Skip to main content

Advertisement

Log in

Inhibition of epithelial-mesenchymal transition in bladder cancer cells via modulation of mTOR signalling

  • Original Article
  • Published:
Tumor Biology

Abstract

Mounting evidence suggests that signalling cross-talk plays a significant role in the regulation of epithelial–mesenchymal transition (EMT) in cancer cells. However, the complex network regulating the EMT in different cancer types has not been fully described yet which affects the development of novel therapeutic strategies. In the present study, we investigated the signalling pathways involved in EMT of bladder cancer cells and demonstrated the effects of two novel agents in the regulation of EMT. Myrtucommulone-A (MC-A) and thymoquinone (TQ) have been shown to possess anti-cancer properties. However, their targets in the regulation of cancer cell behavior are not well defined. Here, we defined the effects of two putative anti-cancer agents on bladder cancer cell migration and their possible intracellular targets in the regulation of EMT. Our results suggest that MC-A or TQ treatment affected N-cadherin, Snail, Slug, and β-catenin expressions and effectively attenuated mTOR activity. The downstream components in mTOR signalling were also affected. MC-A treatment resulted in the concomitant inhibition of extracellular matrix-regulated protein kinases 1 and 2 (ERK 1/2), p38 mitogen-activated protein kinase (MAPK) and Src activity. On the other hand, TQ treatment increased Src activity while exerting no effect on ERK 1/2 or p38 MAPK activity. Given the stronger inhibition of EMT-related markers in MC-A-treated samples, we concluded that this effect might be due to collective inhibition of multiple signalling pathways which result in a decrease in their cross-talk in bladder cancer cells. Overall, the data in this study proposes novel action mechanisms for MC-A or TQ in bladder cancer cells and highlights the potential use of these active compounds in the regulation of EMT.

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

References

  1. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 2014;15(3):178–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Thiery JP et al. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–90.

    Article  CAS  PubMed  Google Scholar 

  3. De Craene B, Berx G. Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013;13(2):97–110.

    Article  CAS  PubMed  Google Scholar 

  4. Moustakas A, Heldin CH. Signaling networks guiding epithelial-mesenchymal transitions during embryogenesis and cancer progression. Cancer Sci. 2007;98(10):1512–20.

    Article  CAS  PubMed  Google Scholar 

  5. Derynck R, Muthusamy BP, Saeteurn KY. Signaling pathway cooperation in TGF-β-induced epithelial-mesenchymal transition. Curr Opin Cell Biol. 2014;31:56–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ikushima H, Miyazono K. TGF-β signal transduction spreading to a wider field: a broad variety of mechanisms for context-dependent effects of TGF-β. Cell Tissue Res. 2012;347(1):37–49.

    Article  CAS  PubMed  Google Scholar 

  7. Lamouille S, Derynck R. Cell size and invasion in TGF-beta-induced epithelial to mesenchymal transition is regulated by activation of the mTOR pathway. J Cell Biol. 2007;178(3):437–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lin G et al. The dual PI3K/mTOR inhibitor NVP-BEZ235 prevents epithelial-mesenchymal transition induced by hypoxia and TGF-β1. Eur J Pharmacol. 2014;729:45–53.

    Article  CAS  PubMed  Google Scholar 

  9. Beauchamp EM, Platanias LC. The evolution of the TOR pathway and its role in cancer. Oncogene. 2013;32(34):3923–32.

    Article  CAS  PubMed  Google Scholar 

  10. Menon S, Manning BD. Common corruption of the mTOR signaling network in human tumors. Oncogene Suppl. 2008;2:S43–51.

    Article  CAS  Google Scholar 

  11. Zaytseva YY et al. mTOR inhibitors in cancer therapy. Cancer Lett. 2012;319(1):1–7.

    Article  CAS  PubMed  Google Scholar 

  12. Guarino M. Src signaling in cancer invasion. J Cell Physiol. 2010;223(1):14–26.

    CAS  PubMed  Google Scholar 

  13. Putzke AP et al. Metastatic progression of prostate cancer and e-cadherin regulation by zeb1 and SRC family kinases. Am J Pathol. 2011;179(1):400–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bromann PA, Korkaya H, Courtneidge SA. The interplay between Src family kinases and receptor tyrosine kinases. Oncogene. 2004;23:7957–68.

    Article  CAS  PubMed  Google Scholar 

  15. Di Florio A et al. Src kinase activity coordinates cell adhesion and spreading with activation of mammalian target of rapamycin in pancreatic endocrine tumour cells. Endocr Relat Cancer. 2011;18(5):541–54.

    Article  CAS  PubMed  Google Scholar 

  16. Gonugunta VK et al. Inhibition of mTOR signaling reduces PELP1-mediated tumor growth and therapy resistance. Mol Cancer Ther. 2014;13(6):1578–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li XY et al. RIG-I modulates Src-mediated AKT activation to restrain leukemic stemness. Mol Cell. 2014;53(3):407–19.

    Article  CAS  PubMed  Google Scholar 

  18. Tanno S et al. AKT activation up-regulates insulin-like growth factor I receptor expression and promotes invasiveness of human pancreatic cancer cells. Cancer Res. 2001;61(2):589–93.

    CAS  PubMed  Google Scholar 

  19. Vojtechová M et al. Regulation of mTORC1 signaling by Src kinase activity is Akt1-independent in RSV-transformed cells. Neoplasia. 2008;10(2):99–107.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sen B, Johnson FM. Regulation of SRC family kinases in human cancers. J Signal Transduct. 2011;2011:865819.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Chen B et al. Rapamycin enhances the anti-cancer effect of dasatinib by suppressing Src/PI3K/mTOR pathway in NSCLC cells. PLoS One. 2015;10(6):e0129663.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Yori JL et al. Combined SFK/mTOR inhibition prevents rapamycin-induced feedback activation of AKT and elicits efficient tumor regression. Cancer Res. 2014;74(17):4762–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Hansel DE et al. Mammalian target of rapamycin (mTOR) regulates cellular proliferation and tumor growth in urothelial carcinoma. Am J Pathol. 2010;176(6):3062–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yuge K et al. Nicotine induces tumor growth and chemoresistance through activation of the PI3K/Akt/mTOR pathway in bladder cancer. Mol Cancer Ther. 2015;14(9):2112–20.

    Article  CAS  PubMed  Google Scholar 

  25. Kyou Kwon J et al. Dual inhibition by S6K1 and Elf4E is essential for controlling cellular growth and invasion in bladder cancer. Urol Oncol. 2014;32(1):51. e27-35.

    PubMed  Google Scholar 

  26. Matsushima M et al. Intravesical dual PI3K/mTOR complex 1/2 inhibitor NVP-BEZ235 therapy in an orthotopic bladder cancer model. Int J Oncol. 2015;47(1):377–83.

    PubMed  Google Scholar 

  27. Moon du G et al. NVP-BEZ235, a dual PI3K/mTOR inhibitor synergistically potentiates the antitumor effects of cisplatin in bladder cancer cells. Int J Oncol. 2014;45(3):1027–35.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Saini S et al. Curcumin modulates microRNA-203-mediated regulation of the Src-Akt axis in bladder cancer. Cancer Prev Res (Phila). 2011;4(10):1698–709.

    Article  CAS  PubMed Central  Google Scholar 

  29. Boyer B, Bourgeois Y, Poupon MF. Src kinase contributes to the metastatic spread of carcinoma cells. Oncogene. 2002;21(15):2347–56.

    Article  CAS  PubMed  Google Scholar 

  30. Chiang GJ et al. The Src-family kinase inhibitor PP2 suppresses the in vitro invasive phenotype of bladder carcinoma cells via modulation of Akt. BJU Int. 2005;96(3):416–22.

    Article  CAS  PubMed  Google Scholar 

  31. Grandjenette C et al. Dual induction of mitochondrial apoptosis and senescence in chronic myelogenous leukemia by myrtucommulone A. Anti Cancer Agents Med Chem. 2015;15(3):363–73.

    Article  CAS  Google Scholar 

  32. Iskender B et al. Myrtucommulone-A treatment decreases pluripotency- and multipotency-associated marker expression in bladder cancer cell line HTB-9. J Nat Med. 2015;69(4):543–54.

    Article  CAS  PubMed  Google Scholar 

  33. Izgi K et al. Myrtucommulone-A induces both extrinsic and intrinsic apoptotic pathways in cancer cells. J Biochem Mol Toxicol. 2015;29(9):432–39. doi:10.1002/jbt.21716.

    Article  CAS  Google Scholar 

  34. Iskender B et al. Priming hMSCs with a putative anti-cancer compound, myrtucommulone-a: a way to harness hMSC cytokine expression via modulating PI3K/Akt pathway? Tumour Biol. 2015. doi:10.1007/s13277-015-3995-9.

    Google Scholar 

  35. Schneider-Stock R et al. Thymoquinone: fifty years of success in the battle against cancer models. Drug Discov Today. 2014;19(1):18–30.

    Article  CAS  PubMed  Google Scholar 

  36. Mu GG et al. Thymoquinone pretreatment overcomes the insensitivity and potentiates the antitumor effect of gemcitabine through abrogation of Notch1, PI3K/Akt/mTOR regulated signaling pathways in pancreatic cancer. Dig Dis Sci. 2015;60(4):1067–80.

    Article  CAS  PubMed  Google Scholar 

  37. Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12(1):9–22.

    Article  CAS  PubMed  Google Scholar 

  38. She QB et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumors. Cancer Cell. 2010;18(1):39–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Bai J, et al. Identification of drugs as single agents or in combination to prevent carcinoma dissemination in a microfluidic 3D environment. Oncotarget. 2015. doi:10.18632/oncotarget.5464.

  40. Zhao L et al. Benzidine induces epithelial-mesenchymal transition in human uroepithelial cells through ERK1/2 pathway. Biochem Biophys Res Commun. 2015;459(4):643–9.

    Article  CAS  PubMed  Google Scholar 

  41. Zeisberg M, Neilson EG. Biomarkers for epithelial-mesenchymal transitions. J Clin Invest. 2009;119(6):1429–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Lindsey S, Langhans SA. Crosstalk of oncogenic signaling pathways during epithelial-mesenchymal transition. Front Oncol. 2014;4:358.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Chang L et al. Acquisition of epithelial-mesenchymal transition and cancer stem cell phenotypes is associated with activation of the PI3K/Akt/mTOR pathway in prostate cancer radioresistance. Cell Death Dis. 2013;4:e875.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Zhang L et al. FTY720 reduces migration and invasion of human glioblastoma cell lines via inhibiting the PI3K/AKT/mTOR/p70S6K signaling pathway. Tumour Biol. 2014;35(11):10707–14.

    Article  CAS  PubMed  Google Scholar 

  45. Gulhati P et al. mTORC1 and mTORC2 regulate EMT, motility, and metastasis of colorectal cancer via RhoA and Rac1 signaling pathways. Cancer Res. 2011;71(9):3246–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Lamouille S et al. TGF-β-induced activation of mTOR complex 2 drives epithelial-mesenchymal transition and cell invasion. J Cell Sci. 2012;125(Pt 5):1259–73.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Pópulo H, Lopes JM, Soares P. The mTOR signalling pathway in human cancer. Int J Mol Sci. 2012;13(2):1886–918.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Chen X et al. mTOR regulate EMT through RhoA and Rac1 pathway in prostate cancer. Mol Carcinog. 2015;54(10):1086–95.

    Article  CAS  PubMed  Google Scholar 

  49. Han B et al. Metformin inhibits thyroid cancer cell growth, migration, and EMT through the mTOR pathway. Tumour Biol. 2015;36(8):6295–304.

    Article  CAS  PubMed  Google Scholar 

  50. Lau MT, So WK, Leung PC. Fibroblast growth factor 2 induces E-cadherin down-regulation via PI3K/Akt/mTOR and MAPK/ERK signaling in ovarian cancer cells. PLoS One. 2013;8(3):e59083.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Zong H et al. Inhibition of mTOR pathway attenuates migration and invasion of gallbladder cancer via EMT inhibition. Mol Biol Rep. 2014;41(7):4507–12.

    Article  CAS  PubMed  Google Scholar 

  52. Islam SS, et al. Simultaneous targeting of bladder tumor growth, survival, and epithelial-to-mesenchymal transition with a novel therapeutic combination of acetazolamide (AZ) and sulforaphane (SFN). Target Oncol. 2015.

  53. Shorning BY, Griffiths D, Clarke AR. Lkb1 and Pten synergise to suppress mTOR-mediated tumorigenesis and epithelial-mesenchymal transition in the mouse bladder. PLoS One. 2011;6(1):e16209.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Cattan N et al. Establishment of two new human bladder carcinoma cell lines, CAL 29 and CAL 185. Comparative study of cell scattering and epithelial to mesenchyme transition induced by growth factors. Br J Cancer. 2001;85(9):1412–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Liang Z et al. Inhibition of tobacco smoke-induced bladder MAPK activation and epithelial-mesenchymal transition in mice by curcumin. Int J Clin Exp Pathol. 2015;8(5):4503–13.

    PubMed  PubMed Central  Google Scholar 

  56. Woo CC et al. Thymoquinone inhibits tumor growth and induces apoptosis in a breast cancer xenograft mouse model: the role of p38 MAPK and ROS. PLoS One. 2013;8(10):e75356.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Yang J et al. Thymoquinone inhibits proliferation and invasion of human nonsmall-cell lung cancer cells via ERK pathway. Tumour Biol. 2015;36(1):259–69.

    Article  CAS  PubMed  Google Scholar 

  58. Green TP et al. Preclinical anticancer activity of the potent, oral Src inhibitor AZD0530. Mol Oncol. 2009;3(3):248–61.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We are grateful to Dr. Maël Charpentier (Institut für Organische Chemie der Universität des Saarlandes in Saarbrücken, Germany) for providing MC-A.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Banu Iskender.

Ethics declarations

Conflicts of interest

None

Grant support

This study was supported by the grants from the The Scientific and Technological Research Council of Turkey (nos 115S042, 114S542 and 113S927).

Electronic supplementary material

Below is the link to the electronic supplementary material.

ESM 1

The effects of MC-A or TQ treatment on clonogenicity of T24 cells. MC-A, TQ or rapamycin treatment reduced colony formation in a dose dependent manner. MC-A, rapamycin and TQ reduced the colony size (a), number (b) and changed colony morphology (c) compared to the control cells (PDF 11713 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Iskender, B., Izgi, K., Hizar, E. et al. Inhibition of epithelial-mesenchymal transition in bladder cancer cells via modulation of mTOR signalling. Tumor Biol. 37, 8281–8291 (2016). https://doi.org/10.1007/s13277-015-4695-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13277-015-4695-1

Keywords

Navigation