DOI QR코드

DOI QR Code

MiR-99a Inhibits Cell Proliferation and Tumorigenesis through Targeting mTOR in Human Anaplastic Thyroid Cancer

  • Huang, Hou-Gang (Department of Anaesthesiology, Yongchuan Hospital of Chongqing Medical University) ;
  • Luo, Xi (Department of Anaesthesiology, Yongchuan Hospital of Chongqing Medical University) ;
  • Wu, Shuai (Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University) ;
  • Jian, Bin (Department of Gastrointestinal Surgery, Yongchuan Hospital of Chongqing Medical University)
  • Published : 2015.07.13

Abstract

MicroRNAs (miRNAs) are emerging as critical regulators in carcinogenesis and tumor progression. Recently, miR-99a has been reported as a tumor suppressor gene in various human cancers, but its functions in the context of anaplastic thyroid cancer (ATC) remain unknown. In this study, we reported that miR-99a was commonly downregulated in ATC tissue specimens and cell lines with important functional consequences. Overexpression of miR-99a not only dramatically reduced ATC cell viability by inducing cell apoptosis and accumulation of cells at G1 phase, but also inhibited tumorigenicity in vivo. We then screened and identified a novel miR-99a target, mammalian target of rapamycin (mTOR), and it was further confirmed by luciferase assay. Up-regulation of miR-99a would markedly reduce the expression of mTOR and its downstream phosphorylated proteins (p-4E-BP1 and p-S6K1). Similar to restoring miR-99a expression, mTOR down-regulation suppressed cell viability and increased cell apoptosis, whereas restoration of mTOR expression significantly reversed the miR-99a antitumor activity and the inhibition of mTOR/p-4E-BP1/p-S6K1 signal pathway profile. In clinical specimens and cell lines, mTOR was commonly overexpressed and its protein levels were statistically inversely correlated with miR-99a expression. Taken together, our results demonstrated for the first time that miR-99a functions as a tumor suppressor and plays an important role in inhibiting the tumorigenesis through targeting the mTOR/p-4E-BP1/p-S6K1 pathway in ATC cells. Given these, miR-99a may serve as a novel prognostic/diagnostic and therapeutic target for treating ATC.

Keywords

References

  1. Ain KB (1998). Anaplastic thyroid carcinoma: behavior, biology, and therapeutic approaches. Thyroid, 8, 715-26. https://doi.org/10.1089/thy.1998.8.715
  2. Bartel DP (2009). MicroRNAs: target recognition and regulatory functions. Cell, 136, 215-33. https://doi.org/10.1016/j.cell.2009.01.002
  3. Bjornsti MA, Houghton PJ (2004). The TOR pathway: a target for cancer therapy. Nat Rev Cancer, 4, 335-48. https://doi.org/10.1038/nrc1362
  4. Calin GA, Croce CM (2006). MicroRNA signatures in human cancers. Nat Rev Cancer, 6, 857-66. https://doi.org/10.1038/nrc1997
  5. Catto JW, Miah S, Owen HC, et al (2009). Distinct microRNA alterations characterize high- and low-grade bladder cancer. Cancer Res, 69, 8472-81. https://doi.org/10.1158/0008-5472.CAN-09-0744
  6. Cui L, Zhou H, Zhao H, et al (2012). MicroRNA-99a induces G1-phase cell cycle arrest and suppresses tumorigenicity in renal cell carcinoma. BMC Cancer, 12, 546. https://doi.org/10.1186/1471-2407-12-546
  7. Doghman M, El Wakil A, Cardinaud B, et al (2010). Regulation of insulin-like growth factor-mammalian target of rapamycin signaling by microRNA in childhood adrenocortical tumors. Cancer Res, 70, 4666-75. https://doi.org/10.1158/0008-5472.CAN-09-3970
  8. Ekman S, Wynes MW, Hirsch FR (2012). The mTOR pathway in lung cancer and implications for therapy and biomarker analysis. J Thorac Oncol, 7, 947-53. https://doi.org/10.1097/JTO.0b013e31825581bd
  9. Esquela-Kerscher A, Slack FJ (2006). Oncomirs - microRNAs with a role in cancer. Nat Rev Cancer, 6, 259-69. https://doi.org/10.1038/nrc1840
  10. Fang Y, Xue JL, Shen Q, et al (2012). MicroRNA-7 inhibits tumor growth and metastasis by targeting the phosphoinositide 3-kinase/Akt pathway in hepatocellular carcinoma. Hepatology, 55, 1852-62. https://doi.org/10.1002/hep.25576
  11. Fingar DC, Richardson CJ, Tee AR, et al (2004). mTOR controls cell cycle progression through its cell growth effectors S6K1 and 4E-BP1/eukaryotic translation initiation factor 4E. Mol Cell Biol, 24, 200-16. https://doi.org/10.1128/MCB.24.1.200-216.2004
  12. Gao W, Shen H, Liu L, et al (2011). MiR-21 overexpression in human primary squamous cell lung carcinoma is associated with poor patient prognosis. J Cancer Res Clin Oncol, 137, 557-66. https://doi.org/10.1007/s00432-010-0918-4
  13. Gera JF, Mellinghoff IK, Shi Y, et al (2004). AKT activity determines sensitivity to mammalian target of rapamycin (mTOR) inhibitors by regulating cyclin D1 and c-myc expression. J Biol Chem, 279, 2737-46. https://doi.org/10.1074/jbc.M309999200
  14. Grabinski N, Ewald F, Hofmann BT, et al (2012). Combined targeting of AKT and mTOR synergistically inhibits proliferation of hepatocellular carcinoma cells. Mol Cancer, 11, 85. https://doi.org/10.1186/1476-4598-11-85
  15. 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
  16. Hara K, Maruki Y, Long X, et al (2002). Raptor, a binding partner of target of rapamycin (TOR), mediates TOR action. Cell, 110, 177-89. https://doi.org/10.1016/S0092-8674(02)00833-4
  17. He H, Jazdzewski K, Li W, et al (2005a). The role of microRNA genes in papillary thyroid carcinoma. Proc Natl Acad Sci U S A, 102, 19075-80. https://doi.org/10.1073/pnas.0509603102
  18. He L, Thomson JM, Hemann MT, et al (2005b). A microRNA polycistron as a potential human oncogene. Nature, 435, 828-33. https://doi.org/10.1038/nature03552
  19. Heinonen H, Nieminen A, Saarela M, et al (2008). Deciphering downstream gene targets of PI3K/mTOR/p70S6K pathway in breast cancer. BMC Genomics, 9, 348. https://doi.org/10.1186/1471-2164-9-348
  20. Hummel R, Hussey DJ, Haier J (2010). MicroRNAs: predictors and modifiers of chemo- and radiotherapy in different tumour types. Eur J Cancer, 46, 298-311. https://doi.org/10.1016/j.ejca.2009.10.027
  21. Iorio MV, Croce CM (2009). MicroRNAs in cancer: small molecules with a huge impact. J Clin Oncol, 27, 5848-56. https://doi.org/10.1200/JCO.2009.24.0317
  22. Jastrzebski K, Hannan KM, Tchoubrieva EB, et al (2007). Coordinate regulation of ribosome biogenesis and function by the ribosomal protein S6 kinase, a key mediator of mTOR function. Growth Factors, 25, 209-26. https://doi.org/10.1080/08977190701779101
  23. Kim DH, Sarbassov DD, Ali SM, et al (2002). mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell, 110, 163-75. https://doi.org/10.1016/S0092-8674(02)00808-5
  24. Kremer CL, Klein RR, Mendelson J, et al (2006). Expression of mTOR signaling pathway markers in prostate cancer progression. Prostate, 66, 1203-12. https://doi.org/10.1002/pros.20410
  25. Li D, Liu X, Lin L, et al (2011). MicroRNA-99a inhibits hepatocellular carcinoma growth and correlates with prognosis of patients with hepatocellular carcinoma. J Biol Chem, 286, 36677-85. https://doi.org/10.1074/jbc.M111.270561
  26. Li JC, Zhu HY, Chen TX, et al (2013). Roles of mTOR and p-mTOR in gastrointestinal stromal tumors. Asian Pac J Cancer Prev, 14, 5925-8. https://doi.org/10.7314/APJCP.2013.14.10.5925
  27. Liu L, Li F, Cardelli JA, et al (2006). Rapamycin inhibits cell motility by suppression of mTOR-mediated S6K1 and 4E-BP1 pathways. Oncogene, 25, 7029-40. https://doi.org/10.1038/sj.onc.1209691
  28. Martelli AM, Evangelisti C, Chiarini F, et al (2009). Targeting the PI3K/AKT/mTOR signaling network in acute myelogenous leukemia. Expert Opin Investig Drugs, 18, 1333-49. https://doi.org/10.1517/14728220903136775
  29. Mothe-Satney I, Yang D, Fadden P, et al (2000). Multiple mechanisms control phosphorylation of PHAS-I in five (S/T)P sites that govern translational repression. Mol Cell Biol, 20, 3558-67. https://doi.org/10.1128/MCB.20.10.3558-3567.2000
  30. Mutallip M, Nohata N, Hanazawa T, et al (2011). Glutathione S-transferase P1 (GSTP1) suppresses cell apoptosis and its regulation by miR-133alpha in head and neck squamous cell carcinoma (HNSCC). Int J Mol Med, 27, 345-52.
  31. Nam EJ, Yoon H, Kim SW, et al (2008). MicroRNA expression profiles in serous ovarian carcinoma. Clin Cancer Res, 14, 2690-5. https://doi.org/10.1158/1078-0432.CCR-07-1731
  32. Nawroth R, Stellwagen F, Schulz WA, et al (2011). S6K1 and 4E-BP1 are independent regulated and control cellular growth in bladder cancer. PLoS ONE, 6, 27509. https://doi.org/10.1371/journal.pone.0027509
  33. Oneyama C, Ikeda J, Okuzaki D, et al (2011). MicroRNA-mediated downregulation of mTOR/FGFR3 controls tumor growth induced by Src-related oncogenic pathways. Oncogene, 30, 3489-501. https://doi.org/10.1038/onc.2011.63
  34. Petroulakis E, Mamane Y, Le Bacquer O, et al (2006). mTOR signaling: implications for cancer and anticancer therapy. Br J Cancer, 94, 195-9. https://doi.org/10.1038/sj.bjc.6602902
  35. Radimerski T, Montagne J, Rintelen F, et al (2002). dS6K-regulated cell growth is dPKB/dPI (3)K-independent, but requires dPDK1. Nat Cell Biol, 4, 251-5. https://doi.org/10.1038/ncb763
  36. Shaha AR (2004). Implications of prognostic factors and risk groups in the management of differentiated thyroid cancer. Laryngoscope, 114, 393-402. https://doi.org/10.1097/00005537-200403000-00001
  37. Shibuya H, Iinuma H, Shimada R, et al (2010). Clinicopathological and prognostic value of microRNA-21 and microRNA-155 in colorectal cancer. Oncology, 79, 313-20. https://doi.org/10.1159/000323283
  38. Song YX, Yue ZY, Wang ZN, et al (2011). MicroRNA-148b is frequently down-regulated in gastric cancer and acts as a tumor suppressor by inhibiting cell proliferation. Mol Cancer, 10, 1. https://doi.org/10.1186/1476-4598-10-1
  39. Soni A, Akcakanat A, Singh G, et al (2008). eIF4E knockdown decreases breast cancer cell growth without activating Akt signaling. Mol Cancer Ther, 7, 1782-8. https://doi.org/10.1158/1535-7163.MCT-07-2357
  40. Sun D, Lee YS, Malhotra A, et al (2011). miR-99 family of MicroRNAs suppresses the expression of prostate-specific antigen and prostate cancer cell proliferation. Cancer Res, 71, 1313-24. https://doi.org/10.1158/0008-5472.CAN-10-1031
  41. Sun J, Chen Z, Tan X, et al (2013). MicroRNA-99a/100 promotes apoptosis by targeting mTOR in human esophageal squamous cell carcinoma. Med Oncol, 30, 411. https://doi.org/10.1007/s12032-012-0411-9
  42. Takakura S, Mitsutake N, Nakashima M, et al (2008). Oncogenic role of miR-17-92 cluster in anaplastic thyroid cancer cells. Cancer Sci, 99, 1147-54. https://doi.org/10.1111/j.1349-7006.2008.00800.x
  43. Tan HK, Moad AI, Tan ML (2014). The mTOR signalling pathway in cancer and the potential mTOR inhibitory activities of natural phytochemicals. Asian Pac J Cancer Prev, 15, 6463-75. https://doi.org/10.7314/APJCP.2014.15.16.6463
  44. Telford WG, King LE, Fraker PJ (1992). Comparative evaluation of several DNA binding dyes in the detection of apoptosis-associated chromatin degradation by flow cytometry. Cytometry, 13, 137-43. https://doi.org/10.1002/cyto.990130205
  45. Valencia-Sanchez MA, Liu J, Hannon GJ, et al (2006). Control of translation and mRNA degradation by miRNAs and siRNAs. Genes Dev, 20, 515-24. https://doi.org/10.1101/gad.1399806
  46. Wang JN, Xu LH, Zeng WG, et al (2015). Treatment of human thyroid carcinoma cells with the g47delta oncolytic herpes simplex virus. Asian Pac J Cancer Prev, 16, 1241-5. https://doi.org/10.7314/APJCP.2015.16.3.1241
  47. Wang JT, Huang R, Kuang AR (2014). Comparison of presentation and clinical outcome between children and young adults with differentiated thyroid cancer. Asian Pac J Cancer Prev, 15, 7271-5. https://doi.org/10.7314/APJCP.2014.15.17.7271
  48. Wong TS, Liu XB, Wong BY, et al (2008). Mature miR-184 as potential oncogenic microRNA of Squamous Cell Carcinoma of Tongue. Clin Cancer Res, 14, 2588-92. https://doi.org/10.1158/1078-0432.CCR-07-0666
  49. Zhang Y, Yang WQ, Zhu H, et al (2014). Regulation of autophagy by miR-30d impacts sensitivity of anaplastic thyroid carcinoma to cisplatin. Biochem Pharmacol, 87, 562-70. https://doi.org/10.1016/j.bcp.2013.12.004
  50. Zhang Z, Liu ZB, Ren WM, et al (2012). The miR-200 family regulates the epithelial-mesenchymal transition induced by EGF/EGFR in anaplastic thyroid cancer cells. Int J Mol Med, 30, 856-62. https://doi.org/10.3892/ijmm.2012.1059
  51. Zhou H, Huang S (2011). Role of mTOR signaling in tumor cell motility, invasion and metastasis. Curr Protein Pept Sci, 12, 30-42. https://doi.org/10.2174/138920311795659407

Cited by

  1. Differentially Expressed MicroRNAs in Meningiomas Grades I and II Suggest Shared Biomarkers with Malignant Tumors vol.8, pp.3, 2016, https://doi.org/10.3390/cancers8030031
  2. Alterations of microRNAs throughout the malignant evolution of cutaneous squamous cell carcinoma: the role of miR-497 in epithelial to mesenchymal transition of keratinocytes pp.1476-5594, 2017, https://doi.org/10.1038/onc.2017.315
  3. Chromosome 21-Encoded microRNAs (mRNAs): Impact on Down’s Syndrome and Trisomy-21 Linked Disease pp.1573-6830, 2018, https://doi.org/10.1007/s10571-017-0514-0
  4. Candidate miRNAs in human breast cancer biomarkers: a systematic review pp.1880-4233, 2017, https://doi.org/10.1007/s12282-017-0814-8
  5. Expression Profile of microRNAs and Their Targeted Pathways in Human Ovaries Detected by Next-Generation Small RNA Sequencing vol.35, pp.5, 2016, https://doi.org/10.1089/dna.2015.3176
  6. Expression analysis of lung miRNAs responding to ovine VM virus infection by RNA-seq vol.20, pp.1, 2019, https://doi.org/10.1186/s12864-018-5416-0