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:

HSF1 critically attunes proteotoxic stress sensing by mTORC1 to combat stress and promote growth

Abstract

To cope with proteotoxic stress, cells attenuate protein synthesis. However, the precise mechanisms underlying this fundamental adaptation remain poorly defined. Here we report that mTORC1 acts as an immediate cellular sensor of proteotoxic stress. Surprisingly, the multifaceted stress-responsive kinase JNK constitutively associates with mTORC1 under normal growth conditions. On activation by proteotoxic stress, JNK phosphorylates both RAPTOR at S863 and mTOR at S567, causing partial disintegration of mTORC1 and subsequent translation inhibition. Importantly, HSF1, the central player in the proteotoxic stress response (PSR), preserves mTORC1 integrity and function by inactivating JNK, independently of its canonical transcriptional action. Thereby, HSF1 translationally augments the PSR. Beyond promoting stress resistance, this intricate HSF1–JNK–mTORC1 interplay, strikingly, regulates cell, organ and body sizes. Thus, these results illuminate a unifying mechanism that controls stress adaptation and growth.

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

Figure 1: Proteotoxic stress activates JNK signalling but suppresses mTORC1 activity.
Figure 2: JNK negatively regulates mTORC1, translation and cell size.
Figure 3: JNK physically associates with mTORC1.
Figure 4: HSF1 maintains mTORC1 activity and integrity through inactivation and sequestration of JNK.
Figure 5: HSF1 suppresses JNK and activates mTORC1, independently of its transcriptional action.
Figure 6: mTORC1 translationally augments the PSR and promotes resistance to proteotoxic stress.
Figure 7: HSF1 positively regulates cell, organ and body sizes through suppression of JNK.
Figure 8: HSF1–JNK interactions regulate liver growth and proliferation.

Similar content being viewed by others

References

  1. Balch, W. E., Morimoto, R. I., Dillin, A. & Kelly, J. W. Adapting proteostasis for disease intervention. Science 319, 916–919 (2008).

    CAS  PubMed  Google Scholar 

  2. Lindquist, S. The heat-shock response. Annu. Rev. Biochem. 55, 1151–1191 (1986).

    CAS  PubMed  Google Scholar 

  3. Morimoto, R. I. The heat shock response: systems biology of proteotoxic stress in aging and disease. Cold Spring. Harb. Symp. Quant. Biol. 76, 91–99 (2011).

    CAS  PubMed  Google Scholar 

  4. Morimoto, R. I. Proteotoxic stress and inducible chaperone networks in neurodegenerative disease and aging. Genes Dev. 22, 1427–1438 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Dai, C., Dai, S. & Cao, J. Proteotoxic stress of cancer: implication of the heat-shock response in oncogenesis. J. Cell. Physiol. 227, 2982–2987 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Min, J. N., Huang, L., Zimonjic, D. B., Moskophidis, D. & Mivechi, N. F. Selective suppression of lymphomas by functional loss of Hsf1 in a p53-deficient mouse model for spontaneous tumors. Oncogene 26, 5086–5097 (2007).

    CAS  PubMed  Google Scholar 

  7. Dai, C., Whitesell, L., Rogers, A. B. & Lindquist, S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 130, 1005–1018 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Meng, L., Gabai, V. L. & Sherman, M. Y. Heat-shock transcription factor HSF1 has a critical role in human epidermal growth factor receptor-2-induced cellular transformation and tumorigenesis. Oncogene 29, 5204–5213 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Jin, X., Moskophidis, D. & Mivechi, N. F. Heat shock transcription factor 1 is a key determinant of HCC development by regulating hepatic steatosis and metabolic syndrome. Cell Metab. 14, 91–103 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Dai, C. et al. Loss of tumor suppressor NF1 activates HSF1 to promote carcinogenesis. J. Clin. Invest. 122, 3742–3754 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Dai, S. et al. Suppression of the HSF1-mediated proteotoxic stress response by the metabolic stress sensor AMPK. EMBO J. 34, 275–293 (2015).

    CAS  PubMed  Google Scholar 

  12. Tang, Z. et al. MEK guards proteome stability and inhibits tumor-suppressive amyloidogenesis via HSF1. Cell 160, 729–744 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Lindquist, S. Regulation of protein synthesis during heat shock. Nature 293, 311–314 (1981).

    CAS  PubMed  Google Scholar 

  14. Laplante, M. & Sabatini, D. M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Inoki, K., Zhu, T. & Guan, K. L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).

    CAS  PubMed  Google Scholar 

  16. Gwinn, D. M. et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 30, 214–226 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Sancak, Y. et al. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320, 1496–1501 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Derijard, B. et al. JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell 76, 1025–1037 (1994).

    CAS  PubMed  Google Scholar 

  20. Kawaguchi, Y. et al. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115, 727–738 (2003).

    CAS  PubMed  Google Scholar 

  21. Leu, J. I., Pimkina, J., Frank, A., Murphy, M. E. & George, D. L. A small molecule inhibitor of inducible heat shock protein 70. Mol. Cell 36, 15–27 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Massey, A. J. et al. A novel, small molecule inhibitor of Hsc70/Hsp70 potentiates Hsp90 inhibitor induced apoptosis in HCT116 colon carcinoma cells. Cancer Chemother. Pharmacol. 66, 535–545 (2010).

    CAS  PubMed  Google Scholar 

  23. Pickart, C. M. & Eddins, M. J. Ubiquitin: structures, functions, mechanisms. Biochim. Biophys. Acta 1695, 55–72 (2004).

    CAS  PubMed  Google Scholar 

  24. Zhang, T. et al. Discovery of potent and selective covalent inhibitors of JNK. Chem. Biol. 19, 140–154 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Manning, A. M. & Davis, R. J. Targeting JNK for therapeutic benefit: from junk to gold? Nat. Rev. Drug Discov. 2, 554–565 (2003).

    CAS  PubMed  Google Scholar 

  26. Kyriakis, J. M. & Avruch, J. Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update. Phys. Rev. 92, 689–737 (2012).

    CAS  Google Scholar 

  27. Geiger, P. C., Wright, D. C., Han, D. H. & Holloszy, J. O. Activation of p38 MAP kinase enhances sensitivity of muscle glucose transport to insulin. Am. J. Physiol. Endocrinol. Metab. 288, E782–E788 (2005).

    CAS  PubMed  Google Scholar 

  28. Kim, D. H. et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 110, 163–175 (2002).

    CAS  PubMed  Google Scholar 

  29. Hara, K. et al. Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell 110, 177–189 (2002).

    CAS  PubMed  Google Scholar 

  30. Soliman, G. A. et al. mTOR Ser-2481 autophosphorylation monitors mTORC-specific catalytic activity and clarifies rapamycin mechanism of action. J. Biol. Chem. 285, 7866–7879 (2010).

    CAS  PubMed  Google Scholar 

  31. Gupta, S. et al. Selective interaction of JNK protein kinase isoforms with transcription factors. EMBO J. 15, 2760–2770 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lei, K. et al. The Bax subfamily of Bcl2-related proteins is essential for apoptotic signal transduction by c-Jun NH(2)-terminal kinase. Mol. Cell. Biol. 22, 4929–4942 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kim, J., Kundu, M., Viollet, B. & Guan, K. L. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat. Cell Biol. 13, 132–141 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Liu, J., Xu, Y., Stoleru, D. & Salic, A. Imaging protein synthesis in cells and tissues with an alkyne analog of puromycin. Proc. Natl Acad. Sci. USA 109, 413–418 (2012).

    CAS  PubMed  Google Scholar 

  35. Fingar, D. C., Salama, S., Tsou, C., Harlow, E. & Blenis, J. Mammalian cell size is controlled by mTOR and its downstream targets S6K1 and 4EBP1/eIF4E. Genes Dev. 16, 1472–1487 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Um, S. H., D’Alessio, D. & Thomas, G. Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1. Cell Metab. 3, 393–402 (2006).

    CAS  PubMed  Google Scholar 

  37. Hirosumi, J. et al. A central role for JNK in obesity and insulin resistance. Nature 420, 333–336 (2002).

    CAS  PubMed  Google Scholar 

  38. Clausson, C. M. et al. Increasing the dynamic range of in situ PLA. Nat. Methods 8, 892–893 (2011).

    CAS  PubMed  Google Scholar 

  39. Rosner, M. & Hengstschlager, M. Cytoplasmic and nuclear distribution of the protein complexes mTORC1 and mTORC2: rapamycin triggers dephosphorylation and delocalization of the mTORC2 components rictor and sin1. Hum. Mol. Genet. 17, 2934–2948 (2008).

    CAS  PubMed  Google Scholar 

  40. Bogoyevitch, M. A. & Kobe, B. Uses for JNK: the many and varied substrates of the c-Jun N-terminal kinases. Microbiol. Mol. Biol. Rev. 70, 1061–1095 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Qian, S. B. et al. mTORC1 links protein quality and quantity control by sensing chaperone availability. J. Biol. Chem. 285, 27385–27395 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Jez, J. M., Chen, J. C., Rastelli, G., Stroud, R. M. & Santi, D. V. Crystal structure and molecular modeling of 17-DMAG in complex with human Hsp90. Chem. Biol. 10, 361–368 (2003).

    CAS  PubMed  Google Scholar 

  43. Jewell, J. L., Russell, R. C. & Guan, K. L. Amino acid signalling upstream of mTOR. Nat. Rev. Mol. Cell. Biol. 14, 133–139 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Xiao, X. et al. HSF1 is required for extra-embryonic development, postnatal growth and protection during inflammatory responses in mice. EMBO J. 18, 5943–5952 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Jorgensen, P. & Tyers, M. How cells coordinate growth and division. Curr. Biol. 14, R1014–R1027 (2004).

    CAS  PubMed  Google Scholar 

  46. Eilers, M. & Eisenman, R. N. Myc’s broad reach. Genes Dev. 22, 2755–2766 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Freeman, A. et al. Improved detection of hepatocyte proliferation using antibody to the pre-replication complex: an association with hepatic fibrosis and viral replication in chronic hepatitis C virus infection. J. Viral Hepat. 10, 345–350 (2003).

    CAS  PubMed  Google Scholar 

  48. Goyal, L., Muzumdar, M. D. & Zhu, A. X. Targeting the HGF/c-MET pathway in hepatocellular carcinoma. Clin. Cancer Res. 19, 2310–2318 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Sancak, Y. et al. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141, 290–303 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Betz, C. & Hall, M. N. Where is mTOR and what is it doing there? J. Cell. Biol. 203, 563–574 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Cui, H. et al. Enhanced expression of asparagine synthetase under glucose-deprived conditions protects pancreatic cancer cells from apoptosis induced by glucose deprivation and cisplatin. Cancer Res. 67, 3345–3355 (2007).

    CAS  PubMed  Google Scholar 

  52. Chaveroux, C. et al. Identification of a novel amino acid response pathway triggering ATF2 phosphorylation in mammals. Mol. Cell. Biol. 29, 6515–6526 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Taipale, M., Jarosz, D. F. & Lindquist, S. HSP90 at the hub of protein homeostasis: emerging mechanistic insights. Nat. Rev. Mol. Cell. Biol. 11, 515–528 (2010).

    CAS  PubMed  Google Scholar 

  54. Trepel, J., Mollapour, M., Giaccone, G. & Neckers, L. Targeting the dynamic HSP90 complex in cancer. Nat. Rev. Cancer 10, 537–549 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhang, H. et al. Hsp90/p50cdc37 is required for mixed-lineage kinase (MLK) 3 signaling. J. Biol. Chem. 279, 19457–19463 (2004).

    CAS  PubMed  Google Scholar 

  56. Chen, J., Miller, E. M. & Gallo, K. A. MLK3 is critical for breast cancer cell migration and promotes a malignant phenotype in mammary epithelial cells. Oncogene 29, 4399–4411 (2010).

    CAS  PubMed  Google Scholar 

  57. Dong, C. et al. Defective T cell differentiation in the absence of Jnk1. Science 282, 2092–2095 (1998).

    CAS  PubMed  Google Scholar 

  58. Yang, D. D. et al. Differentiation of CD4 + T cells to Th1 cells requires MAP kinase JNK2. Immunity 9, 575–585 (1998).

    CAS  PubMed  Google Scholar 

  59. Ventura, A. et al. Restoration of p53 function leads to tumour regression in vivo. Nature 445, 661–665 (2007).

    CAS  PubMed  Google Scholar 

  60. Postic, C. et al. Dual roles for glucokinase in glucose homeostasis as determined by liver and pancreatic beta cell-specific gene knock-outs using Cre recombinase. J. Biol. Chem. 274, 305–315 (1999).

    CAS  PubMed  Google Scholar 

  61. Kwiatkowski, D. J. et al. A mouse model of TSC1 reveals sex-dependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells. Hum. Mol. Genet. 11, 525–534 (2002).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We would like to thank members of the Dai laboratory for discussions and technical assistance. This work was supported by a grant from the NIH (1DP1OD006438) to I.J.B.; and The Jackson Laboratory Cancer Center Support Grant (3P30CA034196), and grants from the NIH (1DP2OD007070) and the Ellison Medical Foundation (AS-NS-0599-09) to C.D.

Author information

Authors and Affiliations

Authors

Contributions

K.-H.S., J.C., Z.T. and S.D. designed and performed the experiments. Y.H. performed statistical analyses and generated graphs. I.J.B. provided mice and cell lines and actively engaged in discussions. C.D. conceived and oversaw this study. S.B.S. and C.D. wrote the manuscript.

Corresponding author

Correspondence to Chengkai Dai.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Perturbation of proteostasis by proteotoxic stressors, related to Fig. 1.

(a) Proteotoxic stressors perturb proteostasis via diverse mechanisms of action. Ub: ubiquitin. (b) HEK293T cells were treated with diverse proteotoxic stressors at the concentrations described in Fig. 1B and 1C for 4 and 8 h. Cell viability was measured by a Guava EasyCyte cytometer using ViaCount reagents (mean of 4 wells of cells per group from one experiment). (c) HEK293T cells were treated with 500 nM MG132 for indicated time. JNK and p38 MAPK phosphorylation were detected by immunoblotting. (d) Following pre-incubation with 10 μM SB202190 for 30 min, HEK293T cells were treated with 500 nM MG132 for indicated time. Phosphorylation of p38 MAPK and mTORC1 signalling were detected by immunoblotting. Uncropped images of blots are shown in Supplementary Fig. 8. Source data for Supplementary Fig. 1b can be found in Supplementary Table 1.

Supplementary Figure 2 JNK is a negative regulator of mTORC1 signalling, translation, and cell size, related to Fig. 2.

(a) HEK293T cells stably transduced with lentiviral scramble or JNK1/2-targeting shRNAs were treated with and without 500 nM MG132 for 4 h. Phosphorylated and total JNK1/2 levels were detected by immunoblotting. (b) Following pre-incubation with 3 μM JNK-IN-8 for 60 min, HEK293T cells were treated with and without 500 nM MG132 for 4 h. Endogenous mTOR-RAPTOR associations and mTORC1 kinase activity were measured by coIP. (c) and (d) Primary MEFs were treated with 200 nM MG132 for 6 h, and cell viability was measured using ViaCount reagents (mean of6 wells of cells per group from one experiment). (e) Following transfection with 10 nM three independent MKK7-targetting siRNAs for 4 days, MKK7 and phosphorylated S6K proteins were detected by immunoblotting in HEK293T cells. (f) Following pre-incubation with 3 μM JNK-IN-8 for 60 min, phosphorylated S6K proteins were detected by immunoblotting in HEK293T cells transfected with LacZ, JNK1CA, or JNK2A2 plasmid. (g) Endogenous mTOR-RAPTOR associations were detected by coIP in HEK293T cells transfected with LacZ, JNK1CA, or JNK2A2 plasmid. (h) Following 1 μM AZD8055 treatment overnight, HEK293T cells were labelled with 6-FAM-dc-puromycin for 30 min and analysed by flow cytometry. Unlabelled cells served as negative controls. (i) Following transfection with JNK1/2-targeting siRNAs (A: siJNK1_2 and siJNK2_2; B: siJNK1_3 and siJNK2_3), total JNK1/2 levels were detected by immunoblotting. (j) Following transfection with indicated plasmids, HeLa cells were treated with DMSO or 1 μM AZD8055 for 2 days and analysed for cell size using a Multisizer 3 Coulter Counter. Changes in cell size distribution, compared to the LacZ control, are statistically significant (Kolmogorov-Smirnov test, P < 0.01). (k) and (l) JNK and mTORC1 signalling were detected by immunoblotting in mouse skin and kidneys. (m)–(o) Whole-body weight and composition were measured in 6-week-old male mice (mean ± s.d., n = 5, 5, or 6 mice per genotype, One-way ANOVA). Statistical significance: P < 0.05; P < 0.01; P < 0.001; n.s.: not significant. Uncropped images of blots are shown in Supplementary Fig. 8. Source data for Supplementary Fig. 2c, d can be found in Supplementary Table 1.

Supplementary Figure 3 JNK physically interacts with RAPTOR and mTOR, related to Fig. 3.

(a)–(c) Validations of mouse monoclonal anti-JNK1 (JM2671), rabbit monoclonal anti-mTOR (7C10), and mouse monoclonal anti-RAPTOR (1H6.2) antibodies by immunostaining in HeLa cells stably expressing shRNAs. Scale bars: 50 μm. Images are representative of 2 independent experiments. (d) Endogenous mTORC1 was precipitated using anti-RAPTOR antibodies from HEK293T cells. Precipitates were resolved on SDS-PAGE under non-reducing conditions. Precipitated mTORC1 components were visualized by silver staining (Silver Stain Kit, Thermo Fisher Scientific). To avoid obscuring of mTOR by the staining of abundant whole IgG, the portion containing IgG was separated from the rest of the gel and stained for less time. The rest of the gel was silver-stained for the same period of time to reveal both mTOR and JNK proteins. The p46 isoforms of JNK proteins could not be clearly identified due to the obscuration by IgG heavy chains (HC). MK: molecular weight marker. The staining intensities of mTOR and JNK proteins were quantitated by ImageJ software, and normalized against their total numbers of amino acids. (e) The consensus JNK phosphorylation sequence is derived from curated JNK substrates (www.phosphosite.org) and plotted using the online WebLoGo software (www.weblogo.berkeley.edu). Predicted JNK phosphorylation sites, RAPTOR Ser863 and mTOR Ser567, are highlighted in red. (f) Endogenous mTORC1 complexes were immunoprecipitated from HEK293T cells. Immediately following addition of 100 ng recombinant GST or GST-JNK1 proteins to equal amounts of precipitates, indicated concentrations of JNK-IN-8 (JIN8) or AZD8055 (AZD) were added to the kinase mixtures and incubated at 30 °C for 30 min for in vitro JNK kinase assays. (g) and (h) Following treatment with 500 nM MG132 for 4 h, endogenous RAPTOR and mTOR proteins were precipitated from HEK293T cells stably expressing scramble or JNK1/2-targeting shRNAs. RAPTOR Ser863 and mTOR Ser567 phosphorylation was detected by immunoblotting. (i) and (j) HEK293T cells depleted of endogenous RAPTOR or mTOR proteins due to stably shRNA expression were transfected with indicated plasmids and treated with DMSO or 500 nM MG132 for 4 h. JNK and mTORC1 signalling were detected by immunoblotting.

Supplementary Figure 4 HSF1 suppresses JNK activation and sustains mTORC1 signalling, related to Figs 4 and 5.

(a) Schematic depiction of the generation of conditional Hsf1fl/fl alleles on the C57BL/6J genetic background. Expression of Cre recombinase results in deletion of exon2-9 (Δ2-9) of the Hsf1 gene. (b) Cells were treated as described in Fig. 4A. Cell viability was measured by a Guava EasyCyte cytometer using ViaCount reagents (mean of4 or 5 wells of cells per group from one experiment). (c) Validations of rabbit polyclonal anti-JNK1/3 (C-17) and mouse monoclonal anti-HSF1 (E-4) antibodies by immunostaining in primary Jnk1+/+ and Jnk1−/− MEFs (left panel), and in immortalized Rosa26-CreERT2; Hsf1fl/fl MEFs treated with 1 μM 4-OHT or EtOH for 7 days (right panel). Scale bars: 50 μm. (d) Endogenous HSF1-JNK1 interactions were detected by PLA in immortalized Rosa26-CreERT2; Hsf1fl/fl MEFs treated with and without 1 μM 4-OHT for 7 days, using mouse anti-HSF1 (E-4) and rabbit anti-JNK1/3 (C-17) antibodies. Scale bars: 10 μm. (e) HEK293T cells were transfected with LacZ or FLAG-tagged HSF1 plasmids. Following IP of endogenous JNK1 proteins, co-precipitated FLAG-tagged HSF1 and endogenous RAPTOR proteins were immunoblotted. (f) Schematic depiction of the proposed sequestration of JNK apart from mTORC1 by HSF1. Uncropped images of blots are shown in Supplementary Fig. 8. Source data for Supplementary Fig. 4b can be found in Supplementary Table 1.

Supplementary Figure 5 mTORC1 inhibition suppresses the PSR, related to Fig. 6.

HEK293T cells were treated with the mTORC1 inhibitor rapamycin (100 nM) or AZD8055 (1 μM) overnight. Translation rates of HSF1 were measured by puromycin labelling as described in Fig. 5b (mean of 6 wells of cells per group per experiment, and this experiment was repeated twice). Source data for Supplementary Fig. 5 can be found in Supplementary Table 1.

Supplementary Figure 6 HSF1 is a positive regulator of cell, organ, and body sizes, related to Fig. 7.

(a) and (b) HEK293T cells were co-transfected with indicated shRNAs and siRNAs. HSF1 and JNK1/2 levels were detected by immunoblotting (a), and protein synthesis rates were measured by puromycin labelling (b). (c) Livers of female mice were weighed at 6 weeks of age (mean ± s.d., n = 6, 7, or 8 mice per genotype, One-way ANOVA). (d) HSP90α, HSP72, and HSP25 proteins were detected by immunoblotting in mouse livers. (e) and (f) Kidneys of male mice were weighed at 6 weeks of age (mean ± s.d., n = 4 mice per genotype, One-way ANOVA). (g) Digital photos of female mice at 6 weeks of age were taken and body surface areas were quantitated by ImageJ software (mean ± s.d., n = 3 mice per genotype, One-way ANOVA). (h) and (i) Body weights were measured in female mice at 6 weeks of age (mean ± s.d., n = 6, 13, 16, or 34 mice per genotype, One-way ANOVA). (j) and (k) Whole-body composition was measured in female mice at 6 weeks of age (mean ± s.d., n = 6, 7, or 18 mice per genotype, One-way ANOVA). Statistical significance: P < 0.01; P < 0.001. Uncropped images of blots are shown in Supplementary Fig. 8. Statistics source data for Supplementary Fig. 6f, g can be found in Supplementary Table 1.

Supplementary Figure 7 HSF1-JNK interactions regulate liver growth and proliferation, related to Fig. 8.

(a) and (b) Liver weights and lean mass of 6-week-old female mice were measured (mean ± s.d., n = number of mice per genotype shown in the graphs, One-way ANOVA). (c) Quantitation of mTORC1 signalling in mouse liver tissues shown in Fig. 7e (mean ± s.d., n = 3 independent experiments, One-way ANOVA). (d) Representative flow cytometry dot plots of co-stained liver cells with PI and anti-BrdU antibodies. (e) and (f) MCM2 and c-MYC levels were detected in mouse liver tissues by immunoblotting. (g) Caspase 3 activities were quantitated in liver tissues of 6-week-old male mice using DEVD-R110 as the substrate (mean ± s.d., n = 3 mice per genotype, One-way ANOVA). This experiment was repeated twice. (h) Hepatocyte-specific deletion of Hsf1 was verified by immunostaining of frozen sections of mouse liver tissues using rat monoclonal anti-HSF1 antibody cocktail (1:100) and CF594 anti-rat IgG secondary antibody conjugates. Arrows indicate hepatocytes and arrowheads indicate non-parenchymal cells. Of note, HSF1 expression was still detected in non-parenchymal cells following Cre-mediated recombination. Scale bars: 50 μm. Representative images from 2 independent experiments. (i) HgfmRNA levels were quantitated by qRT-PCR in primary Jnk1MEFs (mean of 3 wells of cells per genotype per experiment, and this experiment was repeated twice). (j) Hsp mRNA levels were quantitated by qRT-PCR in primary Jnk1 MEFs (mean, of 3 wells of cells per genotype per experiment, and this experiment was repeated twice). (k) HgfmRNA levels were quantitated by qRT-PCR in mouse liver tissues (mean ± s.d., n = 3 mice per genotype, One-way ANOVA). This experiment was repeated twice. (l) and (m) Phosphorylation of c-MET was detected by immunoblotting in mouse liver tissues. Statistical significance: P < 0.05; P < 0.01; P < 0.001; n.s.: not significant. Uncropped images of blots are shown in Supplementary Fig. 8. Statistics source data for Supplementary Fig. 7c, g, i–k can be found in Supplementary Table 1.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2709 kb)

Supplementary Table 1

Supplementary Information (XLSX 65 kb)

Supplementary Table 2

Supplementary Information (XLSX 32 kb)

Supplementary Table 3

Supplementary Information (XLSX 21 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Su, KH., Cao, J., Tang, Z. et al. HSF1 critically attunes proteotoxic stress sensing by mTORC1 to combat stress and promote growth. Nat Cell Biol 18, 527–539 (2016). https://doi.org/10.1038/ncb3335

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3335

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