DOI QR코드

DOI QR Code

Tight junction protein 1 is regulated by transforming growth factor-β and contributes to cell motility in NSCLC cells

  • Lee, So Hee (Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center) ;
  • Paek, A Rome (Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center) ;
  • Yoon, Kyungsil (Lung Cancer Branch, Div. of Translational and Clinical Research I, National Cancer Center) ;
  • Kim, Seok Hyun (Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center) ;
  • Lee, Soo Young (Division of Molecular Life Sciences, Ewha Womans University) ;
  • You, Hye Jin (Cancer Cell and Molecular Biology Branch, Div. of Cancer Biology, National Cancer Center)
  • Received : 2014.02.13
  • Accepted : 2014.07.10
  • Published : 2015.02.28

Abstract

Tight junction protein 1 (TJP1), a component of tight junction, has been reported to play a role in protein networks as an adaptor protein, and TJP1 expression is altered during tumor development. Here, we found that TJP1 expression was increased at the RNA and protein levels in TGF-${\beta}$-stimulated lung cancer cells, A549. SB431542, a type-I TGF-${\beta}$ receptor inhibitor, as well as SB203580, a p38 kinase inhibitor, significantly abrogated the effect of TGF-${\beta}$ on TJP1 expression. Diphenyleneiodonium, an NADPH oxidase inhibitor, also attenuated TJP1 expression in response to TGF-${\beta}$ in lung cancer cells. When TJP1 expression was reduced by shRNA lentiviral particles in A549 cells (A549-sh TJP1), wound healing was much lower than in cells infected with control viral particles. Taken together, these data suggest that TGF-${\beta}$ enhances TJP1 expression, which may play a role beyond structural support in tight junctions during cancer development.

Keywords

References

  1. Bierie B and Moses HL (2006) Tumour microenvironment: TGFbeta: the molecular Jekyll and Hyde of cancer. Nat Rev Cancer 6, 506-520 https://doi.org/10.1038/nrc1926
  2. Gordon KJ and Blobe GC (2008) Role of transforming growth factor-beta superfamily signaling pathways in human disease. Biochim Biophys Acta 1782, 197-228 https://doi.org/10.1016/j.bbadis.2008.01.006
  3. Macias-Silva M, Abdollah S, Hoodless PA, Pirone R, Attisano L and Wrana JL (1996) MADR2 is a substrate of the TGFbeta receptor and its phosphorylation is required for nuclear accumulation and signaling. Cell 87, 1215-1224 https://doi.org/10.1016/S0092-8674(00)81817-6
  4. Massague J, Blain SW and Lo RS (2000) TGFbeta signaling in growth control, cancer, and heritable disorders. Cell 103, 295-309 https://doi.org/10.1016/S0092-8674(00)00121-5
  5. Moustakas A and Heldin CH (2005) Non-Smad TGF-beta signals. J Cell Sci 118, 3573-3584 https://doi.org/10.1242/jcs.02554
  6. Song K, Wang H, Krebs TL and Danielpour D (2006) Novel roles of Akt and mTOR in suppressing TGF-beta/ALK5-mediated Smad3 activation. EMBO J 25, 58-69 https://doi.org/10.1038/sj.emboj.7600917
  7. Watanabe H, de Caestecker MP and Yamada Y (2001) Transcriptional cross-talk between Smad, ERK1/2, and p38 mitogen-activated protein kinase pathways regulates transforming growth factor-beta-induced aggrecan gene expression in chondrogenic ATDC5 cells. J Biol Chem 276, 14466-14473 https://doi.org/10.1074/jbc.M005724200
  8. Heldin CH, Vanlandewijck M and Moustakas A (2012) Regulation of EMT by TGFbeta in cancer. FEBS Lett 586, 1959-1970 https://doi.org/10.1016/j.febslet.2012.02.037
  9. Martin TA and Jiang WG (2009) Loss of tight junction barrier function and its role in cancer metastasis. Biochim Biophys Acta 1788, 872-891 https://doi.org/10.1016/j.bbamem.2008.11.005
  10. Matter K and Balda MS (2007) Epithelial tight junctions, gene expression and nucleo-junctional interplay. J Cell Sci 120, 1505-1511 https://doi.org/10.1242/jcs.005975
  11. Shen L (2012) Tight junctions on the move: molecular mechanisms for epithelial barrier regulation. Ann N Y Acad Sci 1258, 9-18 https://doi.org/10.1111/j.1749-6632.2012.06613.x
  12. Harhaj NS and Antonetti DA (2004) Regulation of tight junctions and loss of barrier function in pathophysiology. Int J Biochem Cell Biol 36, 1206-1237 https://doi.org/10.1016/j.biocel.2003.08.007
  13. Itoh M, Furuse M, Morita K, Kubota K, Saitou M and Tsukita S (1999) Direct binding of three tight junction-associated MAGUKs, ZO-1, ZO-2, and ZO-3, with the COOH termini of claudins. J Cell Biol 147, 1351-1363 https://doi.org/10.1083/jcb.147.6.1351
  14. Tuomi S, Mai A, Nevo J et al (2009) PKCepsilon regulation of an alpha5 integrin-ZO-1 complex controls lamellae formation in migrating cancer cells. Sci Signal 2, ra32 https://doi.org/10.1126/scisignal.2000135
  15. Chen HH, Zhou XL, Shi YL and Yang J (2013) Roles of p38 MAPK and JNK in TGF-beta1-induced human alveolar epithelial to mesenchymal transition. Arch Med Res 44, 93-98 https://doi.org/10.1016/j.arcmed.2013.01.004
  16. Martin TA, Watkins G, Mansel RE and Jiang WG (2004) Loss of tight junction plaque molecules in breast cancer tissues is associated with a poor prognosis in patients with breast cancer. Eur J Cancer 40, 2717-2725 https://doi.org/10.1016/j.ejca.2004.08.008
  17. Hoover KB, Liao SY and Bryant PJ (1998) Loss of the tight junction MAGUK ZO-1 in breast cancer: relationship to glandular differentiation and loss of heterozygosity. Am J Pathol 153, 1767-1773 https://doi.org/10.1016/S0002-9440(10)65691-X
  18. Kaihara T, Kawamata H, Imura J et al (2003) Redifferentiation and ZO-1 reexpression in liver-metastasized colorectal cancer: possible association with epidermal growth factor receptor-induced tyrosine phosphorylation of ZO-1. Cancer Sci 94, 166-172 https://doi.org/10.1111/j.1349-7006.2003.tb01414.x
  19. Kleeff J, Shi X, Bode HP et al (2001) Altered expression and localization of the tight junction protein ZO-1 in primary and metastatic pancreatic cancer. Pancreas 23, 259-265 https://doi.org/10.1097/00006676-200110000-00006
  20. Ni S, Xu L, Huang J et al (2013) Increased ZO-1 expression predicts valuable prognosis in non-small cell lung cancer. Int J Clin Exp Pathol 6, 2887-2895
  21. Smalley KS, Brafford P, Haass NK, Brandner JM, Brown E and Herlyn M (2005) Up-regulated expression of zonula occludens protein-1 in human melanoma associates with N-cadherin and contributes to invasion and adhesion. Am J Pathol 166, 1541-1554 https://doi.org/10.1016/S0002-9440(10)62370-X
  22. Park MK, You HJ, Lee HJ et al (2013) Transglutaminase-2 induces N-cadherin expression in TGF-beta1-induced epithelial mesenchymal transition via c-Jun-N-terminal kinase activation by protein phosphatase 2A down-regulation. Eur J Cancer 49, 1692-1705 https://doi.org/10.1016/j.ejca.2012.11.036
  23. Brown KA, Aakre ME, Gorska AE et al (2004) Induction by transforming growth factor-beta1 of epithelial to mesenchymal transition is a rare event in vitro. Breast Cancer Res 6, R215-R231 https://doi.org/10.1186/bcr778
  24. Halder SK, Beauchamp RD and Datta PK (2005) A specific inhibitor of TGF-beta receptor kinase, SB-431542, as a potent antitumor agent for human cancers. Neoplasia 7, 509-521 https://doi.org/10.1593/neo.04640
  25. Hiraga R, Kato M, Miyagawa S and Kamata T (2013) Nox4-derived ROS signaling contributes to TGF-beta-induced epithelial-mesenchymal transition in pancreatic cancer cells. Anticancer Res 33, 4431-4438
  26. Balda MS and Matter K (2000) The tight junction protein ZO-1 and an interacting transcription factor regulate ErbB-2 expression. EMBO J 19, 2024-2033 https://doi.org/10.1093/emboj/19.9.2024
  27. Brysse A, Mestdagt M, Polette M et al (2012) Regulation of CXCL8/IL-8 expression by zonula occludens-1 in human breast cancer cells. Mol Cancer Res 10, 121-132 https://doi.org/10.1158/1541-7786.MCR-11-0180
  28. Xu J, Lim SB, Ng MY et al (2012) ZO-1 regulates Erk, Smad1/5/8, Smad2, and RhoA activities to modulate self-renewal and differentiation of mouse embryonic stem cells. Stem Cells 30, 1885-1900 https://doi.org/10.1002/stem.1172
  29. Livak KJ and Schmittgen TD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods 25, 402-408 https://doi.org/10.1006/meth.2001.1262
  30. You HJ, Lee JW, Yoo YJ and Kim JH (2004) A pathway involving protein kinase Cdelta up-regulates cytosolic phospholipase A(2)alpha in airway epithelium. Biochem Biophys Res Commun 321, 657-664 https://doi.org/10.1016/j.bbrc.2004.07.022

Cited by

  1. Neolignan and monoterpene glycoside from the seeds of Pharbitis nil vol.20, 2017, https://doi.org/10.1016/j.phytol.2017.04.019
  2. New Triterpenoids from the Stems of Cornus walteri vol.65, pp.7, 2017, https://doi.org/10.1248/cpb.c17-00272
  3. Akt1-Inhibitor of DNA binding2 is essential for growth cone formation and axon growth and promotes central nervous system axon regeneration vol.5, 2016, https://doi.org/10.7554/eLife.20799
  4. TGFBR2-dependent alterations of exosomal cargo and functions in DNA mismatch repair-deficient HCT116 colorectal cancer cells vol.15, pp.1, 2017, https://doi.org/10.1186/s12964-017-0169-y
  5. Tannic acid attenuates TGF-β1-induced epithelial-to-mesenchymal transition by effectively intervening TGF-β signaling in lung epithelial cells 2017, https://doi.org/10.1002/jcp.26127
  6. Cytotoxic Triterpenoids from the Barks ofBetula platyphyllavar.japonica vol.14, pp.4, 2017, https://doi.org/10.1002/cbdv.201600400
  7. Fibroblasts Enhance Migration of Human Lung Cancer Cells in a Paper-Based Coculture System vol.5, pp.6, 2016, https://doi.org/10.1002/adhm.201500709
  8. Akt regulates neurite growth by phosphorylation-dependent inhibition of radixin proteasomal degradation vol.8, pp.1, 2018, https://doi.org/10.1038/s41598-018-20755-w