Skip to main content

Advertisement

Log in

TSC-insensitive Rheb mutations induce oncogenic transformation through a combination of constitutively active mTORC1 signalling and proteome remodelling

  • Original Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

The mechanistic target of rapamycin complex 1 (mTORC1) is an important regulator of cellular metabolism that is commonly hyperactivated in cancer. Recent cancer genome screens have identified multiple mutations in Ras-homolog enriched in brain (Rheb), the primary activator of mTORC1 that might act as driver oncogenes by causing hyperactivation of mTORC1. Here, we show that a number of recurrently occurring Rheb mutants drive hyperactive mTORC1 signalling through differing levels of insensitivity to the primary inactivator of Rheb, tuberous sclerosis complex. We show that two activated mutants, Rheb-T23M and E40K, strongly drive increased cell growth, proliferation and anchorage-independent growth resulting in enhanced tumour growth in vivo. Proteomic analysis of cells expressing the mutations revealed, surprisingly, that these two mutants promote distinct oncogenic pathways with Rheb-T23M driving an increased rate of anaerobic glycolysis, while Rheb-E40K regulates the translation factor eEF2 and autophagy, likely through differential interactions with 5′ AMP-activated protein kinase (AMPK) which modulate its activity. Our findings suggest that unique, personalized, combination therapies may be utilised to treat cancers according to which Rheb mutant they harbour.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

Data availability

All RAW MS-based proteomics data have been deposited to ProteomeXchange via the PRIDE partner repository with the identifier PXD017006 and can be accessed at https://www.ebi.ac.uk/pride/archive/login.

References

  1. Saxton RA, Sabatini DM (2017) mTOR signaling in growth, metabolism, and disease. Cell 168:960–976. https://doi.org/10.1016/j.cell.2017.02.004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Chung J, Kuo CJ, Crabtree GR, Blenis J (1992) Rapamycin-FKBP specifically blocks growth-dependent activation of and signaling by the 70 kd S6 protein kinases. Cell 69:1227–1236

    Article  CAS  Google Scholar 

  3. Beretta L, Gingras AC, Svitkin YV, Hall MN, Sonenberg N (1996) Rapamycin blocks the phosphorylation of 4E-BP1 and inhibits cap-dependent initiation of translation. EMBO J 15:658–664. https://doi.org/10.1002/j.1460-2075.1996.tb00398.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jeno P, Ballou LM, Novak-Hofer I, Thomas G (1988) Identification and characterization of a mitogen-activated S6 kinase. Proc Natl Acad Sci USA 85:406–410. https://doi.org/10.1073/pnas.85.2.406

    Article  CAS  PubMed  Google Scholar 

  5. Dowling RJ, Topisirovic I, Alain T, Bidinosti M, Fonseca BD, Petroulakis E, Wang X, Larsson O, Selvaraj A, Liu Y, Kozma SC, Thomas G, Sonenberg N (2010) mTORC1-mediated cell proliferation, but not cell growth, controlled by the 4E-BPs. Science 328:1172–1176

    Article  CAS  Google Scholar 

  6. Haghighat A, Mader S, Pause A, Sonenberg N (1995) Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. EMBO J 14:5701–5709

    Article  CAS  Google Scholar 

  7. Brunn GJ, Hudson CC, Sekulic A, Williams JM, Hosoi H, Houghton PJ, Lawrence JC Jr, Abraham RT (1997) Phosphorylation of the translational repressor PHAS-I by the mammalian target of rapamycin. Science 277:99–101

    Article  CAS  Google Scholar 

  8. Inoki K, Li Y, Xu T, Guan KL (2003) Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling. Genes Dev 17:1829–1834

    Article  CAS  Google Scholar 

  9. Garami A, Zwartkruis FJ, Nobukuni T, Joaquin M, Roccio M, Stocker H, Kozma SC, Hafen E, Bos JL, Thomas G (2003) Insulin activation of Rheb, a mediator of mTOR/S6K/4E-BP signaling, is inhibited by TSC1 and 2. Mol Cell 11:1457–1466. https://doi.org/10.1016/s1097-2765(03)00220-x

    Article  CAS  PubMed  Google Scholar 

  10. Tee AR, Manning BD, Roux PP, Cantley LC, Blenis J (2003) Tuberous sclerosis complex gene products, Tuberin and Hamartin, control mTOR signaling by acting as a GTPase-activating protein complex toward Rheb. Curr Biol 13:1259–1268. https://doi.org/10.1016/s0960-9822(03)00506-2

    Article  CAS  PubMed  Google Scholar 

  11. Manning BD, Cantley LC (2003) Rheb fills a GAP between TSC and TOR. Trends Biochem Sci 28:573–576. https://doi.org/10.1016/j.tibs.2003.09.003

    Article  CAS  PubMed  Google Scholar 

  12. Demetriades C, Plescher M, Teleman AA (2016) Lysosomal recruitment of TSC2 is a universal response to cellular stress. Nat Commun 7:10662. https://doi.org/10.1038/ncomms10662

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Forbes SA, Beare D, Boutselakis H, Bamford S, Bindal N, Tate J, Cole CG, Ward S, Dawson E, Ponting L, Stefancsik R, Harsha B, Kok CY, Jia M, Jubb H, Sondka Z, Thompson S, De T, Campbell PJ (2017) COSMIC: somatic cancer genetics at high-resolution. Nucleic Acids Res 45:D777-d783. https://doi.org/10.1093/nar/gkw1121

    Article  CAS  PubMed  Google Scholar 

  14. Yan L, Findlay GM, Jones R, Procter J, Cao Y, Lamb RF (2006) Hyperactivation of mammalian target of rapamycin (mTOR) signaling by a gain-of-function mutant of the Rheb GTPase. J Biol Chem 281:19793–19797. https://doi.org/10.1074/jbc.C600028200

    Article  CAS  PubMed  Google Scholar 

  15. Jiang H, Vogt PK (2008) Constitutively active Rheb induces oncogenic transformation. Oncogene 27:5729–5740. https://doi.org/10.1038/onc.2008.180

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mazhab-Jafari MT, Marshall CB, Ho J, Ishiyama N, Stambolic V, Ikura M (2014) Structure-guided mutation of the conserved G3-box glycine in Rheb generates a constitutively activated regulator of mammalian target of rapamycin (mTOR). J Biol Chem 289:12195–12201. https://doi.org/10.1074/jbc.C113.543736

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Armijo ME, Campos T, Fuentes-Villalobos F, Palma ME, Pincheira R, Castro AF (2016) Rheb signaling and tumorigenesis: mTORC1 and new horizons. Int J Cancer 138:1815–1823. https://doi.org/10.1002/ijc.29707

    Article  CAS  PubMed  Google Scholar 

  18. Kim SR, Kareva T, Yarygina O, Kholodilov N, Burke RE (2012) AAV transduction of dopamine neurons with constitutively active Rheb protects from neurodegeneration and mediates axon regrowth. Mol Ther 20:275–286. https://doi.org/10.1038/mt.2011.213

    Article  CAS  PubMed  Google Scholar 

  19. Wang Y, Hong X, Wang J, Yin Y, Zhang Y, Zhou Y, Piao HL, Liang Z, Zhang L, Li G, Xu G, Kwiatkowski DJ, Liu Y (2017) Inhibition of MAPK pathway is essential for suppressing Rheb-Y35N driven tumor growth. Oncogene 36:756–765. https://doi.org/10.1038/onc.2016.246

    Article  CAS  PubMed  Google Scholar 

  20. Heard JJ, Phung I, Potes MI, Tamanoi F (2018) An oncogenic mutant of RHEB, RHEB Y35N, exhibits an altered interaction with BRAF resulting in cancer transformation. BMC Cancer 18:69. https://doi.org/10.1186/s12885-017-3938-5

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xie J, de Souza AV, von der Haar T, O’Keefe L, Lenchine RV, Jensen KB, Liu R, Coldwell MJ, Wang X, Proud CG (2019) Regulation of the elongation phase of protein synthesis enhances translation accuracy and modulates lifespan. Curr Biol 29:737-749.e735. https://doi.org/10.1016/j.cub.2019.01.029

    Article  CAS  PubMed  Google Scholar 

  22. Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 77:248–254

    Article  Google Scholar 

  23. Humphrey SJ, Karayel O, James DE, Mann M (2018) High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform. Nat Protoc 13:1897–1916. https://doi.org/10.1038/s41596-018-0014-9

    Article  CAS  PubMed  Google Scholar 

  24. Harney DJ, Hutchison AT, Hatchwell L, Humphrey SJ, James DE, Hocking S, Heilbronn LK, Larance M (2019) Proteomic analysis of human plasma during Intermittent fasting. J Proteome Res 18:2228–2240. https://doi.org/10.1021/acs.jproteome.9b00090

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Xie J, Shen K, Lenchine RV, Gethings LA, Trim PJ, Snel MF, Zhou Y, Kenney JW, Kamei M, Kochetkova M, Wang X, Proud CG (2018) Eukaryotic elongation factor 2 kinase upregulates the expression of proteins implicated in cell migration and cancer cell metastasis. Int J Cancer 142:1865–1877. https://doi.org/10.1002/ijc.31210

    Article  CAS  PubMed  Google Scholar 

  26. Hein LK, Apaja PM, Hattersley K, Grose RH, Xie J, Proud CG, Sargeant TJ (2017) A novel fluorescent probe reveals starvation controls the commitment of amyloid precursor protein to the lysosome. Biochim Biophys Acta Mol Cell Res 1864:1554–1565. https://doi.org/10.1016/j.bbamcr.2017.06.011

    Article  CAS  PubMed  Google Scholar 

  27. Smith EM, Finn SG, Tee AR, Browne GJ, Proud CG (2005) The tuberous sclerosis protein TSC2 is not required for the regulation of the mammalian target of rapamycin by amino acids and certain cellular stresses. J Biol Chem 280:18717–18727

    Article  CAS  Google Scholar 

  28. Ghosh AP, Marshall CB, Coric T, Shim EH, Kirkman R, Ballestas ME, Ikura M, Bjornsti MA, Sudarshan S (2015) Point mutations of the mTOR-RHEB pathway in renal cell carcinoma. Oncotarget 6:17895–17910. https://doi.org/10.18632/oncotarget.4963

    Article  PubMed  PubMed Central  Google Scholar 

  29. Mosmann T (1983) Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods 65:55–63. https://doi.org/10.1016/0022-1759(83)90303-4

    Article  CAS  PubMed  Google Scholar 

  30. Grabiner BC, Nardi V, Birsoy K, Possemato R, Shen K, Sinha S, Jordan A, Beck AH, Sabatini DM (2014) A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov 4:554–563. https://doi.org/10.1158/2159-8290.cd-13-0929

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Xie J, Wang X, Proud CG (2016) mTOR inhibitors in cancer therapy. F1000Res. https://doi.org/10.12688/f1000research.9207.1

    Article  PubMed  PubMed Central  Google Scholar 

  32. Mossmann D, Park S, Hall MN (2018) mTOR signalling and cellular metabolism are mutual determinants in cancer. Nat Rev Cancer 18:744–757. https://doi.org/10.1038/s41568-018-0074-8

    Article  CAS  PubMed  Google Scholar 

  33. Pike KG, Malagu K, Hummersone MG, Menear KA, Duggan HM, Gomez S, Martin NM, Ruston L, Pass SL, Pass M (2013) Optimization of potent and selective dual mTORC1 and mTORC2 inhibitors: the discovery of AZD8055 and AZD2014. Bioorg Med Chem Lett 23:1212–1216. https://doi.org/10.1016/j.bmcl.2013.01.019

    Article  CAS  PubMed  Google Scholar 

  34. Hamidi H, Ivaska J (2018) Every step of the way: integrins in cancer progression and metastasis. Nat Rev Cancer 18:533–548. https://doi.org/10.1038/s41568-018-0038-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Moore CE, Mikolajek H, Regufe da Mota S, Wang X, Kenney JW, Werner JM, Proud CG (2015) Elongation factor 2 kinase is regulated by proline hydroxylation and protects cells during hypoxia. Mol Cell Biol 35:1788–1804. https://doi.org/10.1128/mcb.01457-14

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu R, Proud CG (2016) Eukaryotic elongation factor 2 kinase as a drug target in cancer, and in cardiovascular and neurodegenerative diseases. Acta Pharmacol Sin 37:285–294. https://doi.org/10.1038/aps.2015.123

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Browne GJ, Finn SG, Proud CG (2004) Stimulation of the AMP-activated protein kinase leads to activation of eukaryotic elongation factor 2 kinase and to its phosphorylation at a novel site, serine 398. J Biol Chem 279:12220–12231. https://doi.org/10.1074/jbc.M309773200

    Article  CAS  PubMed  Google Scholar 

  38. Kim J et al (2016) Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12:1–222. https://doi.org/10.1080/15548627.2015.1100356

    Article  PubMed  PubMed Central  Google Scholar 

  39. Klionsky DJ et al (2021) Autophagy 17:1–382. https://doi.org/10.1080/15548627.2020.1797280

    Article  PubMed  Google Scholar 

  40. Racker E (1972) Bioenergetics and the problem of tumor growth. Am Sci 60:56–63

    CAS  PubMed  Google Scholar 

  41. Huang J, Manning BD (2008) The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J 412:179–190. https://doi.org/10.1042/BJ20080281

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Saucedo LJ, Gao X, Chiarelli DA, Li L, Pan D, Edgar BA (2003) Rheb promotes cell growth as a component of the insulin/mTOR signalling network. Nat Cell Biol 5:566–571

    Article  CAS  Google Scholar 

  43. Stocker H, Radimerski T, Schindelholz B, Wittwer F, Belawat P, Daram P, Breuer S, Thomas G, Hafen E (2003) Rheb is an essential regulator of S6K in controlling cell growth in Drosophila. Nat Cell Biol 5:559–565

    Article  CAS  Google Scholar 

  44. Zhang Y, Gao X, Saucedo LJ, Ru B, Edgar BA, Pan D (2003) Rheb is a direct target of the tuberous sclerosis tumour suppressor proteins. Nat Cell Biol 5:578–581

    Article  CAS  Google Scholar 

  45. Yang H, Jiang X, Li B, Yang HJ, Miller M, Yang A, Dhar A, Pavletich NP (2017) Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40. Nature 552:368–373. https://doi.org/10.1038/nature25023

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lacher MD, Pincheira R, Zhu Z, Camoretti-Mercado B, Matli M, Warren RS, Castro AF (2010) Rheb activates AMPK and reduces p27Kip1 levels in Tsc2-null cells via mTORC1-independent mechanisms: implications for cell proliferation and tumorigenesis. Oncogene 29:6543–6556. https://doi.org/10.1038/onc.2010.393

    Article  CAS  PubMed  Google Scholar 

  47. Choo AY, Kim SG, Vander Heiden MG, Mahoney SJ, Vu H, Yoon SO, Cantley LC, Blenis J (2010) Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply. Mol Cell 38:487–499. https://doi.org/10.1016/j.molcel.2010.05.007

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank Professor Andrew Tee (Cardiff University, Cardiff, UK) and Dr. Brendan Manning (Harvard Medical School, Boston, MA, USA) for their valuable suggestions in regard to the GAP assay.

Funding

This work was initiated with funding from the UK Biotechnology and Biological Sciences Research Council (to CGP) and then supported by SAHMRI. SDP acknowledges support by a Scholarship from the Australian Government Research Training Program (RTP). JX acknowledges a SAHMRI early/mid-career seed funding grant. LAS is supported by a Principal Cancer Research Fellowship awarded by Cancer Council’s Beat Cancer project on behalf of its donors, the state Government through the Department of Health, and the Australian Government through the Medical Research Future Fund.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Christopher G. Proud.

Ethics declarations

Conflict of interest

The authors declare no potential conflicts of interest.

Ethics approval

All animal work was conducted in accordance with the National Health and Medical Research Council’s Care and use of Animals for Scientific Purposes guidelines and with approval (No. SAM339) from the South Australian Health and Medical Research Institute’s animal ethics committee.

Additional information

Publisher's Note

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

Supplementary Information

Below is the link to the electronic supplementary material.

18_2021_3825_MOESM1_ESM.tif

Supplementary file1 (TIF 1892 KB) Supplementary Figure S1 Activation by Rheb mutants of signalling downstream of mTOR is blocked by AZD8055 but not AZD6244. Related to Fig. 1. (a) Immunoblot analysis of HEK293 cells transfected with the indicated Rheb mutants. Cells were treated with 1 µM AZD8055 1 h prior to harvest. (b) Immunoblot of lysates from HEK293 cells that had been transfected with a pcDNA3.1 empty vector (EV) or vector encoding wild-type Rheb or the indicated mutant. After 24 h, cells were treated with either DMSO or 5 µM AZD6244 for 2 h. All figures are representative images of three independent replicate experiments. (c) Immunoblot of lysates from HEK293 cells that had been transfected with a pcDNA3.1 empty vector or vector encoding wild-type Rheb or the indicated mutant. After 24 h, the medium was replaced with fresh medium either with or without FBS. After 16 h, cells were lysed. Immunoblots were probed with antibodies for the indicated proteins or phosphorylation sites. (d) Quantification of Supplementary Fig. S1c. P-ERK and total ERK were normalised to β-actin and the ratio of P-ERK:ERK graphed). Data are means ± S.D.; n = 3. (e) Immunoblots of lysates from HEK293 cells previously transfected with either the indicated Rheb mutant and a pcDNA3.1 empty vector or the indicated Rheb mutant and vectors encoding FLAG-TSC1 and FLAG-TSC2. After 24 h, the medium was replaced with fresh DMEM without FBS for 16 h followed by D-PBS for 1 h. (f) Quantification of Supplementary Fig. S1e. P-S6K1 Thr389 (top) and P-4EBP1 Ser65 (bottom) normalized to Tubulin loading control for means ± S.D.; n = 3. Data were analysed using Student’s t-test where *: 0.01 ≤ P < 0.05; **: 0.001 ≤ P < 0.01; ***: P < 0.001.

18_2021_3825_MOESM2_ESM.tif

Supplementary file2 (TIF 787 KB) Supplementary Figure S2: Rheb Mutants co-immunoprecipitate with TSC1/2 and mTORC1. Related to Fig. 2. (a) HEK293 cells were transfected with indicated vectors for FLAG-Rheb for 48 h, for both (a) and (b) Rheb and associated proteins were pulled down from cell lysates and both immunoprecipitates and input lysates were subjected to immunoblotting analysis for the indicated proteins.

18_2021_3825_MOESM3_ESM.tif

Supplementary file3 (TIF 696 KB) Supplementary Figure S3: Rheb mutants do not drive faster proliferation in fully supplemented media. Related to Fig. 3. (a) MTT assay data for HEK293 cells transiently expressing the indicated Rheb mutants in fully supplemented medium. Data are means ± S.D.; n = 3. (b) BrdU incorporation data for HEK293 cells transiently expressing the indicated Rheb mutants in fully supplemented medium. (c) Table summarizing the phenotypes of various Rheb mutants as determined in Figs. 1d, e, 2c, d, 3a and b.

18_2021_3825_MOESM4_ESM.tif

Supplementary file4 (TIF 1142 KB) Supplementary Figure S4: Characterisation of NIH3T3 cells stably over-expressing Rheb mutants. Related to Figs. 3 and 4. (a) Sanger sequencing results of monoclonal cell lines selected for use in the in vivo study. Cells were grown for 8 weeks before DNA was extracted and sequenced using primers specific to the inserted plasmid. (b) Survival curve showing the number of days post-injection until tumour volume reached 60 mm3. Statistics were calculated using the Mantel-Cox logrank test where @: P < 0.001. (c) Quantification of DAB stain in Fig. 4d for P-rpS6:rpS6 ratio. (d) As in (c) for P-ERK1/2:ERK1/2 ratio. Error bars indicate SD for three independent experiments. For panels c and d, data are means ± S.D.; n = 3. Statistical significance was determined by Student’s t-test where *: 0.01 ≤ P < 0.05; **: 0.001 ≤ P < 0.01.

18_2021_3825_MOESM5_ESM.tif

Supplementary file5 (TIF 2923 KB) Supplementary Figure S5: MS analysis of proteome changes between fully supplemented and serum starved NIH3T3 cells. Related to Fig. 5. NIH3T3 cells stably over-expressing Rheb-WT, T23M or E40K were either cultured in full growth medium (with 10% FBS) or starved of serum for 72 h before being subjected to MS analysis. (a) Heat-map illustrates six proteins whose abundances differ among the cell groups cultured in fully supplemented growth medium. (b) Cluster analysis of proteins that are either upregulated when cultured in full growth medium or when serum starved. (c) Heat-map illustrates four different protein clusters among the treatment groups.

18_2021_3825_MOESM6_ESM.tif

Supplementary file6 (TIF 852 KB) Supplementary Figure S6: Validation of MS data. Related to Fig. 5 a) RT-qPCR analysis of cDNA generated from whole cell RNA extracts from NIH3T3 cells stably expressing the indicated Rheb vector grown in fully supplemented media (+FBS) or starved of serum for 72 h (-FBS) using primers specific for PKM2. b) As in a) with primers for EEF2. c) Immunoblot analysis of NIH3T3 cells stably expressing the indicated Rheb mutant. Cells were grown in fully supplemented media or starved of serum for 72 h, as indicated. d) Quantification of c) normalized to β-actin. e) Proteins from tumour tissue (from Fig. 4) lysates were separated on SDS-PAGE gels, indicated proteins were immunoblotted with their corresponding antibodies. f) The indicated protein ratios in e) were quantified. g) Quantification of Fig. 6a for ratio of P-eEF2:eEF2. For panels XXX, data are means ± S.D.; n = 3. Statistical significance was determined by 2-way ANOVA with multiple comparisons to WT + FBS where *: 0.01 ≤ P < 0.05; **: 0.001 ≤ P < 0.01; ***: P < 0.001.

18_2021_3825_MOESM7_ESM.tif

Supplementary file7 (TIF 1161 KB) Supplementary Figure S7: CA-Rheb mutants drive an increase in migration and invasion in trans-well assays. Related to Fig. 6. a) Transwell migration and invasion assays of 1.5 × 104 HEK293 cells transfected with vectors encoding the indicated Rheb mutant. For invasion assays, transwells were pre-coated with Matrigel. Cells were treated with 0.5 µg/µl for the duration of the experiment. Cells were allowed to migrate or invade for 72 h. b) Quantification of a) for migration. c) Quantification of a) for invasion. d) Immunoblot analysis of NIH3T3 cells. Cells were either grown in fully supplemented media or serum starved for 2h prior to harvest as indicated. As a positive control for AMPK activation, one well had media preplaced for media lacking glucose for 1 h prior to the addition of 2-deoxyglucose (2-DG) for 2 h. e) NIH3T3 cells stably expressing the indicated Rheb mutants were treated with AZD8055 or DMSO for 1h as indicated. f) NIH3T3 cells stably expressing the indicated Rheb mutants were treated with either 1 µM AZD8055 or DMSO, as indicated, for 1 h. Lysates were collected, and western blot analysis performed with the indicated antibodies. For panels b and c, data are means ± S.D.; n = 3. Statistical significance was determined by Student’s t-test where *: 0.01 ≤ P < 0.05; **: 0.001 ≤ P < 0.01; ***: P < 0.001.

Supplementary file8 (DOCX 18 KB)

Supplementary file9 (XLSX 2210 KB)

Supplementary file10 (XLSX 11 KB)

Supplementary file11 (XLSX 19 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Xie, J., De Poi, S.P., Humphrey, S.J. et al. TSC-insensitive Rheb mutations induce oncogenic transformation through a combination of constitutively active mTORC1 signalling and proteome remodelling. Cell. Mol. Life Sci. 78, 4035–4052 (2021). https://doi.org/10.1007/s00018-021-03825-7

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-021-03825-7

Keywords

Navigation