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:

Orosomucoid 2 maintains hepatic lipid homeostasis through suppression of de novo lipogenesis

Abstract

Non-alcoholic fatty liver disease (NAFLD) is caused by imbalance in lipid metabolism. In this study, we show that the hepatokine orosomucoid (ORM) 2 is a key regulator of de novo lipogenesis in the liver. Hepatic and plasma ORM2 levels are markedly decreased in obese murine models and patients with NAFLD. Through multiple loss- and gain-of function studies, we demonstrate that ORM2 is essential to maintain hepatic and systemic lipid homeostasis. At the mechanistic level, ORM2 binds to inositol 1, 4, 5-trisphosphate receptor type 2 to activate AMP-activated protein kinase signaling, thereby inhibiting sterol regulatory element binding protein 1c-mediated lipogenic gene program. Notably, intraperitoneal injections of recombinant ORM2 protein or stabilized ORM2–FC fusion protein markedly improved liver steatosis, steatohepatitis and atherosclerosis in preclinical mouse models, without adverse effects on body weight or food intake. Thus, these findings suggest that ORM2 may serve as a potential target for therapeutic intervention in NAFLD, non-alcoholic steatohepatitis and related lipid 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

Fig. 1: Downregulation of ORM2 in the livers and plasma of mice and patients with NAFLD.
Fig. 2: Both ORM2 KO male and female mice exhibited hepatic TG accumulation under ND conditions.
Fig. 3: ORM2 deficiency exacerbates diet-induced steatosis and steatohepatitis in mice.
Fig. 4: ORM2 overexpression alleviates liver steatosis and hyperlipidemia in obese mice.
Fig. 5: Downregulation of lipogenic genes by ORM2.
Fig. 6: ORM2 activates AMPK signaling to suppress DNL.
Fig. 7: ITPR2 is required for the hepatic benefits of ORM2.
Fig. 8: Recombinant ORM2 protein improves diet-induced liver steatosis and steatohepatitis in mice.

Similar content being viewed by others

Data availability

RNA-seq data have been deposited into the Gene Expression Omnibus database (accession no. GSE186024). Proteomics data have been deposited at the ProteomeXchange Consortium (accession nos. PXD035372 and PXD035373). The data supporting the findings of this study are available in source data files. Source data are provided with this paper.

References

  1. Fabbrini, E., Sullivan, S. & Klein, S. Obesity and nonalcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 51, 679–689 (2010).

    Article  CAS  PubMed  Google Scholar 

  2. Hodson, L. & Gunn, P. J. The regulation of hepatic fatty acid synthesis and partitioning: the effect of nutritional state. Nat. Rev. Endocrinol. 15, 689–700 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Shimano, H. & Sato, R. SREBP-regulated lipid metabolism: convergent physiology-divergent pathophysiology. Nat. Rev. Endocrinol. 13, 710–730 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Xu, X., So, J. S., Park, J. G. & Lee, A. H. Transcriptional control of hepatic lipid metabolism by SREBP and ChREBP. Semin. Liver Dis. 33, 301–311 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Friedman, S. L., Neuschwander-Tetri, B. A., Rinella, M. & Sanyal, A. J. Mechanisms of NAFLD development and therapeutic strategies. Nat. Med. 24, 908–922 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Tilg, H., Adolph, T. E., Dudek, M. & Knolle, P. Non-alcoholic fatty liver disease: the interplay between metabolism, microbes and immunity. Nat. Metab. 3, 1596–1607 (2021).

    Article  CAS  PubMed  Google Scholar 

  7. Smith, G. I. et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J. Clin. Invest. 130, 1453–1460 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Neuschwander-Tetri, B. A. Therapeutic landscape for NAFLD in 2020. Gastroenterology 158, 1984–1998 (2020).

    Article  CAS  PubMed  Google Scholar 

  9. Rinella, M. E. & Sanyal, A. J. Management of NAFLD: a stage-based approach. Nat. Rev. Gastroenterol. Hepatol. 13, 196–205 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Musso, G., Cassader, M. & Gambino, R. Non-alcoholic steatohepatitis: emerging molecular targets and therapeutic strategies. Nat. Rev. Drug Discov. 15, 249–274 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Stefan, N. & Häring, H. U. The role of hepatokines in metabolism. Nat. Rev. Endocrinol. 9, 144–152 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Meex, R. C. R. & Watt, M. J. Hepatokines: linking nonalcoholic fatty liver disease and insulin resistance. Nat. Rev. Endocrinol. 13, 509–520 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Watt, M. J., Miotto, P. M., De Nardo, W. & Montgomery, M. K. The liver as an endocrine organ-linking NAFLD and insulin resistance. Endocr. Rev. 40, 1367–1393 (2019).

    Article  PubMed  Google Scholar 

  14. Inagaki, T. et al. Endocrine regulation of the fasting response by PPARα-mediated induction of fibroblast growth factor 21. Cell Metab. 5, 415–425 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Badman, M. K. et al. Hepatic fibroblast growth factor 21 is regulated by PPARα and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 5, 426–437 (2007).

    Article  CAS  PubMed  Google Scholar 

  16. Lu, Y. et al. Periostin promotes liver steatosis and hypertriglyceridemia through downregulation of PPARα. J. Clin. Invest. 124, 3501–3513 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Wu, J. X. et al. LZP is required for hepatic triacylglycerol transportation through maintaining apolipoprotein B stability. PLoS Genet. 17, e1009357 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Luo, Z., Lei, H., Sun, Y., Liu, X. & Su, D. F. Orosomucoid, an acute response protein with multiple modulating activities. J. Physiol. Biochem. 71, 329–340 (2015).

    Article  CAS  PubMed  Google Scholar 

  19. Schmidt, O. et al. Endosome and Golgi-associated degradation (EGAD) of membrane proteins regulates sphingolipid metabolism. EMBO J. 38, e101433 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Carter, K. C., Post, D. J. & Papaconstantinou, J. Differential expression of the mouse α 1-acid glycoprotein genes (AGP-1 and AGP-2) during inflammation and aging. Biochim. Biophys. Acta 1089, 197–205 (1991).

    Article  CAS  PubMed  Google Scholar 

  21. Jo, M. et al. Astrocytic Orosomucoid-2 modulates microglial activation and neuroinflammation. J. Neurosci. 37, 2878–2894 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wan, J. J. et al. Role of acute-phase protein ORM in a mice model of ischemic stroke. J. Cell. Physiol. 234, 20533–20545 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. Sun, Y. et al. The acute-phase protein orosomucoid regulates food intake and energy homeostasis via leptin receptor signaling pathway. Diabetes 65, 1630–1641 (2016).

    Article  CAS  PubMed  Google Scholar 

  24. Lee, Y. S. et al. Adipocytokine orosomucoid integrates inflammatory and metabolic signals to preserve energy homeostasis by resolving immoderate inflammation. J. Biol. Chem. 285, 22174–22185 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xiong, X. et al. Landscape of intercellular crosstalk in healthy and NASH liver revealed by single-cell secretome gene analysis. Mol. Cell. 75, 644–660 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhou, B. et al. N6 -methyladenosine reader protein YT521-B homology domain-containing 2 suppresses liver steatosis by regulation of mRNA stability of lipogenic genes. Hepatology 73, 91–103 (2021).

    Article  CAS  PubMed  Google Scholar 

  27. Gosis, B. S. et al. Inhibition of nonalcoholic fatty liver disease in mice by selective inhibition of mTORC1. Science 376, eabf8271 (2022).

    Article  CAS  PubMed  Google Scholar 

  28. Liu, B. et al. Sparcl1 promotes nonalcoholic steatohepatitis progression in mice through upregulation of CCL2. J. Clin. Invest. 131, e144801 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Yang, J., Craddock, L., Hong, S. & Liu, Z. M. AMP-activated protein kinase suppresses LXR-dependent sterol regulatory element-binding protein-1c transcription in rat hepatoma McA-RH7777 cells. J. Cell. Biochem. 106, 414–426 (2009).

    Article  CAS  PubMed  Google Scholar 

  30. Li, Y. et al. AMPK phosphorylates and inhibits SREBP activity to attenuate hepatic steatosis and atherosclerosis in diet-induced insulin-resistant mice. Cell Metab. 13, 376–388 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Huang, H. et al. Rho-kinase/AMPK axis regulates hepatic lipogenesis during overnutrition. J. Clin. Invest. 128, 5335–5350 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Lizcano, J. M. et al. LKB1 is a master kinase that activates 13 kinases of the AMPK subfamily, including MARK/PAR-1. EMBO J. 23, 833–843 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Woods, A. et al. LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr. Biol. 13, 2004–2008 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Hawley, S. A. et al. Calmodulin-dependent protein kinase kinase-β is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2, 9–19 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Woods, A. et al. Ca2+/calmodulin-dependent protein kinase kinase-β acts upstream of AMP-activated protein kinase in mammalian cells. Cell Metab. 2, 21–33 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Boudaba, N. et al. AMPK re-activation suppresses hepatic steatosis but its downregulation does not promote fatty liver development. EBioMedicine 28, 194–209 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Smith, B. K. et al. Treatment of nonalcoholic fatty liver disease: role of AMPK. Am. J. Physiol. Endocrinol. Metab. 311, E730–E740 (2016).

    Article  PubMed  Google Scholar 

  38. Fullerton, M. D. et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat. Med. 19, 1649–1654 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lally, J. S. V. et al. Inhibition of acetyl-CoA carboxylase by phosphorylation or the inhibitor ND-654 suppresses lipogenesis and hepatocellular carcinoma. Cell Metab. 29, 174–182 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Taylor, C. W., da Fonseca, P. C. & Morris, E. P. IP(3) receptors: the search for structure. Trends Biochem. Sci. 29, 210–219 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Kerkhofs, M. et al. Pathophysiological consequences of isoform-specific IP3 receptor mutations. Biochim. Biophys. Acta Mol. Cell. Res. 1865, 1707–1717 (2018).

    Article  CAS  PubMed  Google Scholar 

  42. Hirata, K., Pusl, T., O’Neill, A. F., Dranoff, J. A. & Nathanson, M. H. The type II inositol 1,4,5-trisphosphate receptor can trigger Ca2+ waves in rat hepatocytes. Gastroenterology 122, 1088–1100 (2002).

    Article  CAS  PubMed  Google Scholar 

  43. Guerra, M. T. et al. Expression of the type 3 InsP3 receptor is a final common event in the development of hepatocellular carcinoma. Gut 68, 1676–1687 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Wu, T. et al. Feeding-induced hepatokine, Manf, ameliorates diet-induced obesity by promoting adipose browning via p38 MAPK pathway. J. Exp. Med. 218, e20201203 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Montgomery, M. K. et al. SMOC1 is a glucose-responsive hepatokine and therapeutic target for glycemic control. Sci. Transl. Med. 12, eaaz8048 (2020).

    Article  CAS  PubMed  Google Scholar 

  46. Shaw, R. J. et al. The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science 310, 1642–1646 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Racioppi, L. & Means, A. R. Calcium/calmodulin-dependent protein kinase kinase 2: roles in signaling and pathophysiology. J. Biol. Chem. 287, 31658–31665 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Iwabu, M. et al. Adiponectin and AdipoR1 regulate PGC-1α and mitochondria by Ca(2+) and AMPK/SIRT1. Nature 464, 1313–1319 (2010).

    Article  CAS  PubMed  Google Scholar 

  49. Ying, L. et al. Paracrine FGFs target skeletal muscle to exert potent anti-hyperglycemic effects. Nat. Commun. 12, 7256 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Song, L. et al. FGF4 protects the liver from nonalcoholic fatty liver disease by activating the AMP-activated protein kinase-Caspase 6 signal axis. Hepatology https://doi.org/10.1002/hep.32404 (2022).

  51. Marcelo, K. L., Means, A. R. & York, B. The Ca(2+)/calmodulin/CaMKK2 axis: nature’s metabolic CaMshaft. Trends Endocrinol. Metab. 27, 706–718 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Anderson, K. A. et al. Hypothalamic CaMKK2 contributes to the regulation of energy balance. Cell Metab. 7, 377–388 (2008).

    Article  CAS  PubMed  Google Scholar 

  53. Lin, F., Ribar, T. J. & Means, A. R. The Ca2+/calmodulin-dependent protein kinase kinase, CaMKK2, inhibits preadipocyte differentiation. Endocrinology 152, 3668–3679 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Anderson, K. A. et al. Deletion of CaMKK2 from the liver lowers blood glucose and improves whole-body glucose tolerance in the mouse. Mol. Endocrinol. 26, 281–291 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Stork, B. A. et al. Calcium/calmodulin-dependent protein kinase kinase 2 regulates hepatic fuel metabolism. Mol. Metab. 62, 101513 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Sun, Z. & Lazar, M. A. Dissociating fatty liver and diabetes. Trends Endocrinol. Metab. 24, 4–12 (2013).

    Article  CAS  PubMed  Google Scholar 

  57. Vazquez, G., Wedel, B. J., Bird, G. S., Joseph, S. K. & Putney, J. W. An inositol 1,4,5-trisphosphate receptor-dependent cation entry pathway in DT40 B lymphocytes. EMBO J. 21, 4531–4538 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Feriod, C. N. et al. Inositol 1,4,5-trisphosphate receptor type II (InsP3R-II) is reduced in obese mice, but metabolic homeostasis is preserved in mice lacking InsP3R-II. Am. J. Physiol. Endocrinol. Metab. 307, E1057–E1064 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Khamphaya, T. et al. Nonalcoholic fatty liver disease impairs expression of the type II inositol 1,4,5-trisphosphate receptor. Hepatology 67, 560–574 (2018).

    Article  CAS  PubMed  Google Scholar 

  60. Ziegler, D. V. et al. Calcium channel ITPR2 and mitochondria-ER contacts promote cellular senescence and aging. Nat. Commun. 12, 720 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Gimeno, R. E. & Moller, D. E. FGF21-based pharmacotherapy–potential utility for metabolic disorders. Trends Endocrinol. Metab. 25, 303–311 (2014).

    Article  CAS  PubMed  Google Scholar 

  62. Cui, A. et al. The effects of B1344, a novel fibroblast growth factor 21 analog, on nonalcoholic steatohepatitis in nonhuman primates. Diabetes 69, 1611–1623 (2020).

    Article  PubMed  Google Scholar 

  63. Sanyal, A. et al. Pegbelfermin (BMS-986036), a PEGylated fibroblast growth factor 21 analogue, in patients with non-alcoholic steatohepatitis: a randomised, double-blind, placebo-controlled, phase 2a trial. Lancet 392, 2705–2717 (2019).

    Article  PubMed  Google Scholar 

  64. Sun, S. M. et al. AMPK activator C24 inhibits hepatic lipogenesis and ameliorates dyslipidemia in HFHC diet-induced animal models. Acta Pharmacol. Sin. 42, 585–592 (2021).

    Article  CAS  PubMed  Google Scholar 

  65. Zhang, C. S. et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature 548, 112–116 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank J. Li (Shanghai Institute of Materia Medica) for the gift of liver-specific Prkaa1 and Prkaa2 DKO mice and constitutively activated AMPK plasmids, C.-S. Zhang (Xiamen University) for the gift of CaMKK2 KO mice, C. Xie and X. Guo (Shanghai Institute of Materia Medica) for the measurement of lipogenesis rate, G. Shi (Sun Yat-sen University), C. Ruan (Fudan University), Y. Li (Shanghai Jiao Tong University School of Medicine), D. Li (East China Normal University) and J. Wang (Shanghai Jiao Tong University School of Medicine) for helpful suggestions and G. Yan (Shanghai Jiao Tong University School of Medicine) for technical support. This study was supported by grants from the National Key Research and Development Program of China (2018YFA0800402 to Y. Lu), the Shanghai Outstanding Academic Leaders Projects, the Basic Research of Science and Technology Innovation Action Plan and Shanghai Sailing Program by Shanghai Municipal Science and Technology Committee (21XD1423400 and 21JC1401300 to Y. Lu and 22YF1432800 to B.Z.), China Postdoctoral Science Foundation Funded Project (2021M702183 to B.Z.) and Youth Cultivation Project of Shanghai Jiao Tong University Affiliated Sixth People’s Hospital (ynqn202107 to B.Z.).

Author information

Authors and Affiliations

Authors

Contributions

Y. Lu and B.Z. designed and directed the study. B.Z., Y. Luo and N.J. performed all animal and cellular experiments and human sample analysis. C.H. contributed to the discussion. Y. Lu and Z.B. wrote the manuscript.

Corresponding author

Correspondence to Yan Lu.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Kei Sakamoto and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Editor recognition statement Primary Handling Editor: Alfredo Giménez-Cassina, in collaboration with the Nature Metabolism team.

Additional information

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

Extended data

Extended Data Fig. 1 ORM2 deficiency exacerbates TG accumulation.

Related to Figs. 2 and 3. a-i, 8-week-old ORM2 wild-type (WT), heterozygous (HE) and knockout (KO) male mice were fed a high-fat-diet for 8 weeks. (a-c) Plasma LDL-cholesterol (a), ALT (b), AST (c) levels. WT: n = 4 biologically independent mice. HE and KO: n = 6 biologically independent mice per condition. d, Relative mRNA levels of genes related to hepatic inflammation in the liver. n = 4 biologically independent mice per condition. (e-h) Body weight (e), plasma leptin levels (f), iWAT weight (g) and BAT weight (h) of three groups of mice. WT: n = 4 biologically independent mice. HE and KO: n = 6 biologically independent mice per condition. i, H&E staining of WAT and BAT. Scale bar, 50 μm. Representative result from three biologically independent mice. j-n, 8-week-old ORM2 WT, HE and KO female mice were fed a high-fat-diet for 8 weeks. Body weight (j), liver/body weight ratio (k), hepatic (l) and plasma TG (m) levels. WT and HE: n = 6 biologically independent mice per condition. KO: n = 4 biologically independent mice. n, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. Data are shown as mean ± SEM. (a-h, j-m). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (a-d, f-h, j-m) and two-way ANOVA followed by Dunnett’s multiple comparisons test (e).

Source data

Extended Data Fig. 2 The distinct roles of ORM1 and ORM2 in the improvement of liver steatosis.

Related to Fig. 4. a-h, 8-week-old ob/ob male mice were administered with equal amount of Ad-ORM1, Ad-ORM2, or Ad-GFP through tail vein injection. a, b, Relative mRNA levels (a, n = 8 biologically independent mice per condition) and protein expression (b, n = 4 biologically independent mice per condition) of ORM1 and ORM2 in the livers from three groups of mice. c-i, Body weight (c), average daily food intake for 3 consecutive days (d), liver/body weight ratio (e), hepatic TG levels (f), plasma TG (g) and TC (i) levels. (c-i) n = 8 biologically independent mice per condition. h, Macroscopic appearance of liver, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. Data are shown as mean ± SEM (a, c-g, i). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (a, c-g, i).

Source data

Extended Data Fig. 3 ORM2 overexpression attenuates hepatic steatosis and hyperlipidemia in db/db mice and HFD-induced obese mice.

Related to Fig. 4. a-j, 8-week-old db/db male mice were administered with adenovirus containing ORM2 (Ad-ORM2) or GFP (Ad-GFP) through tail vein injection. a, b, Protein ORM2 levels were determined by western blots using the livers (a, top) and plasma (b, bottom) from two groups of mice. n = 6 biologically independent mice per condition. c, Relative mRNA expression levels of ORM1, ORM2 and ORM3 in the liver. n = 6 biologically independent mice per condition. d-i, Body weight (d), average daily food intake for 3 consecutive days (e), liver/body weight ratio (f), hepatic TG (g), plasma TG (h) and TC (i) levels from two groups of mice. (d-i) Ad-GFP: n = 8 biologically independent mice. Ad-ORM2: n = 9 biologically independent mice. j, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. k-q, C57BL/6 J male mice were fed a normal diet or HFD for 12 weeks and then administered with Ad-GFP or Ad-ORM2 for 12 days. k, Protein levels of ORM2 in the livers from three groups of mice. n = 4 biologically independent mice per condition. Liver/body weight ratio (l), hepatic TG (m), plasma TG (n) and TC (p) levels and body weight (q) from three groups of mice. ND + Ad-GFP: n = 4 biologically independent mice. HFD + Ad-GFP: n = 6 biologically independent mice. HFD + Ad-ORM2: n = 7 biologically independent mice. o, Macroscopic appearance of liver, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. Data are shown as mean ± SEM (c-i, l-n, p, q). P values are determined by unpaired two-tailed Student’s t-test (c-i) and one-way ANOVA followed by Dunnett’s multiple comparisons test (l-n, p, q).

Source data

Extended Data Fig. 4 ORM2 has little effects on the expression levels of genes related to fatty acid esterification and oxidation and lipid transport.

Related to Fig. 5. a, b, Relative mRNA level (a, n = 8 biologically independent mice per condition) and protein expression of lipogenic genes (b, n = 4 biologically independent mice per condition) in ob/ob male mice administered with Ad-ORM1, Ad-ORM2 or Ad-GFP. c-e, Relative mRNA levels of genes related to fatty acid esterification (c), fatty acid oxidation (d) and lipid transport (e) in the livers from db/db male mice administered with Ad-ORM2 or Ad-GFP. n = 6 biologically independent mice per condition. f-h, Relative mRNA levels of genes related to fatty acid esterification (f), fatty acid oxidation (g) and lipid transport (h) in the livers from adenovirus-treated HFD male mice as indicated. n = 4 biologically independent mice per condition. i-k, Relative mRNA levels of genes related to fatty acid esterification (i), fatty acid oxidation (j), and lipid transport (k) in the livers from AAV-treated HFD male mice as indicated. n = 4 biologically independent mice per condition. l-n, Relative mRNA levels of genes related to fatty acid esterification (l), fatty acid oxidation (m), and lipid transport (n) in the livers from ORM2 WT, HE and KO male mice fed a normal diet. WT and HE: n = 6 biologically independent mice per condition. KO: n = 4 biologically independent mice. o-q, Relative mRNA levels of genes related to fatty acid esterification (o), fatty acid oxidation (p), and lipid transport (q) in the livers from ORM2 WT, HE and KO male mice fed a high fat diet for 8 weeks. WT: n = 4 biologically independent mice. HE and KO: n = 6 biologically independent mice per condition. Data are shown as mean ± SEM (a, c-q). P values are determined by unpaired two-tailed Student’s t-test (c-e) and one-way ANOVA followed by Dunnett’s multiple comparisons test (a, f-q).

Source data

Extended Data Fig. 5 ORM2 activates CaMKK2-AMPK signaling pathway.

Related to Fig. 6. a, b, Immunoblots of AMPK signaling pathway in MPHs (a) and HepG2 cells (b). Cells were treated with ORM2 recombinant protein (100 ng/ml) or vehicle control (PBS) for 6 hr. n = 3 independent samples per condition. c, d, MPHs were pre-incubated with palmitic acid (200 μM) for 12 hr, and then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr, in the presence or absence of Compound C (20 μM). c, Cellular TG contents. n = 3 independent samples per condition. d, Relative mRNA levels of genes related to de novo lipogenesis. n = 3 independent samples per condition. e, f, HepG2 Cells were pre-incubated with palmitic acid (200 μM) for 12 hr, and then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr, in the presence or absence of Compound C (20 μM). e, Cellular TG contents. n = 3 independent samples per condition. f, Relative mRNA levels of genes related to de novo lipogenesis. n = 3 independent samples per condition. g-i, MPHs from ORM2 WT or KO mice were administered with Ad-GFP, Ad-CA-AMPK α1 (T172D TC312) or Ad-CA-AMPK α2 (T172D TC312). g, Immunoblots of AMPK signaling. h, Cellular TG contents. n = 3 independent samples per condition. i, Relative mRNA levels of genes related to de novo lipogenesis. n = 3 independent samples per condition. j, k, MPHs were pre-incubated with palmitic acid (200 μM) for 12 hr, and then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr, in the presence or absence of CaMKK2 inhibitor (STO-609, 5 μM) or LKB1 inhibitor (PIM1, 10 μM). j, Cellular TG contents in MPHs. n = 3 independent samples per condition. k, Relative mRNA levels of genes related to lipogenesis in MPHs. n = 3 independent samples per condition. l, Immunoblots of CaMKK2 and AMPK signaling in MPHs. Cells were transfected with adenoviral shRNA targeting CaMKK2 or negative control for 24 hr, then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr. m, Immunoblots of LKB1 and AMPK signaling in MPHs. Cells were transfected with adenoviral shRNA targeting LKB1 or negative control for 24 hr, then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr. n, Immunoblots of AMPK in the MPHs from ORM2 WT and KO mice. Cells were treated with ORM2 recombinant protein, Ca2+ ionophore (Ionomycin, 0.5 μM) or vehicle control (DMSO) for 3 hr. o, Genomic sequencing of WT or ACC1 S80A knockin mutation (KI) HepG2 cells. p, Immunoblots of AMPK signaling in WT and KI cells treated with metformin (2 mM) or vehicle control (PBS) for 6 hr. q, r, Cells were incubated for 6 hr with 13C-acetate (1 mM) containing vehicle control (PBS), ORM2 (100 ng/ml), AMPK allosteric activators (A-769662, 10μM and AICAR, 100μM). The rate of de novo lipogenesis in WT cells (q, n = 4 independent samples per condition) and KI cells (r, n = 3 independent samples per condition) were measured. s, t, Relative mRNA levels of genes related to de novo lipogenesis in WT (s) and KI cells (t) were analyzed. n = 3 independent samples per condition. u, v, Mouse primary hepatocytes were transfected with adenoviral shRNA targeting ACC1 or negative control. Cells were then treated with ORM2 (100 ng/ml) or vehicle control (PBS) for 6 hr. ACC1 protein expression (u, n = 3 independent samples per condition) and relative mRNA levels of genes related to de novo lipogenesis (v, n = 3 independent samples per condition) were measured. Data are shown as mean ± SEM (c-f, h-k, q-t, v). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (q-t) and Bonferroni’s multiple comparisons test (c-f, h-k, v).

Source data

Extended Data Fig. 6 ITPR2 is required for the anti-lipogenic role of ORM2 in hepatocytes.

Related to Fig. 7. a, b, Immunoblots of AMPK signaling in MPHs. a, MPHs were transfected with adenoviral shRNA targeting ITPR2 or negative control for 24 hr, and then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr. b, MPHs were transfected with adenoviral shRNA targeting ITPR2 or negative control for 24 hr, then treated with Ca2+ ionophore (Ionomycin, 0.5 μM) or vehicle control (DMSO) for 3 hr. c, Cellular TG contents in MPHs. n = 3 independent samples per condition. d, Relative mRNA levels of genes related to de novo lipogenesis in MPHs. n = 3 independent samples per condition. e, f, Cells were exposed to palmitic acid (200 μM) for 12 hr, and then treated with ORM2 protein (100 ng/ml) or vehicle control (PBS) for 6 hr, in the presence or absence of an ITPR2 inhibitor (2-APB, 50 μM). e, Cellular TG contents. n = 3 independent samples per condition. f, Relative mRNA levels of genes related to de novo lipogenesis. n = 3 independent samples per condition. Data are shown as mean ± SEM (c-f). P values are determined by one-way ANOVA followed by Bonferroni’s multiple comparisons test (c-f).

Source data

Extended Data Fig. 7 ORM2 recombinant protein treatment improves liver steatosis in ob/ob mice.

Related to Fig. 8. a-j, ob/ob male mice were intraperitoneally injected with recombinant ORM2 protein (0.1 mg/kg, 1.0 mg/kg) or vehicle control (PBS, 0) for 10 days. a-d, Body weight (a), average daily food intake for 3 consecutive days (b), iWAT (c) and BAT weight (d). (a-d) n = 8 biologically independent mice per condition. e, H&E staining of WAT and BAT from three groups of mice. Scale bar, 50 μm. Representative result from three biologically independent mice. f-i, Liver TC (f), plasma TC (g), ALT (h) and AST (i) levels from three groups of mice. n = 8 biologically independent mice per condition. j, Relative mRNA levels of genes related to hepatic inflammation in the livers from three groups of mice. n = 8 biologically independent mice per condition. Data are shown as mean ± SEM. (a-d, f-j). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (a-d, f-j).

Source data

Extended Data Fig. 8 ORM2 recombinant protein treatment improves liver steatosis in db/db mice.

Related to Fig. 8. a-j, db/db male mice were intraperitoneally injected with recombinant ORM2 protein (0.1 mg/kg, 1.0 mg/kg) or vehicle control (PBS, 0) for 10 days. a-f, Body weight (a), average daily food intake for 3 consecutive days (b), liver/body weight ratio (c), hepatic TG levels (d), plasma TG (e) and TC levels (f) from three groups of db/db mice. (a-f) n = 8 biologically independent mice per condition. g, Macroscopic appearance of liver, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. h, Relative mRNA levels of genes related to de novo lipogenesis in the livers. n = 8 biologically independent mice per condition. i, Protein levels of lipogenic genes in the livers. n = 4 biologically independent mice per condition. j. Immunoblots of AMPK and mTOR signaling pathways in the livers. n = 4 biologically independent mice per condition. Data are shown as mean ± SEM (a-f, h). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (a-f, h).

Source data

Extended Data Fig. 9 ORM2 recombinant protein treatment improves liver steatosis in HFD-induced obese mice.

Related to Fig. 8. a-i, C57BL/6 J male mice were fed a normal diet or high-fat-diet for 12 weeks, and then treated with ORM2 protein (1.0 mg/kg) or vehicle control (PBS) for 14 days. a-f, Body weight (a), average daily food intake for 3 consecutive days (b), liver/body weight ratio (c), hepatic TG (d), plasma TG (e) and leptin (f) levels from three groups of mice. (a-f) ND + PBS and HFD + PBS: n = 6 biologically independent mice per condition. (a-f) HFD + ORM2: n = 5 biologically independent mice. g, Macroscopic appearance of liver, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. h, Relative mRNA levels of genes related to de novo lipogenesis in the livers. n = 4 biologically independent mice per condition. i, Protein levels of lipogenic genes in the livers. n = 4 biologically independent mice per condition. Data are shown as mean ± SEM (a-f, h). P values are determined by one-way ANOVA followed by Dunnett’s multiple comparisons test (a-f, h).

Source data

Extended Data Fig. 10 ORM2 protein protects against western diet-induced atherosclerotic lesion formation in ApoE null mice.

Related to Fig. 8. a-h, 10-week-old ApoE null male mice were fed a western diet for 8 weeks, and then were intraperitoneally injected with recombinant ORM2 protein (1.0 mg/kg) or vehicle control (PBS) for 14 days. a, b, Liver/body weight ratio (a) and liver TG levels (b). PBS: n = 7 biologically independent mice. OMR2: n = 6 biologically independent mice. c, H&E and Oil Red O staining of liver sections. Scale bar, 50 μm. Representative result from three biologically independent mice. d, Relative mRNA levels of genes related to hepatic TG metabolism in the livers. n = 6 biologically independent mice per condition. e, f, Plasma (e) and hepatic TC (f) levels. PBS: n = 7 biologically independent mice. OMR2: n = 6 biologically independent mice. g, Relative mRNA levels of genes related to hepatic cholesterol metabolism in the livers. n = 6 biologically independent mice per condition. h, Oil Red O staining showing the lipid accumulation in the aorta from two groups of mice. i, Relative mRNA levels of genes related to inflammation in the aorta. n = 6 biologically independent mice per condition. Data are shown as mean ± SEM (a, b, d-g, i). P values are determined by one-way ANOVA followed by unpaired two-tailed Student’s t-test (a, b, d-g, i).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–4, Supplementary Tables 1–6 and unprocessed western blots for Supplementary Figs. 1, 3 and 4.

Reporting Summary

Supplementary Data 1

Statistical Source Data.

Supplementary Data 2

Statistical Source Data.

Supplementary Data 3

Statistical Source Data.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 8

Unprocessed western blots.

Source Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 1

Statistical Source Data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical Source Data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical Source Data.

Source Data Extended Data Fig. 10

Statistical Source Data.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, B., Luo, Y., Ji, N. et al. Orosomucoid 2 maintains hepatic lipid homeostasis through suppression of de novo lipogenesis. Nat Metab 4, 1185–1201 (2022). https://doi.org/10.1038/s42255-022-00627-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00627-4

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