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:

AMP kinase promotes glioblastoma bioenergetics and tumour growth

A Correction to this article was published on 06 September 2018

A Publisher Correction to this article was published on 08 August 2018

This article has been updated

Abstract

Stress is integral to tumour evolution, and cancer cell survival depends on stress management. We found that cancer-associated stress chronically activates the bioenergetic sensor AMP kinase (AMPK) and, to survive, tumour cells hijack an AMPK-regulated stress response pathway conserved in normal cells. Analysis of The Cancer Genome Atlas data revealed that AMPK isoforms are highly expressed in the lethal human cancer glioblastoma (GBM). We show that AMPK inhibition reduces viability of patient-derived GBM stem cells (GSCs) and tumours. In stressed (exercised) skeletal muscle, AMPK is activated to cooperate with CREB1 (cAMP response element binding protein-1) and promote glucose metabolism. We demonstrate that oncogenic stress chronically activates AMPK in GSCs that coopt the AMPK–CREB1 pathway to coordinate tumour bioenergetics through the transcription factors HIF1α and GABPA. Finally, we show that adult mice tolerate systemic deletion of AMPK, supporting the use of AMPK pharmacological inhibitors in the treatment of GBM.

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: AMPK is highly expressed in GBM.
Fig. 2: AMPK is essential for viability of primary GBM lines in vitro.
Fig. 3: AMPK is essential for optimal GSC growth in vivo.
Fig. 4: AMPK regulates GBM bioenergetics.
Fig. 5: AMPK regulates HIF1α transcription in GSCs.
Fig. 6: AMPK regulates GABPA transcription in GSCs.
Fig. 7: AMPK regulates HIF1α and GABPA through CREB1.
Fig. 8: The AMPK–CREB1 transcriptional axis regulates GBM bioenergetics and tumour growth.

Similar content being viewed by others

Change history

  • 06 September 2018

    In the version of this Article originally published, in ref. 34 the first author’s name was spelled incorrectly. The correct reference is: Rodón, L. et al. Active CREB1 promotes a malignant TGFβ2 autocrine loop in glioblastoma. Cancer Discov. 10, 1230–1241 (2014). This has now been amended in all online versions of the Article.

  • 08 August 2018

    In the version of this Article originally published, the competing interests statement was missing. The authors declare no competing interests; this statement has now been added in all online versions of the Article.

References

  1. Hardie, D. G. AMP-activated protein kinase: maintaining energy homeostasis at the cellular and whole-body levels. Annu. Rev. Nutr. 34, 31–55 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Carling, D., Thornton, C., Woods, A. & Sanders, M. J. AMP-activated protein kinase: new regulation, new roles? Biochem. J. 445, 11–27 (2012).

    CAS  PubMed  Google Scholar 

  3. Dasgupta, B. & Chhipa, R. R. Evolving lessons on the complex role of AMPK in normal physiology and cancer. Trends Pharmacol. Sci. 37, 192–206 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Iseli, T. J. et al. AMP-activated protein kinase beta subunit tethers alpha and gamma subunits via its C-terminal sequence (186–270). J. Biol. Chem. 280, 13395–13400 (2005).

    CAS  PubMed  Google Scholar 

  5. Shaw, R. J. et al. The tumor suppressor LKB1 kinase directly activates AMP-activated kinase and regulates apoptosis in response to energy stress. Proc. Natl Acad. Sci. USA 101, 3329–3335 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Hawley, S. A. et al. Calmodulin-dependent protein kinase kinase-beta is an alternative upstream kinase for AMP-activated protein kinase. Cell Metab. 2, 9–19 (2005).

    CAS  PubMed  Google Scholar 

  7. Carling, D., Zammit, V. A. & Hardie, D. G. A common bicyclic protein kinase cascade inactivates the regulatory enzymes of fatty acid and cholesterol biosynthesis. FEBS Lett. 223, 217–222 (1987).

    CAS  PubMed  Google Scholar 

  8. 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 

  9. 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 

  10. Faubert, B. et al. AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo. Cell Metab. 17, 113–124 (2013).

    Article  CAS  PubMed  Google Scholar 

  11. Rios, M., Foretz, M. & Viollet, B. et al. AMPK activation by oncogenesis is required to maintain cancer cell proliferation in astrocytic tumors. Cancer Res. 73, 2628–2638 (2013).

    CAS  PubMed  Google Scholar 

  12. Liu, X. et al. Discrete mechanisms of mTOR and cell cycle regulation by AMPK agonists independent of AMPK. Proc. Natl Acad. Sci. USA 111, E435–E444 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Swinnen, J. V. et al. Mimicry of a cellular low energy status blocks tumor cell anabolism and suppresses the malignant phenotype. Can. Res. 65, 2441–2448 (2005).

    CAS  Google Scholar 

  14. Tang, Y. C., Williams, B. R., Siegel, J. J. & Amon, A. Identification of aneuploidy-selective antiproliferation compounds. Cell 144, 499–512 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Dowling, R. J., Zakikhani, M., Fantus, I. G., Pollak, M. & Sonenberg, N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 67, 10804–10812 (2007).

    CAS  PubMed  Google Scholar 

  16. Guo, D. et al. The AMPK agonist AICAR inhibits the growth of EGFRvIII-expressing glioblastomas by inhibiting lipogenesis. Proc. Natl Acad. Sci. USA 106, 12931–12937 (2009).

    Google Scholar 

  17. Yan., M. et al. The tumor suppressor folliculin regulates AMPK-dependent metabolic transformation. J. Clin. Invest. 124, 2640–2650 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Laderoute., K. R. et al. 5′-AMP-activated protein kinase (AMPK) supports the growth of aggressive experimental human breast cancer tumors. J. Biol. Chem. 289, 22850–22864 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. D’Amico, D. et al. Non-canonical Hedgehog/AMPK-mediated control of polyamine metabolism supports neuronal and nedulloblastoma cell growth. Dev. Cell. 35, 21–35 (2015).

    PubMed  PubMed Central  Google Scholar 

  20. Bungard., D. et al. Signaling kinase AMPK activates stress-promoted transcription via histone H2B phosphorylation. Science 329, 1201–1205 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Laderoute., K. R. et al. 5′-AMP-activated protein kinase (AMPK) is induced by low-oxygen and glucose deprivation conditions found in solid-tumor microenvironments. Mol. Cell. Biol. 26, 5336–5347 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Liu., L. et al. Deregulated MYC expression induces dependence upon AMPK-related kinase 5. Nature 483, 608–612 (2012).

    CAS  PubMed  Google Scholar 

  23. Shackelford., D. B. et al. mTOR and HIF-1alpha-mediated tumor metabolism in an LKB1 mouse model of Peutz-Jeghers syndrome. Proc. Natl Acad. Sci. USA 106, 11137–11142 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kishton., R. J. et al. AMPK is essential to balance glycolysis and mitochondrial metabolism to control T-ALL cell stress and survival. Cell Metab. 23, 649–662 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Wu., S. et al. AMPK-mediated increase of glycolysis as an adaptive response to oxidative stress in human cells: implication of the cell survival in mitochondrial diseases. Biochem. Biophys. Acta 1822, 233–247 (2012).

    CAS  PubMed  Google Scholar 

  26. E. Doménech, E. et al. AMPK and PFKFB3 mediate glycolysis and survival in response to mitophagy during mitotic arrest. Nat. Cell Biol. 17, 1304–1316 (2015).

    Google Scholar 

  27. Almeida, A. et al. Nitric oxide switches on glycolysis through the AMP protein kinase and 6-phosphofructo-2-kinase pathway. Nat. Cell Biol. 6, 45–51 (2004).

    CAS  PubMed  Google Scholar 

  28. Fumarola, C. et al. Effects of sorafenib on energy metabolism in breast cancer cells: role of AMPK-mTORC1 signaling. Breast Cancer Res. Treat. 141, 67–78 (2013).

    CAS  PubMed  Google Scholar 

  29. Reszec, J. et al. The expression of hypoxia-inducible factor-1 in primary brain tumors. Int J. Neurosci. Sep. 123, 657–662 (2013).

    CAS  Google Scholar 

  30. Mayer, A. et al. Differential expression of HIF-1 in glioblastoma multiforme and anaplastic astrocytoma. Int J. Oncol. Oct. 41, 1260–1270 (2012).

    Google Scholar 

  31. Barresi, V. et al. p-CREB expression in human gliomas: potential use in the differential diagnosis between astrocytoma and oligodendroglioma. Human Pathol. 46, 231–238 (2015).

    CAS  Google Scholar 

  32. Li, Z. et al. Hypoxia-inducible factors regulate tumoregenic capacity of glioma stem cells. Cancer Cell 15, 501–513 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Fan, Y. et al. Profilin-1 phosphorylation directs angiocrine expression and glioblastoma progression through HIF1a accumulation. Nat. Cell Biol. 16, 445–456 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Rodón, L. et al. Active CREB1 promotes a malignant TGFβ2 autocrine loop in glioblastoma. Cancer Discov. 10, 1230–1241 (2014).

    Google Scholar 

  35. Chow, L. M. et al. Cooperativity within and among Pten, p53, and Rb pathways induces high-grade astrocytoma in adult brain. Cancer Cell 19, 305–316 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Chae, Y. C. et al. Control of tumor bioenergetics and survival stress signaling by mitochondrial HSP90s. Cancer Cell 22, 331–344 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ros, S. et al. Functional metabolic screen identifies 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 4 as an important regulator of prostate cancer cell survival. Cancer Discov. 2, 328–343 (2012).

    CAS  PubMed  Google Scholar 

  38. Fan, Q. et al. Akt and autophagy cooperate to promote survival of drug-resistance glioma. Sci. Signal. 3, ra81 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Fiedler, T. et al. Arginine deprivation by arginine deiminase of Streptococcus pyogenes controls primary glioblastoma growth in vitro and in vivo. Can. Biol. Ther. 16, 1047–1055 (2015).

    CAS  Google Scholar 

  40. Shirwany, N. A. & Zou, M. H. AMPK: a cellular metabolic and redox sensor. A minireview. Front Biosci. 19, 447–474 (2014).

    CAS  Google Scholar 

  41. Jeon, S. M., Chandel, N. S. & Hay, N. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature 485, 661–665 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Denko, N. C. Hypoxia, HIF1 and glucose metabolism in the solid tumour. Nat. Rev. Cancer 8, 705–713 (2008).

    CAS  PubMed  Google Scholar 

  43. Liu, W., Shen, S. M., Zhao, X. Y. & Chen, G. Q. Targeted genes and interacting proteins of hypoxia inducible factor-1. Int J. Bioche Mol. Biol. 3, 165–178 (2012).

    CAS  Google Scholar 

  44. Benita, Y. et al. An integrative genomics approach identifies Hypoxia Inducible Factor-1 (HIF-1)-target genes that form the core response to hypoxia. Nucleic Acids Res. 37, 4587–4602 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kelly, D. P. & Scarpulla, R. C. Transcriptional regulatory circuits controlling mitochondrial biogenesis and function. Genes Dev. 18, 357–368 (2004).

    CAS  PubMed  Google Scholar 

  46. Bruni, F., Polosa, P. L., Gadaleta, M. N., Cantatore, P. & Roberti, M. Nuclear respiratory factor 2 induces the expression of many but not all human proteins acting in mitochondrial DNA transcription and replication. J. Biol. Chem. 285, 3939–3948 (2010).

    CAS  PubMed  Google Scholar 

  47. Larsson, N. G. et al. Mitochondrial transcription factor A is necessary for mtDNA maintenance and embryogenesis in mice. Nat. Genet. 18, 231–236 (1998).

    CAS  PubMed  Google Scholar 

  48. Cantó, C. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 458, 1056–1060 (2009).

    PubMed  PubMed Central  Google Scholar 

  49. Levesque, M. J. & Raj, A. Single-chromosome transcriptional profiling reveals chromosomal gene expression regulation. Nat. Methods 10, 246–248 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Selak, M. A. et al. Succinate links TCA cycle dysfucntion to oncogenesis by inhibiting HIF-α prolyl hydroxylases. Cancer Cell 1, 77–85 (2005).

    Google Scholar 

  51. Kaelin, W. G. The Von Hippel-Lindau tumor suppressor protein: O2 sensing and cancer. Nat. Rev. Cancer 8, 865–873 (2008).

    CAS  PubMed  Google Scholar 

  52. Seidel, S. et al. A hypoxic niche regulates glioblastoma stem cells through hypoxia inducible factor 2 alpha. Brain 133, 983–995 (2010).

    PubMed  Google Scholar 

  53. Dickinson, A. et al. The regulation of mitochondrial copy number in glioblastoma cells. Cell Death Diff. 20, 1644–1653 (2013).

    CAS  Google Scholar 

  54. Sakamoto, K. et al. Deficiency of LKB1 in skeletal muscle prevents AMPK activation and glucose uptake during contraction. EMBO J. 24, 1810–1820 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Thomson, D. M. et al. Skeletal muscle and heart LKB1 deficiency causes decreased voluntary running and reduced muscle mitochondrial marker enzyme expression in mice. Am. J. Physiol. Endocrinol. Metab. 292, E196–E202 (2007).

    CAS  PubMed  Google Scholar 

  56. McGee, S. L. & Hargreaves, M. AMPK-mediated regulation of transcription in skeletal muscle. Clin. Sci. 118, 507–518 (2010).

    CAS  Google Scholar 

  57. Thomson, D. M. et al. AMP-activated protein kinase phosphorylates transcription factors of the CREB family. J. Appl. Physiol. 104, 429–438 (2008).

    CAS  PubMed  Google Scholar 

  58. Li, Y., Cummings, R. T., Cunningham, B. R., Chen, Y. & Zhou, G. Homogeneous assays for adenosine 5’-monophosphate-activated protein kinase. Anal. Biochem. 321, 151–156 (2003).

    CAS  PubMed  Google Scholar 

  59. Mayr, B. & Montminy, M. Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat. Rev. Mol. Cell Biol. 2, 599–609 (2001).

    CAS  PubMed  Google Scholar 

  60. Jang, T. et al. 5’-AMP-activated protein kinase activity is elevated early during primary brain tumor development in the rat. Int. J. Cancer 128, 2230–2239 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Ferreira, D. et al. in Novel Approaches in Biomarkers Discovery and Therapeutic Targets in Cancer (ed. Lopez-Camarillo, C.) Ch. 6 (IntechOpen, London, 2013).

  62. Rios, M. et al. Lipoprotein internalisation induced by oncogenic AMPK activation is essential to maintain glioblastoma cell growth. Eur. J. Cancer 50, 3187–3197 (2014).

    CAS  PubMed  Google Scholar 

  63. Saito, Y., Chapple, R. H., Lin, A., Kitano, A. & Nakada, D. AMPK protects leukemia-initiating cells in myeloid leukemias from metabolic stress in the bone marrow. Cell Stem Cell 17, 585–596 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Mo, J. S. et al. Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat. Cell Biol. 17, 500–510 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Liu, X. et al. LncRNA NBR2 engages a metabolic checkpoint by regulating AMPK under energy stress. Nat. Cell Biol. 18, 431–442 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Papandreu, I. et al. HIF-1 mediates adaptation to hypoxia by actively downregulating mitochondrial oxygen consumption. Cell Metab. 3, 187–197 (2006).

    Google Scholar 

  67. Dasgupta, B. & Milbrandt, J. AMP-activated protein kinase phosphorylates retinoblastoma protein to control mammalian brain development. Dev. Cell 16, 256–270 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Pooya, S. et al. The tumour suppressor LKB1 regulates myelination through mitochondrial metabolism. Nat. Comm. 5, 4993 (2014).

    CAS  Google Scholar 

  69. Komurov, K., Dursun, S., Erdin, S. & Ram, P. T. NetWalker: a contextual network analysis tool for functional genomics. BMC Genom. 13, 282 (2012).

    Google Scholar 

  70. Ernst, J. et al. Mapping and analysis of chromatin state dynamics in nine human cell types. Nature 473, 43–49 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Kim (University of Alabama) for maintaining and providing primary GSC lines; A. Hinge (Cincinnati Children’s Hospital) for help with flow cytometry; G. Huang (Cincinnati Children’s Hospital) for providing the HRE luciferase reporter construct and for help with ChIP experiments; J. Bridges (Cincinnati Children’s Hospital) for providing constitutive active HIF1α; S. Wells for plenti-CMV-luc plasmid; S. Elledge (Massachusetts Institute of Technology) for providing pInducer plasmid; R. Jones (McGuill University, Canada) for AMPKα2 dominant-negative plasmid; F. Furnari (Lugwig Institute of Cancer Research, San Diego, CA) for pLV EGFRvIII Hygro plasmid; P. Mischel (University of California, San Diego) for U87EGFRvIII glioma cells; D. Carling (Imperial College, London) for providing Compound 991; X. Liu (previously, Cincinnati Children’s Hospital) for assistance with experiments; P. Malik (Cincinnati Children’s Hospital) for providing assistance in determining lentiviral copy number; E. Boscolo (Cincinnati Children’s Hospital) for providing Isolectin B4; B. Viollet (INSERM, Institut Cochin, Paris, France) for providing AMPKα1−/− and AMPKα2lox/lox mice; and A. Kumar (Cincinnati Children’s Hospital) for providing Rosa26 CreER mice (originally from NCI). This work was supported by the CCTST1 Translational Grant Award; Pilot Innovation award, CCHMC; University of Cincinnati Cancer Center Affinity Grant Award, CancerFreeKids and National Institute of Health (1R01NS075291-01A1 and 1R01NS099161-01) (all to B.D.).

Author information

Authors and Affiliations

Authors

Contributions

B.D. and R.C. conceived the experiments, analysed data and wrote the manuscript. R.C. performed most of the experiments. Q.F., J.A. and R.M. performed additional experiments. G.C. helped with electron microscopy. N.R, R.W. and L.M.C. provided valuable reagents and experimental suggestions. N.R. also helped in manuscript editing. Y.H. performed and Z.K. supervised mitochondrial complex activity measurements. M.K. helped with smFISH experiments. M.W. and X.C. provided bioinformatic support for transcription factor analysis. A.K. helped with identification of GBM patient samples. C.M. performed surgery and helped coordinate tissue procurement from the operating room. I.N. provided a few primary GSC lines. N.D. and K.K. provided bioinformatic support for RNA-seq and TCGA data analysis. N.D. also performed statistical analysis.

Corresponding author

Correspondence to Biplab Dasgupta.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 Oncogenic stress-associated high expression of AMPK in glioblastoma.

a, Kaplan-Meier survival plots of LGG patients expressing phosphorylated ACC (Source: TCGA). b, Kaplan-Meier survival plots of GBM patients expressing AMPK subunits (Source: TCGA). c, Western blot (WB) of pAMPK and pACC in Human GBM and normal human brain tissue. X indicates tissue degradation. Actin was used as loading control. d, e, IHC of pACC, DAPI (nuclei) and the microglia marker IBA-1 in human GBM tissue (d), and xenograft of the human GBM line 326 in NSG mice (e). Scale bar = 50 μm (d) and 100μM (e). f, Oncogenesis-associated stress markers are high in primary GSC lines relative to normal human astrocytes (NHA). WB showing markers of genotoxic stress (pATM), and endoplasmic reticulum (ER) stress (BIP and sXBP1). Actin was used as loading control. g, IHC of γH2AX foci in GSC and NHA. Scale bar 100 μm. Nuclei were stained with DAPI. Scale bar = 20 μm h, Box plots Source: (TCGA) showing expression of ER stress transcripts in GBM and normal brain (HSPA5 is the gene name for BIP). The edges on the boxplots indicate the first and 3rd quartile (25th- and 75th percentile) of the data, with the line in the middle being the median. The whiskers on the boxplots extend another 1.5x of the inter-quartile range (between 25%-75% range of data) from the edges of the boxes, respectively. i, j, WB of sXBP1, BIP, CHOP, pATM in NHA treated with tunicamycin and hydroxyurea (HU). k, WB of pAMPK, AMPK, pACC and ACC in NHA treated with indicated reagents. Actin was used as loading control. l, m, Quantification of band intensities in the same order as in (k). n, WB of indicated proteins in NHA expressing oncogenic EGFRvIII, constitutively active KRASG12V or PTEN shRNA. Note: PTEN appears to be significantly downregulated in EGFRvIII and KRASG12V cells. o, p, Quantification of band intensities in (n). All western blots represent data from 2 (f, n, k), or 3 (c, I, j) independent repeats. Scanned images of unprocessed blots are shown in Supplementary Fig. 9.

Supplementary Figure 2 AMPK is essential for optimal growth of primary GBM orthotopic xenografts in mice.

a, b, Luciferase imaging of mice to monitor tumour growth of GSC lines expressing AMPKβ1 or nontarget (NT) shRNA at indicated days. (n = 8-15 mice per line). Primary GSC lines AC17, 1123 were infected with NT or AMPKβ1 shRNA for two days and 10,000 Trypan blue negative live cells were transplanted into the cortex of NSG mice 48 hours after in vitro lentiviral transduction (when AMPK β1shRNA had negligible effect on viability). c, Quantification of tumour luminescence. Total range of tumour luminescence of mice was quantified three weeks after cell transplantation. (n = 20 mice / genotype). *p ≤ 0.0001. d, Luciferase imaging of mice to monitor tumour growth of a GSC line expressing AMPK α1α2 shRNA or nontarget (NT) shRNA at indicated days. (n = 6 mice per genotype). e, Kaplan-Meier survival data of mice in (d). f, Kaplan-Meier survival data of mice transplanted with U87EGFRvIII GBM serum line expressing NT or AMPK β1 shRNA. (n = 4 mice per genotype). g, Expression of cumate-inducible GFP in 293T cells. Scale bar 100μm. h, WB showing expression of cumate-inducible mouse AMPKβ1 (subcloned in the SparQ vector) in 293T cells. i, IHC pf pAMPK and pACC in NT and AMPKβ1 shRNA expressing tumours at indicated days. (T = tumour; N = normal brain). Scale bar 100 μm. j, Q-RTPCR of human AMPK β1 (using human-specific primers) in tumour cells isolated from NT shRNA or AMPKβ1 shRNA tumours. (n = 3). *p = 0.0007. k, WB of AMPKβ1/β2 and pAMPK in tumour-derived cells as in (j). Error bars; mean +/- S.D. Statistical significance in (c, j); two-tailed t-test. n values represent independent experiments. Source data are available in Supplementary Table 4. All western blots represent data from 2 (h) or 3 (k) independent repeats. Unprocessed blots are shown in Supplementary Fig. 9.

Supplementary Figure 3 GSC death occurs independent of AMPK-regulated mTOR and autophagy pathways.

a.WB of pULK1 (AMPK target) and autophagy substrate LC3 in AMPK-silenced GSCs. ULK1 and actin are used as loading controls. b, WB of autophagy substrates LC3 and P62 in NT or AMPKβ1shRNA expressing cells treated with Bafilomycin A showing autophagy flux. c, Viability of GSCs and HepG2 liver cancer line treated with autophagy inhibitors Bafilomycin and Chloroquine, or infected with NT or ATG13 shRNAs. (Average of two independent experiments). d, Superoxide detection by flow cytometry in NT or AMPKβ1 shRNA expressing GSCs using MitoSox Red. (Average of two independent experiments). e, Viability of GSCs expressing NT or AMPKβ1 shRNA treated with the reducing agent N-acetylcysteine (NAC) or the ACC inhibitor TOFA. (Average of two independent experiments). f, ATP levels in GSC lines expressing AMPKβ1 shRNA or NT shRNA at indicated times. ATP value was normalized to protein. (n = 3). *p ≤ 0.002; + ≤ 0.004. g, Viability of GSCs expressing NT or AMPKβ1 shRNA treated with the mTORC1 inhibitor rapamycin (5nM). (n = 3). *p ≤ 0.01; + ≤ 0.004. h, WB of stem cell markers Musashi and Nanog in GSCs expressing NT and AMPKβ1 shRNA. Error bars; mean +/- S/d. Statistical significance in above experiments was assessed using two-tailed t-test. n values represent independent experiments. Source data are available in Supplementary Table 4. All western blots represent data from 2-3 independent repeats. Unprocessed blots are shown in Supplementary Fig. 9.

Supplementary Figure 4 GSC bioenergetics is downregulated in AMPK-silenced GSCs.

a, b, Molecular network of most downregulated glycolysis genes (a) and mitochondrial genes (b) in two AMPKβ1-sienced GSCs. Node colour denotes extent of downregulation (darker = greater downregulation). Ingenuity Pathway was also used to confirm NetWalker analysis. c, Heatmap showing differentially expressed genes in indicated pathways in AMPKβ1 shRNA GSCs relative to NT shRNA expressing GSCs. d. Q-RTPCR analysis of HIF1α, GABPA and their target genes in GSCs using a second AMPKβ1 shRNA (Average of two independent experiments). +p = 0.001; * 0.01; $* 0.002; # 0.006; ** 0.003; $0.02; +* 0.05. e, WB of indicated proteins in GSCs expressing NT or AMPKβ1shRNA. f, Glucose uptake (at 30 min) in GSCs expressing NT or AMPKβ1shRNA. . (n = 3). g, WB of GLUT3 in GSCs expressing NT or AMPKβ1shRNA. h, WB of PGC1α in nontarget and AMPKβ1 shRNA expressing GSCs. i, WB using anti-acetyl antibody following immunoprecipitation with PGC1α antibody in control (NT) and AMPK silenced GSC. j, WB showing overexpression of Myc-tagged PGC1α in GSCs. k, Q-PCR analysis of ND4 and β actin DNA in control and AMPK-silenced GSCs overexpressing PGC1α. (Average of two independent experiments). ns = nonsignificant. l, Oxygen consumption rate (OCR) in control and AMPK-silenced GSCs overexpressing PGC1α. (Average of two independent experiments). m, Q-RT-PCR analysis of ND4 and β actin RNA in control and AMPK-silenced GSCs treated with SIRT1720 for 48h. (Average of two independent experiments). n, OCR in control and AMPK-silenced GSCs treated with SIRT1720. (Average of two independent experiments). Error bars; mean +/- S.D. Statistical significance in above experiments was assessed using two-tailed t-test. n values represent independent experiments. Source data are available in Supplementary Table 4. All western blots represent data from 2 (e, h, I, j) or 3 (e, g) independent repeats. Unprocessed blots are shown in Supplementary Fig. 9.

Supplementary Figure 5 Glucose metabolism is diminished in AMPK-silenced GBM cells.

a-h, GSC10 cells were fed with U13C glucose and 13C carbon incorporation in the metabolites of glycolysis and TCA cycle was quantitated by HPLC/Mass spectrometry. Shown are several metabolites of glycolysis (G6P/F6P, G3P), pentose phosphate pathway (6PG, R5P/X5P, S7P) and TCA cycle (Suc, Mal) that show reduced incorporation of 13C glucose carbon at indicated times. Note: Aspartate is generated from the TCA cycle metabolite oxaloacetate. G6P/F6P, glucose 6-phosphate/fructose 6-phosphate; G3P, glucose 3-phosphate; 6-PG, 6-phosphogluconate; R5P/X5P, ribulose 5-phosphate/xylulose 5-phosphate; S7P, sedoheptulose 7-phosphate; Suc; succinate; Mal, malate; Asp, aspartate. Duplicate samples were processed. ** p = 0.003 (M+6 G6P/F6P; M+6 and M+5 6PG); # p = 0.001 (M+ 2 G3P); * p ≤ 0.05 (M+5 R5P/X5P; M+7 and M+6 S7P; M+2 Suc; M+3 Mal; M+3 and M+2 Asp). Source data are available in Supplementary Table 4.

Supplementary Figure 6 Evidence for CREB1 regulation of GABPA and HIF1α.

a, b, UCSC Genome Browser screenshot showing multiple ENCODE CREB1 ChIP-seq datasets in a variety of cell types. "UCSC Genes" track at the top shows the genomic location of GABPA (a) and HIF1α (b). Thick bars indicate UTRs and coding regions; thin bars indicate introns (arrows show direction of transcription). Coloured tracks below indicate CREB1 ChIP-seq binding signals in various cell types. Higher peaks indicate genomic locations bound by CREB1 in the indicated cell type.

Supplementary Figure 7 Active CREB1 is highly expressed in GBM and is regulated by AMPK.

a,b, WB of CREB1 and pCREB1 in human GBM compared to normal brain tissue (a), and in primary GSC lines compared to normal human astrocytes (NHA) (b). c, Q-RTPCR analysis of CREB1 in NHA and GSC lines. (n = 3). *p ≤ 0.0005. d, High expression of CREB1, CREB1-binding protein EP300, HIF1α, GABPA and TFAM in GBM relative to normal human brain tissue (Source: TCGA). * p ≤ 0.001;** ≤ 0.005; ++ = 0.01. The edges on the boxplots indicate the first and 3rd quartile (25th- and 75th percentile) of the data, with the line in the middle being the median. The whiskers on the boxplots extend another 1.5x of the inter-quartile range (between 25%-75% range of data) from the edges of the boxes, respectively. e, Kaplan-Meier survival plots of LGG patients expressing CREB1 (Source: TCGA). f, Immunocytochemistry using pCREB1 antibody in NHA treated with the AMPK activating small molecule 991 for 30 minutes. Nuclei were detected with DAPI. Scale bar 50 μm. g, Quantification of nuclear pCREB1 signal intensity in vehicle or 991-treated NHA. *p < 0.0001. (n=36 cells/condition). h, IHC of pAMPK and pCREB in GSC326 tumours. Nuclear staining is shown using DAPI. Scale bar 50 μm. Note: Mainly nuclear pCREB but both nuclear and cytoplasmic pAMPK; nuclear pAMPK co-localizes with pCREB (inset). i, WB of pAMPK and pACC and pCREB1 in AMPK α2, β1, γ1 overexpressing NHA. Note: Stabilization of β1 and β2 is increased in cells overexpressing α2 and γ1. j, Viability of NHA overexpressing AMPK β1 subunit. (n = 3). NS = nonsignificant. k, WB of pCREB1, pACC, pPKA substrates, CREB1 and ACC in NT and AMPKβ1 shRNA expressing GSCs treated with the PKA activator forskolin. Error bars; mean +/- S.D. Statistical significance in above experiments was assessed using Student’s two-tailed t-test, except (d) where Welch’s t test was used. n values present independent experiments unless stated otherwise. Source data are available in Supplementary Table 4. All western blots represent data from 2 (i, k) or 3 (a, b) independent repeats. Scanned images of unprocessed blots are shown in Supplementary Fig. 9.

Supplementary Figure 8 Adult mice with global AMPK deletion has normal lifespan.

a, WB showing efficiency of Cre-recombination in the indicated tissues. Expression of total and active AMPK in control and AMPK-deleted tissues in mice are shown. β actin was used as loading control. Note that AMPK α subunits are unstable in the absence of β subunits. b, Kaplan-Meier survival curve of control and AMPK β subunit-deleted mice. (n = 12 mice/genotype). c, Body weight of control and AMPK β subunit-deleted mice are shown. Note: AMPK α subunit deleted mice (genotype #2) also had normal lifespan and bodyweight. (n = 12 mice/genotype). Error bars; mean +/- S.D. Statistical significance in above experiments was assessed using two-tailed t-test.

Supplementary Figure 9

Unprocessed scans of all western blot data.

Supplementary information

Supplementary Information

Supplementary Figures 1–9, Supplementary Table and Supplementary Video legends

Reporting Summary

Supplementary Table 1

Antibody name, vendor, catalogue number and dilution information

Supplementary Table 2

Silencing RNA ID and sequence information

Supplementary Table 3

Primer sequence information

Supplementary Table 4

Supplementary Source data

Supplementary Video 1

Normal proliferation of GSC10 line in the presence of non-target shRNA

Supplementary Video 2

Apoptosis of GSC10 line in the presence of AMPK-β1 shRNA

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chhipa, R.R., Fan, Q., Anderson, J. et al. AMP kinase promotes glioblastoma bioenergetics and tumour growth. Nat Cell Biol 20, 823–835 (2018). https://doi.org/10.1038/s41556-018-0126-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0126-z

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer