Browse > Article
http://dx.doi.org/10.14348/molcells.2015.0007

Cell Proliferation and Motility Are Inhibited by G1 Phase Arrest in 15-kDa Selenoprotein-Deficient Chang Liver Cells  

Bang, Jeyoung (School of Biological Sciences, Seoul National University)
Huh, Jang Hoe (School of Biological Sciences, Seoul National University)
Na, Ji-Woon (School of Biological Sciences, Seoul National University)
Lu, Qiao (School of Biological Sciences, Seoul National University)
Carlson, Bradley A. (Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health)
Tobe, Ryuta (Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health)
Tsuji, Petra A. (Department of Biological Sciences, Towson University)
Gladyshev, Vadim N. (Brigham and Women's Hospital, Harvard Medical School)
Hatfield, Dolph L. (Mouse Cancer Genetics Program, National Cancer Institute, National Institutes of Health)
Lee, Byeong Jae (School of Biological Sciences, Seoul National University)
Abstract
The 15-kDa selenoprotein (Sep15) is a selenoprotein residing in the lumen of the endoplasmic reticulum (ER) and implicated in quality control of protein folding. Herein, we established an inducible RNAi cell line that targets Sep15 mRNA in Chang liver cells. RNAi-induced Sep15 deficiency led to inhibition of cell proliferation, whereas cell growth was resumed after removal of the knockdown inducer. Sep15-deficient cells were arrested at the G1 phase by upregulating p21 and p27, and these cells were also characterized by ER stress. In addition, Sep15 deficiency led to the relocation of focal adhesions to the periphery of the cell basement and to the decrease of the migratory and invasive ability. All these changes were reversible depending on Sep15 status. Rescuing the knockdown state by expressing a silent mutant Sep15 mRNA that is resistant to siRNA also reversed the phenotypic changes. Our results suggest that SEP15 plays important roles in the regulation of the G1 phase during the cell cycle as well as in cell motility in Chang liver cells, and that this selenoprotein offers a novel functional link between the cell cycle and cell motility.
Keywords
15-kDa selenoprotein; cell cycle arrest; cell proliferation; cyclin E; selenium;
Citations & Related Records
Times Cited By KSCI : 2  (Citation Analysis)
연도 인용수 순위
1 Apostolou, S., Klein, J.O., Mitsuuchi, Y., Shetler, J.N., Poulikakos, P.I., Jhanwar, S.C., Kruger, W.D., and Testa, J.R. (2004). Growth inhibition and induction of apoptosis in mesothelioma cells by selenium and dependence on selenoprotein SEP15 genotype. Oncogene 23, 5032-5040.   DOI   ScienceOn
2 Bang, J., Jang, M., Huh, J.H., Na, J., Shim, M., Carlson, B.A., Tobe, R., Tsuji, P.A., Gladyshev, V.N., Hatfield, D.L., et al. (2014, in press). Deficiency of the 15-kDa selenoprotein led to cytoskeleton remodeling and non-apoptotic membrane blebbing through a RhoA/ROCK pathway. Biochem. Biophys. Res. Commun. doi:10.1016/j.bbrc.2014.12.059.   DOI   ScienceOn
3 Boosalis, M.G. (2008). The role of selenium in chronic disease. Nutr. Clin. Pract. 23, 152-160.   DOI   ScienceOn
4 Coqueret, O. (2003). New roles for p21 and p27 cell-cycle inhibitors: a function for each cell compartment? Trends Cell Biol.13, 65-70.   DOI   ScienceOn
5 Davis, C.D., Tsuji, P.A., and Milner, J.A. (2012). Selenoproteins and cancer prevention. Annu. Rev. Nutr. 32, 73-95.   DOI   ScienceOn
6 Ferguson, A.D., Labunskyy, V.M., Fomenko, D.E., Arac, D., Chelliah, Y., Amezcua, C.A., Rizo, J., Gladyshev, V.N., and Deisenhofer, J. (2006). NMR structures of the selenoproteins Sep15 and SelM reveal redox activity of a new thioredoxin-like family. J. Biol. Chem. 281, 3536-3543.   DOI   ScienceOn
7 Flohe, L. (2007). Selenium in mammalian spermiogenesis. Biol. Chem. 388, 987-995.
8 Gladyshev, V.N., Jeang, K.T., Wootton, J.C., and Hatfield, D.L. (1998). A new human selenium-containing protein. Purification, characterization, and cDNA sequence. J. Biol. Chem. 273, 8910-8915.   DOI   ScienceOn
9 Hatfield, D.L., and Gladyshev, V.N. (2002). How selenium has altered our understanding of the genetic code. Mol. Cell. Biol. 22, 3565-3576.   DOI
10 Irons, R., Tsuji, P.A., Carlson, B.A., Ouyang, P., Yoo, M.H., Xu, X.M., Hatfield, D.L., Gladyshev, V.N., and Davis, C.D. (2010). Deficiency in the 15-kDa selenoprotein inhibits tumorigenicity and metastasis of colon cancer cells. Cancer Prev. Res. (Phila) 3, 630-639.   DOI   ScienceOn
11 Jablonska, E., Gromadzinska, J., Sobala, W., Reszka, E., and Wasowicz, W. (2008). Lung cancer risk associated with selenium status is modified in smoking individuals by Sep15 polymorphism. Eur. J. Nutr. 47, 47-54.   DOI
12 Kim, J.Y., Lee, K.H., Shim, M.S., Shin, H., Xu, X.M., Carlson, B.A., Hatfield, D.L., and Lee, B.J. (2010). Human selenophosphate synthetase 1 has five splice variants with unique interactions, subcellular localizations and expression patterns. Biochem. Biophys. Res. Commun. 397, 53-58.   DOI   ScienceOn
13 Labunskyy, V.M., Hatfield, D.L., and Gladyshev, V.N. (2007). The Sep15 protein family: roles in disulfide bond formation and quality control in the endoplasmic reticulum. IUBMB Life 59, 1-5.   DOI   ScienceOn
14 Kim, M., Chen, Z., Shim, M.S., Lee, M.S., Kim, J.E., Kwon, Y.E., Yoo, T.J., Kim, J.Y., Bang, J.Y., Carlson, B.A., et al. (2013). SUMO modification of NZFP mediates transcriptional repression through TBP binding. Mol. Cells 35, 70-78.   DOI
15 Korotkov, K.V., Novoselov, S.V., Hatfield, D.L., and Gladyshev, V.N. (2002). Mammalian selenoprotein in which selenocysteine (Sec) incorporation is supported by a new form of Sec insertion sequence element. Mol. Cell. Biol. 22, 1402-1411.   DOI
16 Kumaraswamy, E., Malykh, A., Korotkov, K.V., Kozyavkin, S., Hu, Y., Kwon, S.Y., Moustafa, M.E., Carlson, B.A., Berry, M.J., Lee, B.J., et al. (2000). Structure-expression relationships of the 15- kDa selenoprotein gene. Possible role of the protein in cancer etiology. J. Biol. Chem. 275, 35540-35547.   DOI   ScienceOn
17 Labunskyy, V.M., Yoo, M.H., Hatfield, D.L., and Gladyshev, V.N. (2009). Sep15, a thioredoxin-like selenoprotein, is involved in the unfolded protein response and differentially regulated by adaptive and acute ER stresses. Biochemistry 48, 8458-8465.   DOI   ScienceOn
18 Low, S.C., and Berry, M.J. (1996). Knowing when not to stop: selenocysteine incorporation in eukaryotes. Trends Biochem. Sci. 21, 203-208.   DOI   ScienceOn
19 Nasr, M.A., Hu, Y.J., and Diamond, A.M. (2003). Allelic loss at the Sep15 locus in breast cancer. Cancer Therapy 1, 293-298.
20 Niculescu III, A.B., Chen, X., Smeets, M., Hengst, L., Prives, C., and Reed, S.I. (1998). Effects of $p21^{Cip1/Waf1}$ at both the G1/S and the G2/M cell cycle transitions: pRb is a critical determinant in blocking DNA replication and in preventing endoreduplication. Mol. Cell. Biol. 18, 629-643.   DOI
21 Papp, L.V., Lu, J., Holmgren, A., and Khanna, K.K. (2007). From selenium to selenoproteins: synthesis, identity, and their role in human health. Antioxid. Redox Signal. 9, 775-806.   DOI   ScienceOn
22 Park, J., Kim, J.S., Jung, K.C., Lee, H.J., Kim, J.I., Kim, J., Lee, J.Y., Park, J.B., and Choi, S.Y. (2003). Exoenzyme Tat-C3 inhibits association of zymosan particles, phagocytosis, adhesion, and complement binding in macrophage cells. Mol. Cells 16, 216-223.
23 Pasapera, A.M., Schneider, I.C., Rericha, E., Schlaepfer, D.D., and Waterman, C.M. (2010). Myosin II activity regulates vinculin recruitment to focal adhesions through FAK-mediated paxillin phosphorylation. J. Cell Biol. 188, 877-890.   DOI   ScienceOn
24 Reed, S.I. (2002). Cell cycling? Check your brakes. Nat. Cell Biol. 4, E199-E201.   DOI   ScienceOn
25 Roman, M., Jitaru, P., and Barbante, C. (2014). Selenium biochemistry and its role for human health. Metallomics 6, 25-54.   DOI
26 Sherr, C.J., and Roberts, J.M. (1999). CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev. 13, 1501-1512.   DOI   ScienceOn
27 Shim, M.S., Kim, J.Y., Jung, H.K., Lee, K.H., Xu, X.M., Carlson, B.A., Kim, K.W., Kim, I.Y., Hatfield, D.L., and Lee, B.J. (2009). Elevation of glutamine level by selenophosphate synthetase 1 knockdown induces megamitochondrial formation in Drosophila cells. J. Biol. Chem. 284, 32881-32894.   DOI   ScienceOn
28 Tsuji, P.A., Carlson, B.A., Naranjo-Suarez, S., Yoo, M.H., Xu, X.M., Fomenko, D.E., Gladyshev, V.N., Hatfield, D.L., and Davis, C.D. (2012). Knockout of the 15 kDa selenoprotein protects against chemically-induced aberrant crypt formation in mice. PLoS One 7, e50574.   DOI
29 Sigoillot, F.D., Lyman, S., Huckins, J.F., Adamson, B., Chung, E., Quattrochi, B., and King, R.W. (2012). A bioinformatics method identifies prominent off-targeted transcript in RNAi screens. Nat. Methods 9, 363-366.   DOI   ScienceOn
30 Sutherland, A., Kim, D.H., Relton, C., Ahn, Y.O., and Hesketh, J. (2010). Polymorphisms in the selenoprotein S and 15-kDa selenoprotein genes are associated with altered susceptibility to colorectal cancer. Genes Nutr. 5, 215-223.   DOI   ScienceOn
31 Tsuji, P.A., Naranjo-Suarez, S., Carlson, B.A., Tobe, R., Yoo, M.H., and Davis, C.D. (2011). Deficiency in the 15 kDa selenoprotein inhibits human colon cancer cell growth. Nutrients 3, 805-817.   DOI