The role of hypoxia response element in TGFβ-induced carbonic anhydrase IX expression in Hep3B human hepatoma cells

Authors

  • Hatice Yildirim University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, 10145 Balikesir
  • Merve Karaman University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, 10145 Balikesir
  • Feray Köçkar University of Balikesir, Faculty of Science and Literature, Department of Molecular Biology and Genetics, 10145 Balikesir

Keywords:

carbonic anhydrase IX (CAIX), hypoxia response element (HRE), transforming growth factor β (TGFβ), mitogen-activated protein kinase (MAPK) signaling, phosphatidylinositol-4, 5-bisphosphate 3-kinase (PI3K) signaling

Abstract

Carbonic anhydrase IX (CAIX) is a hypoxia-regulated gene. It is overexpressed in a variety of cancers, including hepatocellular cancer. Transforming growth factor β (TGFβ) is considered to have an impact on cancer biology dueto its important roles in cell proliferation and differentiation. The effect of the TGFβ on CAIX expression under hypoxia and the mechanism underlying the role of the hypoxia response element (HRE) on this expression areunknown. In this study, we demonstrate that TGFβ upregulates CAIX expression under hypoxic conditions in the Hep3B hepatoma cell line, indicating that the mitogen-activated protein kinase (MAPK)- and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)-signaling pathways might be responsible for this response. Site-directed mutagenesis of the HRE region in CAIX promoter reduced the TGFβ-induced CAIX promoter activity, pointingto the significance of HRE for this response.Upregulation of TGFβ-stimulated CAIX expression was consistent with the upregulation of promoter activity of five different truncated constructs of the CAIX promoter under hypoxia. Our findings show that the HRE region is critical for TGFβ-induced CAIX expression,which is mainly controlled by MAPK and PI3K pathways.

https://doi.org/10.2298/ABS161124003Y

Received: November 24, 2016; Revised: January 4, 2017; Accepted: January 11, 2017; Published online: January 23, 2017

How to cite this article: Yildirim H, Karaman M, Köçkar F. The role of hypoxia response element in TGFβ-induced carbonic anhydrase IX expression in Hep3B human hepatoma cells. Arch Biol Sci. 2017;69(4):593-601.

Downloads

Download data is not yet available.

References

Höckel M, Vaupel P, Tumor hypoxia: definitions and current clinical biologic and molecular aspects. J Natl Cancer Inst.2001;93:266-76.

Xia Y, Choi HK, Lee K. Recent advances in hypoxia-inducible factor (HIF)-1 inhibitors. Eur J Med Chem.2012;49:24-40.

Norman JT, Clark IM, Garcia PL. Hypoxia promotes fibrogenesis in human renal fibroblasts. Kidney Int. 2000;58:2351-66.

Mendonça DB, Mendonça G, Aragão FJ, Cooper LF. NF-κB suppresses HIF-1α response by competing for P300 binding. Biochem Biophys Res Commun. 2011;04:997-1003.

Kaluz S, Kaluzová M, Liao SY, Lerman M, Stanbridg EJ. Transcriptional control of the tumor and hypoxia-marker carbonic anhydrase 9:A one transcription factor (HIF-1) show? Biochim Biophys Acta.2009;1795(2):162-72.

Wykoff CC, Beasley N, Watson PH, Turner KJ, Pastorek J, Sibtain A. Hypoxia-inducible Expression of Tumor-associated Carbonic Anhydrases. Cancer Res. 2000;60(24):7075-83.

Dooley S, Dijke P. TGFβ in progression of liver disease. Cell Tissue Res. 2012;347(1):245-56.

Yoshida K, Murata M, Yamaguchi T, Matsuzaki K. TGFβ/Smad signaling during hepatic fibro-carcinogenesis. Int J Oncol.2014;45(4):1363-71.

Moses HL, Serra R. Regulation of differentiation by TGFβ. Curr Opin Genet Dev. 1996;6(5):581-86.

Bellam N, Pasche B. TGFβ signaling alterations and colon cancer. Cancer Treat Res. 2010;155:85-103.

Hung SP, Yang MH, Tseng KF, Lee OK. Hypoxia-Induced Secretion of TGFβ in Mesenchymal Stem Cell Promotes Breast Cancer Cell Progression. Cell Transplant. 2013;22(10):1869-82.

Zhang H, Akman O, Smith EL, Zhao J, Murphy JE. Cellular response to hypoxia involves signalling via Smad proteins. Blood.2003;101(6):2253-60.

Nishi H, Nakada T, Hokamura M, Osakabe Y, Itokazu O, Huang LE. Hypoxia-inducible factor-1 transactivates transforming growth factor β in trophoblast. Endocrinology. 2004;145(9):4113-18.

Yildirim H, Kockar F. TGFβ upregulates tumor-associated carbonic anhydrase IX gene expression in Hep3B cells. CellBiolInt.2009;33(9):1002-7.

Turkoglu SA, Kockar F. SP1 and USF differentially regulate ADAMTS1 gene expression under normoxic and hypoxic conditions in hepatoma cells. Gene 2016;575(1):48-57.

Alper M, Kockar F. IL-6 upregulates a disintegrin and metalloproteinase with thrombospondin motifs 2 (ADAMTS-2) in human osteosarcoma cells mediated by JNK pathway. Mol CellBiochem.2014;393(1-2):165-75.

Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2 (Delta Delta C(T)). Methods. 2001;25(4):402-8.

Goldberg MA, Dunning SP, Bunn HF.Regulation of the erythropoietin gene: evidence that the oxygen sensor s a heme protein. Science. 1988;242(4884):1412-5.

Wang GL, Semenza GL. Characterization of hypoxia-inducible factor 1 and regulation of DNA binding activity by hypoxia. J Biol Chem. 1993;268(29):21513-8.

Hatipoglu OF, Hirohata S, Cilek MZ, Ogawa H, Miyoshi T, Obika M, Demircan K. ADAMTS1 is a Unique Hypoxic Early Response Gene Expressed by Endothelial Cells. J Biol Chem. 2009;284(24):16325-33.

Wang G, Hazra TK, Mitra S, Lee HM, Englander EW. Mitochondrial DNA damage and a hypoxic response are induced by CoCl2 in rat neuronal PC12 cells. Nucleic Acids Res. 2000;28(10):2135-40.

Huang Y, Du KM, Xue ZH, Yan H, Li D, Liu W. Cobalt chloride and low oxygen tension trigger differentiation of acute myeloid leukemic cells: possible mediation of hypoxia-inducible factor-1-alpha. Leukemia.2003;17:2065-73.

Yuan Y, Hilliard G, Ferguson T, Millhorn DE. Cobalt Inhibits the Interaction between Hypoxia-inducible Factor-a and von Hippel-Lindau Protein by Direct Binding to Hypoxia-inducible Factor. J Biol Chem.2003;278(18):15911-16.

Grasselli F, Basini G, Bussolati S, Bianco F.Cobalt chloride, a hypoxia-mimicking agent, modulates redox status and functional parameters of cultured swine granulosa cells. ReprodFertilDev.2005;17(7):715-20.

Torii S, Okamura N, Suzuki Y, Ishizawa T, Yasumoto K, Sogawa K.Cyclic AMP represses the hypoxic induction of hypoxia-inducible factors in PC12 Cells. J Biochem.2009;146(6):839-44.

Said HM, Supuran CT, Hageman C, Staab A, Polat B, Katzer A.Modulation of Carbonic Anhydrase 9 (CAIX) in Human Brain Cancer. Curr Pharm Des.2010;16(29):3288-99.

Kutz SM, Higgins CE, Samarakoon R, Higgins SP, Allen RR, Qi L. TGFβ1-induced PAI-1 expression is E box/USF-dependentand requires EGFR signaling. Exp Cell Res.2006;312(7):1093-105.

Zvi B, Hayashida T, Hubchak SC, Hanna C, Platanias LC, Schnaper HW. TGFβ/ Smad3 activates mammalian target of rapamycin complex-1 to promote collagen production by increasing HIF-1α expression. Am J Physiol Renal Physiol. 2013;305(4):485-94.

Jeon YJ, Song KS, Han HJ, Park SH, Chang W, Lee MY. Rosmarinic acid inhibits chemical hypoxia-induced cytotoxicity in primary cultured rat hepatocytes. Arch Pharm Res.2014;37(7):907-15.

Jiang L, Lai YK, Zhang JF, Chan CY, Lu G, Lin MC. Transactivation of the TIEG1 confers growth inhibition of transforming growth factor-β-susceptible hepatocellular carcinoma cells. World J Gastroenterol. 2012;18(17):2035-42.

Dzieran J, Fabian J, Feng T, Coulouarn C, Ilkavets I, Kyselova A.Comparative Analysis of TGFβ/Smad Signaling Dependent Cytostasis in Human Hepatocellular Carcinoma Cell Lines. PLoS One. 2013;8(8):e72252.

Mazzocca A, Antonaci S, Giannelli G. The TGF-β Signaling Pathway as a Pharmacological Target in a Hepatocellular Carcinoma. Current Pharmaceutical Design. 2016;18(27):4148-54.

Giannelli G, Mikulits W, Dooley S, Fabregat I, Moustakas A, Dijke P, Portincasa P, Winter P, Janssen R, Leporatti S, Herrera B and Sanchez A. The rationale for targeting TGF-b in chronic liver diseases. Eur J Clin Invest.2016;46:349-61.

Lu W, Qiu J, Z Huang, Liu H. Enhanced circulating transforming growth factor beta 1 is causally associated with an increased risk of hepatocellular carcinoma: a Mendelian randomization meta-analysis. Oncotarget. 2016;7(51):84695-704.

Radwan MI, Pasha HF, Mohamed RH, Hussien HI, El-Khshab MN. Influence of transforming growth factor-β1 and tumor necrosis factor-alpha genes polymorphisms on the development of cirrhosis and hepatocellular carcinoma in chronic hepatitis C patients. Cytokine. 2012;60:271-6.

Kockar F, Yildrim H, Sagkan RI, Hagemann C, Soysal Y, Anacker J. Hypoxia and cytokines regulate carbonic anhydrase 9 expression in hepatocellular carcinoma cells in vitro. World J Clin Oncol. 2012;3(6):82-91.

Salgado C, Peña AB, Novoa JM. Involvement of small Ras GTPases and their effectors in chronic renal disease. CellMolLifeSci.2008;65(3):477-92.

Zhang YE. Non-Smad pathways in TGF-β signaling. Cell Res.2009;19(1):128-39.

Moustakas A, Heldin CH. Non-Smad TGF-β signals. J Cell Sci.2005;118: 3573-84.

Downloads

Published

2017-10-18

How to Cite

1.
Yildirim H, Karaman M, Köçkar F. The role of hypoxia response element in TGFβ-induced carbonic anhydrase IX expression in Hep3B human hepatoma cells. Arch Biol Sci [Internet]. 2017Oct.18 [cited 2024Apr.26];69(4):593-601. Available from: https://serbiosoc.org.rs/arch/index.php/abs/article/view/1236

Issue

Section

Articles