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:

Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA

Abstract

Nuclear receptors regulate gene expression in response to environmental cues, but the molecular events governing the cell type specificity of nuclear receptors remain poorly understood. Here we outline a role for a long noncoding RNA (lncRNA) in modulating the cell type–specific actions of liver X receptors (LXRs), sterol-activated nuclear receptors that regulate the expression of genes involved in cholesterol homeostasis and that have been causally linked to the pathogenesis of atherosclerosis. We identify the lncRNA MeXis as an amplifier of LXR-dependent transcription of the gene Abca1, which is critical for regulation of cholesterol efflux. Mice lacking the MeXis gene show reduced Abca1 expression in a tissue-selective manner. Furthermore, loss of MeXis in mouse bone marrow cells alters chromosome architecture at the Abca1 locus, impairs cellular responses to cholesterol overload, and accelerates the development of atherosclerosis. Mechanistic studies reveal that MeXis interacts with and guides promoter binding of the transcriptional coactivator DDX17. The identification of MeXis as a lncRNA modulator of LXR-dependent gene expression expands understanding of the mechanisms underlying cell type–selective actions of nuclear receptors in physiology and 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: LXR regulates the noncoding RNA MeXis.
Figure 2: MeXis regulates Abca1 expression and function.
Figure 3: Loss of MeXis impairs macrophage Abca1 expression and accelerates atherosclerosis.
Figure 4: MeXis alters chromosome architecture at the Abca1 locus.
Figure 5: Identification of DDX17 as a binding partner of MeXis.
Figure 6: The LXR–MeXis axis in functionally conserved in humans.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Rohatgi, A. et al. HDL cholesterol efflux capacity and incident cardiovascular events. N. Engl. J. Med. 371, 2383–2393 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. de la Llera-Moya, M. et al. The ability to promote efflux via ABCA1 determines the capacity of serum specimens with similar high-density lipoprotein cholesterol to remove cholesterol from macrophages. Arterioscler. Thromb. Vasc. Biol. 30, 796–801 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Libby, P. et al. Inflammation in atherosclerosis: from pathophysiology to practice. J. Am. Coll. Cardiol. 54, 2129–2138 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Cao, Q. et al. The central role of EED in the orchestration of polycomb group complexes. Nat. Commun. 5, 3127 (2014).

    Article  CAS  PubMed  Google Scholar 

  5. Zelcer, N. & Tontonoz, P. Liver X receptors as integrators of metabolic and inflammatory signaling. J. Clin. Invest. 116, 607–614 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Bodzioch, M. et al. The gene encoding ATP-binding cassette transporter 1 is mutated in Tangier disease. Nat. Genet. 22, 347–351 (1999).

    Article  CAS  PubMed  Google Scholar 

  7. Rust, S. et al. Tangier disease is caused by mutations in the gene encoding ATP-binding cassette transporter 1. Nat. Genet. 22, 352–355 (1999).

    Article  CAS  PubMed  Google Scholar 

  8. Rinn, J.L. & Chang, H.Y. Genome regulation by long noncoding RNAs. Annu. Rev. Biochem. 81, 145–166 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Wang, K.C. et al. A long noncoding RNA maintains active chromatin to coordinate homeotic gene expression. Nature 472, 120–124 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kotzin, J.J. et al. The long non-coding RNA Morrbid regulates Bim and short-lived myeloid cell lifespan. Nature 537, 239–243 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Engreitz, J.M. et al. Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature 539, 452–455 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhang, L. et al. Inhibition of cholesterol biosynthesis through RNF145-dependent ubiquitination of SCAP. eLife 6, e28766 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  15. ENCODE Project Consortium. An integrated encyclopedia of DNA elements in the human genome. Nature 489, 57–74 (2012).

  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  PubMed  Google Scholar 

  17. Huarte, M. et al. A large intergenic noncoding RNA induced by p53 mediates global gene repression in the p53 response. Cell 142, 409–419 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Cheng, J.H., Pan, D.Z., Tsai, Z.T. & Tsai, H.K. Genome-wide analysis of enhancer RNA in gene regulation across 12 mouse tissues. Sci. Rep. 5, 12648 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Daniel, B. et al. The active enhancer network operated by liganded RXR supports angiogenic activity in macrophages. Genes Dev. 28, 1562–1577 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Chu, C. et al. Systematic discovery of Xist RNA binding proteins. Cell 161, 404–416 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Auboeuf, D., Hönig, A., Berget, S.M. & O'Malley, B.W. Coordinate regulation of transcription and splicing by steroid receptor coregulators. Science 298, 416–419 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Wortham, N.C. et al. The DEAD-box protein p72 regulates ERα-/oestrogen-dependent transcription and cell growth, and is associated with improved survival in ERα-positive breast cancer. Oncogene 28, 4053–4064 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Nikpay, M. et al. A comprehensive 1000 Genomes–based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Li, W. et al. Functional roles of enhancer RNAs for oestrogen-dependent transcriptional activation. Nature 498, 516–520 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wang, K.C. & Chang, H.Y. Molecular mechanisms of long noncoding RNAs. Mol. Cell 43, 904–914 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Khalil, A.M. et al. Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression. Proc. Natl. Acad. Sci. USA 106, 11667–11672 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Sallam, T. et al. The macrophage LBP gene is an LXR target that promotes macrophage survival and atherosclerosis. J. Lipid Res. 55, 1120–1130 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Rong, X. et al. LXRs regulate ER stress and inflammation through dynamic modulation of membrane phospholipid composition. Cell Metab. 18, 685–697 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Jakobsson, T. et al. GPS2 is required for cholesterol efflux by triggering histone demethylation, LXR recruitment, and coregulator assembly at the ABCG1 locus. Mol. Cell 34, 510–518 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Wongpalee, S.P. et al. Large-scale remodeling of a repressed exon ribonucleoprotein to an exon definition complex active for splicing. eLife 5, e19743 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Shin, K.J. et al. A single lentiviral vector platform for microRNA-based conditional RNA interference and coordinated transgene expression. Proc. Natl. Acad. Sci. USA 103, 13759–13764 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Boyden, E.S., Zhang, F., Bamberg, E., Nagel, G. & Deisseroth, K. Millisecond-timescale, genetically targeted optical control of neural activity. Nat. Neurosci. 8, 1263–1268 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  34. Ito, A. et al. LXRs link metabolism to inflammation through Abca1-dependent regulation of membrane composition and TLR signaling. eLife 4, e08009 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Bhatt, D.M. et al. Transcript dynamics of proinflammatory genes revealed by sequence analysis of subcellular RNA fractions. Cell 150, 279–290 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bradley, M.N. et al. Ligand activation of LXRβ reverses atherosclerosis and cellular cholesterol overload in mice lacking LXRα and apoE. J. Clin. Invest. 117, 2337–2346 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tangirala, R.K., Rubin, E.M. & Palinski, W. Quantitation of atherosclerosis in murine models: correlation between lesions in the aortic origin and in the entire aorta, and differences in the extent of lesions between sexes in LDL receptor–deficient and apolipoprotein E–deficient mice. J. Lipid Res. 36, 2320–2328 (1995).

    CAS  PubMed  Google Scholar 

  38. Feig, J.E. & Fisher, E.A. Laser capture microdissection for analysis of macrophage gene expression from atherosclerotic lesions. Methods Mol. Biol. 1027, 123–135 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Trapnell, C., Pachter, L. & Salzberg, S.L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Trapnell, C. et al. Transcript assembly and quantification by RNA-seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. Zhao, Y. et al. NONCODE 2016: an informative and valuable data source of long non-coding RNAs. Nucleic Acids Res. 44 (D1), D203–D208 (2016).

    Article  CAS  PubMed  Google Scholar 

  43. Koeth, R.A. et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 19, 576–585 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sun, L. et al. Utilizing sequence intrinsic composition to classify protein-coding and long non-coding transcripts. Nucleic Acids Res. 41, e166 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. McLean, C.Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tsai, M.C. et al. Long noncoding RNA as modular scaffold of histone modification complexes. Science 329, 689–693 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Huang, 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 

  48. Buenrostro, J.D., Giresi, P.G., Zaba, L.C., Chang, H.Y. & Greenleaf, W.J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Soneson, C., Love, M.I. & Robinson, M.D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. 4, 1521 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate—a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

Download references

Acknowledgements

We thank members of the Tontonoz, Smale, Black, and Nagy laboratories and the University of California Los Angeles (UCLA) Atherosclerosis Research Unit for technical assistance and useful discussions. This work was supported by the National Institutes of Health grants HL030568, HL066088, HL128822, the Burroughs Wellcome Fund Career Award for Medical Scientists, and the UCLA Cardiovascular Discovery Fund (Lauren B. Leichtman and Arthur E. Levine Investigator Award).

Author information

Authors and Affiliations

Authors

Contributions

T.S. and P.T. conceived and designed the study and guided the interpretation of the results and the preparation of the manuscript. P.T. supervised the study, and T.S. managed the daily experiments. X.W. performed most mouse experiments and data analysis, including for the atherosclerosis study. T.S., M.J., T.G., K.Q., Z.Z., J.S., D.S., P.R., C.H., A.I., and X.L. performed various in vivo and in vitro macrophage experiments and data analysis. Bioinformatic data analysis was performed by D.C. and A.E. B.J.T., X.L., and S.S. assisted with ChIP and ATAC-seq experiments, including data analysis. J.W. performed the mass spectrometry analysis. B.D. and L.N. assisted with ChIP and performed experiments defining enhancer landscape at Abca1. A.C. performed ChIP–seq for LXR. M.C. and A.J.L. provided GWAS data and technical guidance for atherosclerosis analysis. T.S. and P.T. edited the manuscript with input from all authors. All authors discussed the results and approved the final version of the manuscript.

Corresponding authors

Correspondence to Tamer Sallam or Peter Tontonoz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures & Tables

Supplementary Figures 1–14 & Supplementary Tables 1,3–4 (PDF 6140 kb)

Life Sciences Reporting Summary (PDF 171 kb)

Supplementary Table 2

Analysis of lncRNA-adjacent protein-coding genes (XLSX 117 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sallam, T., Jones, M., Thomas, B. et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat Med 24, 304–312 (2018). https://doi.org/10.1038/nm.4479

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4479

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