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:

IL-1 induces mitochondrial translocation of IRAK2 to suppress oxidative metabolism in adipocytes

Abstract

Chronic inflammation is a common feature of obesity, with elevated cytokines such as interleukin-1 (IL-1) in the circulation and tissues. Here, we report an unconventional IL-1R–MyD88–IRAK2–PHB/OPA1 signaling axis that reprograms mitochondrial metabolism in adipocytes to exacerbate obesity. IL-1 induced recruitment of IRAK2 Myddosome to mitochondria outer membranes via recognition by TOM20, followed by TIMM50-guided translocation of IRAK2 into mitochondria inner membranes, to suppress oxidative phosphorylation and fatty acid oxidation, thereby attenuating energy expenditure. Adipocyte-specific MyD88 or IRAK2 deficiency reduced high-fat-diet-induced weight gain, increased energy expenditure and ameliorated insulin resistance, associated with a smaller adipocyte size and increased cristae formation. IRAK2 kinase inactivation also reduced high-fat diet-induced metabolic diseases. Mechanistically, IRAK2 suppressed respiratory super-complex formation via interaction with PHB1 and OPA1 upon stimulation of IL-1. Taken together, our results suggest that the IRAK2 Myddosome functions as a critical link between inflammation and metabolism, representing a novel therapeutic target for patients with obesity.

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: IL-1β inhibits mitochondrial oxidative phosphorylation and super-complex formation in adipocytes.
Fig. 2: Adipocyte-specific MyD88 deficiency (Myd88AKO) improves HFD-induced obesity and energy expenditure.
Fig. 3: IL-1β induces translocation of IRAK2 into inner mitochondrial membranes to suppress oxidative phosphorylation and FAO.
Fig. 4: IRAK2 deficiency improves HFD-induced obesity and energy expenditure.
Fig. 5: IRAK2 deficiency prevents HFD-induced BAT dysfunction.
Fig. 6: IRAK2 disrupts mitochondrial respiratory chain super-complex formation via interaction with PHB–OPA1.
Fig. 7: IRAK2 kinase activity contributes to HFD-induced obesity and BAT dysfunction.
Fig. 8: Adipocyte-specific IRAK2 contributes to HFD-induced obesity and BAT dysfunction.

Similar content being viewed by others

Data availability

The primary data for analysis of all figures and supplementary figures are available upon request. Source data are provided with this paper.

References

  1. Bozkurt, B. et al. Contributory risk and management of comorbidities of hypertension, obesity, diabetes mellitus, hyperlipidemia, and metabolic syndrome in chronic heart failure: a scientific statement from the American Heart Association. Circulation 134, e535–e578 (2016).

    PubMed  Google Scholar 

  2. Grundy, S. M. Metabolic syndrome update. Trends Cardiovasc. Med. 26, 364–373 (2016).

    Article  PubMed  Google Scholar 

  3. Said, S., Mukherjee, D. & Whayne, T. F. Interrelationships with metabolic syndrome, obesity and cardiovascular risk. Curr. Vasc. Pharm. 14, 415–425 (2016).

    Article  CAS  Google Scholar 

  4. Juge-Aubry, C. E. et al. Adipose tissue is a major source of interleukin-1 receptor antagonist: upregulation in obesity and inflammation. Diabetes 52, 1104–1110 (2003).

    Article  CAS  PubMed  Google Scholar 

  5. Greenberg, A. S. & Obin, M. S. Obesity and the role of adipose tissue in inflammation and metabolism. Am. J. Clin. Nutr. 83, 461S–465S (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sun, K., Kusminski, C. M. & Scherer, P. E. Adipose tissue remodeling and obesity. J. Clin. Invest. 121, 2094–2101 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Kershaw, E. E. & Flier, J. S. Adipose tissue as an endocrine organ. J. Clin. Endocrinol. Metab. 89, 2548–2556 (2004).

    Article  CAS  PubMed  Google Scholar 

  9. Ballak, D. B., Stienstra, R., Tack, C. J., Dinarello, C. A. & van Diepen, J. A. IL-1 family members in the pathogenesis and treatment of metabolic disease: focus on adipose tissue inflammation and insulin resistance. Cytokine 75, 280–290 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Cortez, M., Carmo, L. S., Rogero, M. M., Borelli, P. & Fock, R. A. A high-fat diet increases IL-1, IL-6, and TNF-α production by increasing NF-κB and attenuating PPAR-γ expression in bone marrow mesenchymal stem cells. Inflammation 36, 379–386 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Nov, O. et al. Interleukin-1β regulates fat–liver crosstalk in obesity by auto-paracrine modulation of adipose tissue inflammation and expandability. PLoS ONE 8, e53626 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Serena, C. et al. Obesity and type 2 diabetes alters the immune properties of human adipose derived stem cells. Stem Cells 34, 2559–2573 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Krautbauer, S. et al. Free fatty acids, lipopolysaccharide and IL-1α induce adipocyte manganese superoxide dismutase which is increased in visceral adipose tissues of obese rodents. PLoS ONE 9, e86866 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Schaeffler, A. et al. Fatty acid-induced induction of Toll-like receptor-4/nuclear factor-κB pathway in adipocytes links nutritional signalling with innate immunity. Immunology 126, 233–245 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. García, M. C. et al. Mature-onset obesity in interleukin-1 receptor I knockout mice. Diabetes 55, 1205–1213 (2006).

    Article  PubMed  CAS  Google Scholar 

  16. Dascombe, M. J., Hardwick, A., Lefeuvre, R. A. & Rothwell, N. J. Impaired effects of interleukin-1 beta on fever and thermogenesis in genetically obese rats. Int. J. Obes. 13, 367–373 (1989).

    CAS  PubMed  Google Scholar 

  17. Del Rey, A., Monge-Arditi, G., Klusman, I. & Besedovsky, H. O. Metabolic and endocrine effects of interleukin-1 in obese, diabetic Zucker fa/fa rats. Exp. Clin. Endocrinol. Diabetes 104, 317–326 (1996).

    Article  CAS  PubMed  Google Scholar 

  18. Matsuki, T., Horai, R., Sudo, K. & Iwakura, Y. IL-1 plays an important role in lipid metabolism by regulating insulin levels under physiological conditions. J. Exp. Med. 198, 877–888 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Somm, E. et al. Decreased fat mass in interleukin-1 receptor antagonist-deficient mice: impact on adipogenesis, food intake, and energy expenditure. Diabetes 54, 3503–3509 (2005).

    Article  CAS  PubMed  Google Scholar 

  20. Kawai, T. & Akira, S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 34, 637–650 (2011).

    Article  CAS  PubMed  Google Scholar 

  21. Lin, S. C., Lo, Y. C. & Wu, H. Helical assembly in the MyD88–IRAK4–IRAK2 complex in TLR/IL-1R signalling. Nature 465, 885–890 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yao, J. et al. Interleukin-1 (IL-1)-induced TAK1-dependent versus MEKK3-dependent NFκaB activation pathways bifurcate at IL-1 receptor-associated kinase modification. J. Biol. Chem. 282, 6075–6089 (2007).

    Article  CAS  PubMed  Google Scholar 

  23. Wan, Y. et al. Interleukin-1 receptor-associated kinase 2 is critical for lipopolysaccharide-mediated post-transcriptional control. J. Biol. Chem. 284, 10367–10375 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhou, H. et al. IRAK2 directs stimulus-dependent nuclear export of inflammatory mRNAs. Elife 6, e29630 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yu, M. et al. MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases. J. Exp. Med. 211, 887–907 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Everts, B. et al. TLR-driven early glycolytic reprogramming via the kinases TBK1–IKKɛ supports the anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Huang, Y. L. et al. Toll-like receptor agonists promote prolonged triglyceride storage in macrophages. J. Biol. Chem. 289, 3001–3012 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Salerno, F., Guislain, A., Cansever, D. & Wolkers, M. C. TLR-mediated innate production of IFN-γ by CD8+ T cells is independent of glycolysis. J. Immunol. 196, 3695–3705 (2016).

    Article  CAS  PubMed  Google Scholar 

  29. Tannahill, G. M. et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 496, 238–242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. 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 

  31. Cani, P. D. et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 1761–1772 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Shi, H. et al. TLR4 links innate immunity and fatty acid-induced insulin resistance. J. Clin. Invest. 116, 3015–3025 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Juge-Aubry, C. E. et al. Regulatory effects of interleukin (IL)-1, interferon-β, and IL-4 on the production of IL-1 receptor antagonist by human adipose tissue. J. Clin. Endocrinol. Metab. 89, 2652–2658 (2004).

    Article  CAS  PubMed  Google Scholar 

  34. Letts, J. A. & Sazanov, L. A. Clarifying the supercomplex: the higher-order organization of the mitochondrial electron transport chain. Nat. Struct. Mol. Biol. 24, 800–808 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Wesche, H., Henzel, W. J., Shillinglaw, W., Li, S. & Cao, Z. MyD88: an adapter that recruits IRAK to the IL-1 receptor complex. Immunity 7, 837–847 (1997).

    Article  CAS  PubMed  Google Scholar 

  36. Guo, Y. et al. Tim50, a component of the mitochondrial translocator, regulates mitochondrial integrity and cell death. J. Biol. Chem. 279, 24813–24825 (2004).

    Article  CAS  PubMed  Google Scholar 

  37. Rehling, P., Brandner, K. & Pfanner, N. Mitochondrial import and the twin-pore translocase. Nat. Rev. Mol. Cell Biol. 5, 519–530 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Saitoh, T. et al. Tom20 recognizes mitochondrial presequences through dynamic equilibrium among multiple bound states. EMBO J. 26, 4777–4787 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Abe, Y. et al. Structural basis of presequence recognition by the mitochondrial protein import receptor Tom20. Cell 100, 551–560 (2000).

    Article  CAS  PubMed  Google Scholar 

  40. Lee, C. M., Sedman, J., Neupert, W. & Stuart, R. A. The DNA helicase, Hmi1p, is transported into mitochondria by a C-terminal cleavable targeting signal. J. Biol. Chem. 274, 20937–20942 (1999).

    Article  CAS  PubMed  Google Scholar 

  41. Franconi, F. et al. Are the available experimental models of type 2 diabetes appropriate for a gender perspective? Pharm. Res. 57, 6–18 (2008).

    Article  Google Scholar 

  42. Kautzky-Willer, A., Harreiter, J. & Pacini, G. Sex and gender differences in risk, pathophysiology and complications of type 2 diabetes mellitus. Endocr. Rev. 37, 278–316 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Cypess, A. M. et al. Identification and importance of brown adipose tissue in adult humans. N. Engl. J. Med. 360, 1509–1517 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Nijtmans, L. G., Artal, S. M., Grivell, L. A. & Coates, P. J. The mitochondrial PHB complex: roles in mitochondrial respiratory complex assembly, ageing and degenerative disease. Cell Mol. Life Sci. 59, 143–155 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Glytsou, C. et al. Optic atrophy 1 is epistatic to the core MICOS component MIC60 in mitochondrial cristae shape control. Cell Rep. 17, 3024–3034 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Del Dotto, V. et al. OPA1 isoforms in the hierarchical organization of mitochondrial functions. Cell Rep. 19, 2557–2571 (2017).

    Article  CAS  PubMed  Google Scholar 

  47. Sun, X. J. et al. Deletion of interleukin 1 receptor-associated kinase 1 (Irak1) improves glucose tolerance primarily by increasing insulin sensitivity in skeletal muscle. J. Biol. Chem. 292, 12339–12350 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Rosselin, M., Santo-Domingo, J., Bermont, F., Giacomello, M. & Demaurex, N. L-OPA1 regulates mitoflash biogenesis independently from membrane fusion. EMBO Rep. 18, 451–463 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Jian, C. et al. Deficiency of PHB complex impairs respiratory supercomplex formation and activates mitochondrial flashes. J. Cell Sci. 130, 2620–2630 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Galmozzi, A. et al. ThermoMouse: an in vivo model to identify modulators of UCP1 expression in brown adipose tissue. Cell Rep. 9, 1584–1593 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Frezza, C., Cipolat, S. & Scorrano, L. Organelle isolation: functional mitochondria from mouse liver, muscle and cultured fibroblasts. Nat. Protoc. 2, 287–295 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Huynh, F. K., Green, M. F., Koves, T. R. & Hirschey, M. D. Measurement of fatty acid oxidation rates in animal tissues and cell lines. Methods Enzymol. 542, 391–405 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Han, Z. et al. Shear-induced reactive nitrogen species inhibit mitochondrial respiratory complex activities in cultured vascular endothelial cells. Am. J. Physiol. Cell Physiol. 292, C1103–C1112 (2007).

    Article  CAS  PubMed  Google Scholar 

  54. Chen, Y. R., Chen, C. L., Pfeiffer, D. R. & Zweier, J. L. Mitochondrial complex II in the post-ischemic heart: oxidative injury and the role of protein S-glutathionylation. J. Biol. Chem. 282, 32640–32654 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Yeh, S. T. et al. Preservation of mitochondrial function with cardiopulmonary resuscitation in prolonged cardiac arrest in rats. J. Mol. Cell Cardiol. 47, 789–797 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Felgner, P. L., Messer, J. L. & Wilson, J. E. Purification of a hexokinase-binding protein from the outer mitochondrial membrane. J. Biol. Chem. 254, 4946–4949 (1979).

    CAS  PubMed  Google Scholar 

  57. Sottocasa, G. L., Kuylenstierna, B., Ernster, L. & Bergstrand, A. An electron-transport system associated with the outer membrane of liver mitochondria. A biochemical and morphological study. J. Cell Biol. 32, 415–438 (1967).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Pagliarini, D. J. et al. Involvement of a mitochondrial phosphatase in the regulation of ATP production and insulin secretion in pancreatic β cells. Mol. Cell 19, 197–207 (2005).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the NIH (2P01HL029582, R01 AA023722, P01CA062220, R01 HL122283 and P50AA024333) and National Multiple Sclerosis Society (RG5130A2/1). H.Z. was supported by a Postdoctoral Research Fellowship Award (1-16-PDF-138) from the American Diabetes Association.

Author information

Authors and Affiliations

Authors

Contributions

H.Z., H.W., M. Yu, R.C.S., W.Q., F.T., W.L., H.Y., R.E.M. and J.Z. conducted the experiments. J.A.C., J.G. and A.D. performed the proteomics analysis. R.D. collected human adipose tissue samples. M. Yin and J.A.D. performed the electron microscopy analysis. H.Z. and X.L. wrote the manuscript. C.H., Y.-R.C., J.D.S., P.L.F., J.M.B. and X.L. supervised the study.

Corresponding author

Correspondence to Xiaoxia Li.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Extended information related to Fig. 1.

a. Characteristics of patients, related to Fig. 1a. b. Extracellular acidification rate (ECAR), related to Fig. 1d. Data represent mean ± SEM. Data represent one of five independent experiments with similar results. c. Coomassie blue staining of mitochondrial proteins, related to Fig. 1e. d. Mitochondrial proteins were extracted from WT and Il1r1 KO; WT and Irak2 KO newly differentiated primary adipocytes treated with or without IL-1β for indicated time points and analyzed by SDS-PAGE, followed by western blot analysis with indicated antibodies. Data represent one of five independent experiments with similar results. e. Octanoyl-carnitine oxidation-rate in isolated mitochondria from WT and Il1r1 KO primary adipocytes treated with or without IL-1β (n=4). Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 2 IL-1 stimulation did not lead to mitochondria dysfunction.

Newly differentiated primary adipocytes were treated with IL-1β for indicated time points. Cytosolic mtDNA (n=3) (a) and cytosolic Ca2+ (n=4) (b) levels were measured. c. Western blot analysis of cytoplasmic and mitochondrial proteins from IL-1β or TNFα + cycloheximide (CHX) treated newly differentiated primary adipocytes. Data represent one of five independent experiments with similar results. d. Newly differentiated primary adipocytes were treated with IL-1β or trifluoromethoxy carbonylcyanide phenylhydrazone (FCCP) for indicated time points. Cellular reactive oxygen species (ROS) were measured using fluorescent microscopy and microplate reader (n=5). e. Western blot analysis of cytoplasmic and mitochondrial proteins from oligomycin + antimycin (OA) treated newly differentiated WT and Irak2 KO primary adipocytes (pretreated with IL-1β or PBS). Data represent one of five independent experiments with similar results. Densitometric analysis of LC3 I/LC3 III were listed a bar graph (n=3). a, b, e: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM. d: One-way ANOVA was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 3 Extended information related to Fig. 2.

a. Total lysates from iWAT, BAT and liver from Myd88FF and Myd88AKO mice were subjected to western blot analysis with indicated antibodies. Data represent one of five independent experiments with similar results. b. Transmission electron microscopy analysis of iWAT sections from HFD-fed mice. Scale bars, 1 μm. Morphometric analysis of cristae area versus mitochondria area in 40 randomly selected mitochondria per group. c. Coomassie blue staining of mitochondrial proteins, related to Fig. 2i. d. Octanoyl-carnitine oxidation-rate in isolated mitochondria from BAT of HFD-fed mice with indicated genotypes (n=5). e. Rectal temperatures were measured for HFD-fed mice with indicated genotypes (n=5). b, d, e: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 4 Extended information related to Fig. 3.

a. Western blot analysis of immune-precipitated IRAK2 from IL-1β treated mitochondria and subjected to indicated amount of Lambda phosphatase (Lambda PP) treatment for 10 min. b. Wild-type and HA-tagged IRAK2 were restored in Irak2 KO adipocytes. Western blot analysis of cytoplasmic and mitochondrial proteins from IL-1β treated cells. c. Immunogold staining of HA-tagged IRAK2 which was overexpressed in Irak2 KO, Myd88 KO, Il1r1 KO and Phb KD cells with or without IL-1 treatment for 24h. Scale bars, 50 nm. d. Immunogold staining of HA-tagged MyD88 in newly differentiated primary adipocytes from Myd88-HA reporter mice. Mito: mitochondrion; Cyto: cytosol. Scale bar, 200 nm. e. Co-immunoprecipitation (IP) analysis of TIMM50 and TOM20 in mitochondria of in newly differentiated primary adipocytes from Irak2-HA reporter mice treated with IL-1β for indicated time points and followed by western blot analysis. a, b, e: Data represent one of five independent experiments with similar results.

Source data

Extended Data Fig. 5 Extended information related to Fig. 3.

a. Coomassie blue staining of mitochondrial proteins, related to Fig. 3d. b. Expression of indicated mRNAs in WT and Irak2 KO newly differentiated primary adipocytes treated with or without IL-1β for indicated time points (n=3). c. Extracellular acidification rate (ECAR), related to Fig. 3f. Data represent mean ± SEM. Data represent one of five independent experiments with similar results. d. Octanoyl-carnitine oxidation-rate in isolated mitochondria from WT and Irak2 KO primary adipocytes treated with or without IL-1β for 24h (n=5). e. Secondary helical wheel structure of IRAK2 peptide (amino acid: 39–52) which contains mitochondrial localization signal (MLS). The picture was generated by http://lbqp.unb.br/NetWheels/. f. Coomassie blue staining of mitochondrial proteins, related to Fig. 3k. g. Octanoyl-carnitine oxidation-rate in isolated mitochondria from Flag-tagged wild-type and IRAK2 mito-mutant restored Irak2 KO adipocytes treated with or without IL-1β for 24h (n=5). b, d, g: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 6 Extended information related to Fig. 4.

a. Body weight of WT and Irak2 KO mice on HFD feeding (n=6 females per group). b. Glucose tolerance test (GTT) and insulin tolerance test (ITT) were performed on HFD-fed of WT and Irak2 KO mice (n=5 females per group). c. H&E staining of iWAT sections from HFD-fed of WT and Irak2 KO mice. Cell size was quantified (3 views per slide, 3 sections per mouse, n=5). d. H&E staining of BAT sections from HFD-fed of WT and Irak2 KO mice. c, d: scale bars, 150 μm. a, b: Two-way ANOVA, followed by post hoc analysis was performed. Data represent mean ± SEM. c: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 7 Extended information related to Fig. 5.

a. Octanoyl-carnitine oxidation-rate in isolated mitochondria from BAT of HFD-fed mice with indicated genotypes (n=5). b. Various pictures of immunogold staining of HA-tagged IRAK2 in BAT sections of HFD-fed Irak2-HA reporter mice, related to Fig. 5c Scale bars, 200nm. c. Coomassie blue staining of mitochondrial proteins, related to Fig. 5e. d-e. Immunohistochemical staining of RFP in BAT sections of HFD-fed WT and Irak2 KO Ucp1-luc/tdTomato reporter mice. Luciferase enzymatic activity in lysates from BAT (d) and iWAT (e) (n=8). f, g. HFD-fed WT and Irak2 KO Ucp1-luc/tdTomato reporter mice injected with CL-316,243 for 3 days. Immunohistochemical staining of RFP in BAT sections and Luciferase enzymatic activity in lysates from BAT (L) and iWAT (M) (n=6). d, f: Scale bars, 150 nm. a, d–g: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 8 Extended information related to Fig. 6.

a. Co-immunoprecipitation (IP) analysis of HA-tagged IRAK2, in mitochondria of in newly differentiated primary adipocytes from Irak2-HA reporter mice treated with IL-1β for indicated time points and followed by western blot analysis. Data represent one of five independent experiments with similar results. b, c: Densitometric analysis of western blots in Fig. 6a, b. b. Signals corresponding to IP OPA1 were used and normalized to Mito input OPA1 in Fig. 6a (n=3). c. Signals corresponding to IP IRAK2 were used and normalized to Mito input IRAK2 in Fig. 6b (n=3). d, Mitochondrial proteins were extracted Ctrl and Phb KD primary adipocytes treated with IL-1β for indicated time points and analyzed by BN-PAGE, followed by Western blot analysis with anti-OxPhos cocktail antibodies. HMW SCs: high molecular weight super-complexes. e. [1-14C]-palmitic acid oxidation-rate f. Octanoyl-carnitine oxidation-rate in isolated mitochondria from WT and Phb KD primary adipocytes, treated with or without IL-1β for 24h (n=5). g. The activities of respiratory complexes h. Octanoyl-carnitine oxidation-rate in the isolated mitochondria in mitochondria of non-targeting siRNA (WT and Irak2 KO) and PHB siRNA transfected (WT/Phb KD, Irak2 / Phb KD) WT and Irak2 KO primary adipocytes treated with/without IL-1β (n=6). i. The activities of respiratory complexes j. Octanoyl-carnitine oxidation-rate in the isolated mitochondria in mitochondria of empty vector (WT and Irak2 KO) and PHB cDNA transfected (WT/PHB and Irak2 KO/PHB) WT and Irak2 KO primary adipocytes treated with/without IL-1β (n=6). b, c, e–j: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 9 Extended information related to Fig. 6.

a. Immunogold staining of HA-tagged IRAK2 and kinase-inactive (KI) mutant which were overexpressed in Irak2 KO cells with or without IL-1β treatment for 24h. Scale bars, 50 nm. b. Coomassie blue staining of mitochondrial proteins, related to Fig. 6d. c. Extracellular acidification rate (ECAR), related to Fig. 6g. Data represent mean ± SEM. Data represent one of five independent experiments with similar results. d. Octanoyl-carnitine oxidation-rate in isolated mitochondria from WT and Irak2 KI primary adipocytes, treated with or without IL-1β for 24h (n=5). Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Extended Data Fig. 10 Extended information related to Fig. 7 & 8.

a. Rectal temperatures were measured for HFD-fed mice with indicated genotypes (n=5) b. Coomassie blue staining of mitochondrial proteins, related to Fig. 7g. c. Targeting vector design for generation of a novel mouse strain with exon 1 of Irak2 flanked by loxP sites. d. Total lysates from iWAT, BAT and liver from Irak2FF and Irak2AKO mice were subjected to western blot analysis with indicated antibodies. Data represent one of five independent experiments with similar results. e. Rectal temperatures were measured for HFD-fed mice with indicated genotypes (n=5). f. Coomassie blue staining of mitochondrial proteins, related to Fig. 8g. a, c: Student’s t-test (two-tailed) was performed. Data represent mean ± SEM.

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed western blots for Fig. 1c,e.

Source Data Fig. 1

Statistical raw data for Fig. 1a,b,d–g.

Source Data Fig. 2

Unprocessed western blots for Fig. 2i.

Source Data Fig. 2

Statistical raw data for Fig. 2a–k.

Source Data Fig. 3

Unprocessed western blots for Fig. 3a,b,d,i–k.

Source Data Fig. 3

Statistical raw data for Fig. 3d–h,k–n.

Source Data Fig. 4

Statistical raw data for Fig. 4a–i.

Source Data Fig. 5

Unprocessed western blots for Fig. 5e.

Source Data Fig. 5

Statistical raw data for Fig. 5b,d–i.

Source Data Fig. 6

Unprocessed western blots for Fig. 6a–d,i,j.

Source Data Fig. 6

Statistical raw data for Fig. 6d–h.

Source Data Fig. 7

Unprocessed western blots for Fig. 7g.

Source Data Fig. 7

Statistical raw data for Fig. 7a–d,f–i.

Source Data Fig. 8

Unprocessed western blots for Fig. 8g.

Source Data Fig. 8

Statistical raw data for Fig. 8a–d,f–i.

Source Data Extended Data Fig. 1

Unprocessed western blots and gels for Extended Data Fig. 1c,d.

Source Data Extended Data Fig. 1

Statistical raw data for Extended Data Fig. 1b,e.

Source Data Extended Data Fig. 2

Unprocessed western blots for Extended Data Fig. 2c,e.

Source Data Extended Data Fig. 2

Statistical raw data for Extended Data Fig. 2a,b,d,e.

Source Data Extended Data Fig. 3

Unprocessed western blots and gels for Extended Data Fig. 3a,c.

Source Data Extended Data Fig. 3

Statistical raw data for Extended Data Fig. 3b–e.

Source Data Extended Data Fig. 4

Unprocessed western blots for Extended Data Fig. 4a,b,e.

Source Data Extended Data Fig. 5

Unprocessed gels for Extended Data Fig. 5a,f.

Source Data Extended Data Fig. 5

Statistical raw data for Extended Data Fig. 5b–d,g.

Source Data Extended Data Fig. 6

Statistical raw data for Extended Data Fig. 6a–c.

Source Data Extended Data Fig. 7

Unprocessed gels for Extended Data Fig. 7c.

Source Data Extended Data Fig. 7

Statistical raw data for Extended Data Fig. 7a,d–g.

Source Data Extended Data Fig. 8

Unprocessed western blots for Extended Data Fig. 8a,d.

Source Data Extended Data Fig. 8

Statistical raw data for Extended Data Fig. 8b,c,e–i.

Source Data Extended Data Fig. 9

Unprocessed gels for Extended Data Fig. 9b.

Source Data Extended Data Fig. 9

Statistical raw data for Extended Data Fig. 9c,d.

Source Data Extended Data Fig. 10

Unprocessed western blots and gels for Extended Data Fig. 10b,d,f.

Source Data Extended Data Fig. 10

Statistical raw data for Extended Data Fig. 10a,e.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhou, H., Wang, H., Yu, M. et al. IL-1 induces mitochondrial translocation of IRAK2 to suppress oxidative metabolism in adipocytes. Nat Immunol 21, 1219–1231 (2020). https://doi.org/10.1038/s41590-020-0750-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0750-1

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