Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth.
Journal: 2011/May - Proceedings of the National Academy of Sciences of the United States of America
ISSN: 1091-6490
Abstract:
Although aerobic glycolysis (the Warburg effect) is a hallmark of cancer, key questions, including when, how, and why cancer cells become highly glycolytic, remain less clear. For a largely unknown regulatory mechanism, a rate-limiting glycolytic enzyme pyruvate kinase M2 (PKM2) isoform is exclusively expressed in embryonic, proliferating, and tumor cells, and plays an essential role in tumor metabolism and growth. Because the receptor tyrosine kinase/PI3K/AKT/mammalian target of rapamycin (RTK/PI3K/AKT/mTOR) signaling cascade is a frequently altered pathway in cancer, we explored its potential role in cancer metabolism. We identified mTOR as a central activator of the Warburg effect by inducing PKM2 and other glycolytic enzymes under normoxic conditions. PKM2 level was augmented in mouse kidney tumors due to deficiency of tuberous sclerosis complex 2 and consequent mTOR activation, and was reduced in human cancer cells by mTOR suppression. mTOR up-regulation of PKM2 expression was through hypoxia-inducible factor 1α (HIF1α)-mediated transcription activation, and c-Myc-heterogeneous nuclear ribonucleoproteins (hnRNPs)-dependent regulation of PKM2 gene splicing. Disruption of PKM2 suppressed oncogenic mTOR-mediated tumorigenesis. Unlike normal cells, mTOR hyperactive cells were more sensitive to inhibition of mTOR or glycolysis. Dual suppression of mTOR and glycolysis synergistically blunted the proliferation and tumor development of mTOR hyperactive cells. Even though aerobic glycolysis is not required for breach of senescence for immortalization and transformation, the frequently deregulated mTOR signaling during multistep oncogenic processes could contribute to the development of the Warburg effect in many cancers. Components of the mTOR/HIF1α/Myc-hnRNPs/PKM2 glycolysis signaling network could be targeted for the treatment of cancer caused by an aberrant RTK/PI3K/AKT/mTOR signaling pathway.
Relations:
Content
Citations
(182)
References
(54)
Diseases
(1)
Drugs
(1)
Chemicals
(1)
Genes
(2)
Organisms
(3)
Processes
(5)
Similar articles
Articles by the same authors
Discussion board
Proc Natl Acad Sci U S A 108(10): 4129-4134

Mammalian target of rapamycin up-regulation of pyruvate kinase isoenzyme type M2 is critical for aerobic glycolysis and tumor growth

+11 authors

Materials and Methods

For descriptions of reagents, cell culture, cell proliferation assay, immunoblotting, measurements of glucose and lactate, siRNA gene knockdown, generation of stable gene knockdown cell lines, mRNA expression profiling analysis, RT-qPCR, real-time PCR analysis of ChIP DNA, mouse kidney tumor assessment, xenografting tumorigenesis and treatment, and statistics, see SI Materials and Methods.

State Key Laboratory of Medical Molecular Biology, Department of Physiology and Pathophysiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100005, China;
School of Life Sciences, Xia Men University, Xiamen 361005, China;
State Key Laboratory of Molecular Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China;
Division of Translational Medicine, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115;
Mouse Genome Informatics Team, The Jackson Laboratory, Bar Harbor, ME 04609;
Division of Medical Oncology, Department of Internal Medicine, Ohio State University, Columbus, OH 43210; and
Laboratory of Molecular Oncology, Beijing Cancer Hospital/Institute, School of Oncology, Peking University, Beijing 100142, China
Corresponding author.
To whom correspondence may be addressed. E-mail: nc.ude.umx@uoyh or moc.liamg@6002gnahzbh.
Edited by Tak Wah Mak, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute at Princess Margaret Hospital, University Health Network, Toronto, ON, Canada, and approved January 26, 2011 (received for review October 6, 2010)

Author contributions: Q.S., X.C., H. You, and H.Z. designed research; Q.S., X.C., J.M., H.P., F.W., X.Z., Y.W., Y.J., H. Yang, R.C., L.C., Y.Z., and J.G. performed research; H.O., T.C., H. You, and D.K. contributed new reagents/analytic tools; Q.S., X.C., M.-R.W., Y.L., H. You, D.K., and H.Z. analyzed data; and Q.S., X.C., H. You, and H.Z. wrote the paper.

Edited by Tak Wah Mak, The Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute at Princess Margaret Hospital, University Health Network, Toronto, ON, Canada, and approved January 26, 2011 (received for review October 6, 2010)

Abstract

Although aerobic glycolysis (the Warburg effect) is a hallmark of cancer, key questions, including when, how, and why cancer cells become highly glycolytic, remain less clear. For a largely unknown regulatory mechanism, a rate-limiting glycolytic enzyme pyruvate kinase M2 (PKM2) isoform is exclusively expressed in embryonic, proliferating, and tumor cells, and plays an essential role in tumor metabolism and growth. Because the receptor tyrosine kinase/PI3K/AKT/mammalian target of rapamycin (RTK/PI3K/AKT/mTOR) signaling cascade is a frequently altered pathway in cancer, we explored its potential role in cancer metabolism. We identified mTOR as a central activator of the Warburg effect by inducing PKM2 and other glycolytic enzymes under normoxic conditions. PKM2 level was augmented in mouse kidney tumors due to deficiency of tuberous sclerosis complex 2 and consequent mTOR activation, and was reduced in human cancer cells by mTOR suppression. mTOR up-regulation of PKM2 expression was through hypoxia-inducible factor 1α (HIF1α)-mediated transcription activation, and c-Myc–heterogeneous nuclear ribonucleoproteins (hnRNPs)-dependent regulation of PKM2 gene splicing. Disruption of PKM2 suppressed oncogenic mTOR-mediated tumorigenesis. Unlike normal cells, mTOR hyperactive cells were more sensitive to inhibition of mTOR or glycolysis. Dual suppression of mTOR and glycolysis synergistically blunted the proliferation and tumor development of mTOR hyperactive cells. Even though aerobic glycolysis is not required for breach of senescence for immortalization and transformation, the frequently deregulated mTOR signaling during multistep oncogenic processes could contribute to the development of the Warburg effect in many cancers. Components of the mTOR/HIF1α/Myc–hnRNPs/PKM2 glycolysis signaling network could be targeted for the treatment of cancer caused by an aberrant RTK/PI3K/AKT/mTOR signaling pathway.

Keywords: PTEN, tuberous sclerosis 1, hexokinase II, lactate dehydrogenase-B, glyceraldehyde 3-phosphate dehydrogenase
Abstract

Unlike in normal cells, glycolysis is induced by hypoxia, and cancer cells preferentially metabolize glucose by glycolysis, even in an aerobic environment (13). Increased glucose consumption and an elevated rate of lactate production by cancer cells are characteristics of glycolysis, first described by Otto Warburg in the 1920s and thereafter known as the Warburg effect (4). Because this altered metabolism can occur even in the presence of oxygen, glycolysis presumably confers a selective advantage for the survival and proliferation of cancer cells. This catabolic process is, however, inefficient for energy production in that it generates only 2 mol of ATP, instead of an additional 36 mol through the tricarboxylic acid (TCA) cycle, in the presence of oxygen by using 1 mol of glucose (2, 3, 5).

Although the Warburg effect is a well-recognized hallmark of cancer metabolism, its regulatory mechanism is still largely obscure. Critical issues, including how and when cancer cells acquire this highly glycolytic phenotype, and its causal relationship with cancer progression, remain to be addressed. In addition, a cellular adaptation model could not explain the constitutively high rate of glycolysis in cancer cells in tissue culture conditions with 20% oxygen in vitro or in pseudohypoxic tumors in vivo. Nevertheless, changes of rate-limiting glycolytic enzymes are observed during tumor formation. Among these enzymes is pyruvate kinase, which catalyzes the dephosphorylation of phosphoenolpyruvate to pyruvate and yields one molecule of ATP independent of oxygen supply during glycolysis (6). There are four pyruvate kinase isoenzymes: PKL and PKR are encoded by the PKLR gene but under the control of different promoters; and PKM1 and PKM2 are two different splicing forms of the same mRNA transcribed from the PKM gene. The ratio of PKM1 to PKM2 is dictated by heterogeneous nuclear ribonucleoproteins (hnRNPs) (7, 8). PKL, PKR, and PKM1 are tissue-specific isoenzymes, whereas PKM2 is considered an embryonic and cancer cell-specific isoform. By still-unknown regulatory mechanisms, PKM2 is gradually replaced by the respective tissue-specific isoenzyme during embryogenesis. By contrast, tissue-specific isoenzymes are switched to PKM2 during tumorigenesis, and PKM2 plays a critical role in tumor metabolism and growth (6, 9). Because PKM2 is an inactive form of pyruvate kinase for the last step of glycolysis, the buildup of phosphoenolpyruvate is then shunted to an alternative glycolytic pathway for anabolic synthesis and cell growth (10).

The receptor tyrosine kinase/PI3K/AKT/mammalian target of rapamycin (RTK/PI3K/AKT/mTOR) signaling pathway plays a crucial role in regulating cell growth, survival, and metabolism (11, 12). Various alterations of the proto-oncogenes and tumor suppressors along this pathway mark this network as one of the most frequently dysregulated signaling cascades in cancers (1316). A major activated effector of this pathway is mTOR, a serine/threonine protein kinase. mTOR integrates a broad spectrum of input, ranging from growth factor signaling to cellular nutrient status to energy supply, for regulation of protein synthesis and cell growth (14, 17). Even though the downstream events of oncogenic mTOR leading to cancer development are still largely unknown, among numerous mTOR effectors, the proto-oncogene Myc family and hypoxia-inducible factors (HIFs) are often activated in various cancers and have been considered an “axis of evil” in cancer development (5, 18). Although the function of normal c-Myc is inhibited by physiological HIF1α signaling, oncogenic Myc and HIFs collaborate with each other (though the molecular detail of this interaction is less certain) to confer metabolic advantages to cancer cells by induction of the Warburg effect through transcriptional activation of glycolytic enzymes (19).

We initially noticed prominent glycolysis in cells with constitutively active mTOR while characterizing primary and immortalized tuberous sclerosis 1 (Tsc1)- or tuberous sclerosis 2 (Tsc2)-null MEFs. Subsequently we found that mTOR was a major positive regulator of the Warburg effect, not only in cancer cells but also, surprisingly, in benign tumor cells and even in premature senescent primary cells with activated mTOR signaling, under normoxic conditions. We then identified PKM2 as a critical glycolytic enzyme in oncogenic mTOR-induced Warburg effect. Next, we determined that HIF1α and c-Myc–hnRNPs cascades were the transducers of mTOR regulation of PKM2. Moreover, disruption of mTOR signaling, PKM2, and/or glycolysis suppressed the cell proliferation and tumorigenesis caused by oncogenic mTOR signaling. We suggest that PKM2-stimulated glycolysis contributes to the development of tumors caused by hyperactive mTOR, and therefore this cascade may be targeted for the treatment of these cancers.

Click here to view.

Acknowledgments

This study was supported by National Natural Science Foundation of China Grant 30788004, the National Basic Research Program of China 973 Program Grants 2009CB522202 and 2009CB522203, and Ministry of Education of China 111 Project B08007.

Acknowledgments

Footnotes

The authors declare no conflict of interest.

This article is a PNAS Direct Submission.

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1014769108/-/DCSupplemental.

Footnotes

References

  • 1. Pan JG, Mak TWMetabolic targeting as an anticancer strategy: Dawn of a new era? Sci STKE. 2007;2007:pe14.[PubMed][Google Scholar]
  • 2. Hsu PP, Sabatini DMCancer cell metabolism: Warburg and beyond. Cell. 2008;134:703–707.[PubMed][Google Scholar]
  • 3. Vander Heiden MG, Cantley LC, Thompson CBUnderstanding the Warburg effect: The metabolic requirements of cell proliferation. Science. 2009;324:1029–1033.[Google Scholar]
  • 4. Warburg OOn the origin of cancer cells. Science. 1956;123:309–314.[PubMed][Google Scholar]
  • 5. Yoo YG, Hayashi M, Christensen J, Huang LEAn essential role of the HIF-1alpha-c-Myc axis in malignant progression. Ann N Y Acad Sci. 2009;1177:198–204.[PubMed][Google Scholar]
  • 6. Christofk HR, et al The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452:230–233.[PubMed][Google Scholar]
  • 7. David CJ, Chen M, Assanah M, Canoll P, Manley JLHnRNP proteins controlled by c-Myc deregulate pyruvate kinase mRNA splicing in cancer. Nature. 2010;463:364–368.[Google Scholar]
  • 8. Clower CV, et al The alternative splicing repressors hnRNP A1/A2 and PTB influence pyruvate kinase isoform expression and cell metabolism. Proc Natl Acad Sci USA. 2010;107:1894–1899.[Google Scholar]
  • 9. Spoden GA, et al Pyruvate kinase isoenzyme M2 is a glycolytic sensor differentially regulating cell proliferation, cell size and apoptotic cell death dependent on glucose supply. Exp Cell Res. 2009;315:2765–2774.[PubMed][Google Scholar]
  • 10. Vander Heiden MG, et al Evidence for an alternative glycolytic pathway in rapidly proliferating cells. Science. 2010;329:1492–1499.[Google Scholar]
  • 11. Moritz A, et al Akt-RSK-S6 kinase signaling networks activated by oncogenic receptor tyrosine kinases. Sci Signal. 2010;3:ra64.[Google Scholar]
  • 12. Govindarajan B, et al Overexpression of Akt converts radial growth melanoma to vertical growth melanoma. J Clin Invest. 2007;117:719–729.[Google Scholar]
  • 13. Knobbe CB, Lapin V, Suzuki A, Mak TWThe roles of PTEN in development, physiology and tumorigenesis in mouse models: A tissue-by-tissue survey. Oncogene. 2008;27:5398–5415.[PubMed][Google Scholar]
  • 14. Efeyan A, Sabatini DMmTOR and cancer: Many loops in one pathway. Curr Opin Cell Biol. 2010;22:169–176.[Google Scholar]
  • 15. Salmena L, Carracedo A, Pandolfi PPTenets of PTEN tumor suppression. Cell. 2008;133:403–414.[PubMed][Google Scholar]
  • 16. Hill R, et al PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Res. 2010;70:7114–7124.[Google Scholar]
  • 17. Inoki K, et al TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell. 2006;126:955–968.[PubMed][Google Scholar]
  • 18. Podar K, Anderson KCA therapeutic role for targeting c-Myc/Hif-1-dependent signaling pathways. Cell Cycle. 2010;9:1722–1728.[Google Scholar]
  • 19. Dang CV, Kim JW, Gao P, Yustein JThe interplay between MYC and HIF in cancer. Nat Rev Cancer. 2008;8:51–56.[PubMed][Google Scholar]
  • 20. Kwiatkowski DJ, 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. 2002;11:525–534.[PubMed][Google Scholar]
  • 21. Potter CJ, Pedraza LG, Xu TAkt regulates growth by directly phosphorylating Tsc2. Nat Cell Biol. 2002;4:658–665.[PubMed][Google Scholar]
  • 22. Inoki K, Li Y, Zhu T, Wu J, Guan KLTSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling. Nat Cell Biol. 2002;4:648–657.[PubMed][Google Scholar]
  • 23. El-Hashemite N, Zhang H, Henske EP, Kwiatkowski DJMutation in TSC2 and activation of mammalian target of rapamycin signalling pathway in renal angiomyolipoma. Lancet. 2003;361:1348–1349.[PubMed][Google Scholar]
  • 24. Crino PB, Nathanson KL, Henske EPThe tuberous sclerosis complex. N Engl J Med. 2006;355:1345–1356.[PubMed][Google Scholar]
  • 25. Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LCIdentification of the tuberous sclerosis complex-2 tumor suppressor gene product tuberin as a target of the phosphoinositide 3-kinase/akt pathway. Mol Cell. 2002;10:151–162.[PubMed][Google Scholar]
  • 26. Bissler JJ, et al Sirolimus for angiomyolipoma in tuberous sclerosis complex or lymphangioleiomyomatosis. N Engl J Med. 2008;358:140–151.[Google Scholar]
  • 27. Zhang H, et al Loss of Tsc1/Tsc2 activates mTOR and disrupts PI3K-Akt signaling through downregulation of PDGFR. J Clin Invest. 2003;112:1223–1233.[Google Scholar]
  • 28. Keniry M, Parsons RThe role of PTEN signaling perturbations in cancer and in targeted therapy. Oncogene. 2008;27:5477–5485.[PubMed][Google Scholar]
  • 29. Hill R, et al PTEN loss accelerates KrasG12D-induced pancreatic cancer development. Cancer Res. 2010;70:7114–7124.[Google Scholar]
  • 30. Pollizzi K, et al A hypomorphic allele of Tsc2 highlights the role of TSC1/TSC2 in signaling to AKT and models mild human TSC2 alleles. Hum Mol Genet. 2009;18:2378–2387.[Google Scholar]
  • 31. Semenza GLHIF-1 mediates the Warburg effect in clear cell renal carcinoma. J Bioenerg Biomembr. 2007;39:231–234.[PubMed][Google Scholar]
  • 32. Land SC, Tee ARHypoxia-inducible factor 1alpha is regulated by the mammalian target of rapamycin (mTOR) via an mTOR signaling motif. J Biol Chem. 2007;282:20534–20543.[PubMed][Google Scholar]
  • 33. Dekanty A, Lavista-Llanos S, Irisarri M, Oldham S, Wappner PThe insulin-PI3K/TOR pathway induces a HIF-dependent transcriptional response in Drosophila by promoting nuclear localization of HIF-alpha/Sima. J Cell Sci. 2005;118:5431–5441.[PubMed][Google Scholar]
  • 34. Brugarolas J, Kaelin WG., JrDysregulation of HIF and VEGF is a unifying feature of the familial hamartoma syndromes. Cancer Cell. 2004;6:7–10.[PubMed][Google Scholar]
  • 35. Semenza GLHIF-1: Upstream and downstream of cancer metabolism. Curr Opin Genet Dev. 2010;20:51–56.[Google Scholar]
  • 36. Yun J, et al Glucose deprivation contributes to the development of KRAS pathway mutations in tumor cells. Science. 2009;325:1555–1559.[Google Scholar]
  • 37. Ma J, et al Mammalian target of rapamycin regulates murine and human cell differentiation through STAT3/p63/Jagged/Notch cascade. J Clin Invest. 2010;120:103–114.[Google Scholar]
  • 38. Düvel K, et al Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol Cell. 2010;39:171–183.[Google Scholar]
  • 39. Toschi A, et al Phospholipase D-mTOR requirement for the Warburg effect in human cancer cells. Cancer Lett. 2010;299:72–79.[Google Scholar]
  • 40. Blouin MJ, et al Loss of function of PTEN alters the relationship between glucose concentration and cell proliferation, increases glycolysis, and sensitizes cells to 2-deoxyglucose. Cancer Lett. 2010;289:246–253.[PubMed][Google Scholar]
  • 41. Elstrom RL, et al Akt stimulates aerobic glycolysis in cancer cells. Cancer Res. 2004;64:3892–3899.[PubMed][Google Scholar]
  • 42. Khatri S, Yepiskoposyan H, Gallo CA, Tandon P, Plas DRFOXO3a regulates glycolysis via transcriptional control of tumor suppressor TSC1. J Biol Chem. 2010;285:15960–15965.[Google Scholar]
  • 43. Zhang H, et al PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. J Clin Invest. 2007;117:730–738.[Google Scholar]
  • 44. Mazurek SPyruvate kinase type M2: A key regulator of the metabolic budget system in tumor cells. Int J Biochem Cell Biol. 2010 10.1016/j.biocel.2010.02.005. [[PubMed][Google Scholar]
  • 45. Semenza GL, Roth PH, Fang HM, Wang GLTranscriptional regulation of genes encoding glycolytic enzymes by hypoxia-inducible factor 1. J Biol Chem. 1994;269:23757–23763.[PubMed][Google Scholar]
  • 46. Robey IF, Lien AD, Welsh SJ, Baggett BK, Gillies RJHypoxia-inducible factor-1alpha and the glycolytic phenotype in tumors. Neoplasia. 2005;7:324–330.[Google Scholar]
  • 47. Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG., JrTSC2 regulates VEGF through mTOR-dependent and -independent pathways. Cancer Cell. 2003;4:147–158.[PubMed][Google Scholar]
  • 48. El-Hashemite N, Walker V, Zhang H, Kwiatkowski DJLoss of Tsc1 or Tsc2 induces vascular endothelial growth factor production through mammalian target of rapamycin. Cancer Res. 2003;63:5173–5177.[PubMed][Google Scholar]
  • 49. Toschi A, Lee E, Gadir N, Ohh M, Foster DADifferential dependence of hypoxia-inducible factors 1 alpha and 2 alpha on mTORC1 and mTORC2. J Biol Chem. 2008;283:34495–34499.[Google Scholar]
  • 50. Jung JE, et al STAT3 is a potential modulator of HIF-1-mediated VEGF expression in human renal carcinoma cells. FASEB J. 2005;19:1296–1298.[PubMed][Google Scholar]
  • 51. Xu Q, et al Targeting Stat3 blocks both HIF-1 and VEGF expression induced by multiple oncogenic growth signaling pathways. Oncogene. 2005;24:5552–5560.[PubMed][Google Scholar]
  • 52. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, Thompson CBThe biology of cancer: Metabolic reprogramming fuels cell growth and proliferation. Cell Metab. 2008;7:11–20.[PubMed][Google Scholar]
  • 53. Deberardinis RJ, Sayed N, Ditsworth D, Thompson CBBrick by brick: Metabolism and tumor cell growth. Curr Opin Genet Dev. 2008;18:54–61.[Google Scholar]
  • 54. Laplante M, Sabatini DMAn emerging role of mTOR in lipid biosynthesis. Curr Biol. 2009;19:R1046–R1052.[Google Scholar]
  • 55. Choo AY, et al Glucose addiction of TSC null cells is caused by failed mTORC1-dependent balancing of metabolic demand with supply. Mol Cell. 2010;38:487–499.[Google Scholar]
Collaboration tool especially designed for Life Science professionals.Drag-and-drop any entity to your messages.