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:

Cholesterol and fatty acids regulate cysteine ubiquitylation of ACAT2 through competitive oxidation

A Corrigendum to this article was published on 01 December 2017

This article has been updated

Abstract

Ubiquitin linkage to cysteine is an unconventional modification targeting protein for degradation. However, the physiological regulation of cysteine ubiquitylation is still mysterious. Here we found that ACAT2, a cellular enzyme converting cholesterol and fatty acid to cholesteryl esters, was ubiquitylated on Cys277 for degradation when the lipid level was low. gp78–Insigs catalysed Lys48-linked polyubiquitylation on this Cys277. A high concentration of cholesterol and fatty acid, however, induced cellular reactive oxygen species (ROS) that oxidized Cys277, resulting in ACAT2 stabilization and subsequently elevated cholesteryl esters. Furthermore, ACAT2 knockout mice were more susceptible to high-fat diet-associated insulin resistance. By contrast, expression of a constitutively stable form of ACAT2 (C277A) resulted in higher insulin sensitivity. Together, these data indicate that lipid-induced stabilization of ACAT2 ameliorates lipotoxicity from excessive cholesterol and fatty acid. This unconventional cysteine ubiquitylation of ACAT2 constitutes an important mechanism for sensing lipid-overload-induced ROS and fine-tuning lipid homeostasis.

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: ACAT2 is stabilized by sterols and saturated fatty acids.
Figure 2: Lipid-regulated ACAT2 ubiquitylation occurs on Cys277.
Figure 3: ACAT2 is ubiquitylated by Lys48-specific ubiquitin linkages.
Figure 4: The ubiquitylation on Cys277 of ACAT2 is regulated by oxidative stress.
Figure 5: The gp78–Insigs E3 complex mediates the ubiquitylation and degradation of ACTA2.
Figure 6: ACAT2−/− mice are more susceptible to HFD-induced insulin resistance.
Figure 7: The stabilization of hepatic ACAT2 ameliorates lipotoxicity and protects HFD-induced insulin resistance in mice.

Similar content being viewed by others

Change history

  • 13 November 2017

    In the original version of this Article, the procedure for assessing the ubiquitylation assays by western blotting was inadvertently omitted from the Methods. This method was used in Figures 2b,d,g,h, 3a,b,d, 4f and 5h and Supplementary Figures 3b, 5b and 6b, and was designed to omit reagents such as BME and DTT that might break the thioester bond. The full method is given below, and has been added to the Methods section of the Article. Ubiquitylation assay western blot. For Figs 2b,d,g,h, 3a,b, 4f and 5h, and Supplementary Figs 5b and 6b, after the last wash of the beads, the supernatant was discarded and beads were boiled for 10 min in 100 μl 2× SDS loading buffer (75 mM Tris-HCl, pH 6.8, 6% SDS, 15% (v/v) glycerol, 0.01% (w/v) Bromophenol blue) and were then vortexed and centrifuged at 1,000 g for 2 min. 90 μl supernatant was mixed with 90 μl solubilization buffer (62.5 mM Tris-HCl, pH 6.8, 15% SDS, 8 M Urea, 10% glycerol) and incubated at 37 °C for 30 min. For Fig. 3d and Supplementary Fig. 3b, the beads were incubated with 100 μl Myc peptide (1 mg/ml) for 3 hrs at 4 °C, vortexed and centrifuged at 1,000 g for 2 min. 90 μl supernatant was mixed with 90 μl solubilization buffer (62.5 mM Tris-HCl, pH 6.8, 15% SDS, 8 M Urea, 10% glycerol) and 60 μl modified 4× SDS loading buffer (150 mM Tris-HCl, pH 6.8, 12% SDS, 30% (v/v) glycerol, 0.02% (w/v) Bromophenol blue) and incubated at 37 °C for 30 min. Samples were resolved by SDS–PAGE and transferred onto PVDF membranes. Immunoblots were blocked with 5% BSA in TBS containing 0.075% Tween (TBST) and probed with primary antibodies overnight at 4 °C. After washing in TBST 3 times, blots were incubated with secondary antibodies for 1 h at room temperature. After washing in TBST 3 times, bands were visualized by enhanced chemiluminescence (ECL).

References

  1. Cadwell, K. & Coscoy, L. Ubiquitination on nonlysine residues by a viral E3 ubiquitin ligase. Science 309, 127–130 (2005).

    Article  CAS  Google Scholar 

  2. Boden, G. Obesity, insulin resistance and free fatty acids. Curr. Opin. Endocrinol. Diabetes Obes. 18, 139–143 (2011).

    Article  CAS  Google Scholar 

  3. Anavi, S., Hahn-Obercyger, M., Madar, Z. & Tirosh, O. Mechanism for HIF-1 activation by cholesterol under normoxia: a redox signaling pathway for liver damage. Free Radic. Biol. Med. 71, 61–69 (2014).

    Article  CAS  Google Scholar 

  4. Poli, G., Biasi, F. & Leonarduzzi, G. Oxysterols in the pathogenesis of major chronic diseases. Redox Biol. 1, 125–130 (2013).

    Article  CAS  Google Scholar 

  5. Maloney, E. et al. Activation of NF-kappaB by palmitate in endothelial cells: a key role for NADPH oxidase-derived superoxide in response to TLR4 activation. Arterioscler. Thromb. Vasc. Biol. 29, 1370–1375 (2009).

    Article  CAS  Google Scholar 

  6. Brodeur, M. R., Bouvet, C., Barrette, M. & Moreau, P. Palmitic acid increases medial calcification by inducing oxidative stress. J. Vasc. Res. 50, 430–441 (2013).

    Article  CAS  Google Scholar 

  7. Chang, T. Y., Chang, C. C. & Cheng, D. Acyl-coenzyme A:cholesterol acyltransferase. Annu. Rev. Biochem. 66, 613–638 (1997).

    Article  CAS  Google Scholar 

  8. Farese, R. V. J. Acyl-CoA:cholesterol acyltransferase genes and knockout mice. Curr. Opin. Lipidol. 9, 119–124 (1998).

    Article  CAS  Google Scholar 

  9. Chang, T. Y. et al. Roles of acyl-coenzyme A:cholesterol acyltransferase-1 and -2. Curr. Opin. Lipidol. 12, 289–296 (2001).

    Article  CAS  Google Scholar 

  10. Rudel, L., Lee, R. & Cockman, T. Structure, function, and regulation of ACAT. Curr. Opin. Lipidol. 12, 121–127 (2001).

    Article  CAS  Google Scholar 

  11. Liu, J., Chang, C. C., Westover, E. J., Covey, D. F. & Chang, T. Y. Investigating the allosterism of acyl-CoA:cholesterol acyltransferase (ACAT) by using various sterols: in vitro and intact cell studies. Biochem. J. 391, 389–397 (2005).

    Article  CAS  Google Scholar 

  12. Wang, X. et al. Ubiquitination of serine, threonine, or lysine residues on the cytoplasmic tail can induce ERAD of MHC-I by viral E3 ligase mK3. J. Cell. Biol. 177, 613–624 (2007).

    Article  CAS  Google Scholar 

  13. Charles, R. L. et al. Protein sulfenation as a redox sensor: proteomics studies using a novel biotinylated dimedone analogue. Mol. Cell. Proteomics 6, 1473–1484 (2007).

    Article  CAS  Google Scholar 

  14. Poole, L. B. et al. Fluorescent and affinity-based tools to detect cysteine sulfenic acid formation in proteins. Bioconjug. Chem. 18, 2004–2017 (2007).

    Article  CAS  Google Scholar 

  15. Reddie, K. G., Seo, Y. H., Muse III, W. B., Leonard, S. E. & Carroll, K. S. A chemical approach for detecting sulfenic acid-modified proteins in living cells. Mol. Biosyst. 4, 521–531 (2008).

    Article  CAS  Google Scholar 

  16. Song, B. L., Sever, N. & Bose-Boyd, R. A. Gp78, a membrane-anchored ubiquitin ligase, associates with Insig-1 and couples sterol-regulated ubiquitination to degradation of HMG CoA reductase. Mol. Cell. 19, 829–840 (2005).

    Article  CAS  Google Scholar 

  17. Lee, P. C., Sever, N. & Debose-Boyd, R. A. Isolation of sterol-resistant Chinese hamster ovary cells with genetic deficiencies in both Insig-1 and Insig-2. J. Biol. Chem. 280, 25242–25249 (2005).

    Article  CAS  Google Scholar 

  18. Das, A., Davis, M. A. & Rudel, L. L. Identification of putative active site residues of ACAT enzymes. J. Lipid Res. 49, 1770–1781 (2008).

    Article  CAS  Google Scholar 

  19. Shimizu, K. et al. The autocrine motility factor receptor gene encodes a novel type of seven transmembrane protein. FEBS Lett. 456, 295–300 (1999).

    Article  CAS  Google Scholar 

  20. Fang, S. et al. The tumor autocrine motility factor receptor, gp78, is a ubiquitin protein ligase implicated in degradation from the endoplasmic reticulum. Proc. Natl Acad. Sci. USA 98, 14422–14427 (2001).

    Article  CAS  Google Scholar 

  21. Cao, J. et al. Ufd1 is a cofactor of gp78 and plays a key role in cholesterol metabolism by regulating the stability of HMG-CoA reductase. Cell Metab. 6, 115–128 (2007).

    Article  CAS  Google Scholar 

  22. Zhong, X. et al. AAA ATPase p97/valosin-containing protein interacts with gp78, a ubiquitin ligase for endoplasmic reticulum-associated degradation. J. Biol. Chem. 279, 45676–45684 (2004).

    Article  CAS  Google Scholar 

  23. Chen, B. et al. The activity of a human endoplasmic reticulum-associated degradation E3, gp78, requires its Cue domain, RING finger, and an E2-binding site. Proc. Natl Acad. Sci. USA 103, 341–346 (2006).

    Article  CAS  Google Scholar 

  24. Sever, N. et al. Insig-dependent ubiquitination and degradation of mammalian 3-hydroxy-3-methylglutaryl-CoA reductase stimulated by sterols and geranylgeraniol. J. Biol. Chem. 278, 52479–52490 (2003).

    Article  CAS  Google Scholar 

  25. Sharpe, L. J., Cook, E. C., Zelcer, N. & Brown, A. J. The UPS and downs of cholesterol homeostasis. Trends Biochem. Sci. 39, 527–535 (2014).

    Article  CAS  Google Scholar 

  26. Prabhu, A. V., Luu, W., Sharpe, L. J. & Brown, A. J. Cholesterol-mediated degradation of 7-dehydrocholesterol reductase switches the balance from cholesterol to vitamin D synthesis. J. Biol. Chem. 291, 8363–8373 (2016).

    Article  CAS  Google Scholar 

  27. Gill, S., Stevenson, J., Kristiana, I. & Brown, A. J. Cholesterol-dependent degradation of squalene monooxygenase, a control point in cholesterol synthesis beyond HMG-CoA reductase. Cell Metab. 13, 260–273 (2011).

    Article  CAS  Google Scholar 

  28. Brown, J. M., Rudel, L. L. & Yu, L. NPC1L1 (Niemann-Pick C1-like 1) mediates sterol-specific unidirectional transport of non-esterified cholesterol in McArdle-RH7777 hepatoma cells. Biochem. J. 406, 273–283 (2007).

    Article  CAS  Google Scholar 

  29. Ge, L. et al. The cholesterol absorption inhibitor ezetimibe acts by blocking the sterol-induced internalization of NPC1L1. Cell Metab. 7, 508–519 (2008).

    Article  CAS  Google Scholar 

  30. Yu, L., von Bergmann, K., Lutjohann, D., Hobbs, H. H. & Cohen, J. C. Selective sterol accumulation in ABCG5/ABCG8-deficient mice. J. Lipid Res. 45, 301–307 (2004).

    Article  CAS  Google Scholar 

  31. Berge, K. E. et al. Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science 290, 1771–1775 (2000).

    Article  CAS  Google Scholar 

  32. Martins, A. R. et al. Mechanisms underlying skeletal muscle insulin resistance induced by fatty acids: importance of the mitochondrial function. Lipids Health Dis. 11, 30 (2012).

    Article  CAS  Google Scholar 

  33. Miyazaki, M., Kim, Y. C., Gray-Keller, M. P., Attie, A. D. & Ntambi, J. M. The biosynthesis of hepatic cholesterol esters and triglycerides is impaired in mice with a disruption of the gene for stearoyl-CoA desaturase 1. J. Biol. Chem. 275, 30132–30138 (2000).

    Article  CAS  Google Scholar 

  34. Cases, S. et al. ACAT-2, a second mammalian acyl-CoA:cholesterol acyltransferase. Its cloning, expression, and characterization. J. Biol. Chem. 273, 26755–26764 (1998).

    Article  CAS  Google Scholar 

  35. Li, H. et al. A genome-wide association study identifies GRK5 and RASGRP1 as type 2 diabetes loci in Chinese Hans. Diabetes 62, 291–298 (2013).

    Article  CAS  Google Scholar 

  36. Morris, A. P. et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nat. Genet. 44, 981–990 (2012).

    Article  CAS  Google Scholar 

  37. Mahajan, A. et al. Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat. Genet. 46, 234–244 (2014).

    Article  CAS  Google Scholar 

  38. Goldstein, J. L., Basu, S. K. & Brown, M. S. Receptor-mediatedendocytosis of low-density lipoprotein in cultured cells. Methods Enzymol. 98, 241–260 (1983).

    Article  CAS  Google Scholar 

  39. Xie, C. et al. Ezetimibe blocks the internalization of NPC1L1 and cholesterol in mouse small intestine. J. Lipid Res. 53, 2092–2101 (2012).

    Article  CAS  Google Scholar 

  40. Chang, C. C. et al. Recombinant acyl-CoA:cholesterol acyltransferase-1 (ACAT-1) purified to essential homogeneity utilizes cholesterol in mixed micelles or in vesicles in a highly cooperative manner. J. Biol. Chem. 273, 35132–35141 (1998).

    Article  CAS  Google Scholar 

  41. Klingmuller, U. et al. Primary mouse hepatocytes for systems biology approaches: a standardized in vitro system for modelling of signal transduction pathways. Syst. Biol. (Stevenage) 153, 433–447 (2006).

    Article  CAS  Google Scholar 

  42. Gupte, R. S. et al. Activation of glucose-6-phosphate dehydrogenase promotes acute hypoxic pulmonary artery contraction. J. Biol. Chem. 285, 19561–19571 (2010).

    Article  CAS  Google Scholar 

  43. Klomsiri, C. et al. Use of dimedone-based chemical probes for sulfenic acid detection: evaluation of conditions affecting probe incorporation into redox-sensitive proteins. Methods Enzymol. 473, 77–94 (2010).

    Article  CAS  Google Scholar 

  44. Nelson, K. J. et al. Use of dimedone-based chemical probes for sulfenic acid detection; methods to visualize and identify labeled proteins. Methods Enzymol. 473, 95–115 (2010).

    Article  CAS  Google Scholar 

  45. Liu, T. F. et al. Ablation of gp78 in liver improves hyperlipidemia and insulin resistance by inhibiting SREBP to decrease lipid biosynthesis. Cell Metab. 16, 213–225 (2012).

    Article  CAS  Google Scholar 

  46. Kuksis, A., Suomela, J. P., Tarvainen, M. & Kallio, H. Lipidomic analysis of glycerolipid and cholesteryl ester autooxidation products. Mol. Biotechnol. 42, 224–268 (2009).

    Article  CAS  Google Scholar 

  47. Hutchins, P. M., Moore, E. E. & Murphy, R. C. Electrospray MS/MS reveals extensive and nonspecific oxidation of cholesterol esters in human peripheral vascular lesions. J. Lipid Res. 52, 2070–2083 (2011).

    Article  CAS  Google Scholar 

  48. Zong, G. et al. Associations of erythrocyte palmitoleic acid with adipokines, inflammatory markers, and the metabolic syndrome in middle-aged and older Chinese. Am. J. Clin. Nutr. 96, 970–976 (2012).

    Article  CAS  Google Scholar 

  49. Hu, Y. et al. Genome-wide meta-analyses identify novel loci associated with n-3 and n-6 polyunsaturated fatty acid levels in Chinese and European-ancestry populations. Hum. Mol. Genet. 25, 1215–1224 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank W. Qi for helpful discussion and critical reading of the manuscript, and X.-Y. Yang, H.-H. Miao, L. Qian, Y.-X. Qu, J. Xu and J. Qin for technical assistance. This work was supported by the grants from NNSF of China (31690102, 31430044, 31470802, 31230020 and 31271377), MOST of China (2016YFA0500100 and 2014DFG32410), the 111 Project of Ministry of Education of China (B16036) and the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD). T.-Y.C. and C.C.Y.C. are funded by NIH grant AG 037609.

Author information

Authors and Affiliations

Authors

Contributions

B.-L.L. and B.-L.S. conceived the project. Y.-J.W., C.C.Y.C., T.-Y.C., B.-L.L. and B.-L.S. designed the experiments. Y.-J.W., Y.B., M.L., Y.X. and Q.L. performed the experiments. S.-Y.G. and H.-Y.Y. measured the FA compositions. X.L. performed SNP analysis. Y.-J.W., J.L., C.C.Y.C., T.-Y.C., B.-L.L. and B.-L.S. organized and analysed the data. Y.-J.W., J.L., B.-L.L. and B.-L.S. wrote the manuscript with input from all other authors. All authors approved the final version of the manuscript.

Corresponding authors

Correspondence to Bo-Liang Li or Bao-Liang Song.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 ACAT2 is stabilized by sterols and fatty acids.

(a) Structures of different sterols. Red denotes sterols that elevated ACAT2 protein level. (b) CHO/ACAT2-Myc cells were depleted of lipids and treated with different sterols at indicated concentration. 16 hrs later, cells were harvested for western blotting. (c,d) Different cell lines (HepG2, Huh7, Hepa1-6 and Caco2) were depleted of lipids and treated with OA (200 μM) or PA (200 μM). 16 hrs later, cells were harvested for western blotting and RT-qPCR (mean ± s.d., n = 3 independent experiments). The immunoblots are representative of 3 independent experiments. Uncropped blots are shown in Supplementary Fig. 8. Statistics source data for d can be found in Supplementary Table 2.

Supplementary Figure 2 Mapping the ubiquitination site of ACAT2.

(a) Schematic representation of human ACAT2 (WT) and its various truncations. Red box denotes the region (aa 252 and aa 279) essential for sterol-regulated ACAT2 stabilization. (be) CHO cells transiently transfected with plasmids expressing ACAT2 variants were depleted of sterols and then treated with vehicle or 25-HC (3 μg ml−1) for 16 hrs and harvested for western blotting. (f) Schematic representation of human ACAT2 showing all Cs in a broader region (aa 227–328) (WT) and alanine substitution for single or all Cs. (g,h) CHO cells transiently transfected with plasmids expressing Myc-tagged ACAT2 (WT) or Cys mutants were depleted of sterols and then treated with vehicle or 25-HC for 16 hrs and harvested for western blotting. (i) The alignment of mammalian ACAT1 and ACAT2 partial sequences. The asterisk indicates the ubiquitination site. The highly conserved Cys of ACAT2 is marked in red. The immunoblots are representative of 3 independent experiments. Uncropped blots are shown in Supplementary Fig. 8.

Supplementary Figure 3 ACAT2 is ubiquitinated by K48-specific ubiquitin linkages.

(a) 200 ng of M1-linked (M1), K6-linked (K6), K11-linked (K11), K27-linked (K27), K29-linked (K29), K33-linked (K33), K48-linked (K48) or K63-linked (K63) ubiquitin dimers were separated by SDS-PAGE and probed with different anti-ubiquitin antibodies as indicated. The coommassie brilliant blue-stained gel is shown as a loading control. This is an independent repeat of the experiments in Fig. 3c. (b) CHO cells were transfected with pCMV-HA-Ub together with the plasmid expressing Myc-tagged ACAT2 (WT), (K-null) or (C277A) and depleted of lipids for 16 hrs. Cells were treated with or without 25-HC for 11 hrs. Then MG132 was added into medium for another 5 hrs. Cells were harvested and lysed. The ACAT2 proteins were immunoprecipitated by anti-Myc antibodies coupled agarose and eluted with Myc peptides. Western blotting was carried out using different ubiquitin linkage antibodies or anti-Myc antibody as indicated. The di-ubiquitin was loaded about 30 min later than ACAT2 samples and served as controls. This is an independent repeat of the experiments in Fig. 3d. Uncropped blots are shown in Supplementary Fig. 8.

Supplementary Figure 4 The critical concentration and time course of different lipids to induce ROS production and elevate ACAT2 protein level.

(ai) CHO/ACAT2-Myc cells were depleted of lipids and treated with cholesterol (ac), 25-HC (df) or PA (gi) at indicated concentration. 16 hrs later, cells were harvested for western blotting (a,d,g) and ROS measurement (b,e,h). The correlation between these two parameters was analyzed by GraphPad Prism and demonstrated by scatterdiagrams (c,f,i). (jr) CHO/ACAT2-Myc cells were depleted of lipids and treated with cholesterol (10 μg ml−1, jl), 25-HC (5 μg ml−1, mo) or PA (200 μM, pr) for indicated time duration. 16 hrs later, cells were harvested for western blotting analysis (j,m,p) and ROS measurement (k,n,q). The correlation between these two parameters was demonstrated by scatterdiagrams (l,o,r). The immunoblots are representative of 3 independent experiments. Blots were quantified and the mean intensity of ACAT2 normalized to beta-actin relative to control treated cells is indicated. Uncropped blots are shown in Supplementary Fig. 8. Data are presented as mean ± s.d., n = 3 independent experiments. Statistics source data for ar can be found in Supplementary Table 2.

Supplementary Figure 5 Analysis of the ubiquitination, degradation and protein binding of ACAT2.

(a) CHO/ACAT2-Myc cells were depleted of lipids, treated with H2O2 for indicated time durations and harvested for western blotting. (b) CHO/ACAT2-Myc cells were depleted of lipids for 27 hrs. Cells were then treated with H2O2 for indicated times in the presence of 10 μM MG132 and harvested for ubiquitination assay. (c) CHO cells were transfected with plasmids expressing Myc-tagged ACAT2 and HA-tagged gp78 WT or its variants including C356S-RING finger domain mutation (RINGmut), or M467F, F468S, P469S-CUE domain mutation (CUEmut) as indicated. 48 hrs later, cells were harvested for western blotting. (d) CHO cells were transfected with plasmids expressing Myc-tagged ACAT2 (WT) or (C277A) and T7-tagged Insig1 and depleted of lipids for 27 hrs. Cells were then treated with MG132 for another 5 hrs. Cells were harvested and lysed in the non-denaturing IP buffer, and subsequently subjected to co-IP analysis. (e) CHO cells transfected with plasmids expressing Myc-tagged ACAT2, HA tagged-gp78 and T7-tagged-Insig1 were depleted of lipids and treated with 25-HC/PA or menadione as indicated. 11 hrs later, cells were treated with MG132. After incubation for another 5 hrs at 37 °C, cells were harvested, lysed in the absence (-) or presence (+) of 20 mM DTT, and subsequently subjected to co-IP analysis. (f) CHO cells transfected with plasmid expressing Myc-tagged ACAT2 (WT) or (C-null) were depleted of lipids and treated with vehicle or 25-HC/PA. Cells were harvested 16 hrs later for western blotting. (g) CHO cells transfected with plasmids expressing Myc-tagged ACAT2 (WT) or (C-null) in combination with HA tagged-gp78 and T7-tagged-Insig1 were depleted of lipids and treated with vehicle or menadione as indicated. 11 hrs later, cells were treated with MG132. After incubation for another 5 hrs, cells were harvested for co-IP analysis. The immunoblots are representative of 3 independent experiments. Uncropped blots are shown in Supplementary Fig. 8.

Supplementary Figure 6 Schematic representation for the pathway of lipid-induced ACAT2 stabilization via ROS-mediated C277 oxidation.

(a) CHO cells transfected with plasmid expressing ACAT2 or gp78 were depleted of lipids and treated with 25-HC/PA. Cells were harvested 8 hrs later for sulfenic acids detection as described in Methods. (b) CHO-7 (WT) and gp78−/− deficient CHO-7 cell line (gp78 KO) cells were transfected with indicated plasmids. Cells were depleted of lipids and treated with 25-HC/PA or menadione. 11 hrs later, cells were treated with MG132. After incubation for another 5 hrs, cells were harvested, lysed in the non-denaturing buffer, and subsequently subjected to co-IP analysis. (ce) Male ACAT2−/− mice (8–12 weeks old) were randomly grouped (n = 5 mice) and intravenously injected with AAV2/8-ACAT2 (WT) or (C277A) at a dose of 1 × 1012 vg per mouse. Mice were then allowed ad libitum access to water and chow diet or HFD for 12 weeks. (c) The ACAT2 expression in various tissues of mice receiving AAV2/8-ACAT2 injection. (d) The relative mRNA level of ACAT2 in the livers of mice receiving AAV2/8-ACAT2 (WT)- or (C277A)-injections fed on chow diet and HFD normalized by endogenous ACAT2 mRNA level that was defined as 1. (e) Fast performance liquid chromatography (FPLC) analysis of CEs concentrations from the pooled serum of AAV2/8-ACAT2 (WT)- or (C277A)-injected mice fed on HFD. (f) In the presence of low levels of sterols and/or fatty acids, ACAT2 is ubiquitinated at C277 by the gp78/Insig E3 complex and targeted for proteasomal degradation. Lipid overloading, however, triggers ROS generation that further oxidizes C277 and prevents ACAT2 from ubiquitin-mediated enzyme degradation. The stabilized ACAT2 then converts unesterified cholesterol and fatty acids into cholesteryl esters and ameliorates insulin-resistance induced by lipotoxicity. The immunoblots are representative of 3 independent experiments. Uncropped blots are shown in Supplementary Fig. 8. Data are presented as mean ± s.d. Statistics source data for b,c can be found in Supplementary Table 2.

Supplementary Figure 7 Hepatic fatty acid composition in the CEs of WT mice fed on chow diet or HFD.

Male ACAT2−/− WT littermates (8–12 weeks) were randomly grouped and allowed ad libitum access to water and chow diet or HFD for 12 weeks. (ac) Fatty acid composition of the chow diet and HFD revealed by LC/MS (mean, n = 2 independent experiments). (df) Hepatic fatty acid composition of the CEs in WT mice fed on chow diet or HFD revealed by LC/MS (mean ± s.d., n = 6 mice) n.s., no significance; p < 0.05, p < 0.01; unpaired two-tailed Student’s t test. Statistics source data for af can be found in Supplementary Table 2.

Supplementary information

Supplementary Information

Supplementary Information (PDF 7103 kb)

Supplementary Table 1

Supplementary Information (XLS 14 kb)

Supplementary Table 2

Supplementary Information (XLS 190 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wang, YJ., Bian, Y., Luo, J. et al. Cholesterol and fatty acids regulate cysteine ubiquitylation of ACAT2 through competitive oxidation. Nat Cell Biol 19, 808–819 (2017). https://doi.org/10.1038/ncb3551

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3551

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