DOI QR코드

DOI QR Code

LKB1/AMPK/mTOR Signaling Pathway in Non-small-cell Lung Cancer

  • Han, Dong (Respiratory Diseases Research Center, Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University) ;
  • Li, Shao-Jun (Respiratory Diseases Research Center, Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University) ;
  • Zhu, Yan-Ting (Respiratory Diseases Research Center, Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University) ;
  • Liu, Lu (Respiratory Diseases Research Center, Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University) ;
  • Li, Man-Xiang (Respiratory Diseases Research Center, Department of Respiratory Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University)
  • Published : 2013.07.30

Abstract

Links between cancer and metabolism have been suggested for a long time but compelling evidence for this hypothesis came from the recent molecular characterization of the LKB1/AMPK signaling pathway as a tumor suppressor axis. Besides the discovery of somatic mutations in the LKB1 gene in certain type of cancers, a critical emerging point was that the LKB1/AMPK axis remains generally functional and could be stimulated by pharmacological molecules such as metformin in cancer cells. In addition, AMPK plays a central role in the control of cell growth, proliferation and autophagy through the regulation of mTOR activity, which is consistently deregulated in cancer cells. Targeting of AMPK/mTOR is thus an attractive strategy in the development of therapeutic agents against non-small-cell lung cancer (NSCLC). In this review, the LKB1/AMPK/mTOR signaling pathway is described, highlighting its protective role, and opportunities for therapeutic intervention, and clinical trials in NSCLC.

Keywords

References

  1. Chapuis N, Tamburini J, Green AS, et al (2010). Perspectives on inhibiting mTOR as a future treatment strategy for hematological malignancies. Leukemia, 24, 1686-99. https://doi.org/10.1038/leu.2010.170
  2. Choo AY, Yoon SO, Kim SG, Roux PP, Blenis J (2008). Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A, 105, 17414-9. https://doi.org/10.1073/pnas.0809136105
  3. Don AS, Hogg PJ (2004). Mitochondria as cancer drug targets. Trends Mol Med, 10, 372-8. https://doi.org/10.1016/j.molmed.2004.06.005
  4. Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD (2005). Metformin and reduced risk of cancer in diabetic patients. BMJ, 330, 1304-5. https://doi.org/10.1136/bmj.38415.708634.F7
  5. Feldman ME, Apsel B, Uotila A, et al (2009). Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol, 7, e38. https://doi.org/10.1371/journal.pbio.1000038
  6. Guertin DA, Sabatini DM (2007). Defining the role of mTOR in cancer. Cancer Cell, 12, 9-22. https://doi.org/10.1016/j.ccr.2007.05.008
  7. Guertin DA, Stevens DM, Saitoh M, et al (2009). mTOR complex 2 is required for the development of prostate cancer induced by Pten loss in mice. Cancer Cell, 15, 148-59. https://doi.org/10.1016/j.ccr.2008.12.017
  8. Gwinn DM, Shackelford DB, Egan DF, et al (2008). AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell, 30, 214-26. https://doi.org/10.1016/j.molcel.2008.03.003
  9. Hahn-Windgassen A, Nogueira V, Chen CC, et al (2005). Akt activates the mammalian target of rapamycin by regulating cellular ATP level and AMPK activity. J Biol Chem, 280, 32081-9. https://doi.org/10.1074/jbc.M502876200
  10. Han S, Khuri FR, Roman J (2006). Fibronectin stimulates nonsmall cell lung carcinoma cell growth through activation of Akt/mammalian target of rapamycin/S6 kinase and inactivation of LKB1/AMP-activated protein kinase signal pathways. Cancer Res, 66, 315-23. https://doi.org/10.1158/0008-5472.CAN-05-2367
  11. Han S, Roman J (2006). Rosiglitazone suppresses human lung carcinoma cell growth through PPARgamma-dependent and PPARgamma-independent signal pathways. Mol Cancer Ther, 5, 430-7. https://doi.org/10.1158/1535-7163.MCT-05-0347
  12. Hardie DG (2007). AMP-activated/SNF1 protein kinases: conserved guardians of cellular energy. Nat Rev Mol Cell Biol, 8, 774-85. https://doi.org/10.1038/nrm2249
  13. Hardie DG (2011). AMP-activated protein kinase: an energy sensor that regulates all aspects of cell function. Genes Dev, 25, 1895-908. https://doi.org/10.1101/gad.17420111
  14. Hawley SA, Boudeau J, Reid JL, et al (2003). Complexes between the LKB1 tumor suppressor, STRAD alpha/beta and MO25 alpha/beta are upstream kinases in the AMP-activated protein kinase cascade. J Biol, 2, 28. https://doi.org/10.1186/1475-4924-2-28
  15. Hemminki A, Markie D, Tomlinson I, et al (1998). A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature, 391, 184-7. https://doi.org/10.1038/34432
  16. Holz MK, Ballif BA, Gygi SP, Blenis J (2005). mTOR and S6K1 mediate assembly of the translation preinitiation complex through dynamic protein interchange and ordered phosphorylation events. Cell, 123, 569-80. https://doi.org/10.1016/j.cell.2005.10.024
  17. Horman S, Vertommen D, Heath R, et al (2006). Insulin antagonizes ischemia-induced Thr172 phosphorylation of AMP-activated protein kinase alpha-subunits in heart via hierarchical phosphorylation of Ser485/491. J Biol Chem, 281, 5335-40.
  18. Huang J, Manning BD (2008). The TSC1-TSC2 complex: a molecular switchboard controlling cell growth. Biochem J, 412, 179-90. https://doi.org/10.1042/BJ20080281
  19. Huang X, Wullschleger S, Shpiro N, et al (2008). Important role of the LKB1-AMPK pathway in suppressing tumorigenesis in PTEN-deficient mice. Biochem J, 412, 211-21. https://doi.org/10.1042/BJ20080557
  20. Hwang SK, Piao L, Lim HT, et al (2010). Suppression of lung tumorigenesis by leucine zipper/EF hand-containing transmembrane-1. PLoS One, 5.
  21. Inoki K, Zhu T, Guan KL (2003). TSC2 mediates cellular energy response to control cell growth and survival. Cell, 115, 577-90. https://doi.org/10.1016/S0092-8674(03)00929-2
  22. Jacinto E, Loewith R, Schmidt A, et al (2004). Mammalian TOR complex 2 controls the actin cytoskeleton and is rapamycin insensitive. Nat Cell Biol, 6, 1122-8. https://doi.org/10.1038/ncb1183
  23. Jemal A, Bray F, Center M, et al (2011). Global cancer statistics. CA Cancer J Clin, 61, 69-90. https://doi.org/10.3322/caac.20107
  24. Jemal A, Siegel R, Xu J, Ward E (2010). Cancer statistics, 2010. CA Cancer J Clin, 60, 277-300. https://doi.org/10.3322/caac.20073
  25. Jenne DE, Reimann H, Nezu J, et al (1998). Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat Genet, 18, 38-43. https://doi.org/10.1038/ng0198-38
  26. Ji H, Ramsey MR, Hayes DN, et al (2007). LKB1 modulates lung cancer differentiation and metastasis. Nature, 448, 807-10. https://doi.org/10.1038/nature06030
  27. Jin HO, Hong SE, Woo SH, et al (2012). Silencing of Twist1 sensitizes NSCLC cells to cisplatin via AMPK-activated mTOR inhibition. Cell Death Dis, 3, e319. https://doi.org/10.1038/cddis.2012.63
  28. Jin Q, Feng L, Behrens C, et al (2007). Implication of AMPactivated protein kinase and Akt-regulated survivin in lung cancer chemopreventive activities of deguelin. Cancer Res, 67, 11630-9. https://doi.org/10.1158/0008-5472.CAN-07-2401
  29. Jones RG, Plas DR, Kubek S, et al (2005). AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell, 18, 283-93. https://doi.org/10.1016/j.molcel.2005.03.027
  30. Khan N, Afaq F, Khusro FH, et al (2012). Dual inhibition of phosphatidylinositol 3-kinase/Akt and mammalian target of rapamycin signaling in human nonsmall cell lung cancer cells by a dietary flavonoid fisetin. Int J Cancer, 130, 1695-705. https://doi.org/10.1002/ijc.26178
  31. Lee SO, Abdelrahim M, Yoon K, et al (2010). Inactivation of the orphan nuclear receptor TR3/Nur77 inhibits pancreatic cancer cell and tumor growth. Cancer Res, 70, 6824-36. https://doi.org/10.1158/0008-5472.CAN-10-1992
  32. Lee SO, Andey T, Jin UH, et al (2012). The nuclear receptor TR3 regulates mTORC1 signaling in lung cancer cells expressing wild-type p53. Oncogene, 31, 3265-76. https://doi.org/10.1038/onc.2011.504
  33. Liu L, Cash TP, Jones RG, et al (2006). Hypoxia-induced energy stress regulates mRNA translation and cell growth. Mol Cell, 21, 521-31. https://doi.org/10.1016/j.molcel.2006.01.010
  34. Mankouri J, Tedbury PR, Gretton S, et al (2010). Enhanced hepatitis C virus genome replication and lipid accumulation mediated by inhibition of AMP-activated protein kinase. Proc Natl Acad Sci U S A, 107, 11549-54. https://doi.org/10.1073/pnas.0912426107
  35. Matsumoto S, Iwakawa R, Takahashi K, et al (2007). Prevalence and specificity of LKB1 genetic alterations in lung cancers. Oncogene, 26, 5911-8. https://doi.org/10.1038/sj.onc.1210418
  36. Memmott RM, Gills JJ, Hollingshead M, et al (2008). Phosphatidylinositol ether lipid analogues induce AMPactivated protein kinase-dependent death in LKB1-mutant non small cell lung cancer cells. Cancer Res, 68, 580-8. https://doi.org/10.1158/0008-5472.CAN-07-3091
  37. Nanjundan M, Byers LA, Carey MS, et al (2010). Proteomic profiling identifies pathways dysregulated in non-small cell lung cancer and an inverse association of AMPK and adhesion pathways with recurrence. J Thorac Oncol, 5, 1894-904. https://doi.org/10.1097/JTO.0b013e3181f2a266
  38. Rauch A, Schellmoser S, Kraus C, et al (2001). First known microdeletion within the Wolf-Hirschhorn syndrome critical region refines genotype-phenotype correlation. Am J Med Genet, 99, 338-42. https://doi.org/10.1002/ajmg.1203
  39. Rothbart SB, Racanelli AC, Moran RG (2010). Pemetrexed indirectly activates the metabolic kinase AMPK in human carcinomas. Cancer Res, 70, 10299-309. https://doi.org/10.1158/0008-5472.CAN-10-1873
  40. Sanchez-Cespedes M, Parrella P, Esteller M, et al (2002). Inactivation of LKB1/STK11 is a common event in adenocarcinomas of the lung. Cancer Res, 62, 3659-62.
  41. Sanli T, Liu C, Rashid A, et al (2011). Lovastatin sensitizes lung cancer cells to ionizing radiation: modulation of molecular pathways of radioresistance and tumor suppression. J Thorac Oncol, 6, 439-50. https://doi.org/10.1097/JTO.0b013e3182049d8b
  42. Sanli T, Rashid A, Liu C, et al (2010). Ionizing radiation activates AMP-activated kinase (AMPK): a target for radiosensitization of human cancer cells. Int J Radiat Oncol Biol Phys, 78, 221-9. https://doi.org/10.1016/j.ijrobp.2010.03.005
  43. Shackelford DB, Shaw RJ (2009). The LKB1-AMPK pathway: metabolism and growth control in tumour suppression. Nat Rev Cancer, 9, 563-75. https://doi.org/10.1038/nrc2676
  44. Shackelford DB, Vasquez DS, Corbeil J, et al (2009). mTOR and HIF-1alpha-mediated tumor metabolism in an LKB1 mouse model of Peutz-Jeghers syndrome. Proc Natl Acad Sci U S A, 106, 11137-42. https://doi.org/10.1073/pnas.0900465106
  45. Shao JJ, Zhang AP, Qin W, et al (2012). AMP-activated protein kinase (AMPK) activation is involved in chrysin-induced growth inhibition and apoptosis in cultured A549 lung cancer cells. Biochem Biophys Res Commun, 423, 448-53. https://doi.org/10.1016/j.bbrc.2012.05.123
  46. Shaw RJ, Bardeesy N, Manning BD, et al (2004). The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell, 6, 91-9. https://doi.org/10.1016/j.ccr.2004.06.007
  47. Shaw RJ, Cantley LC (2006). Ras, PI(3)K and mTOR signalling controls tumour cell growth. Nature, 441, 424-30. https://doi.org/10.1038/nature04869
  48. Storozhuk Y, Sanli T, Hopmans SN, et al (2012). Chronic modulation of AMP-Kinase, Akt and mTOR pathways by ionizing radiation in human lung cancer xenografts. Radiat Oncol, 7, 71. https://doi.org/10.1186/1748-717X-7-71
  49. Street A, Macdonald A, Crowder K, Harris M (2004). The Hepatitis C virus NS5A protein activates a phosphoinositide 3-kinase-dependent survival signaling cascade. J Biol Chem, 279, 12232-41. https://doi.org/10.1074/jbc.M312245200
  50. Tamm I, Wang Y, Sausville E, et al (1998). IAP-family protein survivin inhibits caspase activity and apoptosis induced by Fas (CD95), Bax, caspases, and anticancer drugs. Cancer Res, 58, 5315-20.
  51. Vaira V, Lee CW, Goel HL, et al (2007). Regulation of survivin expression by IGF-1/mTOR signaling. Oncogene, 26, 2678-84. https://doi.org/10.1038/sj.onc.1210094
  52. Wang HW, Lin CP, Chiu JH, et al (2007). Reversal of inflammation-associated dihydrodiol dehydrogenases (AKR1C1 and AKR1C2) overexpression and drug resistance in nonsmall cell lung cancer cells by wogonin and chrysin. Int J Cancer, 120, 2019-27. https://doi.org/10.1002/ijc.22402
  53. Wang X, Ling MT, Guan XY, et al (2004). Identification of a novel function of TWIST, a bHLH protein, in the development of acquired taxol resistance in human cancer cells. Oncogene, 23, 474-82. https://doi.org/10.1038/sj.onc.1207128
  54. William WN, Kim JS, Liu DD, et al (2012). The impact of phosphorylated AMP-activated protein kinase expression on lung cancer survival. Ann Oncol, 23, 78-85. https://doi.org/10.1093/annonc/mdr036
  55. Woods A, Johnstone SR, Dickerson K, et al (2003). LKB1 is the upstream kinase in the AMP-activated protein kinase cascade. Curr Biol, 13, 2004-8. https://doi.org/10.1016/j.cub.2003.10.031
  56. Wullschleger S, Loewith R, Hall MN (2006). TOR signaling in growth and metabolism. Cell, 124, 471-84. https://doi.org/10.1016/j.cell.2006.01.016
  57. Zhang XK (2007). Targeting Nur77 translocation. Expert Opin Ther Targets, 11, 69-79. https://doi.org/10.1517/14728222.11.1.69
  58. Zheng B, Jeong JH, Asara JM, et al (2009). Oncogenic B-RAF negatively regulates the tumor suppressor LKB1 to promote melanoma cell proliferation. Mol Cell, 33, 237-47. https://doi.org/10.1016/j.molcel.2008.12.026

Cited by

  1. Systemic treatment in EGFR-ALK NSCLC patients: second line therapy and beyond vol.14, pp.7, 2014, https://doi.org/10.1586/14737140.2014.896210
  2. The mTOR Signalling Pathway in Cancer and the Potential mTOR Inhibitory Activities of Natural Phytochemicals vol.15, pp.16, 2014, https://doi.org/10.7314/APJCP.2014.15.16.6463
  3. Recently Emerging Signaling Landscape of Ataxia-Telangiectasia Mutated (ATM) Kinase vol.15, pp.16, 2014, https://doi.org/10.7314/APJCP.2014.15.16.6485
  4. Genetic Variations in STK11, PRKAA1, and TSC1 Associated with Prognosis for Patients with Colorectal Cancer vol.21, pp.S4, 2014, https://doi.org/10.1245/s10434-014-3729-z
  5. Identification of Specific Gene Modules in Mouse Lung Tissue Exposed to Cigarette Smoke vol.16, pp.10, 2015, https://doi.org/10.7314/APJCP.2015.16.10.4251
  6. Metformin Addition to Chemotherapy in Stage IV Non-Small Cell Lung Cancer: an Open Label Randomized Controlled Study vol.16, pp.15, 2015, https://doi.org/10.7314/APJCP.2015.16.15.6621
  7. Metformin and lung cancer risk of patients with type 2 diabetes mellitus: A meta-analysis vol.3, pp.2, 2015, https://doi.org/10.3892/br.2015.417
  8. A Single-Nucleotide Polymorphism in Serine-Threonine Kinase 11, the Gene Encoding Liver Kinase B1, Is a Risk Factor for Multiple Sclerosis vol.7, pp.1, 2015, https://doi.org/10.1177/1759091415568914
  9. Expression and clinical significance of mammalian target of rapamycin/P70 ribosomal protein S6 kinase signaling pathway in human colorectal carcinoma tissue pp.1792-1082, 2015, https://doi.org/10.3892/ol.2015.3228
  10. Reciprocal expression of p-AMPKa and p-S6 is strongly associated with the prognosis of gastric cancer vol.37, pp.4, 2016, https://doi.org/10.1007/s13277-015-4193-5
  11. Lycorine Induces Apoptosis of A549 Cells via AMPK-Mammalian Target of Rapamycin (mTOR)-S6K Signaling Pathway vol.23, pp.1643-3750, 2017, https://doi.org/10.12659/MSM.900742
  12. DDX5 promotes gastric cancer cell proliferation in vitro and in vivo through mTOR signaling pathway vol.7, pp.2045-2322, 2017, https://doi.org/10.1038/srep42876
  13. Apoptosis triggered by isoquercitrin in bladder cancer cells by activating the AMPK-activated protein kinase pathway vol.8, pp.10, 2017, https://doi.org/10.1039/C7FO00778G
  14. Differential regulation of spermatogenic process by Lkb1 isoforms in mouse testis vol.8, pp.10, 2017, https://doi.org/10.1038/cddis.2017.527
  15. Metformin in Lung Cancer: Review of in Vitro and in Vivo Animal Studies vol.9, pp.5, 2017, https://doi.org/10.3390/cancers9050045
  16. Role of Autophagy and Apoptosis in Non-Small-Cell Lung Cancer vol.18, pp.2, 2017, https://doi.org/10.3390/ijms18020367
  17. Progress in the application and mechanism of metformin in treating non-small cell lung cancer vol.13, pp.5, 2017, https://doi.org/10.3892/ol.2017.5862
  18. Cytoplasmic liver kinase B1 promotes the growth of human lung adenocarcinoma by enhancing autophagy vol.109, pp.10, 2018, https://doi.org/10.1111/cas.13746
  19. Sodium butyrate induces autophagy in colorectal cancer cells through LKB1/AMPK signaling pp.1877-8755, 2018, https://doi.org/10.1007/s13105-018-0651-z
  20. Exendin-4 enhances radiation response of prostate cancer vol.78, pp.15, 2018, https://doi.org/10.1002/pros.23687
  21. Comparative Proteomics of Chromium-Transformed Beas-2B Cells by 2D-DIGE and MALDI-TOF/TOF MS vol.185, pp.1, 2018, https://doi.org/10.1007/s12011-017-1222-9
  22. (−)-Guaiol regulates autophagic cell death depending on mTOR signaling in NSCLC vol.19, pp.8, 2018, https://doi.org/10.1080/15384047.2018.1451277