DOI QR코드

DOI QR Code

Hypoxia Inducible Factor-1α Directly Regulates Nuclear Clusterin Transcription by Interacting with Hypoxia Response Elements in the Clusterin Promoter

  • Park, Jeongsook (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Park, So Yun (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Shin, Eunkyung (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Lee, Sun Hee (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Kim, Yoon Sook (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Lee, Dong Hoon (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Roh, Gu Seob (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Kim, Hyun Joon (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Kang, Sang Soo (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Cho, Gyeong Jae (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Jeong, Bo-Young (Department of Food & Nutrition, College of Natural Sciences, Gyeongsang National University) ;
  • Kim, Hwajin (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University) ;
  • Choi, Wan Sung (Department of Anatomy and Neurobiology, Institute of Health Science, Medical Research Center for Neural Dysfunction, School of Medicine, Gyeongsang National University)
  • Received : 2013.11.21
  • Accepted : 2013.12.30
  • Published : 2014.02.28

Abstract

Differential transcription of the clusterin (CLU) gene yields two CLU isoforms, a nuclear form (nCLU) and a secretory form (sCLU), which play crucial roles in prostate tumorigenesis. Pro-apoptotic nCLU and anti-apoptotic sCLU have opposite effects and are differentially expressed in normal and cancer cells; however, their regulatory mechanisms at the transcriptional level are not yet known. Here, we examined the transcriptional regulation of nCLU in response to hypoxia. We identified three putative hypoxia response elements (HREs) in the human CLU promoter between positions -806 and +51 bp. Using a luciferase reporter, electrophoretic gel mobility shift, and chromatin immunoprecipitation assays, we further showed that hypoxia-inducible factor-$1{\alpha}$ (HIF-$1{\alpha}$) bound directly to these sites and activated transcription. Exposure to the hypoxia-mimetic compound $CoCl_2$, incubation under 1% $O_2$ conditions, or overexpression of HIF-$1{\alpha}$ enhanced nCLU expression and induced apoptosis in human prostate cancer PC3M cells. However, LNCaP prostate cancer cells were resistant to hypoxia-induced cell death. Methylation-specific PCR analysis revealed that the CLU promoter in PC3M cells was not methylated; in contrast, the CLU promoter in LNCap cells was methylated. Co-treatment of LNCaP cells with $CoCl_2$ and a demethylating agent promoted apoptotic cell death through the induction of nCLU. We conclude that nCLU expression is regulated by direct binding of HIF-$1{\alpha}$ to HRE sites and is epigenetically controlled by methylation of its promoter region.

Keywords

References

  1. Andersen, C.L., Schepeler, T., Thorsen, K., Birkenkamp-Demtroder, K., Mansilla, F., Aaltonen, L.A., Laurberg, S., and Orntoft, T.F. (2007). Clusterin expression in normal mucosa and colorectal cancer. Mol. Cell. Proteomics 6, 1039-1048. https://doi.org/10.1074/mcp.M600261-MCP200
  2. Bettuzzi, S. (2009). Chapter 1: Introduction. Adv. Cancer Res. 104, 1-8. https://doi.org/10.1016/S0065-230X(09)04001-9
  3. Bettuzzi, S., Scorcioni, F., Astancolle, S., Davalli, P., Scaltriti, M., and Corti, A. (2002). Clusterin (SGP-2) transient overexpression decreases proliferation rate of SV40-immortalized human prostate epithelial cells by slowing down cell cycle progression. Oncogene 21, 4328-4334. https://doi.org/10.1038/sj.onc.1205594
  4. Chen, N., Chen, X., Huang, R., Zeng, H., Gong, J., Meng, W., Lu, Y., Zhao, F., Wang, L., and Zhou, Q. (2009). BCL-xL is a target gene regulated by hypoxia-inducible factor-1{alpha}. J. Biol. Chem. 284, 10004-10012. https://doi.org/10.1074/jbc.M805997200
  5. Cochrane, D.R., Wang, Z., Muramaki, M., Gleave, M.E., and Nelson, C.C. (2007). Differential regulation of clusterin and its isoforms by androgens in prostate cells. J. Biol. Chem. 282, 2278-2287. https://doi.org/10.1074/jbc.M608162200
  6. de Silva, H.V., Harmony, J.A., Stuart, W.D., Gil, C.M., and Robbins, J. (1990). Apolipoprotein J: structure and tissue distribution. Biochemistry 29, 5380-5389. https://doi.org/10.1021/bi00474a025
  7. Forsythe, J.A., Jiang, B.H., Iyer, N.V., Agani, F., Leung, S.W., Koos, R.D., and Semenza, G.L. (1996). Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell. Biol. 16, 4604-4613. https://doi.org/10.1128/MCB.16.9.4604
  8. Gleave, M., and Miyake, H. (2005). Use of antisense oligonucleotides targeting the cytoprotective gene, clusterin, to enhance androgen- and chemo-sensitivity in prostate cancer. World J. Urol. 23, 38-46. https://doi.org/10.1007/s00345-004-0474-0
  9. Higashimura, Y., Nakajima, Y., Yamaji, R., Harada, N., Shibasaki, F., Nakano, Y., and Inui, H. (2011). Up-regulation of glyceraldehyde-3-phosphate dehydrogenase gene expression by HIF-1 activity depending on Sp1 in hypoxic breast cancer cells. Arch. Biochem. Biophys. 509, 1-8. https://doi.org/10.1016/j.abb.2011.02.011
  10. Holmquist-Mengelbier, L., Fredlund, E., Lofstedt, T., Noguera, R., Navarro, S., Nilsson, H., Pietras, A., Vallon-Christersson, J., Borg, A., Gradin, K., et al. (2006). Recruitment of HIF-1alpha and HIF-2alpha to common target genes is differentially regulated in neuroblastoma: HIF-2alpha promotes an aggressive phenotype. Cancer Cell 10, 413-423. https://doi.org/10.1016/j.ccr.2006.08.026
  11. Kase, S., He, S., Sonoda, S., Kitamura, M., Spee, C., Wawrousek, E., Ryan, S.J., Kannan, R., and Hinton, D.R. (2010). alphaBcrystallin regulation of angiogenesis by modulation of VEGF. Blood 115, 3398-3406. https://doi.org/10.1182/blood-2009-01-197095
  12. Kevans, D., Gorman, S., Tosetto, M., Sheahan, K., O'Donoghue, D., Mulcahy, H., and O'Sullivan, J. (2012). Clusterin and chemotherapy sensitivity under normoxic and graded hypoxic conditions in colorectal cancer. J. Gastrointest. Cancer 43, 305-313. https://doi.org/10.1007/s12029-011-9277-x
  13. Kim, N., and Choi, W.S. (2011). Proapoptotic role of nuclear clusterin in brain. Anat. Cell Biol. 44, 169-175. https://doi.org/10.5115/acb.2011.44.3.169
  14. Kim, N., Yoo, J.C., Han, J.Y., Hwang, E.M., Kim, Y.S., Jeong, E.Y., Sun, C.H., Yi, G.S., Roh, G.S., Kim, H.J., et al. (2011). Human nuclear clusterin mediates apoptosis by interacting with Bcl-XL through C-terminal coiled coil domain. J. Cell Physiol. 227, 1157-1167.
  15. Kim, N., Han, J.Y., Roh, G.S., Kim, H.J., Kang, S.S., Cho, G.J., Park, J.Y., and Choi, W.S. (2012). Nuclear clusterin is associated with neuronal apoptosis in the developing rat brain upon ethanol exposure. Alcohol Clin. Exp. Res. 36, 72-82. https://doi.org/10.1111/j.1530-0277.2011.01588.x
  16. Lee, D.H., Ha, J.H., Kim, Y., Bae, K.H., Park, J.Y., Choi, W.S., Yoon, H.S., Park, S.G., Park, B.C., Yi, G.S., et al. (2011). Interaction of a putative BH3 domain of clusterin with anti-apoptotic Bcl-2 family proteins as revealed by NMR spectroscopy. Biochem. Biophys. Res. Commun. 408, 541-547. https://doi.org/10.1016/j.bbrc.2011.04.054
  17. Leskov, K.S., Klokov, D.Y., Li, J., Kinsella, T.J., and Boothman, D.A. (2003). Synthesis and functional analyses of nuclear clusterin, a cell death protein. J. Biol. Chem. 278, 11590-11600. https://doi.org/10.1074/jbc.M209233200
  18. Liao, F.T., Lee, Y.J., Ko, J.L., Tsai, C.C., Tseng, C.J., and Sheu, G.T. (2009). Hepatitis delta virus epigenetically enhances clusterin expression via histone acetylation in human hepatocellular carcinoma cells. J. Gen. Virol. 90, 1124-1134. https://doi.org/10.1099/vir.0.007211-0
  19. Loboda, A., Jozkowicz, A., and Dulak, J. (2010). HIF-1 and HIF-2 transcription factors--similar but not identical. Mol. Cells 29, 435-442. https://doi.org/10.1007/s10059-010-0067-2
  20. Lund, P., Weisshaupt, K., Mikeska, T., Jammas, D., Chen, X., Kuban, R.J., Ungethum, U., Krapfenbauer, U., Herzel, H.P., Schafer, R., et al. (2006). Oncogenic HRAS suppresses clusterin expression through promoter hypermethylation. Oncogene 25, 4890-4903. https://doi.org/10.1038/sj.onc.1209502
  21. Millar, N.L., Reilly, J.H., Kerr, S.C., Campbell, A.L., Little, K.J., Leach, W.J., Rooney, B.P., Murrell, G.A., and McInnes, I.B. (2012). Hypoxia: a critical regulator of early human tendinopathy. Ann. Rheum. Dis. 71, 302-310. https://doi.org/10.1136/ard.2011.154229
  22. Nakamura, E., Abreu-e-Lima, P., Awakura, Y., Inoue, T., Kamoto, T., Ogawa, O., Kotani, H., Manabe, T., Zhang, G.J., Kondo, K., et al. (2006). Clusterin is a secreted marker for a hypoxiainducible factor-independent function of the von Hippel-Lindau tumor suppressor protein. Am. J. Pathol. 168, 574-584. https://doi.org/10.2353/ajpath.2006.050867
  23. Otrock, Z.K., Hatoum, H.A., Awada, A.H., Ishak, R.S., and Shamseddine, A.I. (2009). Hypoxia-inducible factor in cancer angiogenesis: structure, regulation and clinical perspectives. Crit. Rev. Oncol. Hematol. 70, 93-102. https://doi.org/10.1016/j.critrevonc.2009.01.001
  24. Park, S.Y., Kim, J.B., and Han, Y.M. (2007). REST is a key regulator in brain-specific homeobox gene expression during neuronal differentiation. J. Neurochem. 103, 2565-2574.
  25. Piret, J.P., Mottet, D., Raes, M., and Michiels, C. (2002). $CoCl_2$, a chemical inducer of hypoxia-inducible factor-1, and hypoxia reduce apoptotic cell death in hepatoma cell line HepG2. Ann. N Y Acad. Sci. 973, 443-447. https://doi.org/10.1111/j.1749-6632.2002.tb04680.x
  26. Poulios, E., Trougakos, I.P., and Gonos, E.S. (2006). Comparative effects of hypoxia on normal and immortalized human diploid fibroblasts. Anticancer Res. 26, 2165-2168.
  27. Pucci, S., Bonanno, E., Pichiorri, F., Angeloni, C., and Spagnoli, L.G. (2004). Modulation of different clusterin isoforms in human colon tumorigenesis. Oncogene 23, 2298-2304. https://doi.org/10.1038/sj.onc.1207404
  28. Rauhala, H.E., Porkka, K.P., Saramaki, O.R., Tammela, T.L., and Visakorpi, T. (2008). Clusterin is epigenetically regulated in prostate cancer. Int. J. Cancer 123, 1601-1609. https://doi.org/10.1002/ijc.23658
  29. Rizzi, F., and Bettuzzi, S. (2009). Clusterin (CLU) and prostate cancer. Adv. Cancer Res. 105, 1-19. https://doi.org/10.1016/S0065-230X(09)05001-5
  30. Rizzi, F., and Bettuzzi, S. (2010). The clusterin paradigm in prostate and breast carcinogenesis. Endocr. Relat. Cancer 17, R1-17. https://doi.org/10.1677/ERC-09-0140
  31. Rizzi, F., Coletta, M., and Bettuzzi, S. (2009). Chapter 2: Clusterin (CLU): From one gene and two transcripts to many proteins. Adv. Cancer Res. 104, 9-23. https://doi.org/10.1016/S0065-230X(09)04002-0
  32. Rosemblit, N., and Chen, C.L. (1994). Regulators for the rat clusterin gene: DNA methylation and cis-acting regulatory elements. J. Mol. Endocrinol. 13, 69-76. https://doi.org/10.1677/jme.0.0130069
  33. Rosenberg, M.E., and Silkensen, J. (1995). Clusterin: physiologic and pathophysiologic considerations. Int. J. Biochem. Cell Biol. 27, 633-645. https://doi.org/10.1016/1357-2725(95)00027-M
  34. Sala, A., Bettuzzi, S., Pucci, S., Chayka, O., Dews, M., and Thomas-Tikhonenko, A. (2009). Regulation of CLU gene expression by oncogenes and epigenetic factors implications for tumorigenesis. Adv. Cancer Res. 105, 115-132. https://doi.org/10.1016/S0065-230X(09)05007-6
  35. Salnikow, K., Donald, S.P., Bruick, R.K., Zhitkovich, A., Phang, J.M., and Kasprzak, K.S. (2004). Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J. Biol. Chem. 279, 40337-40344. https://doi.org/10.1074/jbc.M403057200
  36. Schepeler, T., Mansilla, F., Christensen, L.L., Orntoft, T.F., and Andersen, C.L. (2007). Clusterin expression can be modulated by changes in TCF1-mediated Wnt signaling. J. Mol. Signal. 2, 6. https://doi.org/10.1186/1750-2187-2-6
  37. Semenza, G.L. (1998). Hypoxia-inducible factor 1: master regulator of $O_2$ homeostasis. Curr. Opin. Genet. Dev. 8, 588-594. https://doi.org/10.1016/S0959-437X(98)80016-6
  38. Semenza, G.L. (2001). HIF-1, $O_2$, and the 3 PHDs: how animal cells signal hypoxia to the nucleus. Cell 107, 1-3. https://doi.org/10.1016/S0092-8674(01)00518-9
  39. Serrano, A., Redondo, M., Tellez, T., Castro-Vega, I., Roldan, M.J., Mendez, R., Rueda, A., and Jimenez, E. (2009). Regulation of clusterin expression in human cancer via DNA methylation. Tumour. Biol. 30, 286-291. https://doi.org/10.1159/000259912
  40. Shannan, B., Seifert, M., Leskov, K., Willis, J., Boothman, D., Tilgen, W., and Reichrath, J. (2006). Challenge and promise: roles for clusterin in pathogenesis, progression and therapy of cancer. Cell Death Differ. 13, 12-19. https://doi.org/10.1038/sj.cdd.4401779
  41. Trougakos, I.P., and Gonos, E.S. (2002). Clusterin/apolipoprotein J in human aging and cancer. Int. J. Biochem. Cell Biol. 34, 1430-1448. https://doi.org/10.1016/S1357-2725(02)00041-9
  42. Trougakos, I.P., So, A., Jansen, B., Gleave, M.E., and Gonos, E.S. (2004). Silencing expression of the clusterin/apolipoprotein j gene in human cancer cells using small interfering RNA induces spontaneous apoptosis, reduced growth ability, and cell sensitization to genotoxic and oxidative stress. Cancer Res. 64, 1834-1842. https://doi.org/10.1158/0008-5472.CAN-03-2664
  43. Wiesener, M.S., Jurgensen, J.S., Rosenberger, C., Scholze, C.K., Horstrup, J.H., Warnecke, C., Mandriota, S., Bechmann, I., Frei, U.A., Pugh, C.W., et al. (2003). Widespread hypoxia-inducible expression of HIF-2alpha in distinct cell populations of different organs. FASEB J. 17, 271-273. https://doi.org/10.1096/fj.02-0445fje
  44. Wilson, M.R., and Easterbrook-Smith, S.B. (2000). Clusterin is a secreted mammalian chaperone. Trends Biochem. Sci. 25, 95-98. https://doi.org/10.1016/S0968-0004(99)01534-0
  45. Yamaji, R., Fujita, K., Takahashi, S., Yoneda, H., Nagao, K., Masuda, W., Naito, M., Tsuruo, T., Miyatake, K., Inui, H., et al. (2003). Hypoxia up-regulates glyceraldehyde-3-phosphate dehydrogenase in mouse brain capillary endothelial cells: involvement of $Na^+/Ca^{2+}$ exchanger. Biochim. Biophys. Acta. 1593, 269-276. https://doi.org/10.1016/S0167-4889(02)00397-X

Cited by

  1. Distinct promoters, subjected to epigenetic regulation, drive the expression of two clusterin mRNAs in prostate cancer cells vol.1849, pp.1, 2015, https://doi.org/10.1016/j.bbagrm.2014.11.003
  2. Bcl-2 silencing attenuates hypoxia-induced apoptosis resistance in pulmonary microvascular endothelial cells vol.21, pp.1, 2016, https://doi.org/10.1007/s10495-015-1184-3
  3. Regulation of fatty acid binding proteins by hypoxia inducible factors 1α and 2α in the placenta: Relevance to pre-eclampsia vol.93, 2015, https://doi.org/10.1016/j.plefa.2014.09.004
  4. Transcriptome-Based Analysis of Molecular Pathways for Clusterin Functions in Kidney Cells vol.231, pp.12, 2016, https://doi.org/10.1002/jcp.25415
  5. Molecular mechanisms of the interaction between the processes of the cell response to mechanical stress and neuronal apoptosis in primary open-angle glaucoma vol.7, pp.5, 2017, https://doi.org/10.1134/S2079059717050173
  6. Thirty Minutes of Hypobaric Hypoxia Provokes Alterations of Immune Response, Haemostasis, and Metabolism Proteins in Human Serum vol.18, pp.9, 2017, https://doi.org/10.3390/ijms18091882
  7. Hypoxia Response Elements Can Cause the Overexpression of the BAX mRNA Under Hypoxic Condition vol.In Press, pp.In Press, 2016, https://doi.org/10.17795/ijcp-4554
  8. Proteomic Profiling of the Retinas in a Neonatal Rat Model of Oxygen-Induced Retinopathy with a Reproducible Ion-Current-Based MS1 Approach vol.14, pp.5, 2015, https://doi.org/10.1021/pr501238m
  9. Clusterin gene is predominantly regulated by histone modifications in human colon cancer and ectopic expression of the nuclear isoform induces cell death vol.1852, pp.8, 2015, https://doi.org/10.1016/j.bbadis.2015.04.021
  10. Requirement of clusterin expression for prosurvival autophagy in hypoxic kidney tubular epithelial cells vol.310, pp.2, 2016, https://doi.org/10.1152/ajprenal.00304.2015
  11. Association Between Clusterin Gene Polymorphisms and Epilepsy in a Han Chinese Population vol.21, pp.11, 2017, https://doi.org/10.1089/gtmb.2017.0032
  12. Proteotranscriptomic Measurements of E6-Associated Protein (E6AP) Targets in DU145 Prostate Cancer Cells vol.17, pp.6, 2018, https://doi.org/10.1074/mcp.RA117.000504
  13. Hypoperfusion is a potential inducer of immunosuppressive network in Alzheimer's disease vol.142, pp.None, 2014, https://doi.org/10.1016/j.neuint.2020.104919
  14. Circulating levels of clusterin and complement factor H in patients with obstructive sleep apnea vol.15, pp.5, 2014, https://doi.org/10.2217/bmm-2020-0533
  15. Contribution of the Transcription Factors Sp1/Sp3 and AP-1 to Clusterin Gene Expression during Corneal Wound Healing of Tissue-Engineered Human Corneas vol.22, pp.22, 2014, https://doi.org/10.3390/ijms222212426