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:

mTORC1 regulates a lysosome-dependent adaptive shift in intracellular lipid species

Abstract

The mechanistic target of rapamycin complex 1 (mTORC1) senses and relays environmental signals from growth factors and nutrients to metabolic networks and adaptive cellular systems to control the synthesis and breakdown of macromolecules; however, beyond inducing de novo lipid synthesis, the role of mTORC1 in controlling cellular lipid content remains poorly understood. Here we show that inhibition of mTORC1 via small molecule inhibitors or nutrient deprivation leads to the accumulation of intracellular triglycerides in both cultured cells and a mouse tumor model. The elevated triglyceride pool following mTORC1 inhibition stems from the lysosome-dependent, but autophagy-independent, hydrolysis of phospholipid fatty acids. The liberated fatty acids are available for either triglyceride synthesis or β-oxidation. Distinct from the established role of mTORC1 activation in promoting de novo lipid synthesis, our data indicate that mTORC1 inhibition triggers membrane phospholipid trafficking to the lysosome for catabolism and an adaptive shift in the use of constituent fatty acids for storage or energy production.

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: Lipidomic analysis of cells treated with mTORC1 inhibitors.
Fig. 2: Cells accumulate TGs following mTORC1 inhibition.
Fig. 3: mTORC1 suppresses endogenous lipid remodeling.
Fig. 4: TG accumulation is linked to other changes in lipid metabolism.
Fig. 5: Changes in intracellular lipid species following mTORC1 inhibition require lysosomal function.
Fig. 6: TG and other lipid species accumulate independently of autophagy following mTORC1 inhibition.
Fig. 7: Induction of endosomal delivery to the lysosome is required for the changes in cellular lipid species following mTORC1 inhibition.
Fig. 8: Model of the differential effects of mTORC1 activation and inhibition on cellular lipid metabolism.

Similar content being viewed by others

Data availability

All data generated or analyzed during this study are included in this published article (and its supplementary information files). Source data are provided with this paper.

References

  1. Rousseau, A. & Bertolotti, A. An evolutionarily conserved pathway controls proteasome homeostasis. Nature 536, 184–189 (2016).

    Article  CAS  Google Scholar 

  2. Ottens, F., Franz, A. & Hoppe, T. Build-UPS and break-downs: metabolism impacts on proteostasis and aging. Cell Death Differ. 28, 505–521 (2021).

    Article  CAS  Google Scholar 

  3. Ben-Sahra, I. & Manning, B. D. mTORC1 signaling and the metabolic control of cell growth. Curr. Opin. Cell Biol. 45, 72–82 (2017).

    Article  CAS  Google Scholar 

  4. Muller, M. et al. The coordinated action of the MVB pathway and autophagy ensures cell survival during starvation. eLife 4, e07736 (2015).

    Article  Google Scholar 

  5. Saxton, R. A. & Sabatini, D. M. mTOR signaling in growth, metabolism, and disease. Cell 168, 960–976 (2017).

    Article  CAS  Google Scholar 

  6. Kim, J. & Guan, K. L. mTOR as a central hub of nutrient signalling and cell growth. Nat. Cell Biol. 21, 63–71 (2019).

    Article  CAS  Google Scholar 

  7. Torrence, M. E. & Manning, B. D. Nutrient sensing in cancer. Annu. Rev. Cancer Biol. 2, 251–269 (2018).

    Article  Google Scholar 

  8. Duvel, K. et al. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol. Cell 39, 171–183 (2010).

    Article  Google Scholar 

  9. Peterson, T. R. et al. mTOR complex 1 regulates lipin 1 localization to control the SREBP pathway. Cell 146, 408–420 (2011).

    Article  CAS  Google Scholar 

  10. Porstmann, T. et al. SREBP activity is regulated by mTORC1 and contributes to Akt-dependent cell growth. Cell Metab. 8, 224–236 (2008).

    Article  CAS  Google Scholar 

  11. Li, S., Brown, M. S. & Goldstein, J. L. Bifurcation of insulin signaling pathway in rat liver: mTORC1 required for stimulation of lipogenesis, but not inhibition of gluconeogenesis. Proc. Natl Acad. Sci. USA 107, 3441–3446 (2010).

    Article  CAS  Google Scholar 

  12. Condon, K. J. & Sabatini, D. M. Nutrient regulation of mTORC1 at a glance. J. Cell Sci. 132, jcs222570 (2019).

    Article  CAS  Google Scholar 

  13. Kim, E., Goraksha-Hicks, P., Li, L., Neufeld, T. P. & Guan, K. L. Regulation of TORC1 by Rag GTPases in nutrient response. Nat. Cell Biol. 10, 935–945 (2008).

    Article  CAS  Google Scholar 

  14. Castellano, B. M. et al. Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science 355, 1306–1311 (2017).

    Article  CAS  Google Scholar 

  15. Dikic, I. & Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 19, 349–364 (2018).

    Article  CAS  Google Scholar 

  16. Palm, W. et al. The utilization of extracellular proteins as nutrients is suppressed by mTORC1. Cell 162, 259–270 (2015).

    Article  CAS  Google Scholar 

  17. Caron, A., Richard, D. & Laplante, M. The Roles of mTOR complexes in lipid metabolism. Annu. Rev. Nutr. 35, 321–348 (2015).

    Article  CAS  Google Scholar 

  18. Yecies, J. L. et al. Akt stimulates hepatic SREBP1c and lipogenesis through parallel mTORC1-dependent and independent pathways. Cell Metab. 14, 21–32 (2011).

    Article  CAS  Google Scholar 

  19. Ricoult, S. J., Yecies, J. L., Ben-Sahra, I. & Manning, B. D. Oncogenic PI3K and K-Ras stimulate de novo lipid synthesis through mTORC1 and SREBP. Oncogene 35, 1250–1260 (2016).

    Article  CAS  Google Scholar 

  20. Horton, J. D., Goldstein, J. L. & Brown, M. S. SREBPs: Activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109, 1125–1131 (2002).

    Article  CAS  Google Scholar 

  21. Hagen, R. M., Rodriguez-Cuenca, S. & Vidal-Puig, A. An allostatic control of membrane lipid composition by SREBP1. FEBS Lett. 584, 2689–2698 (2010).

    Article  CAS  Google Scholar 

  22. Quinn, W. J. 3rd et al. mTORC1 stimulates phosphatidylcholine synthesis to promote triglyceride secretion. J. Clin. Invest. 127, 4207–4215 (2017).

    Article  Google Scholar 

  23. Sengupta, S., Peterson, T. R., Laplante, M., Oh, S. & Sabatini, D. M. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468, 1100–1104 (2010).

    Article  CAS  Google Scholar 

  24. Soliman, G. A., Acosta-Jaquez, H. A. & Fingar, D. C. mTORC1 inhibition via rapamycin promotes triacylglycerol lipolysis and release of free fatty acids in 3T3-L1 adipocytes. Lipids 45, 1089–1100 (2010).

    Article  CAS  Google Scholar 

  25. Paolella, L. M. et al. mTORC1 restrains adipocyte lipolysis to prevent systemic hyperlipidemia. Mol. Metab. 32, 136–147 (2020).

    Article  CAS  Google Scholar 

  26. Morrisett, J. D. et al. Effects of sirolimus on plasma lipids, lipoprotein levels, and fatty acid metabolism in renal transplant patients. J. Lipid Res. 43, 1170–1180 (2002).

    Article  CAS  Google Scholar 

  27. Li, Z. et al. Cancer cells depend on environmental lipids for proliferation when electron acceptors are limited. Nat. Metab. 4, 711–723 (2020).

    Article  Google Scholar 

  28. Ackerman, D. et al. Triglycerides promote lipid homeostasis during hypoxic stress by balancing fatty acid saturation. Cell Rep. 24, 2596–2605 (2018).

    Article  CAS  Google Scholar 

  29. Lien, E. C. et al. Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 599, 302–307 (2021).

    Article  CAS  Google Scholar 

  30. Young, R. M. et al. Dysregulated mTORC1 renders cells critically dependent on desaturated lipids for survival under tumor-like stress. Genes Dev. 27, 1115–1131 (2013).

    Article  CAS  Google Scholar 

  31. Qiu, B. et al. HIF2α-dependent lipid storage promotes endoplasmic reticulum homeostasis in clear-cell renal cell carcinoma. Cancer Disco. 5, 652–667 (2015).

    Article  CAS  Google Scholar 

  32. Nguyen, T. B. et al. DGAT1-dependent lipid droplet biogenesis protects mitochondrial function during starvation-induced autophagy. Dev. Cell 42, 9–21 (2017).

    Article  CAS  Google Scholar 

  33. Rambold, A. S., Cohen, S. & Lippincott-Schwartz, J. Fatty acid trafficking in starved cells: regulation by lipid droplet lipolysis, autophagy, and mitochondrial fusion dynamics. Dev. Cell 32, 678–692 (2015).

    Article  CAS  Google Scholar 

  34. Filippakis, H. et al. Lysosomal regulation of cholesterol homeostasis in tuberous sclerosis complex is mediated via NPC1 and LDL-R. Oncotarget 8, 38099–38112 (2017).

    Article  Google Scholar 

  35. Abu-Remaileh, M. et al. Lysosomal metabolomics reveals V-ATPase- and mTOR-dependent regulation of amino acid efflux from lysosomes. Science 358, 807–813 (2017).

    Article  CAS  Google Scholar 

  36. Showalter, M. R. et al. The emerging and diverse roles of bis(monoacylglycero) phosphate lipids in cellular physiology and disease. Int. J. Mol. Sci. 21, 8067 (2020).

    Article  CAS  Google Scholar 

  37. Ohsaki, Y., Suzuki, M., Shinohara, Y. & Fujimoto, T. Lysosomal accumulation of mTOR is enhanced by rapamycin. Histochem. Cell Biol. 134, 537–544 (2010).

    Article  CAS  Google Scholar 

  38. Shayman, J. A. & Tesmer, J. J. G. Lysosomal phospholipase A2. Biochim. Biophys. Acta Mol. Cell. Biol. Lipids 1864, 932–940 (2019).

    Article  CAS  Google Scholar 

  39. Tsuboyama, K. et al. The ATG conjugation systems are important for degradation of the inner autophagosomal membrane. Science 354, 1036–1041 (2016).

    Article  CAS  Google Scholar 

  40. Kim, Y. M. et al. mTORC1 phosphorylates UVRAG to negatively regulate autophagosome and endosome maturation. Mol. Cell 57, 207–218 (2015).

    Article  CAS  Google Scholar 

  41. Yuan, H. X., Russell, R. C. & Guan, K. L. Regulation of PIK3C3/VPS34 complexes by MTOR in nutrient stress-induced autophagy. Autophagy 9, 1983–1995 (2013).

    Article  CAS  Google Scholar 

  42. Napolitano, G. & Ballabio, A. TFEB at a glance. J. Cell Sci. 129, 2475–2481 (2016).

    CAS  Google Scholar 

  43. Macia, E. et al. Dynasore, a cell-permeable inhibitor of dynamin. Dev. Cell 10, 839–850 (2006).

    Article  CAS  Google Scholar 

  44. Bago, R. et al. Characterization of VPS34-IN1, a selective inhibitor of Vps34, reveals that the phosphatidylinositol 3-phosphate-binding SGK3 protein kinase is a downstream target of class III phosphoinositide 3-kinase. Biochem. J. 463, 413–427 (2014).

    Article  CAS  Google Scholar 

  45. Rennick, J. J., Johnston, A. P. R. & Parton, R. G. Key principles and methods for studying the endocytosis of biological and nanoparticle therapeutics. Nat. Nanotechnol. 16, 266–276 (2021).

    Article  CAS  Google Scholar 

  46. Filippakis, H. et al. Vps34-mediated macropinocytosis in tuberous sclerosis complex 2-deficient cells supports tumorigenesis. Sci. Rep. 8, 14161 (2018).

    Article  Google Scholar 

  47. Dauner, K., Eid, W., Raghupathy, R., Presley, J. F. & Zha, X. mTOR complex 1 activity is required to maintain the canonical endocytic recycling pathway against lysosomal delivery. J. Biol. Chem. 292, 5737–5747 (2017).

    Article  CAS  Google Scholar 

  48. Madeira, J. B. et al. TORC1 inhibition induces lipid droplet replenishment in yeast. Mol. Cell. Biol. 35, 737–746 (2015).

    Article  Google Scholar 

  49. Bjedov, I. et al. Mechanisms of life span extension by rapamycin in the fruit fly Drosophila melanogaster. Cell Metab. 11, 35–46 (2010).

    Article  CAS  Google Scholar 

  50. Wilcock, D. J. et al. Oxidative stress from DGAT1 oncoprotein inhibition in melanoma suppresses tumor growth when ROS defenses are also breached. Cell Rep. 39, 110995 (2022).

    Article  CAS  Google Scholar 

  51. Chitraju, C. et al. Triglyceride synthesis by DGAT1 protects adipocytes from lipid-induced ER stress during lipolysis. Cell Metab. 26, 407–418 (2017).

    Article  CAS  Google Scholar 

  52. Piccolis, M. et al. Probing the global cellular responses to lipotoxicity caused by saturated fatty acids. Mol. Cell 74, 32–44 (2019).

    Article  CAS  Google Scholar 

  53. Fredrikson, G., Stralfors, P., Nilsson, N. O. & Belfrage, P. Hormone-sensitive lipase of rat adipose tissue. Purification and some properties. J. Biol. Chem. 256, 6311–6320 (1981).

    Article  CAS  Google Scholar 

  54. Priolo, C. et al. Tuberous sclerosis complex 2 loss increases lysophosphatidylcholine synthesis in lymphangioleiomyomatosis. Am. J. Respir. Cell Mol. Biol. 53, 33–41 (2015).

    Article  CAS  Google Scholar 

  55. Li, C. et al. Rapamycin-insensitive up-regulation of adipocyte phospholipase A2 in tuberous sclerosis and lymphangioleiomyomatosis. PLoS ONE 9, e104809 (2014).

    Article  Google Scholar 

  56. Naslavsky, N. & Caplan, S. The enigmatic endosome - sorting the ins and outs of endocytic trafficking. J. Cell Sci. 131, jcs216499 (2018).

    Article  Google Scholar 

  57. Bissig, C. & Gruenberg, J. Lipid sorting and multivesicular endosome biogenesis. Cold Spring Harb. Perspect. Biol. 5, a016816 (2013).

    Article  Google Scholar 

  58. Nnah, I. C. et al. TFEB-driven endocytosis coordinates MTORC1 signaling and autophagy. Autophagy 15, 151–164 (2019).

    Article  CAS  Google Scholar 

  59. Alesi, N. et al. TSC2 regulates lysosome biogenesis via a non-canonical RAGC and TFEB-dependent mechanism. Nat. Commun. 12, 4245 (2021).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  61. Oren, Y. et al. Cycling cancer persister cells arise from lineages with distinct programs. Nature 596, 576–582 (2021).

    Article  CAS  Google Scholar 

  62. Huang, J., Dibble, C. C., Matsuzaki, M. & Manning, B. D. The TSC1-TSC2 complex is required for proper activation of mTOR complex 2. Mol. Cell. Biol. 28, 4104–4115 (2008).

    Article  CAS  Google Scholar 

  63. Breitkopf, S. B. et al. A relative quantitative positive/negative ion switching method for untargeted lipidomics via high resolution LC-MS/MS from any biological source. Metabolomics 13, 30 (2017).

    Article  Google Scholar 

  64. Byles, V. et al. Hepatic mTORC1 signaling activates ATF4 as part of its metabolic response to feeding and insulin. Mol. Metab. 53, 101309 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Manning laboratory for helpful input, in particular S. Schrötter for her guidance in microscopy studies. We thank D. Kwiatkowski (Brigham and Women’s Hospital), E. Henske (Brigham and Women’s Hospital), D. Sabatini and W. Harper (Harvard Medical School) for providing cell lines and other reagents. We thank A. Kreutzberger, A. Sanyal and T. Kirchausen for helpful conversations and initial guidance in studying endocytosis and Z. W. Lai and Y. Ambaw from the Harvard-Chan Advanced Multi-omics Platform for guidance in piloting the Lyso-IP lipidomics experiments. Cell sorting was performed with the Harvard Medical School Systems Biology Flow Cytometry Core. A.M.H. was supported by a postdoctoral fellowship from the Jane Coffin Childs Memorial Fund for Medical Research. This study was funded by National Institutes of Health grants R35-CA197459 (B.D.M) and P01-CA120964 (B.D.M. and J.M.A.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization was the responsibility of A.M.H. and B.D.M. Investigation was the responsibility of A.M.H., M.E.W, M.C.M., K.C.K., M.E.T. and J.M.A. Writing of the manuscript was conducted by A.M.H. and B.D.M. Funding was acquired by A.M.H., J.M.A. and B.D.M. Resources were the responsibility of B.D.M. Supervision was carried out by B.D.M.

Corresponding author

Correspondence to Brendan D. Manning.

Ethics declarations

Competing interests

B.D.M. is a member of the scientific advisory board and a shareholder of Navitor Pharmaceuticals. All other authors declare no competing interests.

Peer review

Peer review information

Nature Metabolism thanks Martin Giera, Mathieu Laplante and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary Handling Editor: Yanina-Yasmin Pesch, 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 Lipidomic analysis of cells treated with mTORC1 inhibitors.

(a–c) Immunoblots for AKT and pAKT corresponding to Fig. 1a–c. (d, e) Fold changes of individual lipids grouped by class from the lipidomics experiment shown in Fig. 1d. (d) Non-ether and (e) ether lipids are separated. Dots represent the fold change of each individual lipid species relative its abundance in vehicle-treated cells. Bars represent the mean fold change for that class, graphed ± SD for n = 3 replicates. (f, g) Triglyceride species from the results in Fig. 1c stratified by the total number of double bonds (f) or carbon atoms (g) in their side chains. Each point represents an individual triglyceride. Group mean ± SD is shown in (f). Pearson’s r and line of best fit are shown in (g). Data represent n = 3 replicates. (h, i) Immunoblots for AKT and pAKT corresponding to Fig. 1g–j. (j) Changes in the abundance of individual lipid species from the experiment in Fig. 1h, with neutral lipids color coded, for mouse ES cells treated with torin1. Dotted lines indicate a 2-fold change (FC) and an adjusted p-value of 0.1. Statistical analysis by one-way ANOVA (f); n.s., not significant.

Source data

Extended Data Fig. 2 Intracellular triglycerides accumulate following mTORC1 inhibition.

(a) Triglyceride accumulation measured by enzyme assay and normalized to protein content from the same sample in Tsc2−/− MEFs serum-starved for 16 hrs, followed by 0–8 hrs of vehicle, rapamycin (20 nM), or torin1 (250 nM) treatment, graphed as mean ± SD relative to the 0 hr time point, n = 3. (b) Immunoblot for AKT and pAKT corresponding to Fig. 2m. (c) Immunoblot for the tumors shown in Fig. 2o. (d) Tumor TG versus plasma TG for individual mice shown in Fig. 2o, p. (e) Plasma non-esterified fatty acids (NEFA) measured for the mice shown in Fig. 2o, graphed as mean ± SD, n = 5 mice. (f) Immunoblot corresponding to the experiment shown in Fig. 2q. (g, h) Cell cycle profile for PC3 (g) and H1299 (H) cells treated for 16 hrs with vehicle, palbociclib (CDK4/6i, 100 nM), and/or torin1 (250 nM), n = 1. (i,j) Triglyceride levels quantified by enzyme assay and normalized to protein content from the same sample for cells treated as in (g, h), graphed as mean ± SD relative to vehicle-treated cells, n = 3. Statistical analysis by two-way ANOVA (i, j) and two-tailed Student’s t-test (e); n.s., not significant.

Source data

Extended Data Fig. 3 mTORC1 suppresses endogenous lipid remodeling and activates de novo lipid synthesis.

(a) SREBP1 target gene expression in Tsc2+/+ and Tsc2−/− MEFs treated for 16 hrs with vehicle, rapamycin (20 nM), or torin1 (250 nM), graphed as mean ± SD relative to vehicle-treated cells, n = 3. (B) Representative thin layer chromatogram for Tsc2−/− MEFs labeled with [1-14C]-oleate and treated with vehicle, rapamycin (20 nM), or torin1 (250 nM) for 16 hrs, developed to separate neutral lipids. ChE, cholesteryl-esters; TG, triglycerides; FA, fatty acids; DG, diglycerides; MG, monoglycerides. (c,d) Immunoblot (c) and TGs (d) assayed by pulse-chase with [1-14C]-oleate tracer (6 h) in Tsc2+/+ MEFs followed by chase in cold medium for 16 h with vehicle, 20 nM rapamycin, 250 nM torin1, 500 nM AZD2014, or 5 nM RapaLink-1, graphed as mean ± SD relative to vehicle-treated cells, n = 3. (e, f) Immunoblot (e) and TGs (f) assayed by pulse-chase with [1-14C]-oleate tracer (6 h) in MCF7 cells followed by chase in cold medium for 16 h with vehicle, 250 nM torin1, or 1 µM BYL719 treatment, graphed as mean ± SD relative to vehicle-treated cells, n = 3. NS denotes a non-specific band. (g–j) Immunoblot (g), de novo lipogenesis of all lipids (h), TG labeled from de novo lipogenesis (i), and lipid remodeling (j), assayed as in Fig. 3h,i in primary mouse hepatocytes serum-starved for 16 hrs, pretreated for 30 min with vehicle, rapamycin (20 nM), or torin1 (250 nM), and stimulated with insulin (100 nM) for 6 hrs, graphed as mean ± SD relative to vehicle-treated unstimulated cells, n = 3. Statistical analysis by one-way ANOVA (a, d, f, h–j); n.s., not significant.

Source data

Extended Data Fig. 4 TG accumulation is linked to other changes in lipid metabolism.

(a) 14C-TG turnover in PC3 cells treated with vehicle, torin1, or ATGLi, measured as in Fig. 4c. 14C-TG abundance is graphed over time as mean ± SD relative to 0 hrs, n = 3. (b) Immunoblots for ATGL and DGAT1 levels from two separate experiments in cell lines treated with vehicle, rapamycin, or torin1 for 16 hrs. NS denotes a non-specific band. (c) 14C-Fatty acid levels in Tsc2−/− MEFs labeled with [1-14C]-palmitate, treated with vehicle, rapamycin, or torin1 for 16 hrs, graphed as mean ± SD relative to vehicle, n = 3. (d) Acyl-carnitine species in Tsc2−/− MEFs in the experiment in Fig. 1d, graphed as mean ± SD relative to vehicle, n = 3. (e, f) Acyl-carnitine species detected in mouse embryonic stem (ES) cells (e) and wild-type, primary MEFs (f) in the experiments in Fig. 1h, j, graphed as mean ± SD relative to vehicle, n = 3. (g) Representative thin layer chromatogram for Tsc2−/− MEFs traced with [methyl-14C]-choline and treated with vehicle, rapamycin, or torin1 for 16 hrs, developed to separate phospholipids. PC, phosphatidylcholine; SM, sphingomyelin; LPC, lysophosphatidylcholine. (h) Ratio of lysophosphatidylcholine to phosphatidylcholine in PC3 cells pulse-labeled with [methyl-14C]-choline as in (g); mean ± SD relative to vehicle, n = 3. (i,j) Glycerophosphocholine levels in HEK-293T (i) and PC3 (j) cells treated for 16 hrs with vehicle, rapamycin, or torin1, graphed as mean ± SD relative to vehicle, n = 3. (k, l) 14C-TG (k) and 14C-fatty acids (L) in PC3 cells measured as in Fig. 4i, j and graphed as mean ± SD, n = 3. (m) Fatty acid accumulation in Tsc2−/− MEFs labeled with [1-14C]-oleate following vehicle (DMSO) treatment with 1 hr pretreatment with DGAT inhibitors and/or etomoxir; mean ± SD relative to 0 hrs, n = 3. (n) Triglyceride ion counts in the experiment shown in Fig. 4l, m; mean ± SD relative to vehicle, n = 3. (o) Carnitine palmitoyl transferase (CPT) gene expression in MEFs treated for 16 hrs with inhibitors, graphed as mean ± SD relative to vehicle, n = 6. Statistics: by one-way ANOVA (c, h–j, n), two-way ANOVA (d–f, k–m, o), and extra sum-of-squares F-test (A). n.s., not significant.

Source data

Extended Data Fig. 5 Changes in intracellular lipid species following mTORC1 inhibition require lysosomal function.

(a, b) Immunoblot (a) and fatty acids (b) corresponding from the experiment shown in Fig. 5a. Fatty acids quantification is graphed as mean ± SD relative to vehicle-treated cells, n = 3. (c, d) Immunoblots corresponding to the experiments shown in Fig. 5b, c. (e–g) Triglyceride and acyl-carnitine levels from the experiment shown in Fig. 5d–g. (h) Immunoblot for organelle markers in whole cell lysates or in HA-tag immunopurified lysosomes (IP) from HEK-293T cells expressing TMEM192-2xFLAG or 3xHA. (i) Immunoblot corresponding to the experiment shown in Fig. 5k, l. (j) Immunoblot corresponding to the experiment shown in Fig. 5m, n. LC3B is indicated in its unmodified (LC3B-I) and lipidated (LC3B-II) forms. Statistical analysis by two-way ANOVA (b, e–g). n.s., not significant.

Source data

Extended Data Fig. 6 Regulation of intracellular lipids by mTORC1 is inhibited by MAFP but not by loss of LPLA2.

(a–f) Lipid class sums (a–e) and glycerophosphocholine levels (f) detected in Tsc2−/− MEFs treated with vehicle, MAFP (a non-specific serine hydrolase inhibitor, 10 µM), and/or torin1 (250 nM) for 16 hrs, graphed as mean ± SD relative to vehicle-treated cells, n = 3. (g) Phospholipase A1 (PLA1) and A2 (PLA2) gene expression in Tsc2+/+ and Tsc2−/− MEFs treated for 16 hrs with vehicle, rapamycin (20 nM), or torin1 (250 nM), calculated relative to vehicle-treated Tsc2−/− MEFs, n = 3. cPLA2, cytosolic PLA2; iPLA2, calcium-independent PLA2; PA-PLA1, phosphatidic acid-preferring PLA1; PLB, phospholipase B. (h, i) Immunoblot (h) and TGs measured by pulse-chase with [1-14C]-oleate tracer (i) in parental and Pla2g15 knockout Tsc2−/− MEFs expressing enhanced green fluorescent protein (EGFP) or wild-type LPLA2 (PLA2G15) treated for 16 hrs with vehicle or torin1 (250 nM), graphed as mean ± SD relative to vehicle-treated cells, n = 3. Statistical analysis by two-way ANOVA (A-F,I). n.s., not significant.

Source data

Extended Data Fig. 7 TG and other lipid species accumulate independently of autophagy following mTORC1 inhibition.

(a, b) Immunoblot (a) and TGs measured by pulse-chase with [1-14C]-oleate tracer (b) in Atg5+/+ and −/− MEFs treated for 16 hrs with vehicle or BafA1 (250 nM), graphed as mean ± SD relative to vehicle-treated cells, n = 3. (c,d) Immunoblot (c) and triglyceride levels measured by enzyme assay (D) and normalized to protein content from the same sample for parental, ATG7 knockout, and FIP200 knockout HEK-293T cells treated with vehicle or bafilomycin A1 (250 nM) for 16 hrs, graphed as mean ± SD relative to vehicle-treated cells, n = 3. (e, f) Immunoblot (e) and triglyceride levels (f) corresponding to the results shown in Fig. 6e, f. Triglyceride levels are graphed as mean ± SD relative to vehicle-treated cells, n = 3. (g, h) Immunoblot (g) and triglyceride levels measured by enzyme assay (h) and normalized to protein content from the same sample for parental and FIP200 knockout HEK-293T cells treated with vehicle, torin1 (250 nM), and/or bafilomycin A1 (250 nM) for 16 hrs, graphed as mean ± SD relative to vehicle-treated cells, n = 3. (i) EGFR degradation assay in parental Tsc2−/− MEFs and sgUvrag or sgAtg14 clones. Cells were serum-starved for 16 hrs and stimulated with EGF (10 ng/mL) for the indicated times. (j) Immunoblot for autophagy markers in parental Tsc2−/− MEFs and sgAtg14 clone after 4 hrs in amino acid replete or free medium. Statistical analysis by two-way ANOVA (B,D,F,H). n.s., not significant.

Source data

Extended Data Fig. 8 TGs accumulate independently of TFEB and TFE3 following mTORC1 inhibition.

(a) Immunoblot for TFEB, TFE3, and proteins corresponding to their transcriptional target genes in Tsc2−/− MEFs treated for two days with control siRNA, siTfeb, or siTfe3. (b,c) 14C-TGs assayed by pulse-chase with [1-14C]-oleate tracer (6 h) in Tsc2−/− MEFs treated as in (A) followed by chase in cold medium for 16 hrs with vehicle or 250 nM torin1, graphed as mean ± SD relative to control siRNA-treated cells (b) and relative to vehicle-treated cells (C), n = 3. Statistical analysis by one-way ANOVA (b) and two-way ANOVA (c). n.s., not significant.

Source data

Extended Data Fig. 9 Induction of endosomal delivery to the lysosome is required for the changes in cellular lipid species following mTORC1 inhibition.

(a) Immunoblot corresponding to the experiment shown in Fig. 7a, b. (b–d) Immunoblots (b) and 14C-TGs (c, d) assayed by pulse-chase with [1-14C]-oleate tracer (6 h) in Tsc2−/− MEFs followed by chase in cold medium for 16 hrs with vehicle, 250 nM torin1, 5 µM VPS34-IN1, or 80 µM Dynasore, graphed as mean ± SD relative to vehicle-treated cells (c) and relative to no-torin1 controls (d), n = 3. (e) Immunoblot corresponding to the experiment shown in Fig. 7c. (f) Fatty acid accumulation measured by pulse-chase with [1-14C]-oleate tracer in the presence of DGAT inhibitors (3 µM each) and etomoxir (20 µM) in Tsc2−/− MEFs following a 4 hr treatment with endocytosis inhibitors (5 µM VPS34-IN1, 1 µM PIK-FYVEi, 80 µM Dynasore, or 20 µM PitStop 2). Graphed as mean ± SD relative to vehicle-treated cells at time 0, n = 3. (g, h) Binding of transferrin to Tsc2−/− MEFs (g) and PC3 (h) cells treated for 4 hrs with vehicle or torin1 (250 nM) and kept on ice while labeling with transferrin, confirming that the amount of plasma membrane transferrin receptor is not regulated by mTOR and that surface-bound transferrin is eliminated following an acid wash. Graphed as mean ± SD, n = 3. (i, j) Endocytosis assays in PC3 cells, measuring the uptake of transferrin (i) or 10 kDa dextran (j), following 4 hrs of vehicle, rapamycin (20 nM), or torin1 (250 nM) treatment, graphed as mean ± SD, n = 3. a.u., Arbitrary Units. Statistical analysis by one-way ANOVA (c, f, j), and two-way ANOVA (d, g–i). n.s., not significant.

Source data

Extended Data Fig. 10 EIPA and protease inhibitors serve as controls for BSA uptake and lysosomal cleavage assays.

(a) BSA uptake in Tsc2−/− MEFs treated with torin1 (250 nM) and vehicle or EIPA (20 µM) for 1 hr prior to labeling with 10 µg/mL BSA Alexa Fluor 647 (uptake, red) for 3 hrs. Nuclei are shown in blue. (b) BSA cleavage in Tsc2−/− MEFs treated with torin1 (250 nM) and vehicle or protease inhibitors (2 µM E64d, 2 µM Pepstatin A, and 10 µM Leupeptin) for 1 hr prior to labeling with 10 µg/mL each of BSA Alexa Fluor 647 (red, uptake) and DG-Green BSA (green, cleavage) for 3 hrs. Nuclei are shown in blue. (c, d) Quantification of the effect of EIPA on BSA uptake (c) and protease inhibitors on BSA cleavage (d) for cells treated as in (a, b), graphed as mean ± SD relative to vehicle-treated cells, n = 57–84 cells. (e) Co-localization of hydrolyzed DQ-Green BSA (green) and LysoTracker Deep Red (red) in Tsc2−/− MEFs pretreated with torin1 (250 nM) for 1 hr prior to labeling with 10 µg/mL DQ-Green BSA for 3 hrs. Nuclei are shown in blue. These images are representative of two replicate experiments in which similar results were obtained.

Source data

Supplementary information

Supplementary Information

Supplementary Methods, Supplementary Tables 1–4 and Supplementary Fig. 1.

Reporting Summary

Supplementary Data 1

Supplementary lipidomics data.

Supplementary Data 2

Supplementary lipidomics data.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed western blots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed western blots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hosios, A.M., Wilkinson, M.E., McNamara, M.C. et al. mTORC1 regulates a lysosome-dependent adaptive shift in intracellular lipid species. Nat Metab 4, 1792–1811 (2022). https://doi.org/10.1038/s42255-022-00706-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-022-00706-6

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