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:

Hepatocyte-derived exosomes from early onset obese mice promote insulin sensitivity through miR-3075

Abstract

In chronic obesity, hepatocytes become insulin resistant and exert important effects on systemic metabolism. Here we show that in early onset obesity (4 weeks high-fat diet), hepatocytes secrete exosomes that enhance insulin sensitivity both in vitro and in vivo. These beneficial effects were due to exosomal microRNA miR-3075, which is enriched in these hepatocyte exosomes. FA2H is a direct target of miR-3075 and small interfering RNA depletion of FA2H in adipocytes, myocytes and primary hepatocytes leads to increased insulin sensitivity. In chronic obesity (16–18 weeks of a high-fat diet), hepatocyte exosomes promote a state of insulin resistance. These chronic obese hepatocyte exosomes do not directly cause impaired insulin signalling in vitro but do promote proinflammatory activation of macrophages. Taken together, these studies show that in early onset obesity, hepatocytes produce exosomes that express high levels of the insulin-sensitizing miR-3075. In chronic obesity, this compensatory effect is lost and hepatocyte-derived exosomes from chronic obese mice promote insulin resistance.

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: Hepatocyte-derived exosomes from early stage obese mice enhance insulin sensitivity.
Fig. 2: miRNAs are the key cargo causing the hepatocyte exos effects.
Fig. 3: Effects of hepatocyte-specific Rab27KO on exos secretion and miRNA content.
Fig. 4: miR-3075 is an insulin sensitizer within 4wk HFD Exos.
Fig. 5: Fa2h is a miR-3075 target gene that regulates cellular insulin responses.
Fig. 6: Chronic obesity induces hepatocytes to secrete pathogenic exosomal miRNAs that cause tissue inflammation and insulin resistance.
Fig. 7: Hepatocyte-derived exos from chronic obese mice stimulate the proinflammatory M1-like state in macrophages.

Similar content being viewed by others

Data availability

The datasets generated in current study are available from the corresponding authors upon reasonable request. There are no restrictions on data availability. All mouse lines in this study are available from the Jackson Laboratory. The miRNA-seq data have been deposited in the NCBI Gene Expression Omnibus under accession number GSE164310Source data are provided with this paper.

References

  1. Kahn, S. E., Hull, R. L. & Utzschneider, K. M. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature 444, 840–846 (2006).

    Article  CAS  Google Scholar 

  2. Lee, Y. S., Wollam, J. & Olefsky, J. M. An integrated view of immunometabolism. Cell 172, 22–40 (2018).

    Article  CAS  Google Scholar 

  3. Roden, M. & Shulman, G. I. The integrative biology of type 2 diabetes. Nature 576, 51–60 (2019).

    Article  CAS  Google Scholar 

  4. Romeo, G. R., Lee, J. & Shoelson, S. E. Metabolic syndrome, insulin resistance, and roles of inflammation—mechanisms and therapeutic targets. Arter. Thromb. Vasc. Biol. 32, 1771–1776 (2012).

    Article  CAS  Google Scholar 

  5. Mathieu, M., Martin-Jaular, L., Lavieu, G. & Thery, C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat. Cell Biol. 21, 9–17 (2019).

    Article  CAS  Google Scholar 

  6. Mori, M. A., Ludwig, R. G., Garcia-Martin, R., Brandao, B. B. & Kahn, C. R. Extracellular miRNAs: from biomarkers to mediators of physiology and disease. Cell Metab. https://doi.org/10.1016/j.cmet.2019.07.011 (2019).

  7. Kalluri, R. & LeBleu, V. S. The biology, function, and biomedical applications of exosomes. Science https://doi.org/10.1126/science.aau6977 (2020).

  8. Castano, C., Kalko, S., Novials, A. & Parrizas, M. Obesity-associated exosomal miRNAs modulate glucose and lipid metabolism in mice. Proc. Natl Acad. Sci. USA 115, 12158–12163 (2018).

    Article  CAS  Google Scholar 

  9. Deng, Z. B. et al. Adipose tissue exosome-like vesicles mediate activation of macrophage-induced insulin resistance. Diabetes 58, 2498–2505 (2009).

    Article  CAS  Google Scholar 

  10. Ferrante, S. C. et al. Adipocyte-derived exosomal miRNAs: a novel mechanism for obesity-related disease. Pediatr. Res 77, 447–454 (2015).

    Article  CAS  Google Scholar 

  11. Flaherty, S. E. 3rd et al. A lipase-independent pathway of lipid release and immune modulation by adipocytes. Science 363, 989–993 (2019).

    Article  CAS  Google Scholar 

  12. Ying, W. et al. Adipose tissue macrophage-derived exosomal miRNAs can modulate in vivo and in vitro insulin sensitivity. Cell 171, 372–384 (2017).

    Article  CAS  Google Scholar 

  13. Ying, W. et al. MiR-690, an exosomal-derived miRNA from M2-polarized macrophages, improves insulin sensitivity in obese mice. Cell Metab. https://doi.org/10.1016/j.cmet.2020.12.019 (2021).

  14. Crewe, C. et al. An endothelial-to-adipocyte extracellular vesicle axis governed by metabolic state. Cell 175, 695–708 e613 (2018).

    Article  CAS  Google Scholar 

  15. Thomou, T. et al. Adipose-derived circulating miRNAs regulate gene expression in other tissues. Nature 542, 450–455 (2017).

    Article  CAS  Google Scholar 

  16. Baek, D. et al. The impact of microRNAs on protein output. Nature 455, 64–71 (2008).

    Article  CAS  Google Scholar 

  17. Bartel, D. P. MicroRNAs: target recognition and regulatory functions. Cell 136, 215–233 (2009).

    Article  CAS  Google Scholar 

  18. Selbach, M. et al. Widespread changes in protein synthesis induced by microRNAs. Nature 455, 58–63 (2008).

    Article  CAS  Google Scholar 

  19. Scheja, L. & Heeren, J. Metabolic interplay between white, beige, brown adipocytes and the liver. J. Hepatol. 64, 1176–1186 (2016).

    Article  CAS  Google Scholar 

  20. Turner, N. et al. Distinct patterns of tissue-specific lipid accumulation during the induction of insulin resistance in mice by high-fat feeding. Diabetologia 56, 1638–1648 (2013).

    Article  CAS  Google Scholar 

  21. Povero, D. et al. Lipid-induced hepatocyte-derived extracellular vesicles regulate hepatic stellate cell via microRNAs targeting PPAR-gamma. Cell Mol. Gastroenterol. Hepatol. 1, 646–663 e644 (2015).

    Article  Google Scholar 

  22. Lee, Y. S. et al. Exosomes derived from palmitic acid-treated hepatocytes induce fibrotic activation of hepatic stellate cells. Sci. Rep. 7, 3710 (2017).

    Article  Google Scholar 

  23. Chen, L., Chen, R., Kemper, S., Charrier, A. & Brigstock, D. R. Suppression of fibrogenic signaling in hepatic stellate cells by Twist1-dependent microRNA-214 expression: role of exosomes in horizontal transfer of Twist1. Am. J. Physiol. Gastrointest. Liver Physiol. 309, G491–G499 (2015).

    Article  CAS  Google Scholar 

  24. Pescador, N. et al. Serum circulating microRNA profiling for identification of potential type 2 diabetes and obesity biomarkers. PLoS ONE 8, e77251 (2013).

    Article  CAS  Google Scholar 

  25. Chen, L., Brenner, D. A. & Kisseleva, T. Combatting fibrosis: exosome-based therapies in the regression of liver fibrosis. Hepatol. Commun. 3, 180–192 (2019).

    Article  Google Scholar 

  26. Shurtleff, M. J., Temoche-Diaz, M. M., Karfilis, K. V., Ri, S. & Schekman, R. Y-box protein 1 is required to sort microRNAs into exosomes in cells and in a cell-free reaction. eLife https://doi.org/10.7554/eLife.19276 (2016).

  27. Shurtleff, M. J. et al. Broad role for YBX1 in defining the small noncoding RNA composition of exosomes. Proc. Natl Acad. Sci. USA 114, E8987–E8995 (2017).

    Article  CAS  Google Scholar 

  28. Ran, F. A. et al. In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186–U198 (2015).

    Article  CAS  Google Scholar 

  29. Platt, R. J. et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 159, 440–455 (2014).

    Article  CAS  Google Scholar 

  30. Liu, P. S. et al. Alpha-ketoglutarate orchestrates macrophage activation through metabolic and epigenetic reprogramming. Nat. Immunol. 18, 985–994 (2017).

    Article  CAS  Google Scholar 

  31. Ostrowski, M. et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nat. Cell Biol. 12, 19–30 (2010).

    Article  CAS  Google Scholar 

  32. Song, L. et al. KIBRA controls exosome secretion via inhibiting the proteasomal degradation of Rab27a. Nat. Commun. 10, 1639 (2019).

    Article  Google Scholar 

  33. Bobrie, A. et al. Rab27a supports exosome-dependent and -independent mechanisms that modify the tumor microenvironment and can promote tumor progression. Cancer Res. 72, 4920–4930 (2012).

    Article  CAS  Google Scholar 

  34. Agarwal, V., Bell, G. W., Nam, J. W. & Bartel, D. P. Predicting effective microRNA target sites in mammalian mRNAs. eLife https://doi.org/10.7554/eLife.05005 (2015).

  35. Zhao, Y. et al. Liver governs adipose remodelling via extracellular vesicles in response to lipid overload. Nat. Commun. 11, 719 (2020).

    Article  CAS  Google Scholar 

  36. Alderson, N. L. et al. The human FA2H gene encodes a fatty acid 2-hydroxylase. J. Biol. Chem. 279, 48562–48568 (2004).

    Article  CAS  Google Scholar 

  37. Eckhardt, M., Yaghootfam, A., Fewou, S. N., Zoller, I. & Gieselmann, V. A mammalian fatty acid hydroxylase responsible for the formation of alpha-hydroxylated galactosylceramide in myelin. Biochem. J. 388, 245–254 (2005).

    Article  CAS  Google Scholar 

  38. Shang, M. et al. Macrophage-derived glutamine boosts satellite cells and muscle regeneration. Nature 587, 626–631 (2020).

    Article  CAS  Google Scholar 

  39. Lignitto, L. et al. Nrf2 activation promotes lung cancer metastasis by inhibiting the degradation of Bach1. Cell 178, 316–329 (2019).

    Article  CAS  Google Scholar 

  40. Yan, S. et al. IRF3 reduces adipose thermogenesis via ISG15-mediated reprogramming of glycolysis. J. Clin. Invest. https://doi.org/10.1172/JCI144888 (2021).

  41. Seo, J. B. et al. Knockdown of Ant2 reduces adipocyte hypoxia and improves insulin resistance in obesity. Nat. Metab. 1, 86–97 (2019).

    Article  CAS  Google Scholar 

  42. Li, P. et al. Hematopoietic-derived galectin-3 causes cellular and systemic insulin resistance. Cell 167, 973–984 e912 (2016).

    Article  CAS  Google Scholar 

  43. Ying, W., Cheruku, P. S., Bazer, F. W., Safe, S. H. & Zhou, B. Investigation of macrophage polarization using bone marrow derived macrophages. J. Vis. Exp. https://doi.org/10.3791/50323 (2013).

  44. Li, P. et al. LTB4 promotes insulin resistance in obese mice by acting on macrophages, hepatocytes and myocytes. Nat. Med. 21, 239–247 (2015).

    Article  CAS  Google Scholar 

  45. Garcia, D. M. et al. Weak seed-pairing stability and high target-site abundance decrease the proficiency of lsy-6 and other microRNAs. Nat. Struct. Mol. Biol. 18, 1139–1146 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the UCSD IGM Genomics Center for RNA-seq, the UCSD electron microscopy core for electron microscopic analysis, and the Vector Development Core Laboratory for lentivirus preparation. This study was funded by the US National Institute of Diabetes and Digestive and Kidney Diseases (grant nos. P30 DK063491, R00DK115998 and R01DK125560 to W.Y.) and the US National Institute of Diabetes and Digestive and Kidney Diseases (grant nos. P30 DK063491 and DK101395 to J.M.O.).

Author information

Authors and Affiliations

Authors

Contributions

W.Y. designed the studies, and Y.J. and Z.L. performed most of the experiments. F.C.G.D.R., K.A.M. and H.G. assisted with in vivo insulin stimulation assays, cell culture, qPCR analysis and western blot analysis. Z.J. and H.G. assisted with bone marrow cell isolation and differentiation. R.I., M.Y., W.F. and Z.J. assisted with tissue collection. G.B. assisted with hepatocyte isolation and glucose output assays. W.Y. and J.M.O. supervised the project. W.Y. and J.M.O. analysed and interpreted the data and cowrote the paper.

Corresponding authors

Correspondence to Wei Ying or Jerrold M. Olefsky.

Ethics declarations

Competing interests

W.Y. and J.M.O. are coinventors on a provisional patent covering the use of miR-3075 as an insulin sensitizer. Outside this, the other authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Jan-Wilhelm Kornfeld, Anja Zeigerer and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary handling editor: Isabella Samuelson.

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

Extended data

Extended Data Fig. 1 Hepatocyte exosomes from early stage obese mice improve cellular insulin sensitivity.

The marker characteristics (a) and production (b) of exosomes (Exos) derived from lean, 4wk or 16wk HFD WT hepatocytes. c, The appearance of red fluorescence in key metabolic tissues of 12wk HFD WT mice after tail vein injection with PKH26-labeled 4wk HFD hepatocyte Exos. 4wk HFD hepatocyte Exos without PKH26 labeling were injected to evaluate the level of autofluorescence. d, Effect of empty liposome treatment on metabolic phenotypes of 16wks HFD/obese WT mice. e, Effect of hepatocyte Exos treatment on body weight. f, the population and activation status of adipose tissue macrophages by flow cytometry analysis. Adipose tissue macrophages (ATM)=CD11b+F4/80+, M1 ATM=CD11c+CD206-, and M2 ATM=CD11c-CD206+. g, Effect of 4wk HFD Exos on proinflammatory cytokine abundance in epididymal fat of 18wk HFD mice by qPCR analysis. Effects of 4wk HFD Exos on glucose uptake of 3T3-L1 adipocytes (h), L6 myocytes (i), and glucose production of 12wk HFD/obese hepatocytes (j). k, The levels of insulin-stimulated AKT phosphorylation in 3T3-L1 adipocytes, L6 myocytes, and obese hepatocytes. e-k, empty liposomes were used in the control groups. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=7 (b), n=6 (d), n=5-10 (e), n=5 (f, h), n=3 (g), n=6 (i), n=8 (j) per group. Two-sided Student’s t-test.

Source data

Extended Data Fig. 2 Effect of hepatocyte-specific YBX1 knockout on the ability of hepatocyte Exos to regulate metabolic responses.

a, Hepatocyte YBX1 levels in the hepatocyte-specific Cas9 transgenic mice after 4 weeks of injection of lentivirus harbouring gRNA-YBX1 vectors. Control mice were treated with a lentiviral empty vector. b, Expression of TSG101 and CD63 in Exos derived from both 4wk HFD WT and YBX1KO hepatocytes. c, Body weight of both WT and YBX1KO mice after 4 weeks HFD feeding. d, Effect of hepatocyte Exos on body weight of 18wks HFD/obese recipient mice. Empty liposomes were used in the control HFD WT mice. e, Effect of 4wk HFD WT Exos or YBX1KO Exos on cellular phosphorylated AKT levels. Control cells were treated with empty liposomes. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=8 (c and d) per group.

Source data

Extended Data Fig. 3 Effect of hepatocyte-specific Rab27 knockout on metabolic phenotypes.

a, Hepatocyte Rab27 levels in the hepatocyte-specific Cas9 transgenic mice after 4 weeks of injection of lentivirus harboring gRNA Rab27 vectors. Control mice were treated with a lentiviral empty vector. b, Effect of 4wk HFD Rab27KO hepatocyte Exos on glucose production in 16wk HFD/obese primary hepatocytes. Empty liposomes were used in the control cells. c, Body weight of both WT and hepatocyte-specific Rab27KO mice after 4 weeks HFD feeding. d, Hepatic glucose production after Rab27 knockout. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=4 (b), n=6 (c), n=8 (d) per group. Two-sided Student’s t-test.

Source data

Extended Data Fig. 4 Effect of miR-3075 on insulin sensitivity.

miR-3075 abundance in liver (a), skeletal muscle (SKL, b), and eWAT (c) of 18wk HFD WT recipient mice after treatment with 4wk HFD Exos. qPCR analysis of miR-3075 abundance (d), hepatic glucose production (e), and insulin-stimulated AKT phosphorylation (f) in lean, 4wk or 16wk HFD WT hepatocytes. g, Effect of the miR-3075 antagomir on AKT phosphorylation level in 4wk HFD hepatocytes. h, miR-3075 abundance in key metabolic tissues of lean WT mice, 3T3-L1 adipocytes, and L6 myocytes. i, Effect of 4wk HFD Exos, 4wk HFD YBX1KO Exos, or combination treatment with 4wk HFD YBX1KO plus miR-3075 mimic on cellular insulin responses. j, Glucose tolerance of NCD, 4wk HFD, and 16wk HFD WT mice. k, qPCR analysis of miR-3075 abundance within Exos derived from lean hepatocytes transfected with the miR-3075 mimic. l, Effect of miR-3075-enriched hepatocyte (miR-3075oe) Exos on 3T3-L1 adipocyte glucose uptake. m, The levels of phosphorylated AKT in metabolic tissues of 4wks HFD WT mice after treatment with the miR-3075 antagomir. Effects of miR-3075 antagomir on glucose tolerance (n) and insulin sensitivity (o) of lean WT mice. n=4 (a-c, h, k), n=6-8 (d), n=8 (e), n=6 (i), n=8 (j, l), n=5-6 (n and o) per group. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. Two-sided Student’s t-test.

Source data

Extended Data Fig. 5 miR-3075 represses FA2H expression.

a, qPCR analysis of possible target genes of miR-3075. b, FA2H abundance after transfection with the miR-3075 mimic. c, The expression of miR-3075 in human HepG2 cells. d, Effect of miR-3075 overexpression on FA2H abundance in human HepG2 cells. e, Validation of FA2H knockdown in 3T3-L1 adipocytes, L6 myocytes, and obese hepatocytes. f, Effect of Fa2h knockdown on AKT phosphorylation in the cells without insulin stimulation. b and c, The control cells were transfected with a scramble miRNA mimic. e and f, non-targeting control siRNA was used in the control cells. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=5-6 (a, c) per group. Two-sided Student’s t-test.

Source data

Extended Data Fig. 6 16wk HFD Exos treatment did not affect cellular insulin sensitivity.

a-c, Effect of 16wk HFD Exos on cellular insulin responses. Empty liposomes were used in the control groups. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=5-6 (a), n=6 (b), n=4 (c) per group.

Extended Data Fig. 7 Effect of miR-434-3p on macrophage activation.

a, Gating strategy of flow cytometry analysis of adipose tissue macrophages. F4/80+CD11b+ ATMs were gated from viable CD45+ cells, and M1 (CD11c+CD206-) and M2 (CD11c-CD206+) cells were then gated from F4/80+CD11b+ ATMs. b, miR-434-3p abundance in epididymal fat of NCD recipient mice after treatment of 16wk HFD Exos for 5 weeks. c, qPCR analysis of miR-434-3p abundance in BMDMs after transfection with the miR-434-3p mimic. d, Effect of miR-434-3p overexpression on proinflammatory activation in naïve M0 BMDMs. e-g, Effect of miR-434-3p on glucose uptake in 3T3-L1 adipocytes and L6 myocytes and primary hepatocyte glucose production. c-g, A scramble miRNA mimic was used in control cells. All experiments were repeated at least twice with similar results. Data are presented as mean ± SEM. n=4 (a, e, g), n=5-6 (c), n=6-7 (d), n=12 (f) per group. Two-sided Student’s t-test.

Supplementary information

Supplementary Information

Supplementary Table 1: information on reagents and resources.

Reporting Summary

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ji, Y., Luo, Z., Gao, H. et al. Hepatocyte-derived exosomes from early onset obese mice promote insulin sensitivity through miR-3075. Nat Metab 3, 1163–1174 (2021). https://doi.org/10.1038/s42255-021-00444-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00444-1

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