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:

TFEB–GDF15 axis protects against obesity and insulin resistance as a lysosomal stress response

Abstract

TFEB, a key regulator of lysosomal biogenesis and autophagy, is induced not only by nutritional deficiency but also by organelle stress. Here, we find that Tfeb and its downstream genes are upregulated together with lipofuscin accumulation in adipose tissue macrophages (ATMs) of obese mice or humans, suggestive of obesity-associated lysosomal dysfunction/stress in ATMs. Macrophage-specific TFEB-overexpressing mice display complete abrogation of diet-induced obesity, adipose tissue inflammation and insulin resistance, which is independent of autophagy, but dependent on TFEB-induced GDF15 expression. Palmitic acid induces Gdf15 expression through lysosomal Ca2+-mediated TFEB nuclear translocation in response to lysosomal stress. In contrast, mice fed a high-fat diet with macrophage-specific Tfeb deletion show aggravated adipose tissue inflammation and insulin resistance, accompanied by reduced GDF15 level. Finally, we observe activation of TFEB–GDF15 in ATMs of obese humans as a consequence of lysosomal stress. These findings highlight the importance of the TFEB–GDF15 axis as a lysosomal stress response in obesity or metabolic syndrome and as a promising therapeutic target for treatment of these conditions.

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: Increased TFEB expression in adipose tissue macrophages from obese mice and humans.
Fig. 2: Protection against diet-induced obesity and insulin resistance in mice with macrophage TFEB overexpression.
Fig. 3: Reduced adipose tissue inflammation and enhanced lipid catabolism/thermogenesis in TFEBMac-Tg mice.
Fig. 4: Autophagy-independent metabolic improvement in TFEBMac-Tg mice.
Fig. 5: Identification of GDF15 as a TFEB target, and activation of the TFEB–GDF15 axis in ATMs of humans.
Fig. 6: TFEB-dependent GDF15 expression downstream of lysosomal stress.
Fig. 7: GDF15-dependent improvement of metabolic profile in TFEBMac-Tg mice.
Fig. 8: Aggravated adipose inflammation and insulin resistance in TfebΔMac obese mice.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon request. Source data are provided with this paper.

References

  1. Levine, B. & Kroemer, G. Autophagy in the pathogenesis of disease. Cell 132, 27–42 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Settembre, C. et al. TFEB links autophagy to lysosomal biogenesis. Science 332, 1429–1433 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Roczniak-Ferguson, A. et al. The transcription factor TFEB links mTORC1 signaling to transcriptional control of lysosome homeostasis. Sci. Signal 5, ra42 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Settembre, C. et al. A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. EMBO J. 31, 1095–1108 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Medina, D. L. et al. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat. Cell Biol. 17, 288–299 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. González-Muniesa, P. et al. Obesity. Nat. Rev. Dis. Primers 3, 17034 (2012).

    Article  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lynch, L. et al. Adipose tissue invariant NKT cells protect against diet-induced obesity and metabolic disorder through regulatory cytokine production. Immunity 37, 574–587 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nishimura, S. et al. CD8+ effector T cells contribute to macrophage recruitment and adipose tissue inflammation in obesity. Nat. Med. 15, 914–920 (2009).

    Article  CAS  PubMed  Google Scholar 

  10. Talukdar, S. et al. Neutrophils mediate insulin resistance in mice fed a high-fat diet through secreted elastase. Nat. Med. 18, 1407–1412 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Weisberg, S. P. et al. Obesity is associated with macrophage accumulation in adipose tissue. J. Clin. Invest. 112, 1797–1808 (2003).

    Article  CAS  Google Scholar 

  12. Winer, D. A. et al. B cells promote insulin resistance through modulation of T cells and production of pathogenic IgG antibodies. Nat. Med. 17, 610–617 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jung, S. B. et al. Reduced oxidative capacity in macrophages results in systemic insulin resistance. Nat. Commun. 9, 1551 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mansueto, G. et al. Transcription factor EB controls metabolic flexibility during exercise. Cell Metab. 25, 182–196 (2015).

    Article  CAS  Google Scholar 

  16. Settembre, C. et al. TFEB controls cellular lipid metabolism through a starvation-induced autoregulatory loop. Nat. Cell Biol. 15, 647–658 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Martina, J. A., Diab, H. I., Brady, O. A. & Puertollano, R. TFEB and TFE3 are novel components of the integrated stress response. EMBO J. 35, 479–495 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Martina, J. A. & Puertollano, R. Protein phosphatase 2A stimulates activation of TFEB and TFE3 transcription factors in response to oxidative stress. J. Biol. Chem. 293, 12525–12534 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Nezich, C. L., Wang, C., Fogel, A. & Youle, R. J. MiT/TFE transcription factors are activated during mitophagy downstream of Parkin and Atg5. J. Cell Biol. 210, 435–450 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Pastore, N. et al. TFEB and TFE3 cooperate in the regulation of the innate immune response in activated macrophages. Autophagy 12, 1240–1258 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Fu, S., Watkins, S. M. & Hotamisligil, G. S. The role of endoplasmic reticulum in hepatic lipid homeostasis and stress signaling. Cell Metab. 15, 623–634 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Szendroedi, J., Phielix, E. & Roden, M. The role of mitochondria in insulin resistance and type 2 diabetes mellitus. Nat. Rev. Endocrinol. 8, 92–103 (2011).

    Article  PubMed  CAS  Google Scholar 

  23. Lawrence, R. E. & Zoncu, R. The lysosome as a cellular centre for signalling, metabolism and quality control. Nat. Cell Biol. 21, 133–142 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Gabriel, T. L. et al. Lysosomal stress in obese adipose tissue macrophages contributes to MITF-dependent Gpnmb induction. Diabetes 63, 3310–3323 (2014).

    Article  CAS  PubMed  Google Scholar 

  25. Feng, B. et al. Clodronate liposomes improve metabolic profile and reduce visceral adipose macrophage content in diet-induced obese mice. PLoS ONE 6, e24358 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Patsouris, D. et al. Ablation of CD11c-positive cells normalizes insulin sensitivity in obese insulin resistant animals. Cell Metab. 8, 301–309 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Palmieri, M. et al. Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum. Mol. Genet. 20, 3852–3866 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Lê, K. A. et al. Subcutaneous adipose tissue macrophage infiltration is associated with hepatic and visceral fat deposition, hyperinsulinemia and stimulation of NF-κB stress pathway. Diabetes 60, 2802–2809 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Silva, H. M. et al. Vasculature-associated fat macrophages readily adapt to inflammatory and metabolic challenges. J. Exp. Med. 216, 786–806 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Xu, X. et al. Obesity activates a program of lysosomal-dependent lipid metabolism in adipose tissue macrophages independently of classic activation. Cell Metab. 18, 816–830 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Björntorp, P. & Sjöström, L. Number and size of adipose tissue fat cells in relation to metabolism in human obesity. Metabolism 20, 703–713 (1971).

    Article  PubMed  Google Scholar 

  32. Hirsch, J. & Batchelor, B. Adipose tissue cellularity in human obesity. Clin. Endocrinol. Metab. 5, 299–311 (1976).

    Article  CAS  PubMed  Google Scholar 

  33. Kang, Y. H. et al. Impaired macrophage autophagy induces systemic insulin resistance in obesity. Oncotarget 7, 35577–35591 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Lee, H.-Y. et al. Autophagy deficiency in myeloid cells increases susceptibility to obesity-induced diabetes and experimental colitis. Autophagy 12, 1390–1403 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Danforth, E. J. & Burger, A. The role of thyroid hormones in the control of energy expenditure. Clin. Endocrinol. Metab. 13, 581–595 (1984).

    Article  CAS  PubMed  Google Scholar 

  36. Fisher, F. M. et al. FGF21 regulates PGC-1α and browning of white adipose tissues in adaptive thermogenesis. Genes Dev. 126, 271–281 (2012).

    Article  CAS  Google Scholar 

  37. Halaas, J. L. et al. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269, 543–546 (1995).

    Article  CAS  PubMed  Google Scholar 

  38. Yamauchi, T. et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat. Med. 7, 941–946 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Kim, K. H. et al. Autophagy deficiency leads to protection from obesity and insulin resistance by inducing Fgf21 as a mitokine. Nat. Med. 19, 83–92 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Yang, L., Li, P., Fu, S., Calay, E. S. & Hotamisligil, G. S. Defective hepatic autophagy in obesity promotes ER stress and causes insulin resistance. Cell Metab. 11, 467–478 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Koo, B. K. et al. Growth differentiation factor 15 predicts advanced fibrosis in biopsy-proven non-alcoholic fatty liver disease. Liver Int. 38, 695–705 (2017).

    Article  PubMed  CAS  Google Scholar 

  42. Chung, S. et al. Modulation of N-type calcium currents by presynaptic imidazoline receptor activation in rat superior cervical ganglion neurons. Exp. Physiol. 95, 982–993 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Shen, D. et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat. Commun. 3, 731 (2012).

    Article  PubMed  CAS  Google Scholar 

  44. Zhang, X. et al. MCOLN1 is a ROS sensor in lysosomes that regulates autophagy. Nat. Commun. 7, 12109 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang, W. et al. Upregulation of lysosomal TRPML1 channels is essential for lysosomal adaptation to nutrient starvation. Proc. Natl Acad. Sci. USA 112, E1373–E1381 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Yasuda, M. et al. Fatty acids are novel nutrient factors to regulate mTORC1 lysosomal localization and apoptosis in podocytes. Biochim. Biophys. Acta 1842, 1097–1108 (2014).

    Article  CAS  PubMed  Google Scholar 

  47. Cereghetti, G. M. et al. Dephosphorylation by calcineurin regulates translocation of Drp1 to mitochondria. Proc. Natl Acad. Sci. USA 105, 15803–15808 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Klimcakoova, E. et al. Worsening of obesity and metabolic status yields similar molecular adaptations in human subcutaneous and visceral adipose tissue: decreased metabolism and increased immune response. J. Clin. Endocrinol. Metab. 96, E73–E82 (2011).

    Article  CAS  Google Scholar 

  49. Kursawe, R. et al. A role of the inflammasome in the sow storage capacity of the abdominal subcutaneous adipose tissue in obese adolescents. Diabetes 65, 610–618 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Grijalva, A., Xu, X. & Ferrante, A. W. Autophagy is dispensable for macrophage-mediated lipid homeostasis in adipose tissue. Diabetes 65, 967–980 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chrysovergis, K. et al. NAG-1/GDF-15 prevents obesity by increasing thermogenesis, lipolysis and oxidative metabolism. Int. J. Obes. 38, 1555–1564 (2014).

    Article  CAS  Google Scholar 

  52. Chung, H. K. et al. Growth differentiation factor 15 is a myomitokine governing systemic energy homeostasis. J. Cell Biol. 216, 149–165 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Wang, X., Chrysovergis, K., Kosak, J. & Eling, T. E. Lower NLRP3 inflammasome activity in NAG-1 transgenic mice is linked to a resistance to obesity and increased insulin sensitivity. Obesity 22, 1256–1263 (2014).

    Article  CAS  PubMed  Google Scholar 

  54. Luan, H. H. et al. GDF15 is an inflammation-induced central mediator of tissue tolerance. Cell 178, 1232–1244 (2019).

    Article  CAS  Google Scholar 

  55. Sater, Z. A. et al. Pheochromocytoma and paraganglioma patients with poor survival often show brown adipose tissue activation. J. Clin. Endocrinol. Metab. 105, 1176–1185 (2020).

    Article  PubMed Central  Google Scholar 

  56. Tsoli, M. et al. Activation of thermogenesis in brown adipose tissue and dysregulated lipid metabolism associated with cancer cachexia in mice. Cancer Res. 72, 4372–43282 (2012).

    Article  CAS  PubMed  Google Scholar 

  57. Reihani, N. et al. Unexpected macrophage-independent dyserythropoiesis in Gaucher disease. Hematologica 101, 1489–1498 (2016).

    Article  CAS  Google Scholar 

  58. Kim, K. H. et al. Growth differentiation factor 15 ameliorates nonalcoholic steatohepatitis and related metabolic disorders in mice. Sci. Rep. 8, 6789 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Hotamisligil, G. S. & Davis, R. J. Cell signaling and stress responses. Cold Spring Harb. Perspect. Biol. 8, a006072 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Patel, S. et al. GDF15 provides an endocrine signal of nutritional stress in mice and humans. Cell Metab. 29, 707–718 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Hsiao, E. C. et al. Characterization of growth-differentiation factor 15, a transforming growth factor beta superfamily member induced following liver injury. Mol. Cell. Biol. 20, 3742–3751 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Komatsu, M. et al. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J. Cell Biol. 169, 425–434 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Chang, E. J. et al. IL-34 is associated with obesity, chronic inflammation and insulin resistance. J. Clin. Endocrinol. Metab. 99, E1263–E1271 (2014).

    Article  CAS  PubMed  Google Scholar 

  64. Song, Y. S. et al. Association between low SIRT1 expression in visceral and subcutaneous adipose tissues and metabolic abnormalities in women with obesity and type 2 diabetes. Diabetes Res. Clin. Pract. 101, 341–348 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Heng, T. S. P. & Painter, M. W., Immunological Genome Project Consortium. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

  66. Fitzgibbons, T. P. et al. Similarity of mouse perivascular and brown adipose tissues and their resistance to diet-induced inflammation. Am. J. Physiol. 301, H1425–H1437 (2011).

    CAS  Google Scholar 

  67. Baek, S. J., Horowitz, J. M. & Eling, T. E. Molecular cloning and characterization of human nonsteroidal anti-inflammatory drug-activated gene promoter. Basal transcription is mediated by Sp1 and Sp3. J. Biol. Chem. 276, 33384–33392 (2001).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank S. J. Baek (Seoul National University) for human GDF15-promoter luciferase vectors, H. Xu (University of Michigan) for GCaMP3-ML1 plasmid and R. J. Youle (National Institute of Neurological Disorders and Stroke) for TFEB-KO and TFEB/TFE3/MITF triple-KO HeLa cell lines. This work was supported by the Basic Science Research Program through the National Research Foundation of Korea funded by the Ministry of Education (2013R1A6A3A04065825 to K.H.K.), a National Research Foundation of Korea grant funded by the Korean government (2019R1A2C3002924 to M.-S.L.), the Bio & Medical Technology Development Program (2017M3A9G7073521 to M.-S.L.), a grant from the Faculty Research Assistance Program of Yonsei University College of Medicine (6-2016-0055 to M.-S.L.), the Korea Mouse Phenotyping Project of the Ministry of Science, ICT and Future Planning through the National Research Foundation (NRF-2014M3A9D5A01073886 to C.S.C.) and the Basic Science Research Program, through the National Research Foundation of Korea funded by the Ministry of Science, ICT & Future Planning (2017R1A2B4008673 to Y.J.J.).

Author information

Authors and Affiliations

Authors

Contributions

K.H.K. and M.-S.L. conceived the experiments. K.H.K., J.K. and S.H.K. designed and conducted all experiments except the portions indicated below. K.H.K., J.K., S.H.K. and M.-S.L. analysed/interpreted the data. S.-Y.P., Y.C. and C.S.C. measured and analysed body composition and indirect calorimetry. H.K. measured intracellular/lysosomal Ca2+ and mitochondrial ROS contents. J.W.A., Y.-H.K. and S.C. conducted electrophysiology experiments. S.L. conducted experiments with BMDMs. Y.-M.L. measured the lysosomal pH. Y.J.J., H.S.P. and Y.H. provided human adipose tissues and analysed the data. K.H.K., J.K. and M.-S.L. wrote the manuscript.

Corresponding authors

Correspondence to Kook Hwan Kim or Myung-Shik Lee.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Metabolism thanks Carmine Settembre and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editors: Elena Bellafante; George Caputa.

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

Extended data

Extended Data Fig. 1 Tfeb and Gdf15 expression in immune cells and adipose tissue.

a, A heatmap showing the relative expression of Tfeb and Gdf15 mRNA in various immune cell types of C57BL/6N mice. b, Tfeb mRNA expression in macrophage and non-macrophage fractions separated by magnetic-activated cell sorting (MACS). c, Tfeb mRNA expression in M1-like and M2-like macrophages estimated by real-time RT-PCR after FACS sorting. d, A heatmap showing relative expression of Tfeb and its target genes in ATM subsets of C57BL/6N mice fed chow diet or HFD (GSE126407). DP-ATM and VA-ATM indicate monocyte-derived CD11c+CD64+ double-positive ATM and vasculature-associated ATM, respectively. VA-ATM1 and VA-ATM2 are distinguished by expression level of MHCII. e, Correlation between VAT TFEB mRNA level and body weight, fasting glucose level or VAT adipocyte size in pooled human subjects (lean, obese and obese T2DM) (n=54). f, Correlation between TFEB mRNA level in SAT and body weight, fasting glucose level or adipocyte sizes of SAT in pooled human subjects (n=54). All data are shown as means ± SEM. *P < 0.05 and **P < 0.01 by one-way ANOVA with Tukey’s test (b,c) or Pearson’s correlation analysis (e,f).

Source data

Extended Data Fig. 2 Generation and characterization of TFEBMac-Tg mouse.

a, A schematic diagram of generation of macrophage-specific TFEB-Tg mice. Excision of LoxP sites by Cre recombinase expressed under the control of the LysM promoter leads to expression of constitutively active TFEB transgene in myeloid cells. b, Relative mRNA expression of TFEB/Tfeb in peritoneal macrophages from TFEBMac-Tg and control mice. c, Immunoblots of peritoneal macrophages from TFEBMac-Tg and control mice using antibodies specific for human TFEB (hTFEB), Flag, LC3, p62 or HSP90 (a loading control). Numbers below immunoblot bands represent densitometric values. d, Representative images of hTFEB immunofluorescence in peritoneal macrophages from TFEBMac-Tg and control mice. Scale bars, 20 μm. e, Immunofluorescence images showing TFEB colocalized with F4/80 in the brain, liver, pancreas and adipose tissues. Occasional F4/80+ cells without nuclear TFEB were seen because efficiency of Cre-mediated excision of LoxP sequences was not 100%. Scale bars, 20 μm. f, Fasting blood glucose and serum insulin level in TFEBMac-Tg and control mice on chow diet (n=11). g, Serum CHO, FFA and TG levels in TFEBMac-Tg and control mice on chow diet (n=11). All data are shown as means ± SEM. *P < 0.05 and ***P < 0.001 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 3 Depletion of macrophages by clodronate administration.

a-e, GTT (a), fasting blood glucose (b), body weight (c), immunohistochemistry of adipose tissue using F4/80 antibody (d), and real-time RT-PCR analysis of F4/80 or Cd11b expression (e) in adipose tissue from HFD-fed TFEBMac-Tg and control mice with or without clodronate treatment (Clod) (n=4 each group). Scale bars, 200 μm. All data are shown as means ± SEM. *P < 0.05, **P < 0.01 and ***P < 0.001 by two-way ANOVA with Bonferroni’s test (a) or one-way ANOVA with Tukey’s test (b,c,e). (In a, red asterisk, comparison between untreated TFEBMac-Tg and untreated TFEBMac-Con mice; black asterisk, comparison between treated TFEBMac-Con and untreated TFEBMac-Con mice). (n.s., not significant).

Source data

Extended Data Fig. 4 Characterization of HFD-fed TFEBMac-Tg mice.

a, Representative gross images of adipose tissues from TFEBMac-Tg and control mice fed HFD for 12 weeks. b,c, Distribution of adipocyte size (diameter) in eWAT (b) and iWAT (c) of TFEBMac-Tg and control mice fed HFD for 12 weeks (n=4-6). d,e, Quantification of CLS (d) and F4/80 immunohistochemistry (e) in eWAT of TFEBMac-Tg and control mice fed HFD for 12 weeks (n=4-6). f, Relative expression of inflammatory markers in iWAT of TFEBMac-Tg and control mice fed HFD for 12 weeks (n=4-6). g, Average EE during day and night time periods in TFEBMac-Tg and control mice fed HFD for 8 weeks (n=8-10). h,i, Average food intake (h) and locomotor activity (i) during day and night time periods in TFEBMac-Tg and control mice fed HFD for 8 weeks (n=8-10). j-n, T3 (j), T4 (k), fibroblast growth factor 21 (FGF21) (l), adiponectin (m) and leptin (n) levels in TFEBMac-Tg and control mice fed HFD for 12 weeks (n=8-15). o, serum GDF15 in TFEBMac-Con/Atg7W/W, TFEBMac-Tg/Atg7W/W, TFEBMac-Con/Atg7DMac and TFEBMac-Tg/Atg7DMac mice fed HFD for 12 weeks (n=7-11) p, Relative Gdf15 mRNA levels in adipose tissues from C57BL/6N mice fed chow diet or HFD for 12 weeks (left, n=3-6) and from ob/ob mice (right, n=5). q, Relative Gdf15 mRNA level in mature adipocytes and SVCs of eWAT from C57BL/6N mice fed chow diet or HFD for 12 weeks estimated by real-time RT-PCR (n=3) (left). Expression of Adipoq and Cd45 mRNA level showing successful fractionation of adipocytes and SVCs (middle and right). r, Relative Gdf15 mRNA level in macrophage and non-macrophage fractions of SVCs of eWAT estimated by real-time RT-PCR after MACS separation. All data are shown as means ± SEM. *P < 0.05, **P < 0.01 and ***P < 0.001 by two-tailed Student’s t-test (d,e,f,g,n,p) or one-way ANOVA with Tukey’s test (o,q,r). (n.s., not significant).

Source data

Extended Data Fig. 5 Metabolic profile in thermoneutrality condition.

a-e, Body weight (a), body weight gain (b), adipose tissue weight (c), H&E staining of adipose tissues (d) and serum GDF15 (e) in HFD-fed TFEBMac-Tg and control mice housed at normal ambient (22 °C) or thermoneutral temperature (30 °C) (n=6-12). Scale bars, 200 μm. All data are shown as means ± SEM. *P < 0.05, **P < 0.01 and ***P < 0.001 by two-way ANOVA with Bonferroni’s test (a) or one-way ANOVA with Tukey’s test (b,c,e). (n.s., not significant).

Source data

Extended Data Fig. 6 Gdf15 mRNA expression in tissues with TFEB overexpression and metabolic profile of HFD-fed TFEBMac-Tg/Gdf15–/– mice.

a, Relative Gdf15 mRNA level in BMDMs of TFEBMac-Tg and control mice (n=3). b, Relative Gdf15 mRNA expression in skeletal muscle of mice injected intramuscularly with AAV2.1-TFEB (GSE62975) and in the liver of mice injected intravenously with HDAd-Pepck-TFEB (GSE41139). c,d, Relative Gdf15 mRNA levels in eWAT, iWAT and BAT of TFEBMac-Tg and control mice fed HFD (c, n=4-6) or chow diet (d, n=3-4) for 12 weeks. e,f, Real-time RT-PCR analysis of GDF15 expression (e) and serum GDF15 (f) in HFD-fed TFEBMac-Tg and control mice with or without clodronate treatment (Clod) (n=4 each group). g-i, Fasting blood glucose (g), fasting serum insulin (h) and serum CHO/FFA/TG levels (i) in TFEBMac-Con/Gdf15+/+, TFEBMac-Tg/Gdf15+/+, TFEBMac-Con/Gdf15–/– and TFEBMac-Tg/Gdf15–/– mice fed HFD for 12 weeks (n=7-12). All data are shown as means ± SEM. *P < 0.05, **P < 0.01 and ***P < 0.001 by two-tailed Student’s t-test (a,b,c,d) or one-way ANOVA with Tukey’s test (e,f,g,h,i).

Source data

Extended Data Fig. 7 Metabolic profile of chow- or HFD-fed GDF15-Tg mice.

a-c, Body weight (a), GTT (b) and ITT (insulin 1.5 U/kg) (c) in GDF15-Tg and control mice fed chow diet or HFD for 12 weeks (n=3-4). d-h, Food intake (d), tissue weight (e), representative H&E staining of eWAT, iWAT, BAT and the liver (f), serum ALT/AST (g) and serum CHO/FFA/TG levels (h) in GDF15-Tg and control mice fed chow diet or HFD for 12 weeks (n=3-4). Scale bars, 200 μm. i, Representative TH immunohistochemistry of BAT and iWAT in GDF15-Tg and control mice fed HFD for 12 weeks. Arrows indicate cells with positive TH staining. Scale bars, 50 μm for BAT and 200 μm for iWAT. All data are shown as means ± SEM. *P < 0.05, **P < 0.01, and ***P < 0.001 by two-way ANOVA with Bonferroni’s test (a,b,c) or one-way ANOVA with Tukey’s test (e,g,h). (In a-c, *P < 0.05, **P < 0.01 and ***P < 0.001 between GDF15-Tg and control mice fed chow diet. #P < 0.05, ##P < 0.01, and ###P < 0.001 between GDF15-Tg and control mice fed HFD). (n.s., not significant).

Source data

Extended Data Fig. 8 GDF15 increases sympathetic tone.

a-b, Action potential (AP) firing number under various stimulus intensity (a), and relationship between AP firing number and stimulus intensity (b) in mice treated with vehicle (Veh) or GDF15. c-d, Voltage tracing under various stimulus intensity (c), and AP threshold stimuli (d) in mice treated with vehicle or GDF15 (n=6). All data are shown as means ± SEM. **P < 0.01 and ***P < 0.001 by two-tailed Student’s t-test.

Source data

Extended Data Fig. 9 Gdf15 mRNA expression in cells treated with lysosomal stressors, and lysosomal stress in adipose tissues of obese mice and human subjects.

a, Quantification of nuclear TFEB in peritoneal macrophages treated with NH4Cl (20 μM), CQ (25 μM), BAF (50 nM) or ultrapure water (as vehicle) for 12 h (right). Representative immunofluorescence pictures are presented (left). Scale bars, 20 μm. b, Relative GDF15 (Gdf15) mRNA levels in BMDMs, HeLa, FaO and AML12 cells treated with CQ (25 μM) or NH4Cl (20 μM) for 12 h (n=3). c, Relative GDF15 mRNA levels in WT HeLa, TFEB-KO HeLa and TKO HeLa cells treated with NH4Cl (20 μM) for 12 h (n=3). d, Representative F4/80-GPNMB double immunofluorescence of eWAT from C57BL/6N mice fed chow diet or HFD. Scale bars, 20 μm. e, Relative GPNMB mRNA levels in VAT or SAT from lean subjects and obese subjects with or without T2DM (n=14-24). f, Representative CD68/GPNMB double immunohistochemistry in serial VAT or SAT sections from lean subjects and obese subjects with or without T2DM. Arrows indicate positive cells stained with each antibody. Scale bars, 100 μm. g, Relative Gpnmb mRNA levels in peritoneal macrophages treated with BSA or PA (400 μM) for 16 h (n=3). All data are shown as means ± SEM. *P < 0.05, **P < 0.01 and ***P < 0.001 by one-way ANOVA with Tukey’s test (a,b,c,e) or two-tailed Student’s t-test (g).

Source data

Extended Data Fig. 10 Cellular events associated with PA-induced TFEB activation.

a, Effect of ML-SI3, a TRPML1 inhibitor, on PA-induced TFEB nuclear translocation (right). Representative immunofluorescence pictures are presented (left). Scale bars, 5 μm. b, Mitochondrial ROS stained with MitoSOX in peritoneal macrophages treated with BSA or PA (400 μM) for 1 h in the presence or absence of Mito-Tempo (500 μM) pretreatment (lower). Gating strategy and representative scattergrams are presented (upper and middle). c, Macrophages were treated with PA or BSA control, and cell extract was subjected to immunoblot analysis to study phosphorylation of mTOR and its target, 4EBP1. Representative immunoblot (left). Densitometric ratio of phospho-mTOR or -4EBP1 band to HSP90 band (right). d, Macrophages were treated with PA or BSA control were subjected to immunofluorescence, and colocalization between mTOR and LAMP1 was examined (right). Representative immunofluorescence pictures are presented (left). Scale bars, 5 μm. e, Raw264.7 cells transfected with pcDNA 3.1-HA or HA-DCnA-H151Q were treatment with 400 μM PA or BSA. After immunostaining with anti-HA and TFEB antibodies, cells were analyzed by confocal microscopy (yellow arrow head, HA-DCnA-H151Q-transfected cells showing no TFEB translocation after PA treatment; red arrow head, untransfected cells showing TFEB translocation after PA treatment). Scale bars, 5 μm. All data are shown as means ± SEM. ***P < 0.001 by one-way ANOVA with Tukey’s test. (n.s., not significant).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2 and Tables 1 and 2

Reporting Summary

Supplementary Data 1

Statistical source data for Supplementary Fig. 1

Supplementary Data 2

Statistical source data for Supplementary Fig. 2

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 immunoblots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed immunoblots.

Source Data Fig. 7

Statistical source data.

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. 2

Unprocessed immunoblots.

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.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed immunoblots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kim, J., Kim, S.H., Kang, H. et al. TFEB–GDF15 axis protects against obesity and insulin resistance as a lysosomal stress response. Nat Metab 3, 410–427 (2021). https://doi.org/10.1038/s42255-021-00368-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-021-00368-w

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
-