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:

Brown adipose tissue-derived MaR2 contributes to cold-induced resolution of inflammation

Abstract

Obesity induces chronic inflammation resulting in insulin resistance and metabolic disorders. Cold exposure can improve insulin sensitivity in humans and rodents, but the mechanisms have not been fully elucidated. Here, we find that cold resolves obesity-induced inflammation and insulin resistance and improves glucose tolerance in diet-induced obese mice. The beneficial effects of cold exposure on improving obesity-induced inflammation and insulin resistance depend on brown adipose tissue (BAT) and liver. Using targeted liquid chromatography with tandem mass spectrometry, we discovered that cold and β3-adrenergic stimulation promote BAT to produce maresin 2 (MaR2), a member of the specialized pro-resolving mediators of bioactive lipids that play a role in the resolution of inflammation. Notably, MaR2 reduces inflammation in obesity in part by targeting macrophages in the liver. Thus, BAT-derived MaR2 could contribute to the beneficial effects of BAT activation in resolving obesity-induced inflammation and may inform therapeutic approaches to combat obesity and its complications.

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

Fig. 1: Cold exposure reduces inflammation and insulin resistance and improves glucose tolerance in DIO mice.
Fig. 2: Cold resolves obesity-induced inflammation in BAT and liver of DIO mice.
Fig. 3: Cold increases DHA-derived MaR2 and related structural isomers in BAT and liver of DIO mice.
Fig. 4: Cold specifically enhances the expression of 12-LOX and sEH in BAT, but not in the liver of DIO mice.
Fig. 5: BAT secretes MaR2 isomers in circulation.
Fig. 6: Mirabegron increases maresin pathway products in humans.
Fig. 7: BAT-specific loss of Alox12 increases inflammation in the liver of DIO mice.
Fig. 8: MaR2 resolves inflammation in obesity in part by targeting macrophages.

Similar content being viewed by others

Data availability

The authors declare that the data supporting the findings of this study are available within the paper and the Supplementary Information files. The raw data that support the findings of this study are available as source data files. Source data are provided with this paper.

References

  1. Blüher, M. Obesity: global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 15, 288–298 (2019).

    Article  PubMed  Google Scholar 

  2. Weisberg, S. P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Investig. 112, 1796–1808 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. Hotamisligil, G. S., Arner, P., Caro, J. F., Atkinson, R. L. & Spiegelman, B. M. Increased adipose tissue expression of tumor necrosis factor-α in human obesity and insulin resistance. J. Clin. Investig. 95, 2409–2415 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Esser, N., Legrand-Poels, S., Piette, J., Scheen, A. J. & Paquot, N. Inflammation as a link between obesity, metabolic syndrome and type 2 diabetes. Diabetes Res. Clin. Pract. 105, 141–150 (2014).

    Article  CAS  PubMed  Google Scholar 

  6. Serhan, C. N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510, 92–101 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Serhan, C. N. et al. Macrophage proresolving mediator maresin 1 stimulates tissue regeneration and controls pain. FASEB J. 26, 1755–1765 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hellmann, J. et al. Biosynthesis of D-series resolvins in skin provides insights into their role in tissue repair. J. Invest. Dermatol. 138, 2051–2060 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Spite, M., Claria, J. & Serhan, C. N. Resolvins, specialized proresolving lipid mediators, and their potential roles in metabolic diseases. Cell Metab. 19, 21–36 (2014).

    Article  CAS  PubMed  Google Scholar 

  10. Pal, A., Gowdy, K. M., Oestreich, K. J., Beck, M. & Shaikh, S. R. Obesity-driven deficiencies of specialized pro-resolving mediators may drive adverse outcomes during SARS-CoV-2 infection. Front. Immunol. 11, 1997 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Serhan, C. N. & Levy, B. D. Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J. Clin. Investig. 128, 2657–2669 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Serhan, C. N. et al. Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions. J. Exp. Med. 206, 15–23 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dalli, J. et al. The novel 13S,14S-epoxy-maresin is converted by human macrophages to maresin 1 (MaR1), inhibits leukotriene A4 hydrolase (LTA4H), and shifts macrophage phenotype. FASEB J. 27, 2573–2583 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Deng, B. et al. Maresin biosynthesis and identification of maresin 2, a new anti-inflammatory and pro-resolving mediator from human macrophages. PLoS ONE 9, e102362 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Han, Y. H. et al. A maresin 1/RORα/12-lipoxygenase autoregulatory circuit prevents inflammation and progression of nonalcoholic steatohepatitis. J. Clin. Investig. 129, 1684–1698 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Laiglesia, L. M. et al. Maresin 1 mitigates liver steatosis in ob/ob and diet-induced obese mice. Int. J. Obes. (2005) 42, 572–579 (2018).

    Article  CAS  Google Scholar 

  17. Jung, T. W., Kim, H. C., Abd El-Aty, A. M. & Jeong, J. H. Maresin 1 attenuates NAFLD by suppression of endoplasmic reticulum stress via AMPK-SERCA2b pathway. J. Biol. Chem. 293, 3981–3988 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Chiang, N., Libreros, S., Norris, P. C., de la Rosa, X. & Serhan, C. N. Maresin 1 activates LGR6 receptor promoting phagocyte immunoresolvent functions. J. Clin. Investig. 129, 5294–5311 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hanssen, M. J. et al. Short-term cold acclimation improves insulin sensitivity in patients with type 2 diabetes mellitus. Nat. Med. 21, 863–865 (2015).

    Article  CAS  PubMed  Google Scholar 

  20. Iwen, K. A. et al. Cold-induced brown adipose tissue activity alters plasma fatty acids and improves glucose metabolism in men. J. Clin. Endocrinol. Metab. 102, 4226–4234 (2017).

    Article  PubMed  Google Scholar 

  21. Bartelt, A. et al. Brown adipose tissue activity controls triglyceride clearance. Nat. Med. 17, 200–205 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Ouellet, V. et al. Brown adipose tissue oxidative metabolism contributes to energy expenditure during acute cold exposure in humans. J. Clin. Investig. 122, 545–552 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  23. van der Lans, A. A. et al. Cold acclimation recruits human brown fat and increases nonshivering thermogenesis. J. Clin. Investig. 123, 3395–3403 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Yoneshiro, T. et al. Recruited brown adipose tissue as an antiobesity agent in humans. J. Clin. Investig. 123, 3404–3408 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Becher, T. et al. Brown adipose tissue is associated with cardiometabolic health. Nat. Med. 27, 58–65 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Villarroya, F., Cereijo, R., Villarroya, J. & Giralt, M. Brown adipose tissue as a secretory organ. Nat. Rev. Endocrinol. 13, 26–35 (2017).

    Article  CAS  PubMed  Google Scholar 

  27. Scheele, C. & Wolfrum, C. Brown adipose crosstalk in tissue plasticity and human metabolism. Endocr. Rev. 41, 53–65 (2020).

    Article  Google Scholar 

  28. Leiria, L. O. et al. 12-Lipoxygenase regulates cold adaptation and glucose metabolism by producing the omega-3 lipid 12-HEPE from brown fat. Cell Metab. 30, 768–783 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lynes, M. D. et al. The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue. Nat. Med. 23, 631–637 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Hotamisligil, G. S., Budavari, A., Murray, D. & Spiegelman, B. M. Reduced tyrosine kinase activity of the insulin receptor in obesity-diabetes. Central role of tumor necrosis factor-α. J. Clin. Investig. 94, 1543–1549 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Miyazaki, Y., Pipek, R., Mandarino, L. J. & DeFronzo, R. A. Tumor necrosis factor-α and insulin resistance in obese type 2 diabetic patients. Int. J. Obes. Relat. Metab. Disord. 27, 88–94 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Olson, N. C. et al. Circulating levels of TNF-α are associated with impaired glucose tolerance, increased insulin resistance, and ethnicity: the Insulin Resistance Atherosclerosis study. J. Clin. Endocrinol. Metab. 97, 1032–1040 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Plomgaard, P. et al. Associations between insulin resistance and TNF-α in plasma, skeletal muscle and adipose tissue in humans with and without type 2 diabetes. Diabetologia 50, 2562–2571 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Ayina, C. N. et al. Association of serum leptin and adiponectin with anthropomorphic indices of obesity, blood lipids and insulin resistance in a Sub-Saharan African population. Lipids Health Dis. 15, 96 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Kennedy, A. et al. The metabolic significance of leptin in humans: gender-based differences in relationship to adiposity, insulin sensitivity, and energy expenditure. J. Clin. Endocrinol. Metab. 82, 1293–1300 (1997).

    CAS  PubMed  Google Scholar 

  36. Francisco, V. et al. Obesity, fat mass and immune system: role for leptin. Front. Physiol. 9, 640 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Santos-Alvarez, J., Goberna, R. & Sánchez-Margalet, V. Human leptin stimulates proliferation and activation of human circulating monocytes. Cell. Immunol. 194, 6–11 (1999).

    Article  CAS  PubMed  Google Scholar 

  38. Zhao, J., Unelius, L., Bengtsson, T., Cannon, B. & Nedergaard, J. Coexisting β-adrenoceptor subtypes: significance for thermogenic process in brown fat cells. Am. J. Physiol. 267, C969–C979 (1994).

    Article  CAS  PubMed  Google Scholar 

  39. Ghorbani, M. et al. Anti-diabetic effect of CL 316,243 (a β3-adrenergic agonist) by down regulation of tumour necrosis factor (TNF-α) expression. PLoS ONE 7, e45874 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Himms-Hagen, J. et al. Effect of CL-316,243, a thermogenic β3-agonist, on energy balance and brown and white adipose tissues in rats. Am. J. Physiol. 266, R1371–R1382 (1994).

    CAS  PubMed  Google Scholar 

  41. Vandanmagsar, B. et al. The NLRP3 inflammasome instigates obesity-induced inflammation and insulin resistance. Nat. Med. 17, 179–188 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kazankov, K. et al. The role of macrophages in nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Nat. Rev. Gastroenterol. Hepatol. 16, 145–159 (2019).

    Article  CAS  PubMed  Google Scholar 

  43. Xu, H. et al. Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance. J. Clin. Invest. 112, 1821–1830 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zatterale, F. et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Front. Physiol. 10, 1607 (2019).

    Article  PubMed  Google Scholar 

  45. Lumeng, C. N., Bodzin, J. L. & Saltiel, A. R. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J. Clin. Investig. 117, 175–184 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Seki, E. et al. TLR4 enhances TGF-β signaling and hepatic fibrosis. Nat. Med. 13, 1324–1332 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Yang, L. & Seki, E. Toll-like receptors in liver fibrosis: cellular crosstalk and mechanisms. Front. Physiol. 3, 138 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Kim, W. R., Flamm, S. L., Di Bisceglie, A. M. & Bodenheimer, H. C. Serum activity of alanine aminotransferase (ALT) as an indicator of health and disease. Hepatology 47, 1363–1370 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Chen, G., Ni, Y., Nagata, N., Xu, L. & Ota, T. Micronutrient antioxidants and nonalcoholic fatty liver disease. Int. J. Mol. Sci. 17, 1379 (2016).

    Article  PubMed Central  Google Scholar 

  50. Shamsi, F., Wang, C. H. & Tseng, Y. H. The evolving view of thermogenic adipocytes - ontogeny, niche and function. Nat. Rev. Endocrinol. 17, 726–744 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Goldenberg, M. M. Pharmaceutical approval update. J. Formul. Manag. 37, 668–708 (2012).

    Google Scholar 

  52. Cypess, A. M. et al. Activation of human brown adipose tissue by a β3-adrenergic receptor agonist. Cell Metab. 21, 33–38 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Seidman, J. S. et al. Niche-Specific reprogramming of epigenetic landscapes drives myeloid cell diversity in nonalcoholic steatohepatitis. Immunity 52, 1057–1074 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Daemen, S. et al. Dynamic shifts in the composition of resident and recruited macrophages influence tissue remodeling in NASH. Cell Rep. 34, 108626 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jaitin, D. A. et al. Lipid-associated macrophages control metabolic homeostasis in a TREM2-dependent manner. Cell 178, 686–698 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Perugorria, M. J. et al. Non-parenchymal TREM-2 protects the liver from immune-mediated hepatocellular damage. Gut 68, 533–546 (2019).

    Article  CAS  PubMed  Google Scholar 

  57. Hou, J. et al. TREM2 sustains macrophage-hepatocyte metabolic coordination in nonalcoholic fatty liver disease and sepsis. J. Clin. Investig. 131, e135197 (2021).

    Article  CAS  PubMed Central  Google Scholar 

  58. Deczkowska, A., Weiner, A. & Amit, I. The physiology, pathology, and potential therapeutic applications of the TREM2 signaling pathway. Cell 181, 1207–1217 (2020).

    Article  CAS  PubMed  Google Scholar 

  59. Hamerman, J. A. et al. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J. Immunol. 177, 2051–2055 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. Turnbull, I. R. et al. Cutting edge: TREM-2 attenuates macrophage activation. J. Immunol. 177, 3520–3524 (2006).

    Article  CAS  PubMed  Google Scholar 

  61. Wen, H. et al. Fatty acid-induced NLRP3-ASC inflammasome activation interferes with insulin signaling. Nat. Immunol. 12, 408–415 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wang, G. X. et al. The brown fat-enriched secreted factor Nrg4 preserves metabolic homeostasis through attenuation of hepatic lipogenesis. Nat. Med. 20, 1436–1443 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Mills, E. L. et al. UCP1 governs liver extracellular succinate and inflammatory pathogenesis. Nat. Metab. 3, 604–617 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Qing, H. et al. Origin and function of stress-induced IL-6 in murine models. Cell https://doi.org/10.1016/j.cell.2020.05.054 (2020).

  66. Simcox, J. et al. Global analysis of plasma lipids identifies liver-derived acylcarnitines as a fuel source for brown fat thermogenesis. Cell Metab. 26, 509–522 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Titos, E. et al. Signaling and immunoresolving actions of resolvin D1 in inflamed human visceral adipose tissue. J. Immunol. 197, 3360–3370 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Félix-Soriano, E. et al. Changes in brown adipose tissue lipid mediator signatures with aging, obesity, and DHA supplementation in female mice. FASEB J. 35, e21592 (2021).

    Article  PubMed  Google Scholar 

  69. Serhan, C. N. et al. Anti-inflammatory actions of neuroprotectin D1/protectin D1 and its natural stereoisomers: assignments of dihydroxy-containing docosatrienes. J. Immunol. 176, 1848–1859 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Shimizu, T., Rådmark, O. & Samuelsson, B. Enzyme with dual lipoxygenase activities catalyzes leukotriene A4 synthesis from arachidonic acid. PNAS 81, 689–693 (1984).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Musso, G., Gambino, R., Cassader, M., Paschetta, E. & Sircana, A. Specialized proresolving mediators: enhancing nonalcoholic steatohepatitis and fibrosis resolution. Trends Pharmacol. Sci. 39, 387–401 (2018).

    Article  CAS  PubMed  Google Scholar 

  72. Barden, A. et al. The effects of alcohol on plasma lipid mediators of inflammation resolution in patients with type 2 diabetes mellitus. Prostaglandins Leukot. Essent. Fatty Acids 133, 29–34 (2018).

    Article  CAS  PubMed  Google Scholar 

  73. Colas, R. A., Shinohara, M., Dalli, J., Chiang, N. & Serhan, C. N. Identification and signature profiles for pro-resolving and inflammatory lipid mediators in human tissue. Am. J. Physiol. Cell Physiol. 307, C39–C54 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Gomez, E. A. et al. Blood pro-resolving mediators are linked with synovial pathology and are predictive of DMARD responsiveness in rheumatoid arthritis. Nat. Commun. 11, 5420 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  75. López-Vicario, C. et al. Leukocytes from obese individuals exhibit an impaired SPM signature. FASEB J. 33, 7072–7083 (2019).

    Article  PubMed  Google Scholar 

  76. Welty, F. K. et al. Regression of human coronary artery plaque is associated with a high ratio of (18-hydroxy-eicosapentaenoic acid + resolvin E1) to leukotriene B(4). FASEB J. 35, e21448 (2021).

    Article  CAS  PubMed  Google Scholar 

  77. Burguillos, M. A. Use of meso-scale discovery™ to examine cytokine content in microglia cell supernatant. Methods Mol. Biol. 1041, 93–100 (2013).

    Article  CAS  PubMed  Google Scholar 

  78. Choi, S. H. et al. Combined adult neurogenesis and BDNF mimic exercise effects on cognition in an Alzheimer’s mouse model. Science https://doi.org/10.1126/science.aan8821 (2018).

  79. Dalli, J., Colas, R. A., Walker, M. E. & Serhan, C. N. Lipid mediator metabolomics via LC–MS/MS profiling and analysis. Methods Mol. Biol. 1730, 59–72 (2018).

    Article  CAS  PubMed  Google Scholar 

  80. Daemen, S., Chan, M. M. & Schilling, J. D. Comprehensive analysis of liver macrophage composition by flow cytometry and immunofluorescence in murine NASH. STAR Protoc. 2, 100511 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Tseng, Y. H. et al. New role of bone morphogenetic protein 7 in brown adipogenesis and energy expenditure. Nature 454, 1000–1004 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Xue, R. et al. Clonal analyses and gene profiling identify genetic biomarkers of the thermogenic potential of human brown and white preadipocytes. Nat. Med. 21, 760–768 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Kriszt, R. et al. Optical visualisation of thermogenesis in stimulated single-cell brown adipocytes. Sci. Rep. 7, 1383 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  84. Sansbury, B. E. et al. Myeloid ALX/FPR2 regulates vascularization following tissue injury. PNAS 117, 14354–14364 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported in part by US National Institutes of Health grants R01DK122808 (to Y.-H.T. and M.S.), R01DK077097 and R01DK102898 (to Y.-H.T.), R01HL106173 (to M.S.), R01DK099511 and R01DK112283 (to L.J.G.), P30DK036836 (to Joslin Diabetes Center’s Diabetes Research Center) and by US Army Medical Research grant W81XWH-17-1-0428 (to Y.-H.T.). S.S. was supported by the Manpei Suzuki Diabetes Foundation in Japan. G.P. was supported by grant 2019/20554-7 from The Sao Paulo Research Foundation, Sao Paulo Research Foundation. L.O.L. was supported by an American Diabetes Association post-doctoral fellowship (1-16-PDF-063) and by the Sao Paulo Research Foundation grants 2017/02684 and 2019/26008-4. We thank M. Lynes, F. Shamsi and Y. Zhang for providing general technical support and helpful advice. We thank S. Dong for helping with animal work and A. Clermont, A. Dean and M. Halpin of the Joslin Diabetes Research Center Animal Physiology core for assisting with the studies using the diurnal incubators. We thank H. Takahashi for providing helpful advice related to the clinical aspects of this work. We thank the laboratory of B. Hammock for providing the sEH antibody. All the schematics of the experimental design were created with BioRender.com.

Author information

Authors and Affiliations

Authors

Contributions

S.S., H.A.M., B.E.S., S.K., M.S. and Y.-H.T. developed the study concept and designed the experiments. S.S., H.A.M., B.E.S., S.K., T.T., C.H.W., X.Y., T.L.H., J.K., S.D.K., J.D., G.P., N.P., R.E.T., C.Z. and L.O.L. performed the research. E.K., L.J.G., T.S. and A.M.C. provided research advice. A.M.C. provided the human plasma samples. S.S., H.A.M., B.E.S., S.K., T.T., M.S. and Y.-H.T. contributed to data analysis. S.S., H.A.M., M.S. and Y.-H.T. wrote the paper. M.S. and Y.-H.T. directed the research. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Matthew Spite or Yu-Hua Tseng.

Ethics declarations

Competing interests

M.S. and Y.-H.T. are inventors of a pending provisional patent application related to maresin 2 and metabolic therapeutics.

Peer review

Peer review information

Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary handling editor: Isabella Samuelson, 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 Cold exposure reduces body weight, inflammation and insulin resistance in obese mice, related to Fig. 1.

(a) C57BL6/J male mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 14 weeks. The mice were then put in thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. Plasma levels of IFNγ, IL-6 and IL-1β at day 7 following TN or cold exposure (related to Fig. 1, n = 5 biologically independent animals per group, from 1 independent experiment). (b–e) C57BL6/J female mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 19 weeks. The mice were then put in thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. (b) Body weight on day 0 or day 7 following TN or cold exposure (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 6 for HF-TN, from 1 independent experiment). (c) Total caloric intake during the exposure period (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 6 for HF-TN, from 1 independent experiment). (d) Fasting blood glucose, plasma insulin and homeostatic model assessment of insulin resistance (HOMA-IR) (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 6 for HF-TN, from 1 independent experiment). (e) Plasma levels of TNF-α (n = 5 biologically independent animals per group, from 1 independent experiment, statistical significance was determined by two-tailed unpaired Student’s t-test). One-Way ANOVA followed by a Tukey’s post hoc test. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 2 Two days of cold exposure reduces insulin resistance and inflammation in DIO mice, related to Fig. 1.

C57BL6/J male mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 14 weeks to create DIO mice. (a-f) The DIO mice were exposed to thermoneutral (30 °C, TN) or cold (5 °C) conditions for 2 days. (a) Body weight on day 0 or day 2 following TN or cold exposure (n = 5 biologically independent animals for NC-TN and NC-cold, n = 10 for HF-TN and HF-cold, from 2 independent experiments). (b) Total caloric intake during the exposure period (n = 5 biologically independent animals for NC-TN and NC-cold, n = 10 for HF-TN and HF-cold, from 2 independent experiments). (c) Tissue weights at the end of the exposure period (epiWAT, ingWAT, Liver, Muscle: n = 5 biologically independent animals for NC-TN and NC-cold, n = 10 for HF-TN and HF-cold, from 2 independent experiments; BAT: n = 5 per group, from 1 independent experiment). (d) Fasting blood glucose, plasma insulin and homeostatic model assessment of insulin resistance (HOMA-IR) (n = 5 biologically independent animals for NC-TN and NC-cold, n = 10 for HF-TN and HF-cold, from 2 independent experiments). (e) Plasma levels of TNF-α (n = 5 biologically independent animals per group, from 1 independent experiment). (f) Plasma leptin levels (n = 5 biologically independent animals per group, from 1 independent experiment). (g-k) The DIO mice fed a HF diet for 15 weeks were treated with CL316243 (1 mg/kg/day, daily, i.p) for 9 days. (g, h) Body weight on day 0 and day 9 (n = 5 biologically independent animals per group, from 1 independent experiment). (i) Total caloric intake between day 0 and day 9 (n = 5 biologically independent animals per group, from 1 independent experiment). (j) Glucose levels during IPGTT after 7 days of CL316243 treatment (n = 5 biologically independent animals per group, from 1 independent experiment, Two-Way ANOVA followed by a Tukey’s post hoc). (k) Plasma TNF-α levels in mice treated with CL316243 for 9 days (n = 5 biologically independent animals for Vehicle, n = 6 for CL316243, from 1 independent experiment). Two-tailed Unpaired Student’s t-tests were performed to compare only 2 groups and One-Way ANOVA followed by a Tukey’s post hoc test was performed to compare 4 groups. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 3 Cold does not resolve inflammation in white fat of DIO mice, related to Fig. 2.

C57BL6/J male mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 14 weeks. The mice were then put in thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. (a) Relative mRNA expression of proinflammatory and NLRP3 inflammasome-related genes in ingWAT (n = 5 biologically independent animals per group, from 1 independent experiment). (b) Representative image of F4/80 staining of epiWAT sections from each group (Scale bar=100 μm) (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 4 for HF-TN, from 1 independent experiment). (c) The number of crown-like structures in epiWAT was determined by counting F4/80 positive areas per mm2 (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 4 for HF-TN, from 1 independent experiment). (d) The percentage of CD11c+ cells or CD206+ cells within the CD45+ F4/80+ population from ingWAT of DIO mice (n = 4 biologically independent animals per group, from 1 independent experiment). (e, f) Gating strategy for identification of CD11c+ and CD206+ cells in the epiWAT, ingWAT and BAT (e) and in the liver (f). One-Way ANOVA followed by a Tukey’s post hoc test. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 4 Cold-induced resolution of inflammation precedes changes in lipid accumulation in the liver of DIO mice, related to Fig. 2.

(a-c) C57BL6/J male mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 14 weeks. The mice were then exposed to thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. (a) Relative mRNA expression of lipogenesis-related genes in the liver (n = 5 biologically independent animals per group, from 1 independent experiment). (b) Liver TG levels (n = 5 biologically independent animals per group, from 1 independent experiment). (c) Representative images of hematoxylin and eosin (H&E) staining of liver sections from NC-TN, HF-TN and HF-cold groups (Scale bar=100 μm). (d-g) C57BL6/J male mice were fed with normal chow (NC) or high-fat (HF) diet at room temperature for 14 weeks. The mice were then put in thermoneutral temperature (30 °C, TN) or cold (5 °C) for 2 days. (d) Relative mRNA expression of lipogenesis-related genes in the liver (n = 5 biologically independent animals for NC-TN and NC-cold and HF-cold, n = 4 for HF-TN, from 1 independent experiment). (e) Liver TG levels (n = 5 biologically independent animals per group, from 1 independent experiment). (f) Representative images of hematoxylin and eosin (H&E) staining of liver sections from HF-TN and HF-cold groups (Scale bar=100 μm). (g) Relative mRNA expression of proinflammatory, NLRP3 inflammasome-related- and fibrosis-related genes in the liver (n = 5 biologically independent animals for NC-TN, NC-cold and HF-cold, n = 4 for HF-TN, Casp1 NC-cold, Casp1 HF-cold, Il18 HF-cold, Tlr4 HF-cold, from 1 independent experiment). One-Way ANOVA followed by a Tukey’s post hoc test. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 5 Identification and quantification of maresin pathway products in BAT and liver, related to Fig. 3.

(a, d) MRM peaks (top) and MS/MS spectra with diagnostic ions labeled (bottom) of authentic MaR2 standard (blue) and of MaR2 and a structural isomer (denoted isomer II, red) identified in a selected sample from BAT (a) and liver (d) of 14 weeks high-fat (HF)-fed mice exposed to cold (5 °C) or thermoneutral (30 °C, TN) conditions for 7 days. (b, c) Quantification of 14-HDHA, MaR2, and MaR2 isomer II in BAT of C57BL6/J mice fed a high-fat diet (HF; b; normalized to protein concentration; n = 5 biologically independent animals per group, from 1 independent experiment) or normal chow (NC; 14 weeks; c; n = 5 biologically independent animals for NC-TN and n = 4 for NC-cold, from 1 independent experiment) exposed to thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. ND: not detected. (e, f) Quantification of 14-HDHA, MaR2, and MaR2 isomer II in the liver of C57BL6/J mice fed HF (e; normalized to protein concentration; n = 5 biologically independent animals per group, except n = 4 for 14-HDHA HF-cold, from 1 independent experiment) or NC (f; n = 5 biologically independent animals per group, from 1 independent experiment) exposed to thermoneutral temperature (30 °C, TN) or cold (5 °C) for 7 days. (g) Quantification of 14-HDHA, MaR2, and MaR2 Isomer II (normalized to protein concentration) in the liver of HF-fed (15 weeks) male C57BL6/J mice treated with vehicle or CL316243 (1 mg/kg/day, daily i.p.) for 9 days (n = 5 biologically independent animals per group, from 1 independent experiment). Two-tailed Unpaired Student’s t-tests. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 6 Identification of maresin pathway products in ingWAT of obese mice exposed to cold and in plasma of mice with BAT removal, related to Fig. 4 and Fig. 5.

(a) Relative expression of Ucp1 mRNA in ingWAT of HF-fed (14 weeks) C57BL6/J mice exposed to TN or cold for 7 days (n = 5 biologically independent animals per group, from 1 independent experiment). (b, c) Quantification of 14-HDHA, MaR1, and MaR2 in ingWAT and normalized to tissue weight (n = 6 biologically independent animals for HF-TN, n = 5 for HF-cold, from 1 independent experiment; b) or protein concentration (n = 6 biologically independent animals for HF-TN, n = 5 for HF-cold, from 1 independent experiment; c). (d, e) MRM peaks (d) and MS/MS spectra with diagnostic ions labeled (e) of authentic MaR2 standard and of two MaR2 isomers (denoted isomer I and II) identified in a selected sample of plasma of HF-fed mice following BAT removal. ND: not detected. Two-tailed Unpaired Student’s t-tests. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 7 Identification of maresin pathway products in plasma of humans and expression of 12-LOX and sEH in human brown adipocytes, related to Fig. 6.

(a, b) MRM peaks (a) and MS/MS spectra with diagnostic ions labeled (b) of authentic 14-HDHA and MaR2 standards (top) and of 14-HDHA and MaR2 Isomer I identified in a selected sample of human plasma (bottom) following mirabegron administration. (c) Schematic of the treatment of human brown adipocytes with vehicle control or Forskolin (10μM), followed by the collection of the cells for qPCR (d) Relative mRNA expression of Alox12 and Ephx2 in human brown adipocytes treated with vehicle or Forskolin for indicated times (Alox12: n = 6 biologically independent cells for Vehicle, n = 4 biologically independent cells for 12 h, 18 h and 36 h, n = 3 biologically independent cells for 24 h, Ephx2: n = 6 biologically independent cells for Vehicle, n = 4 biologically independent cells for 12 h, 24 h and 36 h, n = 5 biologically independent cells for 18 h). One-Way ANOVA followed by a Tukey’s post hoc test. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 8 MaR2 resolves inflammation systemically and in the liver of DIO mice, related to Fig. 8.

(a) Schematic of the experimental design of MaR2 treatment. C57BL6/J male mice were fed with a high-fat (HF) diet at room temperature for 14 weeks. Then the mice were administered vehicle or MaR2 (5 μg/kg/day; daily i.p.) for 28 days. (b) Body weight at day 0 and day 28 post-treatment (n = 5 biologically independent animals per group, from 1 independent experiment). (c) Plasma TNF-α levels post-treatment (n = 5 biologically independent animals per group, from 1 independent experiment). (d) Relative mRNA expression of proinflammatory, NLRP3-inflammasome-related- and fibrosis-related genes in the liver post-treatment (n = 5 biologically independent animals per group, from 1 independent experiment). (e) Schematic of the experimental design of MaR2 treatment. C57BL6/J male mice were fed with a high-fat (HF) diet at room temperature for 16 weeks. Then, the mice were administered vehicle or MaR2 (10 μg/kg/day; daily i.p.) for 26 days. (f) Body weight at day 0 and day 25 post-treatment, and the body weight change from baseline (n = 6 biologically independent animals per group, from 1 independent experiment). (g) Total caloric intake (n = 6 biologically independent animals per group, from 1 independent experiment). (h, i) Liver weight and TG levels in liver (n = 6 biologically independent animals per group, from 1 independent experiment). (j) Relative mRNA expression of lipogenesis-related genes in liver (n = 6 biologically independent animals per group, from 1 independent experiment). (k) Representative liver H&E staining from each group (Scale bar=100 μm). (l) Plasma levels of ALT in mice treated with vehicle or MaR2 (n = 6 biologically independent animals per group, from 1 independent experiment). (m, n) Relative mRNA expression of inflammation and NLRP3-inflammasome-related genes in the BAT (n = 6 biologically independent animals per group, from 1 independent experiment, m) and epiWAT (n = 5 for Vehicle, except Casp1 n = 4, n = 6 for MaR2, from 1 independent experiment, n) of mice treated with vehicle or MaR2. Two-tailed Unpaired Student’s t-tests. Data are presented as mean ± SEM.

Source data

Extended Data Fig. 9 Absorption kinetics of d5-MaR2 and regulation of liver monocytes/macrophages by MaR2, related to Fig. 8.

(a) Recovery of d5-MaR2 spiked into murine plasma and subjected to solid phase extraction and LC-MS/MS analysis, with mean d5-MaR2 recovered indicated along with the calculated coefficient of variation (CV) from 4 individual replicates. (b) Schematic of the administration of d5-MaR2 to male mice fed a high-fat diet for 15 weeks, followed by collection of blood at the indicated time points. The red circle indicates the position of the deuterium (D) atoms at the omega end of MaR2. (c) Quantification of d5-MaR2 in plasma after intraperitoneal administration to obese mice, as determined by LC-MS/MS (n = 3 biologically independent animals per group, from 1 independent experiment, Kruskal-Wallis test, followed by Dunn’s multiple comparisons post-tests). (d) Flow cytometry gating strategy of monocyte and macrophage populations in the liver. Representative dot plots showing Single cells, Live cells and CD45 + cells (related to Fig. 8e). (e, f) Quantification of monocyte (e) and macrophage (f) populations in the liver of mice treated with vehicle or MaR2 (10 μg/kg/day; daily i.p.) for 5 days (related to Fig. 8; n = 5 biologically independent animals per group, from 1 independent experiment) (g) Primary rat Kupffer cells were incubated with vehicle or MaR2 (50 nM) for 18 hours, then RNA was harvested for qPCR to measure relative mRNA expression of proinflammatory, NLRP3-inflammasome-related- and fibrosis-related genes (n = 3 technical replicates per group). Two-tailed Unpaired Student’s t-tests, except c. Data are presented as mean ± SEM.

Source data

Supplementary information

Supplementary Information

Supplementary Tables 1 and 2

Reporting Summary

Source data

Source Data Fig. 1

Statistical Source Data.

Source Data Fig. 2

Statistical Source Data.

Source Data Fig. 3

Statistical Source Data.

Source Data Fig. 4

Statistical Source Data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical Source Data.

Source Data Fig. 6

Statistical Source Data.

Source Data Fig. 7

Statistical Source Data.

Source Data Fig. 7

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

Statistical Source Data.

Source Data Extended Data Fig. 3

Statistical Source Data.

Source Data Extended Data Fig. 4

Statistical Source Data.

Source Data Extended Data Fig. 5

Statistical Source Data.

Source Data Extended Data Fig. 6

Statistical Source Data.

Source Data Extended Data Fig. 7

Statistical Source Data.

Source Data Extended Data Fig. 8

Statistical Source Data.

Source Data Extended Data Fig. 9

Statistical Source Data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sugimoto, S., Mena, H.A., Sansbury, B.E. et al. Brown adipose tissue-derived MaR2 contributes to cold-induced resolution of inflammation. Nat Metab 4, 775–790 (2022). https://doi.org/10.1038/s42255-022-00590-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00590-0

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