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.

mTORC1 directly inhibits AMPK to promote cell proliferation under nutrient stress

Abstract

Highly conserved signalling pathways controlled by mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) are central to cellular metabolism and cell proliferation1,2, and their dysregulation is implicated in the pathogenesis of major human diseases such as cancer and type 2 diabetes. AMPK pathways leading to reduced cell proliferation are well established and, in part, act through inhibition of TOR complex 1 (TORC1) activity. Here we demonstrate reciprocal regulation, specifically that TORC1 directly downregulates AMPK signalling by phosphorylating the evolutionarily conserved residue S367 in the fission yeast AMPK catalytic subunit Ssp2 and AMPK α1 S347 and α2 S345 in the mammalian homologs, which is associated with reduced phosphorylation of activation loop T172. Genetic or pharmacological inhibition of TORC1 signalling led to AMPK activation in the absence of increased AMP/ATP ratios, which under nutrient stress conditions was associated with growth limitation in both yeast and human cell cultures. Our findings reveal fundamental bidirectional regulation between two major metabolic signalling networks and uncover new opportunities for cancer treatment strategies aimed at suppressing cell proliferation in the nutrient-poor tumour microenvironment.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Ssp2 S367A is associated with increased activating phosphorylation and reduced cell growth under nutrient stress.
Fig. 2: TORC1-dependent regulation of Ssp2 S367 phosphorylation.
Fig. 3: mTORC1 phosphorylation of mammalian AMPK α subunit at S345 negatively regulates cellular phosphorylation at T172 and AMPK signalling.
Fig. 4: α2 S345 phosphorylation promotes cell proliferation under conditions of nutrient stress.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. Source data for Figs. 14 and Extended Data Figs. 110 are presented with the paper.

References

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

    Article  CAS  Google Scholar 

  2. Hardie, D. G. AMPK—sensing energy while talking to other signaling pathways. Cell Metab. 20, 939–952 (2014).

    Article  CAS  Google Scholar 

  3. Oakhill, J. S., Scott, J. W. & Kemp, B. E. AMPK functions as an adenylate charge-regulated protein kinase. Trends Endocrinol. Metab. 23, 125–132 (2012).

    Article  CAS  Google Scholar 

  4. Zhang, S.-C. et al. The lysosomal v-ATPase–Ragulator complex is a common activator for AMPK and mTORC1, acting as a switch between catabolism and anabolism. Cell Metab. 20, 526–540 (2014).

    Article  CAS  Google Scholar 

  5. Hanyu, Y. et al. Schizosaccharomyces pombe cell division cycle under limited glucose requires Ssp1 kinase, the putative CaMKK, and Sds23, a PP2A-related phosphatase inhibitor. Genes Cells 14, 539–554 (2009).

    Article  CAS  Google Scholar 

  6. Davie, E., Forte, G. M. & Petersen, J. Nitrogen regulates AMPK to control TORC1 signalling. Curr. Biol. 25, 445–454 (2015).

    Article  CAS  Google Scholar 

  7. Koch, A., Krug, K., Pengelley, S., Macek, B. & Hauf, S. Mitotic substrates of the kinase Aurora with roles in chromatin regulation identified through quantitative phosphoproteomics of fission yeast. Sci. Signal. 4, rs6 (2011).

    Article  CAS  Google Scholar 

  8. Wilson-Grady, J. T., Villén, J. & Gygi, S. P. Phosphoproteome analysis of fission yeast. J. Proteome Res. 7, 1088–1097 (2008).

    Article  CAS  Google Scholar 

  9. Wissing, J. et al. Proteomics analysis of protein kinases by target class-selective prefractionation and tandem mass spectrometry. Mol. Cell. Proteom. 6, 537–547 (2007).

    Article  CAS  Google Scholar 

  10. Parker, B. L. et al. Targeted phosphoproteomics of insulin signaling using data-independent acquisition mass spectrometry. Sci. Signal. 8, rs6 (2015).

    Article  Google Scholar 

  11. Chen, L. et al. Conserved regulatory elements in AMPK. Nature 472, 230–233 (2011).

    Article  Google Scholar 

  12. Xiao, B. et al. Structure of mammalian AMPK and its regulation by ADP. Nature 472, 230–233 (2011).

    Article  CAS  Google Scholar 

  13. Petersen, J. & Nurse, P. TOR signalling regulates mitotic commitment through the stress MAP kinase pathway and the Polo and Cdc2 kinases. Nat. Cell Biol. 9, 1263–1272 (2007).

    Article  CAS  Google Scholar 

  14. Matsuzawa, T., Fujita, Y., Tohda, H. & Takegawa, K. Snf1-like protein kinase Ssp2 regulates glucose derepression in Schizosaccharomyces pombe. Eukaryot. Cell 11, 159–167 (2012).

    Article  CAS  Google Scholar 

  15. Du, W., Hálová, L., Kirkham, S., Atkin, J. & Petersen, J. TORC2 and the AGC kinase Gad8 regulate phosphorylation of the ribosomal protein S6 in fission yeast. Biol. Open 1, 884–888 (2012).

    Article  CAS  Google Scholar 

  16. Álvarez, B. & Moreno, S. Fission yeast Tor2 promotes cell growth and represses cell differentiation. J. Cell Sci. 119, 4475–4485 (2005).

    Article  Google Scholar 

  17. Wolfson, R. L. & Sabatini, D. M. The dawn of the age of amino acid sensors for the mTORC1 pathway. Cell Metab. 26, 301–309 (2017).

    Article  CAS  Google Scholar 

  18. Dite, T. A. et al. The autophagy initiator ULK1 sensitizes AMPK to allosteric drugs. Nat. Commun. 18, 571 (2017).

    Article  Google Scholar 

  19. Cybulski, N., Zinzalla, V. & Hall, M. N. Inducible raptor and rictor knockout mouse embryonic fibroblasts. Methods Mol. Biol. 821, 267–278 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  21. de Souza Almeida Matos, A. L. et al. Allosteric regulation of AMP-activated protein kinase by adenylate nucleotides and small-molecule drugs. Biochem. Soc. Trans. 30, 733–741 (2019).

    Article  Google Scholar 

  22. Liu, X. et al. Discrete mechansism of mTOR and cell cycle regulation by AMPK agonists independent of AMPK. Proc. Natl Acad. Sci. USA 111, E435–E444 (2013).

    Article  Google Scholar 

  23. Hülsmann, H. J. et al. Activation of AMP-activated protein kinase sensitizes lung cancer cells and H1299 xenografts to erlotinib. Lung Cancer 86, 151–157 (2014).

    Article  Google Scholar 

  24. Lopez-Mejia, I. C. et al. CDK4 phosphorylates AMPKα2 to inhibit its activity and repress fatty acid oxidation. Mol. Cell 68, 336–349 (2017).

    Article  CAS  Google Scholar 

  25. Zhang, C.-S. et al. Fructose-1,6-bisphophate and aldolase mediate glucose sensing by AMPK. Nature 548, 112–116 (2017).

    Article  CAS  Google Scholar 

  26. Barban, S. & Schulze, H. O. The effects of 2-deoxyglucose on the growth and metabolism of cultured cells. J. Biol. Chem. 236, 1887–1890 (1961).

    CAS  PubMed  Google Scholar 

  27. Egan, D. F. et al. Small molecule inhibition of the autophagy kinase ULK1 and identification of ULK1 substrates. Mol. Cell 59, 285–297 (2015).

    Article  CAS  Google Scholar 

  28. Dite, T. A. et al. AMP-activated protein kinase selectively inhibited by the type II inhibitor SBI-0206965. J. Biol. Chem. 293, 8874–8885 (2018).

    Article  CAS  Google Scholar 

  29. Jeon, S.-M. Regulation and function of AMPK in physiology and diseases. Exp. Mol. Med. 48, e245 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  31. Fantes, P. A. Control of cell size and cycle time in Schizosaccharomyces pombe. J. Cell Sci. 24, 51–67 (1977).

    CAS  PubMed  Google Scholar 

  32. Petersen, J. & Russell, P. Growth and the environment of Schizosaccharomyces pombe. Cold Spring Harb. Protoc. 2016, pdb.top079764 (2016).

    Article  Google Scholar 

  33. Caspari, T. et al. Characterization of Schizosaccharomyces pombe Hus1: a PCNA-related protein that associates with Rad1 and Rad9. Mol. Cell. Biol. 20, 1254–1262 (2000).

    Article  CAS  Google Scholar 

  34. Oakhill, J. S. et al. β-Subunit myristoylation is the gatekeeper for initiating metabolic stress sensing by AMP-activated protein kinase (AMPK). Proc. Natl Acad. Sci. USA 107, 19237–19241 (2010).

    Article  CAS  Google Scholar 

  35. Langendorf, C. G. et al. Structural basis of allosteric and synergistic activation of AMPK by furan-2-phosphonic derivative C2 binding. Nat. Commun. 7, 10912 (2016).

    Article  CAS  Google Scholar 

  36. Smiles, W. J. et al. Modulation of autophagy signaling with resistance exercise and protein ingestion following short-term energy deficit. Am. J. Physiol. Regul. Integr. Comp. Physiol. 309, R603–R612 (2015).

    Article  CAS  Google Scholar 

  37. Scott, J. W. et al. Small molecule drug A-769662 and AMP synergistically activate naive AMPK independent of upstream kinase signaling. Chem. Biol. 21, 619–627 (2014).

    Article  CAS  Google Scholar 

  38. Scott, J. W. et al. Thienopyridone drugs are selective activators of AMP-activated protein kinase β1-containing complexes. Chem. Biol. 15, 1220–1230 (2008).

    Article  CAS  Google Scholar 

  39. Scott, J. W. et al. Inhibition of AMP-activated protein kinase at the allosteric drug-binding site promotes islet insulin release. Chem. Biol. 22, 705–711 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank K. Gull (Oxford University, UK) and S. Lim (St. Vincent’s Institute, Australia) for antibodies, M. Balasubramanian (Warwick University, UK), K. Shiozaki (Nara University, Japan) and S. Moreno (IBFG Salamanca, Spain) for yeast strains, M. Hall (University of Basel, Switzerland) for iRapKO MEFs, I. Hagan for stimulating discussions and C. Proud and J. Murphy for critical evaluation of the manuscript. C.G.L. was supported by an Early Career Fellowship from the National Health and Medical Research Council (NHMRC; 1143080). J.S.O. was supported by a Future Fellowship from the Australian Research Council (ARC; FT130100988), the NHMRC (1098459), St Vincent’s Institute of Medical Research (Australia) and in part by the Victorian Government’s Operational Infrastructure Support Program. J.P. was supported by a Cancer Research UK senior fellowship (C10888/A11178), Cancer Council Australia (1125662), Worldwide Cancer Research (16-0052), the NHMRC (1161262), the ARC (DP180101682), a Flinders Foundation seeding grant, Manchester (UK), and Flinders University (Australia).

Author information

Authors and Affiliations

Authors

Contributions

N.X.Y.L., A.K., A.H., E.D., K.R.W.N., K.R.M., W.J.S., G.M.F., T.W., S.L., T.A.D., J.S.O. and J.P. performed the experiments. C.G.L. and J.W.S. provided reagents and intellectual input. J.S.O. and J.P. designed and coordinated the study and wrote the manuscript. All authors read and approved the manuscript.

Corresponding authors

Correspondence to Jonathan S. Oakhill or Janni Petersen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Primary Handling Editors: Ana Mateus; Christoph Schmitt.

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

Extended data

Extended Data Fig. 1 Analysis of S. pombe Ssp2-S367A/D KI mutants and validation of S. pombe temperature-sensitive TORC1 expression mutant tor2.ts.

Ssp2 dependent phosphorylation of the Scr1 transcription factor is elevated in S. pombe expressing Ssp2-S367A mutant. a) Lysates were prepared from WT S. pombe and indicated ssp2 mutants in which Scr1 was tagged with GPF and immunoblotted for anti-GFP, Ponceau S staining shows total protein. Error bars, mean Ssp2 specific phosphorylation (signal absent in the kinase inactive Ssp2-T189A mutant) vs. WT ± s.e.m., n = 4. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. b) Lysates from WT S. pombe and and S367A/D mutants, treated as indicated, were prepared and immunoblotted for Maf1-PK. Increased hypo-phosphorylation is indicated by lower arrow. Error bars, mean fold change in phosphorylation ± s.e.m., n = 3. Statistical significance was calculated using by two-way ANOVA with Sidak’s multiple comparisons test. n represent independent experiments. c) The yeast S6 ribosomal protein Rps6 is not phosphorylated in the temperature-sensitive TORC1 mutant tor2.ts at restricted temperature or when torin1 is added to WT cells. Lysates were prepared from WT S. pombe and indicated mutants, under conditions indicated, and immunoblotted for pRps6 and total Rps6. Error bars, mean fold change in pRps6 ± s.e.m., n = 3 independent experiments. Statistical significance was calculated using by one-way ANOVA with Sidak’s multiple comparisons test. d) Heat stress of wild type cells at 37 ˚C for 3 h does not affect phosphorylation of Ssp2-S367. Lysates were prepared from WT S. pombe and immunoblotted for pS367 and total Ssp2. e) Lysates were prepared from WT S. pombe and indicated mutants and immunoblotted for Maf1 with anti-PK antibodies. MAF1 hypo-phosphorylation is indicated by lower arrow. For d, e) similar results were obtained from 3 independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 2 AMPK α2-S345 is basally phosphorylated in a variety of mammalian cell lines and tissues and is an mTORC1 substrate using purified enzymes.

a) Validation of the AMPK α2-S345 phospho-specific antibody. Lysates were prepared from FLAG-α2- (WT and S345A/E mutants) expressing α1-/-/α2-/- MEFs and immunoblotted as indicated. α2-S345 is phosphorylated under nutrient-replete/basal conditions in b) mouse liver and a range of mammalian cell lines (negative control: α2β2γ1 expressed and purified from E. coli, which is not phosphorylated on α2-S345), c) human vastus lateralis skeletal muscle, and d) all GST-fusion α2 AMPK complexes expressed in COS7 mammalian cells. e) mTORC1 phosphorylates α-S345 on purified, bacterial expressed recombinant AMPK (α1β2γ1 and α2β1γ1). Kinase inactive (KI) AMPK was used as the substrate to exclude autophosphorylation. f) LC-MS/MS analysis of mTORC1 treated KIα2β1γ1. The masses of the b and y ion series provide direct evidence for phosphate incorporation onto α2-S345. M(O): oxidised methionine; pS: pS345. For a-e), similar results were obtained from 3 independent experiments; for f), results were obtained from a single experiment. Representative immunoblots are shown.

Source Data

Extended Data Fig. 3 Elevated AMPKα2 signalling in MEFs in response to pharmacological mTOR inhibition is mediated by α2-S345.

a) Lysates were prepared from FLAG-α2-(WT and S345A/E mutants) expressing α1-/-/α2-/- MEFs and immunoblotted for AMPK substrates TSC2-pT1387 (n = 6), raptor-pS792 (n = 6) and ULK1-pS555 (n = 3). Error bars, mean fold change in phosphorylation ± s.e.m. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. n represent independent experiments. b) Lysates were prepared from FLAG-α2- (WT or S345A/E mutants) expressing α1-/-/α2-/- MEFs, following incubation with mTOR inhibitors AZD8055 or INK128, and immunoblotted as indicated. Error bars, mean fold change in phosphorylation ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. Black P values vs. vehicle; red P values vs. 1 μM AZD8055 treated; blue P values vs. 1 μM INK128. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 4 Elevated AMPKα2 signalling in MEFs in response to pharmacological mTOR inhibition is independent of AMP/ATP and ADP/ATP ratios but is synergistic with energy stress.

a) Adenine nucleotides were perchlorate extracted from lysates of FLAG-α2-expressing α1-/-/α2-/- MEFs, incubated for 1 h with rapamycin, AZD8055 or INK128, and measured by LC-MS. Error bars, mean adenylate energy charge ± s.e.m., n = 3. b) torin1 (2 h) treatment of FLAG-α2-expressing α1-/-/α2-/- MEFs. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. c) Lysates were prepared from FLAG-α2-expressing α1-/-/α2-/- MEFs, following 2 h incubation with torin1 ± phenformin, and immunoblotted as indicated. Error bars, mean fold change in phosphorylation ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. Black P values vs. vehicle; red P values vs. 1 μM torin1 treated. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 5 Serum starvation suppresses mTORC1-mediated α2-S345 phosphorylation in MEFs.

FLAG-α2-expressing α1-/-/α2-/- MEFs were serum starved for 4 h, and α2-pS345 and α-pT172 tracked for 1 h following a) serum re-addition, or b) 100 nM insulin incubation. Error bars, mean fold change in phosphorylation ± s.e.m., n = 3. For a), statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. Black P values vs. basal α2-pS345; red P values vs. basal α-pT172. For b), statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. Black P values vs. serum-starved α2-pS345; red P values vs. serum-starved α-pT172. c) WT and inducible Raptor KO (iRapKO) MEFs were treated with 4-OHT and transduced with AMPK FLAG-α2 lentivirus. Cells at full confluence were serum starved overnight (DMEM only), followed by 20 min complete nutrient starvation (1xPBS only) ± subsequent serum/nutrient re-addition (DMEM, 10% FBS) for 60 min. Prepared lysates were immunoblotted for α2-pS345. Error bars, mean fold change in phosphorylation vs. serum/nutrient starved ± s.e.m., n = 4. Statistical significance was calculated using unpaired, two-tailed Student’s t test. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 6 mTORC1 inhibition in MEFs induces more robust dephosphorylation of α1-pS347 compared to α2-pS345.

a) Rapamycin or torin1 (2 h) treatment of FLAG-α-expressing α1-/-/α2-/- MEFs. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test (α1-pS347 vehicle vs. rapamycin/torin1 treatment) or unpaired, two-tailed Student’s t test (α1-pS347 vs. α2-pS345). b) Rapamycin and/or phenformin (1 h) treatment of FLAG-α1-expressing α1-/-/α2-/- MEFs. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. c) Phenformin (1 h) treatment of FLAG-α2(S345E)-expressing α1-/-/α2-/- MEFs. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 3. Statistical significance was calculated using unpaired, two-tailed Student’s t test. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 7 Regulation of endogenous AMPK signalling in HEK293T cells in response to mTOR inhibitors and AMPK activating conditions.

a) Lysates were prepared from HEK293T cells, following incubation for 1-24 h with 0.25 or 1 μM INK128, and immunoblotted as indicated. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. Black P values vs. vehicle α2-pS345; red P values vs. vehicle α-pT172. b) Lysates were prepared from HEK293T cells, treated with direct AMPK activators (A-769662, SC4) or indirect AMPK activating agents (phenformin, 2-deoxyglucose (2-DG), H2O2) as detailed, and immunoblotted as indicated. Error bars, mean fold change in phosphorylation vs. vehicle ± s.e.m., n = 4. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 8 S. pombe Ssp2-S367 is not regulated by a range of CDKs.

Lysates were prepared from WT S. pombe and indicated cdk mutants and immunoblotted for Ssp2-pS367 and total Ssp2. Error bars, mean fold change in phosphorylation vs. WT 25 °C ± s.e.m., n = 3 independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 9 Phosphorylation of AMPK α2-S345 does not influence AMP sensitivity and is not dependent on β-subunit myristoylation in HEK293T cells.

a) α2-pS345 does not affect AMP allosteric activation of AMPK. Lysates were prepared from FLAG-α2- (WT and S345A mutant) expressing α1-/-/α2-/- MEFs and AMPK immunoprecipitated using FLAG-agarose. AMPK activity was assayed ± AMP (0-50 μM). Error bars, fold change in AMPK activity vs. basal ± s.e.m., n = 3. α2-pS345 does not affect T172 phosphorylation by b) LKB1, or c) CaMKK2. Bacterial expressed α1β1γ1, incubated ± mTORC1 during purification (see Extended Data Fig. 2e) was treated with upstream kinase and immunoblotted for α-pT172. For b), Error bars, mean fold change in pT172 ± s.e.m., n = 6. For c), Error bars, mean pT172 (arbitrary units) ± s.e.m., n = 3. Statistical analyses performed by unpaired, two-tailed, Student’s t test. d) α2-pS345 does not affect rate of pT172 dephosphorylation by phosphatase PP2c. Bacterial expressed α1β1γ1, incubated with CaMKK2 and ± mTORC1 during purification, was incubated with PP2c and residual pT172 measured by immunoblot. Error bars, mean % residual pT172 ± s.e.m., n = 4. Analyses performed by unpaired, two-tailed, Student’s t test. e) Loss of AMPK β-subunit myristoylation (G2A mutant) does not affect basal or rapamycin-induced reductions in α2-S345 phosphorylation in HEK293T cells. Lysates were prepared from GST-α2β1γ1 AMPK- (WT or β1-G2A mutant) expressing HEK293T cells, following 1 h incubation with rapamycin, and immunoblotted for AMPK α2-pS345. Error bars, mean phosphorylation (arbitrary units) ± s.e.m., n = 3. Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. n represent independent experiments. Representative immunoblots are shown.

Source Data

Extended Data Fig. 10 Arg/Lys starvation suppresses AMPK α2-S345 phosphorylation in HEK293 cells and affects the ability of 2-DG to enhance cell proliferation.

See main Fig. 4. a–d, i, j) Real time proliferation analysis of HEK293 cells (untransfected or transiently expressing GFP-α2β1γ1 (WT or α2-S345A mutant)), treated as indicated. Error bars, relative confluence ± s.e.m. For a), n = 3; for b-d, i, j), n = 8. For c), ns P = 0.1819; for d), ns P > 0.9999. Statistical significance was calculated using two-way ANOVA with Sidak’s multiple comparisons test. e–h) Lysates were prepared from GFP-α2β1γ1 AMPK- (WT or α2-S345A mutant) expressing HEK293 cells (treated as indicated) and immunoblotted for AMPKα2-pS345, S6K-pT389 and ACC-pS79/212. Error bars, mean fold change in phosphorylation vs. -Arg/Lys ± s.e.m., n = 3 (WT) and 4 (S345A). Statistical significance was calculated using one-way ANOVA with Dunnett’s multiple comparisons test. n represent independent experiments. Representative immunoblots are shown.

Source Data

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Supplementary Table 1

Reporting Summary

Supplementary Data 1

Unprocessed immunoblots for Supplementary Fig. 1.

Source data

Source Data Fig. 1

Unprocessed immunoblots.

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Unprocessed immunoblots.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Unprocessed immunoblots.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Unprocessed immunoblots.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed immunoblots.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed immunoblots.

Source Data Extended Data Fig. 3

Unprocessed immunoblots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed immunoblots.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed immunoblots.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed immunoblots.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed immunoblots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed immunoblots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed immunoblots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed immunoblots.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ling, N.X.Y., Kaczmarek, A., Hoque, A. et al. mTORC1 directly inhibits AMPK to promote cell proliferation under nutrient stress. Nat Metab 2, 41–49 (2020). https://doi.org/10.1038/s42255-019-0157-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s42255-019-0157-1

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing