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:

Reducing endoplasmic reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis

An Erratum to this article was published on 01 February 2010

This article has been updated

Abstract

Macrophages show endoplasmic reticulum (ER) stress when exposed to lipotoxic signals associated with atherosclerosis, although the pathophysiological importance and the underlying mechanisms of this phenomenon remain unknown. Here we show that mitigation of ER stress with a chemical chaperone results in marked protection against lipotoxic death in macrophages and prevents macrophage fatty acid–binding protein-4 (aP2) expression. Using genetic and chemical models, we show that aP2 is the predominant regulator of lipid-induced macrophage ER stress. The absence of lipid chaperones incites an increase in the production of phospholipids rich in monounsaturated fatty acids and bioactive lipids that render macrophages resistant to lipid-induced ER stress. Furthermore, the impact of aP2 on macrophage lipid metabolism and the ER stress response is mediated by upregulation of key lipogenic enzymes by the liver X receptor. Our results demonstrate the central role for lipid chaperones in regulating ER homeostasis in macrophages in atherosclerosis and show that ER responses can be modified, genetically or chemically, to protect the organism against the deleterious effects of hyperlipidemia.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: PBA treatment protects against macrophage ER stress and reduces vascular disease progression.
Figure 2: Requirement for aP2 in lipid-induced ER stress and apoptosis.
Figure 3: aP2 deficiency protects from hypercholesteremia-induced macrophage ER stress and apoptosis in atherosclerotic lesions.
Figure 4: Regulation of macrophage lipid composition by aP2.
Figure 5: A central role for SCD and C16:1n7 in aP2 mediated lipotoxic signaling.
Figure 6: Linking toxic lipids to ER stress and atherosclerosis through aP2–LXR-α crosstalk.

Similar content being viewed by others

Change history

  • 04 February 2010

    In the version of this article initially published, the official symbol for the gene encoding the aP2 protein was misidentified as Tcfap2a (the gene symbol for the transcription factor AP-2). The correct gene symbol is Fabp4. In no instances anywhere in the study was AP-2 examined. Additionally, Supplementary Figure 2a should also have been cited where Figure 2c was cited. The errors have been corrected in the HTML and PDF versions of the article.

References

  1. Hotamisligil, G.S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).

    Article  CAS  Google Scholar 

  2. Gregor, M.F. & Hotamisligil, G.S. Thematic review series: Adipocyte Biology. Adipocyte stress: the endoplasmic reticulum and metabolic disease. J. Lipid Res. 48, 1905–1914 (2007).

    Article  CAS  Google Scholar 

  3. Feng, B. et al. The endoplasmic reticulum is the site of cholesterol-induced cytotoxicity in macrophages. Nat. Cell Biol. 5, 781–792 (2003).

    Article  CAS  Google Scholar 

  4. Tabas, I. Consequences and therapeutic implications of macrophage apoptosis in atherosclerosis: the importance of lesion stage and phagocytic efficiency. Arterioscler. Thromb. Vasc. Biol. 25, 2255–2264 (2005).

    Article  CAS  Google Scholar 

  5. Ozcan, U. et al. Endoplasmic reticulum stress links obesity, insulin action and type 2 diabetes. Science 306, 457–461 (2004).

    Article  Google Scholar 

  6. Ozawa, K. et al. The endoplasmic reticulum chaperone improves insulin resistance in type 2 diabetes. Diabetes 54, 657–663 (2005).

    Article  CAS  Google Scholar 

  7. Ozcan, U. et al. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 313, 1137–1140 (2006).

    Article  Google Scholar 

  8. Myoishi, M. et al. Increased endoplasmic reticulum stress in atherosclerotic plaques associated with acute coronary syndrome. Circulation 116, 1226–1233 (2007).

    Article  Google Scholar 

  9. Ron, D. & Walter, P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat. Rev. Mol. Cell Biol. 8, 519–529 (2007).

    Article  CAS  Google Scholar 

  10. Hotamisligil, G.S. & Erbay, E. Nutrient sensing and inflammation in metabolic diseases. Nat. Rev. Immunol. 8, 923–934 (2008).

    Article  CAS  Google Scholar 

  11. Sriburi, R., Jackowski, S., Mori, K. & Brewer, J.W. XBP1: a link between the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J. Cell Biol. 167, 35–41 (2004).

    Article  CAS  Google Scholar 

  12. Brookheart, R.T., Michel, C.I. & Schaffer, J.E. As a matter of fat. Cell Metab. 10, 9–12 (2009).

    Article  CAS  Google Scholar 

  13. Ferré, P. & Foufelle, F. SREBP-1c transcription factor and lipid homeostasis: clinical perspective. Horm. Res. 68, 72–82 (2007).

    Google Scholar 

  14. Oyadomari, S., Harding, H.P., Zhang, Y., Oyadomari, M. & Ron, D. Dephosphorylation of translation initiation factor 2α enhances glucose tolerance and attenuates hepatosteatosis in mice. Cell Metab. 7, 520–532 (2008).

    Article  CAS  Google Scholar 

  15. Rutkowski, D.T. et al. UPR pathways combine to prevent hepatic steatosis caused by ER stress–mediated suppression of transcriptional master regulators. Dev. Cell 15, 829–840 (2008).

    Article  CAS  Google Scholar 

  16. Ramanadham, S. et al. Apoptosis of insulin-secreting cells induced by endoplasmic reticulum stress is amplified by overexpression of group VIA calcium–independent phospholipase A2 (iPLA2 β) and suppressed by inhibition of iPLA2 β. Biochemistry 43, 918–930 (2004).

    Article  CAS  Google Scholar 

  17. Tessitore, A. et al. GM1-ganglioside–mediated activation of the unfolded protein response causes neuronal death in a neurodegenerative gangliosidosis. Mol. Cell 15, 753–766 (2004).

    Article  CAS  Google Scholar 

  18. Furuhashi, M. & Hotamisligil, G.S. Fatty acid–binding proteins: role in metabolic diseases and potential as drug targets. Nat. Rev. Drug Discov. 7, 489–503 (2008).

    Article  CAS  Google Scholar 

  19. Shum, B.O. et al. The adipocyte fatty acid–binding protein aP2 is required in allergic airway inflammation. J. Clin. Invest. 116, 2183–2192 (2006).

    Article  CAS  Google Scholar 

  20. Hotamisligil, G.S. et al. Uncoupling of obesity from insulin resistance through a targeted mutation in aP2, the adipocyte fatty acid binding protein. Science 274, 1377–1379 (1996).

    Article  CAS  Google Scholar 

  21. Uysal, K.T., Scheja, L., Wiesbrock, S.M., Bonner-Weir, S. & Hotamisligil, G.S. Improved glucose and lipid metabolism in genetically obese mice lacking aP2. Endocrinology 141, 3388–3396 (2000).

    Article  CAS  Google Scholar 

  22. Makowski, L. et al. Lack of macrophage fatty-acid–binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nat. Med. 7, 699–705 (2001).

    Article  CAS  Google Scholar 

  23. Cao, H. et al. Regulation of metabolic responses by adipocyte/macrophage fatty acid–binding proteins in leptin-deficient mice. Diabetes 55, 1915–1922 (2006).

    Article  CAS  Google Scholar 

  24. Ozcan, U. et al. Loss of the tuberous sclerosis complex tumor suppressors triggers the unfolded protein response to regulate insulin signaling and apoptosis. Mol. Cell 29, 541–551 (2008).

    Article  CAS  Google Scholar 

  25. Borradaile, N.M. et al. Disruption of endoplasmic reticulum structure and integrity in lipotoxic cell death. J. Lipid Res. 47, 2726–2737 (2006).

    Article  CAS  Google Scholar 

  26. Lai, E., Bikopoulos, G., Wheeler, M.B., Rozakis-Adcock, M. & Volchuk, A. Differential activation of ER stress and apoptosis in response to chronically elevated free fatty acids in pancreatic beta-cells. Am. J. Physiol. Endocrinol. Metab. 294, E540–E550 (2008).

    Article  CAS  Google Scholar 

  27. Schaffer, J.E. Lipotoxicity: when tissues overeat. Curr. Opin. Lipidol. 14, 281–287 (2003).

    Article  CAS  Google Scholar 

  28. Tuncman, G. et al. A genetic variant at the fatty acid–binding protein aP2 locus reduces the risk for hypertriglyceridemia, type 2 diabetes and cardiovascular disease. Proc. Natl. Acad. Sci. USA 103, 6970–6975 (2006).

    Article  CAS  Google Scholar 

  29. Furuhashi, M. et al. Treatment of diabetes and atherosclerosis by inhibiting fatty-acid–binding protein aP2. Nature 447, 959–965 (2007).

    Article  CAS  Google Scholar 

  30. Sha, R.S., Kane, C.D., Xu, Z., Banaszak, L.J. & Bernlohr, D.A. Modulation of ligand binding affinity of the adipocyte lipid-binding protein by selective mutation. Analysis in vitro and in situ. J. Biol. Chem. 268, 7885–7892 (1993).

    CAS  PubMed  Google Scholar 

  31. Makowski, L., Brittingham, K.C., Reynolds, J.M., Suttles, J. & Hotamisligil, G.S. The fatty acid-binding protein, aP2, coordinates macrophage cholesterol trafficking and inflammatory activity. Macrophage expression of aP2 impacts peroxisome proliferator-activated receptor γ and IκB kinase activities. J. Biol. Chem. 280, 12888–12895 (2005).

    Article  CAS  Google Scholar 

  32. Enoch, H.G., Catala, A. & Strittmatter, P. Mechanism of rat liver microsomal stearyl-CoA desaturase. Studies of the substrate specificity, enzyme-substrate interactions, and the function of lipid. J. Biol. Chem. 251, 5095–5103 (1976).

    CAS  PubMed  Google Scholar 

  33. Heinemann, F.S. & Ozols, J. Stearoyl-CoA desaturase, a short-lived protein of endoplasmic reticulum with multiple control mechanisms. Prostaglandins Leukot. Essent. Fatty Acids 68, 123–133 (2003).

    Article  CAS  Google Scholar 

  34. Cao, H. et al. Identification of a lipokine, a lipid hormone linking adipose tissue to systemic metabolism. Cell 134, 933–944 (2008).

    Article  CAS  Google Scholar 

  35. Joseph, S.B. et al. Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by liver X receptors. J. Biol. Chem. 277, 11019–11025 (2002).

    Article  CAS  Google Scholar 

  36. Chu, K., Miyazaki, M., Man, W.C. & Ntambi, J.M. Stearoyl-coenzyme A desaturase 1 deficiency protects against hypertriglyceridemia and increases plasma high-density lipoprotein cholesterol induced by liver X receptor activation. Mol. Cell. Biol. 26, 6786–6798 (2006).

    Article  CAS  Google Scholar 

  37. Venkateswaran, A. et al. Human white/murine ABC8 mRNA levels are highly induced in lipid-loaded macrophages. A transcriptional role for specific oxysterols. J. Biol. Chem. 275, 14700–14707 (2000).

    Article  CAS  Google Scholar 

  38. Repa, J.J. et al. Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 289, 1524–1529 (2000).

    Article  CAS  Google Scholar 

  39. Joseph, S.B. et al. LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 119, 299–309 (2004).

    Article  CAS  Google Scholar 

  40. 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  Google Scholar 

  41. Aqel, N.M., Ball, R.Y., Waldmann, H. & Mitchinson, M.J. Monocytic origin of foam cells in human atherosclerotic plaques. Atherosclerosis 53, 265–271 (1984).

    Article  CAS  Google Scholar 

  42. Han, S. et al. Macrophage insulin receptor deficiency increases ER stress-induced apoptosis and necrotic core formation in advanced atherosclerotic lesions. Cell Metab. 3, 257–266 (2006).

    Article  CAS  Google Scholar 

  43. Boord, J.B. et al. Adipocyte fatty acid–binding protein, aP2, alters late atherosclerotic lesion formation in severe hypercholesterolemia. Arterioscler. Thromb. Vasc. Biol. 22, 1686–1691 (2002).

    Article  CAS  Google Scholar 

  44. Listenberger, L.L. et al. Triglyceride accumulation protects against fatty acid–induced lipotoxicity. Proc. Natl. Acad. Sci. USA 100, 3077–3082 (2003).

    Article  CAS  Google Scholar 

  45. Brown, J.M. et al. Inhibition of stearoyl-coenzyme A desaturase 1 dissociates insulin resistance and obesity from atherosclerosis. Circulation 118, 1467–1475 (2008).

    Article  CAS  Google Scholar 

  46. Rolph, M.S. et al. Regulation of dendritic cell function and T cell priming by the fatty acid–binding protein aP2. J. Immunol. 177, 7794–7801 (2006).

    Article  CAS  Google Scholar 

  47. Erbay, E., Cao, H. & Hotamisligil, G.S. Adipocyte/macrophage fatty acid binding proteins in metabolic syndrome. Curr. Atheroscler. Rep. 9, 222–229 (2007).

    Article  CAS  Google Scholar 

  48. Gregor, M.F. et al. Endoplasmic reticulum stress is reduced in tissues of obese subjects after weight loss. Diabetes 58, 693–700 (2009).

    Article  CAS  Google Scholar 

  49. Joseph, S.B. et al. LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 119, 299–309 (2004).

    Article  CAS  Google Scholar 

  50. Furuhashi, M. et al. Treatment of diabetes and atherosclerosis by inhibiting fatty-acid-binding protein aP2. Nature 447, 959–965 (2007).

    Article  CAS  Google Scholar 

  51. Venkateswaran, A. et al. Human white/murine ABC8 mRNA levels are highly induced in lipid-loaded macrophages. A transcriptional role for oxysterols. J. Biol. Chem. 275, 14700–14707 (2000).

    Article  CAS  Google Scholar 

  52. Pan, W. On the use of permutation in and the performance of a class of nonparametric methods to detect differential gene expression. Bioinformatics 19, 1333–1340 (2003).

    Article  CAS  Google Scholar 

  53. Kuehl, R.O . Design of Experiments: Statistical Principles of Research Design and Analysis. 2nd edn., 56–58 (Duxbury Press, Pacific Grove, California, 2000).

Download references

Acknowledgements

This project has been supported by grants from the US National Institutes of Health DK DK52539 (to G.S.H.), HL65405 (to M.F.L. and G.S.H.) and DK59637 (Lipid, Lipoprotein and Atherosclerosis Core of the Vanderbilt Mouse Metabolic Phenotype Center). E.E. is supported by the Ruth Kirschstein National Research Award (F32 HL090258). We are grateful to the members of the Hotamisligil lab, J. Chen and R. Bachman for their scientific input and contributions, to A. Onur for technical assistance, to R. Foote and K. Gilbert for administrative support, to D. Mangelsdorf (University of Texas Southwestern) for TK-LXRE-X3luc reporter and Nr1h3−/− mice, and A. Edgar (Fournier) for the ACAT inhibitor. The pGEX-aP2-LM (R126L, Y128F) plasmid was a generous gift from D. Bernlohr (University of Minnesota).

Author information

Authors and Affiliations

Authors

Contributions

E.E. developed the hypothesis, designed and performed the bulk of the experiments, analyzed all data and wrote the manuscript; V.R.B. contributed to the in vivo studies; J.R.M. contributed to the in vitro studies and conducted data analysis, K.N.C. and M.E.S. contributed to the in vitro studies; L.M., M.M.W. and S.M.W. contributed to the analysis of lipidomic data; S.F. and M.F.L. contributed to the analysis of in vivo data and assisted with writing; and G.S.H. developed the hypothesis, designed and analyzed all data, wrote the manuscript and supervised the project and the peer review process.

Corresponding author

Correspondence to Gökhan S Hotamisligil.

Ethics declarations

Competing interests

G.S.H. is on the scientific advisory board of Syndexa Pharmaceuticals. M.M.W. and S.M.W. work for Lipomics, an organization for profit.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Table 1 (PDF 969 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Erbay, E., Babaev, V., Mayers, J. et al. Reducing endoplasmic reticulum stress through a macrophage lipid chaperone alleviates atherosclerosis. Nat Med 15, 1383–1391 (2009). https://doi.org/10.1038/nm.2067

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2067

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