–232 (2018).

    Article  Google Scholar 

  6. Feinberg, M. W. & Moore, K. J. microRNA regulation of atherosclerosis. Circ. Res. 118, 703–720 (2016).

    Article  CAS  Google Scholar 

  7. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  9. Ouimet, M. et al. miRNA targeting of oxysterol-binding protein-like 6 regulates cholesterol trafficking and efflux. Arterioscler. Thromb. Vasc. Biol. 36, 942–951 (2016).

    Article  CAS  Google Scholar 

  10. Allen, R. M. et al. miR-33 controls the expression of biliary transporters, and mediates statin- and diet-induced hepatotoxicity. EMBO Mol. Med. 4, 882–895 (2012).

    Article  CAS  Google Scholar 

  11. Goedeke, L. et al. miR-27b inhibits LDLR and ABCA1 expression but does not influence plasma and hepatic lipid levels in mice. Atherosclerosis 243, 499–509 (2015).

    Article  CAS  Google Scholar 

  12. de Aguiar Vallim, T. Q. et al. microRNA-144 regulates hepatic ATP binding cassette transporter A1 and plasma high-density lipoprotein after activation of the nuclear receptor farnesoid X receptor. Circ. Res. 112, 1602–1612 (2013).

    Article  Google Scholar 

  13. Ramirez, C. M. et al. Control of cholesterol metabolism and plasma high-density lipoprotein levels by microRNA-144. Circ. Res. 112, 1592–1601 (2013).

    Article  CAS  Google Scholar 

  14. Liu, X. H. et al. lnc RNA HOTAIR functions as a competing endogenous RNA to regulate HER2 expression by sponging miR-331-3p in gastric cancer. Mol. Cancer 13, 92 (2014).

    Article  CAS  Google Scholar 

  15. Wagschal, A. et al. Genome-wide identification of microRNAs regulating cholesterol and triglyceride homeostasis. Nat. Med. 21, 1290–1297 (2015).

    Article  CAS  Google Scholar 

  16. Quinn, J. J. & Chang, H. Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 17, 47–62 (2016).

    Article  CAS  Google Scholar 

  17. Palazzo, A. F. & Lee, E. S. Non-coding RNA: what is functional and what is junk? Front. Genet. 6, 2 (2015).

    Article  Google Scholar 

  18. Schmitz, S. U., Grote, P. & Herrmann, B. G. Mechanisms of long noncoding RNA function in development and disease. Cell. Mol. Life Sci. 73, 2491–2509 (2016).

    Article  CAS  Google Scholar 

  19. Freedman, J. E. & Miano, J. M. Challenges and opportunities in linking long noncoding RNAs to cardiovascular, lung, and blood diseases. Arterioscler. Thromb. Vasc. Biol. 37, 21–25 (2017).

    Article  CAS  Google Scholar 

  20. Yan, C., Chen, J. & Chen, N. Long noncoding RNA MALAT1 promotes hepatic steatosis and insulin resistance by increasing nuclear SREBP-1c protein stability. Sci. Rep. 6, 22640 (2016).

    Article  CAS  Google Scholar 

  21. Liu, C. et al. Long noncoding RNA H19 interacts with polypyrimidine tract-binding protein 1 to reprogram hepatic lipid homeostasis. Hepatology 67, 1768–1783 (2018).

    Article  CAS  Google Scholar 

  22. Li, D. et al. Identification of a novel human long non-coding RNA that regulates hepatic lipid metabolism by inhibiting SREBP-1c. Int. J. Biol. Sci. 13, 349–357 (2017).

    Article  CAS  Google Scholar 

  23. Sallam, T. et al. Feedback modulation of cholesterol metabolism by the lipid-responsive non-coding RNA LeXis. Nature 534, 124–128 (2016).

    Article  CAS  Google Scholar 

  24. Sallam, T. et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat. Med. 24, 304–312 (2018).

    Article  CAS  Google Scholar 

  25. Pasmant, E., Sabbagh, A., Vidaud, M. & Bieche, I. ANRIL, a long, noncoding RNA, is an unexpected major hotspot in GWAS. FASEB J. 25, 444–448 (2011).

    Article  CAS  Google Scholar 

  26. Holdt, L. M. & Teupser, D. Recent studies of the human chromosome 9p21 locus, which is associated with atherosclerosis in human populations. Arterioscler. Thromb. Vasc. Biol. 32, 196–206 (2012).

    Article  CAS  Google Scholar 

  27. Nsengimana, J. et al. Enhanced linkage of a locus on chromosome 2 to premature coronary artery disease in the absence of hypercholesterolemia. Eur. J. Hum. Genet. 15, 313–319 (2007).

    Article  CAS  Google Scholar 

  28. North, K. E., Martin, L. J., Dyer, T., Comuzzie, A. G. & Williams, J. T. HDL cholesterol in females in the Framingham Heart Study is linked to a region of chromosome 2q. BMC Genet. 4, S98 (2003).

    Article  Google Scholar 

  29. Kapusta, A. et al. Transposable elements are major contributors to the origin, diversification, and regulation of vertebrate long noncoding RNAs. PLoS Genet. 9, e1003470 (2013).

    Article  CAS  Google Scholar 

  30. Perisic, L. et al. Profiling of atherosclerotic lesions by gene and tissue microarrays reveals PCSK6 as a novel protease in unstable carotid atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 33, 2432–2443 (2013).

    Article  CAS  Google Scholar 

  31. Moore, K. J. & Tabas, I. Macrophages in the pathogenesis of atherosclerosis. Cell 145, 341–355 (2011).

    Article  CAS  Google Scholar 

  32. Cabili, M. N. et al. Localization and abundance analysis of human lncRNAs at single-cell and single-molecule resolution. Genome Biol. 16, 20 (2015).

    Article  Google Scholar 

  33. Lennox, K. A. & Behlke, M. A. Cellular localization of long non-coding RNAs affects silencing by RNAi more than by antisense oligonucleotides. Nucleic Acids Res. 44, 863–877 (2016).

    Article  CAS  Google Scholar 

  34. John, S. et al. Kinetic complexity of the global response to glucocorticoid receptor action. Endocrinology 150, 1766–1774 (2009).

    Article  CAS  Google Scholar 

  35. Davalos, 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  CAS  Google Scholar 

  36. 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  Google Scholar 

  37. Yoon, J. H., Srikantan, S. & Gorospe, M. MS2-TRAP (MS2-tagged RNA affinity purification): tagging RNA to identify associated miRNAs. Methods 58, 81–87 (2012).

    Article  CAS  Google Scholar 

  38. Liu, J., Valencia-Sanchez, M. A., Hannon, G. J. & Parker, R. microRNA-dependent localization of targeted mRNAs to mammalian P-bodies. Nat. Cell Biol. 7, 719–723 (2005).

    Article  CAS  Google Scholar 

  39. Hubstenberger, A. et al. P-body purification reveals the condensation of repressed mRNA regulons. Mol. Cell 68, 144–157 (2017).

    Article  CAS  Google Scholar 

  40. Ulitsky, I. Evolution to the rescue: using comparative genomics to understand long non-coding RNAs. Nat. Rev. Genet. 17, 601–614 (2016).

    Article  CAS  Google Scholar 

  41. Rayner, K. J. & Moore, K. J. microRNA control of high-density lipoprotein metabolism and function. Circ. Res. 114, 183–192 (2014).

    Article  CAS  Google Scholar 

  42. Liang, B. et al. microRNA-20a/b regulates cholesterol efflux through post-transcriptional repression of ATP-binding cassette transporter A1. Biochim. Biophys. Acta 1862, 929–938 (2017).

    Article  CAS  Google Scholar 

  43. Marquart, T. J., Allen, R. M., Ory, D. S. & Baldan, A. miR-33 links SREBP-2 induction to repression of sterol transporters. Proc. Natl Acad. Sci. USA 107, 12228–12232 (2010).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  45. 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  Google Scholar 

  46. Abumrad, N. A. & Davidson, N. O. Role of the gut in lipid homeostasis. Physiol. Rev. 92, 1061–1085 (2012).

    Article  CAS  Google Scholar 

  47. Westerterp, M. et al. ATP-binding cassette transporters, atherosclerosis, and inflammation. Circ. Res. 114, 157–170 (2014).

    Article  CAS  Google Scholar 

  48. Ameres, S. L. et al. Target RNA-directed trimming and tailing of small silencing RNAs. Science 328, 1534–1539 (2010).

    Article  CAS  Google Scholar 

  49. Cajigas, I. J. et al. The local transcriptome in the synaptic neuropil revealed by deep sequencing and high-resolution imaging. Neuron 74, 453–466 (2012).

    Article  CAS  Google Scholar 

  50. Jens, M. & Rajewsky, N. Competition between target sites of regulators shapes post-transcriptional gene regulation. Nat. Rev. Genet. 16, 113–126 (2015).

    Article  CAS  Google Scholar 

  51. Ouimet, M. et al. microRNA-33 regulates macrophage autophagy in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 37, 1058–1067 (2017).

    Article  CAS  Google Scholar 

  52. Montenont, E. et al. Platelet WDR1 suppresses platelet activity and associates with cardiovascular disease. Blood 128, 2033–2042 (2016).

    Article  CAS  Google Scholar 

  53. Perisic, L. et al. Gene expression signatures, pathways and networks in carotid atherosclerosis. J. Intern. Med. 279, 293–308 (2016).

    Article  CAS  Google Scholar 

  54. Thasler, W. E. et al. Charitable state-controlled foundation human tissue and cell research: ethic and legal aspects in the supply of surgically removed human tissue for research in the academic and commercial sector in Germany. Cell Tissue Bank 4, 49–56 (2003).

    Article  Google Scholar 

  55. Johnson, C. V., Singer, R. H. & Lawrence, J. B. Fluorescent detection of nuclear RNA and DNA: implications for genome organization. Methods Cell Biol. 35, 73–99 (1991).

    Article  CAS  Google Scholar 

  56. Tam, R., Smith, K. P. & Lawrence, J. B. The 4q subtelomere harboring the FSHD locus is specifically anchored with peripheral heterochromatin unlike most human telomeres. J. Cell. Biol. 167, 269–279 (2004).

    Article  CAS  Google Scholar 

  57. Ihaka, R. & Gentleman, R. A language for data analysis and graphics. J. Comput. Graph. Stat. 5, 299–314 (1996).

    Google Scholar 

  58. Baran-Gale, J., Fannin, E. E., Kurtz, C. L. & Sethupathy, P. Beta cell 5′-shifted isomiRs are candidate regulatory hubs in type 2 diabetes. PLoS ONE 8, e73240 (2013).

    Article  CAS  Google Scholar 

  59. Listenberger, L. L. & Brown, D. A. Fluorescent detection of lipid droplets and associated proteins. Curr. Protoc. Cell. Biol. 24, 24.2.1–24.2.11 (2007).

    Article  Google Scholar 

  60. Folch, J., Lees, M. & Sloane Stanley, G. H. A simple method for the isolation and purification of total lipides from animal tissues. J. Biol. Chem. 226, 497–509 (1957).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the NIH (R01HL119047 (K.J.M.), R35HL135799 (K.J.M.), R01HL117226 (M.J.G.), T32HL098129 (E.J.H., C.v.S.), R01HL114978 (J.S.B.), R00HL088528 (R.E.T.), R01HL111932 (R.E.T.), R01HL128996 (K.C.V.), P01HL116263 (K.C.V.), R01DK105965 (P.S.)), the American Heart Association (14POST20180018 (C.v.S.), 13CRP14410042 (J.S.B.)), FINOVI (E.P.R.), the Swedish Society for Medical Research (L.P.M.) and the Heart and Lung Foundation (L.P.M.), the German Research Foundation CRC 1123 Project B1 (D.T. and L.M.H.), German Biobank Alliance BMBF 01EY1711C (German Ministry of Education and Research to D.T. and L.M.H.) and the Leducq Foundation CAD genomics (D.T. and L.M.H.). The BiKE study was supported by the Swedish Heart and Lung Foundation, the Swedish Research Council (K2009-65X-2233-01-3, K2013-65X-06816-30-4, 349-2007-8703), Uppdrag Besegra Stroke (P581/2011-123), the Strategic Cardiovascular Programs of Karolinska Institutet and Stockholm County Council, the Foundation for Strategic Research and the European Commission (CarTarDis, AtheroRemo, VIA, AtheroFlux projects). We thank E. A. Fisher (New York University) for helpful discussions, and S. Zhao and Q. Sheng (Vanderbilt University) for their efforts in sequencing-data analysis.

Author information

Authors and Affiliations

Authors

Contributions

E.J.H., C.v.S. and K.J.M. designed the study, guided the interpretation of the results and prepared the manuscript, with input from all authors. E.J.H. and C.v.S. performed experiments and data analyses. K.R.S., M.O., M.S.A., J.P., G.J.K., M.S., B.R., K.C.V., M.K. and P.S. contributed to experiments and data analyses. S.C. created and analyzed stable cell lines. A.B. and M.O. performed in situ hybridization of human plaques. E.C., L.P.M., U.H. and L.M. processed and analyzed BiKE datasets. B.E.C. performed RNAcofold and RNAhybrid analyses. E.P.R. performed polysome fractionation experiments. R.E.T. supervised nonhuman primate studies. M.A.H. and M.J.G. performed chromatin immunoprecipitation experiments. J.S.B. provided human plasma samples and assisted in data interpretation. D.T. and L.M.H. performed human liver RNA and lipoprotein analyses.

Corresponding author

Correspondence to Kathryn J. Moore.

Ethics declarations

Competing interests

K.J.M. and New York University hold a patent (US 9241950, status: issued 26 January 2016) on the use of miR-33 inhibitors to treat inflammation. All other authors have no competing interests.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hennessy, E.J., van Solingen, C., Scacalossi, K.R. et al. The long noncoding RNA CHROME regulates cholesterol homeostasis in primates. Nat Metab 1, 98–110 (2019). https://doi.org/10.1038/s42255-018-0004-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-018-0004-9

Keywords

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