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:

Genome-wide identification of microRNAs regulating cholesterol and triglyceride homeostasis

Subjects

Abstract

Genome-wide association studies (GWASs) have linked genes to various pathological traits. However, the potential contribution of regulatory noncoding RNAs, such as microRNAs (miRNAs), to a genetic predisposition to pathological conditions has remained unclear. We leveraged GWAS meta-analysis data from >188,000 individuals to identify 69 miRNAs in physical proximity to single-nucleotide polymorphisms (SNPs) associated with abnormal levels of circulating lipids. Several of these miRNAs (miR-128-1, miR-148a, miR-130b, and miR-301b) control the expression of key proteins involved in cholesterol-lipoprotein trafficking, such as the low-density lipoprotein (LDL) receptor (LDLR) and the ATP-binding cassette A1 (ABCA1) cholesterol transporter. Consistent with human liver expression data and genetic links to abnormal blood lipid levels, overexpression and antisense targeting of miR-128-1 or miR-148a in high-fat diet–fed C57BL/6J and Apoe-null mice resulted in altered hepatic expression of proteins involved in lipid trafficking and metabolism, and in modulated levels of circulating lipoprotein-cholesterol and triglycerides. Taken together, these findings support the notion that altered expression of miRNAs may contribute to abnormal blood lipid levels, predisposing individuals to human cardiometabolic disorders.

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: Identification of miRNAs located in genomic loci that are enriched for SNPs associated with abnormal circulating total cholesterol, triglyceride, LDL-C and HDL-C levels.
Figure 2: Expression of LDLR and ABCA1 is regulated by miRNAs located in genomic loci associated with abnormal circulating blood lipid levels.
Figure 3: miR-128-1, miR-148a, miR-130b and miR-301b regulate ABCA1 expression and cholesterol efflux in mouse macrophages.
Figure 4: Overexpression of miR-128-1 and miR-148a in C57BL/6J mice decreases circulating levels of HDL-C, whereas antisense antagonism of these miRNAs increases LDL-C clearance.
Figure 5: Antisense inhibition of miR-148a in Apoe−/− mice fed a Western-type diet results in altered circulating lipoprotein/lipid levels.
Figure 6: Antisense inhibition of miR-128-1 in Apoe−/− mice fed a Western-type diet results in altered circulating lipoprotein/lipid levels, improved glucose homeostasis/insulin signaling and decreased hepatic steatosis.

Similar content being viewed by others

Accession codes

Accessions

Gene Expression Omnibus

References

  1. Cornier, M.A. et al. The metabolic syndrome. Endocr. Rev. 29, 777–822 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Go, A.S. et al. Heart disease and stroke statistics–2013 update: a report from the American Heart Association. Circulation 127, e6–e245 (2013).

    PubMed  Google Scholar 

  3. Quiat, D. & Olson, E.N. MicroRNAs in cardiovascular disease: from pathogenesis to prevention and treatment. J. Clin. Invest. 123, 11–18 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Rottiers, V. & Naar, A.M. MicroRNAs in metabolism and metabolic disorders. Nat. Rev. Mol. Cell Biol. 13, 239–250 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. 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 

  7. 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  PubMed  PubMed Central  Google Scholar 

  8. Horie, T. et al. MicroRNA-33 encoded by an intron of sterol regulatory element-binding protein 2 (Srebp2) regulates HDL in vivo. Proc. Natl. Acad. Sci. USA 107, 17321–17326 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  9. 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  PubMed  PubMed Central  Google Scholar 

  10. Goedeke, L., Aranda, J.F. & Fernandez-Hernando, C. microRNA regulation of lipoprotein metabolism. Curr. Opin. Lipidol. 25, 282–288 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Willer, C.J. et al. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Merino, D.M., Ma, D.W. & Mutch, D.M. Genetic variation in lipid desaturases and its impact on the development of human disease. Lipids Health Dis. 9, 63 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Charles, M.A. & Kane, J.P. New molecular insights into CETP structure and function: a review. J. Lipid Res. 53, 1451–1458 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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 

  15. Dennis, G. Jr. et al. DAVID: Database for Annotation, Visualization, and Integrated Discovery. Genome Biol. 4, P3 (2003).

    Article  PubMed  Google Scholar 

  16. Ishibashi, S. et al. Hypercholesterolemia in low density lipoprotein receptor knockout mice and its reversal by adenovirus-mediated gene delivery. J. Clin. Invest. 92, 883–893 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Brown, M.S. & Goldstein, J.L. How LDL receptors influence cholesterol and atherosclerosis. Sci. Am. 251, 58–66 (1984).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  19. Navab, M., Reddy, S.T., Van Lenten, B.J. & Fogelman, A.M. HDL and cardiovascular disease: atherogenic and atheroprotective mechanisms. Nat. Rev. Cardiol. 8, 222–232 (2011).

    Article  CAS  PubMed  Google Scholar 

  20. Duffy, D. & Rader, D.J. Update on strategies to increase HDL quantity and function. Nat. Rev. Cardiol. 6, 455–463 (2009).

    Article  PubMed  Google Scholar 

  21. Sparks, J.D., Sparks, C.E. & Adeli, K. Selective hepatic insulin resistance, VLDL overproduction, and hypertriglyceridemia. Arterioscler. Thromb. Vasc. Biol. 32, 2104–2112 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Stein, S. & Matter, C.M. Protective roles of SIRT1 in atherosclerosis. Cell Cycle 10, 640–647 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Yoon, Y.S., Seo, W.Y., Lee, M.W., Kim, S.T. & Koo, S.H. Salt-inducible kinase regulates hepatic lipogenesis by controlling SREBP-1c phosphorylation. J. Biol. Chem. 284, 10446–10452 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Hardie, D.G. Organismal carbohydrate and lipid homeostasis. Cold Spring Harb. Perspect. Biol. 4 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bonnefont, J.P. et al. Carnitine palmitoyltransferases 1 and 2: biochemical, molecular and medical aspects. Mol. Aspects Med. 25, 495–520 (2004).

    Article  CAS  PubMed  Google Scholar 

  26. Goldstein, J.L., DeBose-Boyd, R.A. & Brown, M.S. Protein sensors for membrane sterols. Cell 124, 35–46 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Goldstein, J.L. & Brown, M.S. The LDL receptor. Arterioscler. Thromb. Vasc. Biol. 29, 431–438 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Soutar, A.K. & Naoumova, R.P. Mechanisms of disease: genetic causes of familial hypercholesterolemia. Nat. Clin. Pract. Cardiovasc. Med. 4, 214–225 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Lu, Y.C. et al. ELAVL1 modulates transcriptome-wide miRNA binding in murine macrophages. Cell Rep. 9, 2330–2343 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhang, S.H., Reddick, R.L., Piedrahita, J.A. & Maeda, N. Spontaneous hypercholesterolemia and arterial lesions in mice lacking apolipoprotein E. Science 258, 468–471 (1992).

    Article  CAS  PubMed  Google Scholar 

  31. Nakashima, Y., Plump, A.S., Raines, E.W., Breslow, J.L. & Ross, R. ApoE-deficient mice develop lesions of all phases of atherosclerosis throughout the arterial tree. Arterioscler. Thromb. 14, 133–140 (1994).

    Article  CAS  PubMed  Google Scholar 

  32. Teslovich, T.M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hatoum, I.J. et al. Melanocortin-4 receptor signaling is required for weight loss after gastric bypass surgery. J. Clin. Endocrinol. Metab. 97, E1023–E1031 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Greenawalt, D.M. et al. A survey of the genetics of stomach, liver, and adipose gene expression from a morbidly obese cohort. Genome Res. 21, 1008–1016 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Hatoum, I.J. et al. Weight loss after gastric bypass is associated with a variant at 15q26.1. Am. J. Hum. Genet. 92, 827–834 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Raymond, C.K., Roberts, B.S., Garrett-Engele, P., Lim, L.P. & Johnson, J.M. Simple, quantitative primer-extension PCR assay for direct monitoring of microRNAs and short-interfering RNAs. RNA 11, 1737–1744 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bates, D. et al. Fitting linear mixed-effects models using lme4. arXiv, http://arxiv.org/abs/1406.5823 (2014).

  38. Liu, E.Y., Li, M., Wang, W. & Li, Y. MaCH-admix: genotype imputation for admixed populations. Genet. Epidemiol. 37, 25–37 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).

    Article  CAS  PubMed  Google Scholar 

  40. Shabalin, A.A. Matrix eQTL: ultra fast eQTL analysis via large matrix operations. Bioinformatics 28, 1353–1358 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Weischenfeldt, J. & Porse, B. Bone marrow-derived macrophages (BMM): isolation and applications. CSH Protoc 2008 doi:10.1101/pdb.prot5080 (2010).

  42. Hafner, M. et al. Identification of microRNAs and other small regulatory RNAs using cDNA library sequencing. Methods 44, 3–12 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Hafner, M. et al. Transcriptome-wide identification of RNA-binding protein and microRNA target sites by PAR-CLIP. Cell 141, 129–141 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Corcoran, D.L. et al. PARalyzer: definition of RNA binding sites from PAR-CLIP short-read sequence data. Genome Biol. 12, R79 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. 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 

  46. 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 

  47. Lillie, R.D. & Fulmer, H.M. Histopathologic Technique and Practical Histochemistry. (McGraw-Hill, 1976).

  48. Ljungberg, O. & Tibblin, S. Peroperative fat staining of frozen sections in primary hyperparathyroidism. Am. J. Pathol. 95, 633–641 (1979).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Galabova, G. et al. Peptide-based anti-PCSK9 vaccines - an approach for long-term LDLc management. PLoS ONE 9, e114469 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by the following grants: US National Institutes of Health (NIH) grants R21DK084459 and R01DK094184 (A.M.N); R37DK048873 and R01DK056626 to D.E.C.; K24DK078772 to R.T.C.; and R01HL107953 and R01HL106063 to C.F.-H. A.M.N. was supported by a scholar award from the Massachusetts General Hospital (MGH). R.E.G. was supported by an Established Investigator Award from the American Heart Association. C.F.-H. was supported by a Fondation Leducq Transatlantic Network of Excellence in Cardiovascular Research Award. T.H. acknowledges the support of NIH grant R01HL49094 and a Fondation Leducq Transatlantic Network of Excellence in Cardiovascular Research Award. N.S. and J.S.T. were supported by the Intramural Research Program of the US National Institute of Allergy and Infectious Diseases. C.M.R. was supported by the American Heart Association SDG Grant 15SDG23000025. A.W. was supported by a fellowship from the MGH Executive Committee on Research. We thank C. Molony of Merck Research Laboratories for help with genotyping data quality assessment. We thank the Harvard Medical School Neurobiology Department and the Neurobiology Imaging Facility for consultation and instrument availability that supported this work. This facility is supported in part by the Neural Imaging Center as part of a National Institutes on Neurological Disorders and Stroke P30 Core Center grant no. NS072030, and by the Harvard Digestive Diseases Center (P30 DK034854).

Author information

Authors and Affiliations

Authors

Contributions

S.H.N.-S. and A.M.N. conceived and carried out the initial miR-128-1 studies that provided support for the expanded project. A.W. and A.M.N. conceived and designed the expanded, published studies, interpreted the data, and wrote the manuscript, which was commented on by all authors. A.W., S.H.N.-S., L.W., S.S., Y. Li, F.K., N.P., D.E.C. and R.E.G. performed experiments and analyzed data in Apoe deficient mice and in C57BL/6 mice. L.G., C.M.R. and C.F.-H. quantified miRNAs in Rhesus monkeys and mice fed with different diets and performed efflux experiments in mouse peritoneal macrophages. N.S., Y. Lu, J.S.T., E.S., R.T., I.H., P.C.S. and L.M.K. contributed to the human liver miR-QTL analysis. Y.-C.L. and T.H. performed the Ago2 PAR-CLIP analysis in BMDMs. A.S.d. and R.T.C. performed the liver histology analysis. V.V. injected the lentivirus in C57BL/6 mice. J.C.B. and J.W. performed DNA microarray analysis from mouse liver samples prepared by A.W. S.K. and A.B. analyzed the GWAS data. R.d.C. carried out the non-human primate studies.

Corresponding author

Correspondence to Anders M Näär.

Ethics declarations

Competing interests

A.M.N., A.W. and S.H.N.-S. have issued and pending patents (US Patent nos. 9,045,749 and US 61/865,327) on microRNA-targeting therapeutics for the treatment of cardiometabolic diseases.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 3650 kb)

Supplementary Table 1

69 miRNAs are associated with abnormal levels of total cholesterol (TC), LDL-C, HDL-C, and TAGs. (XLSX 65 kb)

Supplementary Table 2

Gene Ontology analysis of miRNA target genes. (XLSX 37 kb)

Supplementary Table 3

List of SNPs associated with total cholesterol, LDL-C, HDL- C and triglycerides in the miR-128-1, miR-148a, miR-130b and miR-301b loci. (XLSX 121 kb)

Supplementary Table 4

Selected metabolism genes predicted to be targeted by miR- 128-1, miR-148a, miR-130b and miR-301b. (XLSX 46 kb)

Supplementary Table 5

Liver cis miR-QTL data. (XLSX 1002 kb)

Supplementary Table 6

Expression changes of predicted miR-128-1 targets in the livers of mice treated with antimiR-128-1 versus control scrambled LNA antimiR over 5 days. (XLSX 91 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wagschal, A., Najafi-Shoushtari, S., Wang, L. et al. Genome-wide identification of microRNAs regulating cholesterol and triglyceride homeostasis. Nat Med 21, 1290–1297 (2015). https://doi.org/10.1038/nm.3980

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.3980

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