Selenoproteins and their emerging roles in signaling pathways

Keywords: functions of selenoproteins; cytoplasm selenoproteins; thioredoxin reductases; glutathione peroxidases; methionine sulfoxide reductases; carcinogenesis

Abstract

The functional activity of selenoproteins has a wide range of effects on complex pathogenetic processes, including teratogenesis, immuno-inflammatory, neurodegenerative. Being active participants and promoters of many signaling pathways, selenoproteins support the lively interest of a wide scientific community. This review is devoted to the analysis of recent data describing the participation of selenoproteins in various molecular interactions mediating important signaling pathways. Data processing was carried out by the method of complex analysis. For convenience, all selenoproteins were divided into groups depending on their location and function. Among the group of selenoproteins of the ER membrane, selenoprotein N affects the absorption of Ca2+ by the endoplasmic reticulum mediated by oxidoreductin (ERO1), a key player in the CHOP/ERO1 branch, a pathogenic mechanism that causes myopathy. Another selenoprotein of the ER membrane selenoprotein K binding to the DHHC6 protein affects the IP3R receptor that regulates Ca2+ flux. Selenoprotein K is able to affect another protein of the endoplasmic reticulum CHERP, also appearing in Ca2+ transport. Selenoprotein S, associated with the lumen of ER, is able to influence the VCP protein, which ensures the incorporation of selenoprotein K into the ER membrane. Selenoprotein M, as an ER lumen protein, affects the phosphorylation of STAT3 by leptin, which confirms that Sel M is a positive regulator of leptin signaling. Selenoprotein S also related to luminal selenoproteins ER is a modulator of the IRE1α-sXBP1 signaling pathway. Nuclear selenoprotein H will directly affect the suppressor of malignant tumours, p53 protein, the activation of which increases with Sel H deficiency. The same selenoprotein is involved in redox regulation. Among the cytoplasmic selenoproteins, abundant investigations are devoted to SelP, which affects the PI3K/Akt/Erk signaling pathway during ischemia/reperfusion, is transported into the myoblasts through the plasmalemma after binding to the apoER2 receptor, and into the neurons to the megaline receptor and in general, selenoprotein P plays the role of a pool that stores the necessary trace element and releases it, if necessary, for vital selenoproteins. The thioredoxin reductase family plays a key role in the invasion and metastasis of salivary adenoid cystic carcinoma through the influence on the TGF-β-Akt/GSK-3β pathway during epithelial-mesenchymal transition. The deletion of thioredoxin reductase 1 affects the levels of messengers of the Wnt/β-catenin signaling pathway. No less studied is the glutathione peroxidase group, of which GPX3 is able to inhibit signaling in the Wnt/β-catenin pathway and thereby inhibit thyroid metastasis, as well as suppress protein levels in the PI3K/Akt/c-fos pathway. A key observation is that in cases of carcinogenesis, a decrease in GPX3 and its hypermethylation are almost always found. Among deiodinases, deiodinase 3 acts as a promoter of the oncogenes BRAF, MEK or p38, while stimulating a decrease in the expression of cyclin D1. The dependence of the level of deiodinase 3 on the Hedgehog (SHH) signaling pathway is also noted. Methionine sulfoxide reductase A can compete for the uptake of ubiquitin, reduce p38, JNK and ERK promoters of the MAPK signaling pathway; methionine sulfoxide reductase B1 suppresses MAPK signaling messengers, and also increases PARP and caspase 3.

References

Anouar, Y., Lihrmann, I., Falluel-Morel, A., & Boukhzar, L. (2018). Selenoprotein T is a key player in ER proteostasis, endocrine homeostasis and neuroprotection. Free Radical Biology and Medicine, 127, 145–152.

Appenzeller-Herzog, C., & Simmen, T. (2016). ER-luminal thiol/selenol-mediated regulation of Ca2+ signalling. Biochemical Society Transactions, 44(2), 452–459.

Bachmann, C., Noreen, F., Voermans, N. C., Schär, P. L., Vissing, J., Fock, J. M., Bulk, S., Kusters, B., Moore, S. A., Beggs, A. H., Mathews, K. D., Meyer, M., Genetti, C. A., Meola, G., Cardani, R., Mathews, E., Jungbluth, H., Muntoni, F., Zorzato, F., & Treves, S. (2019). Aberrant regulation of epigenetic modifiers contributes to the pathogenesis in patients with selenoprotein N-related myopathies. Human Mutation, 40(7), 962–974.

Bang, J., Huh, J. H., Na, J. W., Lu, Q., Carlson, B. A., Tobe, R., Tsuji, P. A., Gladyshev, V. N., Hatfield, D. L., & Lee, B. J. (2015). Cell proliferation and motility are inhibited by G1 phase arrest in 15-kDa selenoprotein-deficient chang liver cells. Molecules and Cells, 38(5), 457–465.

Bang, J., Jang, M., Huh, J. H., Na, J. W., Shim, M., Carlson, B. A., Tobe, R., Tsuji, P. A., Gladyshev, V. N., Hatfield, D. L., & Lee, B. J. (2015). Deficiency of the 15-kDa selenoprotein led to cytoskeleton remodeling and non-apoptotic membrane blebbing through a RhoA/ROCK pathway. Biochemical and Biophysical Research Communications, 456(4), 884–890.

Barrett, C. W., Ning, W., Chen, X., Smith, J. J., Washington, M. K., Hill, K. E., Coburn, L. A., Peek, R. M., Chaturvedi, R., Wilson, K. T., Burk, R. F., & Williams, C. S. (2013). Tumor suppressor function of the plasma glutathione peroxidase gpx3 in colitis-associated carcinoma. Cancer Research, 73(3), 1245–1255.

Barrett, C. W., Reddy, V. K., Short, S. P., Motley, A. K., Lintel, M. K., Bradley, A. M., Freeman, T., Vallance, J., Ning, W, Parang, B., Poindexter, S. V., Fingleton, B., Chen, X., Washington, M. K., Wilson, K. T., Shroyer, N. F., Hill, K. E., Burk, R. F., & Williams, C. S. (2015). Selenoprotein P influences colitis-induced tumorigenesis by mediating stemness and oxidative damage. Journal of Clinical Investigation, 125(7), 2646–2660.

Ben, S. B., Peng, B., Wang, G. C., Li, C., Gu, H. F., Jiang, H., Meng, X. L., Lee, B. J., & Chen, C. L. (2015). Overexpression of selenoprotein SelK in BGC-823 cells inhibits cell adhesion and migration. Biochemistry (Moscow), 80(10), 1344–1353.

Bertz, M., Kühn, K., Koeberle, S. C., Müller, M. F., Hoelzer, D., Thies, K., Deubel, S., Thierbach, R., & Kipp, A. P. (2018). Selenoprotein H controls cell cycle progression and proliferation of human colorectal cancer cells. Free Radical Biology and Medicine, 2018, S0891-5849(18)30020-0.

Brigelius-Flohé, R., Müller, M., Lippmann, D., & Kipp, A. P. (2012). The yin and yang of nrf2-regulated selenoproteins in carcinogenesis. International Journal of Cell Biology, 2012, 486147.

Burk, R. F., & Hill, K. E. (2015). Regulation of selenium metabolism and transport. Annual Review of Nutrition, 35, 109–134.

Burk, R. F., Hill, K. E., Motley, A. K., Winfrey, V. P., Kurokawa, S., Mitchell, S. L., & Zhang, W. (2014). Selenoprotein P and apolipoprotein E receptor-2 interact at the blood-brain barrier and also within the brain to maintain an essential selenium pool that protects against neurodegeneration. FASEB Journal, 28(8), 3579–3588.

Chadani, H., Usui, S., Inoue, O., Kusayama, T., Takashima, S. I., Kato, T., Murai, H., Furusho, H., Nomura, A., Misu, H., Takamura, T., Kaneko, S., & Takamura, M. (2018). Endogenous selenoprotein P, a liver-derived secretory protein, mediates myocardial ischemia/reperfusion injury in mice. International Journal of Molecular Sciences, 19(3), e878.

Chang, S. N., Lee, J. M., Oh, H., & Park, J. H. (2016). Glutathione peroxidase 3 inhibits prostate tumorigenesis in TRAMP mice. The Prostate, 76(15), 1387–1398.

Cox, A. G., Tsomides, A., Kim, A. J., Saunders, D., Hwang, K. L., Evason, K. J., Heidel, J., Brown, K. K., Yuan, M., Lien, E. C., Lee, B. C., Nissim, S., Dickinson, B., Chhangawala, S., Chang, C. J., Asara, J. M., Houvras, Y., Gladyshev, V. N., & Goessling, W. (2016). Selenoprotein H is an essential regulator of redox homeostasis that cooperates with p53 in development and tumorigenesis. Proceedings of the National Academy of Sciences of the United States of America, 113, E5562–E5571.

Cui, S., Men, L., Li, Y., Zhong, Y., Yu, S., Li, F., & Du, J. (2018). Selenoprotein S attenuates tumor necrosis factor-α-induced dysfunction in endothelial cells. Mediators of Inflammation, 2018, 1625414.

Dagnell, M., Schmidt, E. E., & Arnér, E. S. J. (2018). The A to Z of modulated cell patterning by mammalian thioredoxin reductases. Free Radical Biology and Medicine, 115, 484–496.

Dai, J., Liu, H., Zhou, J., & Huang, K. (2016). Selenoprotein R protects human lens epithelial cells against D-galactose-induced apoptosis by regulating oxidative stress and endoplasmic reticulum stress. International Journal of Molecular Sciences, 17(2), 231.

Deng, Y., Jiang, B., Rankin, C. L., Toyo-Oka, K., Richter, M. L., Maupin-Furlow, J. A., & Moskovitz, J. (2018). Methionine sulfoxide reductase A (MsrA) mediates the ubiquitination of 14-3-3 protein isotypes in brain. Free Radical Biology and Medicine, 129, 600–607.

Dong, C., Zhang, L., Sun, R., Liu, J., Yin, H., Li, X., Zheng, X., & Zeng, H. (2016). Role of thioredoxin reductase 1 in dysplastic transformation of human breast epithelial cells triggered by chronic oxidative stress. Scientific Reports, 6, 36860.

Du, X., Li, H., Wang, Z., Qiu, S., Liu, Q., & Ni, J. (2013). Selenoprotein P and selenoprotein M block Zn2+-mediated Aβ42 aggregation and toxicity. Metallomics, 5(7), 861–870.

Emmink, B. L., Laoukili, J., Kipp, A. P., Koster, J., Govaert, K. M., Fatrai, S., Verheem, A., Steller, E. J., Brigelius-Flohé, R., Jimenez, C. R., Borel Rinkes, I. H., & Kranenburg, O. (2014). GPx2 suppression of H2O2 stress links the formation of differentiated tumor mass to metastatic capacity in colorectal cancer. Cancer Research, 74(22), 6717–6730.

Fan, H., Wu, P. F., Zhang, L., Hu, Z. L., Wang, W., Guan, X. L., Luo, H., Ni, M., Yang, J. W., Li, M. X., Chen, J. G., & Wang, F. (2015). Methionine sulfoxide reductase A negatively controls microglia-mediated neuroinflammation via inhibiting ROS/MAPKs/NF-κB signaling pathways through a catalytic antioxidant function. Antioxidants and Redox Signaling, 22(10), 832–847.

Fan, R. F., Cao, C. Y., Chen, M. H., Shi, Q. X., & Xu, S. W. (2018). Gga-let-7f-3p promotes apoptosis in selenium deficiency-induced skeletal muscle by targeting selenoprotein K. Metallomics, 10(7), 941–952.

Fredericks, G. J., Hoffmann, F. W., Rose, A. H., Osterheld, H. J., Hess, F. M., Mercier, F., & Hoffmann, P. R. (2014). Stable expression and function of the inositol 1, 4, 5-triphosphate receptor requires palmitoylation by a DHHC6/selenoprotein K complex. Proceedings of the National Academy of Sciences of the USA, 111(46), 16478–16483.

Gan, F., Hu, Z., Zhou, Y., & Huang, K. (2017). Overexpression and low expression of selenoprotein S impact ochratoxin A-induced porcine cytotoxicity and apoptosis in vitro. Journal of Agricultural and Food Chemistry, 65(32), 6972–6981.

Gladyshev, V. N. (2014). Selenium and methionine sulfoxide reduction. Free Radical Biology and Medicine, 75(1), S8–S9.

Gong, T., Hashimoto, A. C., Sasuclark, A. R., Khadka, V. S., Gurary, A., & Pitts, M. W. (2019). Selenoprotein M promotes hypothalamic leptin signaling and thioredoxin antioxidant activity. Antioxidants and Redox Signaling, 2019, in print.

Gong, T., Torres, D. J., Berry, M. J., & Pitts, M. W. (2018). Hypothalamic redox balance and leptin signaling – Emerging role of selenoproteins. Free Radical Biology and Medicine, 127, 172–181.

Guerriero, E., Accardo, M., Capone, F., Colonna, G., Castello, G., & Costantini, S. (2014). Assessment of the selenoprotein M (SELM) over-expression on human hepatocellular carcinoma tissues by immunohistochemistry. European Journal of Histochemistry, 58(4), 287–290.

Hamieh, A., Cartier, D., Abid, H., Calas, A., Burel, C., Bucharles, C., Jehan, C., Grumolato, L., Landry, M., Lerouge, P., Anouar, Y., & Lihrmann, I. (2017). Selenoprotein T is a novel OST subunit that regulates UPR signaling and hormone secretion. EMBO Reports: European Molecular Biology Organization reports, 18(11), 1935–1946.

Han, L., Yang, X., Sun, W., Li, Z., Ren, H., Li, B., Zhang, R., Zhang, D., Shi, Z., Liu, J., Cao, J., Zhang, J., & Xiong, Y. (2018). The study of GPX3 methylation in patients with Kashin-Beck Disease and its mechanism in chondrocyte apoptosis. Bone, 117, 15–22.

He, Q., Li, H., Meng, F., Sun, X., Feng, X., Chen, J., Li, L., & Liu, J. (2018). Methionine sulfoxide reductase B1 regulates hepatocellular carcinoma cell proliferation and invasion via the mitogen-activated protein kinase pathway and epithelial-mesenchymal transition. Oxidative Medicine and Cellular Longevity, 2018, 5287971.

Hu, X., Luo, J., Lai, H., Li, M., Zheng, X., Nie, T., Li, F., & Li, H. (2018). Knockdown of Trnau1ap inhibits the proliferation and migration of NIH3T3, JEG-3 and Bewo cells via the PI3K/Akt signaling pathway. Biochemical and Biophysical Research Communications, 503(2), 521–527.

Hudson, T. S., Carlson, B. A., Hoeneroff, M. J., Young, H. A., Sordillo, L., Muller, W. J., Hatfield, D. L., & Green, J. E. (2012). Selenoproteins reduce susceptibility to DMBA-induced mammary carcinogenesis. Carcinogenesis, 33(6), 1225–1230.

Ishikura, K., Misu, H., Kumazaki, M., Takayama, H., Matsuzawa-Nagata, N., Tajima, N., Chikamoto, K., Lan, F., Ando, H., Ota, T., Sakurai, M., Takeshita, Y., Kato, K., Fujimura, A., Miyamoto, K., Saito, Y., Kameo, S., Okamoto, Y., Takuwa, Y., Takahashi, K., Kidoya, H., Takakura, N., Kaneko, S., & Takamura, T. (2014). Selenoprotein P as a diabetes-associated hepatokine that impairs angiogenesis by inducing VEGF resistance in vascular endothelial cells. Diabetologia, 57(9), 1968–1976.

Jiang, H., Shi, Q. Q., Ge, L. Y., Zhuang, Q. F., Xue, D., Xu, H. Y., & He, X. Z. (2019). Selenoprotein M stimulates the proliferative and metastatic capacities of renal cell carcinoma through activating the PI3K/AKT/mTOR pathway. Cancer Medicine, 8(10), 4836–4844.

Jiang, Y. Y., Huang, J. Q., Lin, G. C., Guo, H. Y., Ren, F. Z., & Zhang, H. (2015). Characterization and expression of chicken selenoprotein U. Biological Trace Element Research, 166(2), 216–224.

Jiang, Y., Feng, X., Zheng, L., Li, S. L., Ge, X. Y., & Zhang, J. G. (2015). Thioredoxin 1 mediates TGF-β-induced epithelial-mesenchymal transition in salivary adenoid cystic carcinoma. Oncotarget, 6(28), 25506–25519.

Kaushal, N., Kudva, A. K., Patterson, A. D., Chiaro, C., Kennett, M. J., Desai, D., Amin, S., Carlson, B. A., Cantorna, M. T., & Prabhu, K. S. (2014). Crucial role of macrophage selenoproteins in experimental colitis. Journal of Immunology, 193(7), 3683–3692.

Kikuchi, N., Satoh, K., Kurosawa, R., Yaoita, N., Elias-Al-Mamun, M., Siddique, M. A. H., Omura, J., Satoh, T., Nogi, M., Sunamura, S., Miyata, S., Saito, Y., Hoshikawa, Y., Okada, Y., & Shimokawa, H. (2018). Selenoprotein P promotes the development of pulmonary arterial hypertension. Circulation, 138(6), 600–623.

Klutho, P. J., Pennington, S. M., Scott, J. A., Wilson, K. M., Gu, S. X., Doddapattar, P., Xie, L., Venema, A. N., Zhu, L. J., Chauhan, A. K., Lentz, S. R., & Grumbach, I. M. (2015). Deletion of methionine sulfoxide reductase A does not affect atherothrombosis but promotes neointimal hyperplasia and extracellular signal-regulated kinase signaling. Arteriosclerosis Thrombosis and Vascular Biology, 35(12), 2594–2604.

Kumaraswamy, E., Carlson, B. A., Morgan, F., Miyoshi, K., Robinson, G. W., Su, D., Wang, S., Southon, E., Tessarollo, L., Lee, B. J., Gladyshev, V. N., Hennighausen, L., & Hatfield, D. L. (2003). Selective removal of the selenocysteine tRNA [Ser]Sec gene (Trsp) in mouse mammary epithelium. Molecular and Cellular Biology, 23(5), 1477–1488.

Kuras, R., Reszka, E., Wieczorek, E., Jablonska, E., Gromadzinska, J., Malachowska, B., Kozlowska, L., Stanislawska, M., Janasik, B., & Wasowicz, W. (2018). Biomarkers of selenium status and antioxidant effect in workers occupationally exposed to mercury. Journal of Trace Elements in Medicine and Biology, 49, 43–50.

Kurokawa, S., Bellinger, F. P., Hill, K. E., Burk, R. F., & Berry, M. J. (2014). Isoform-specific binding of selenoprotein P to the β-propeller domain of apolipoprotein E receptor 2 mediates selenium supply. Journal of Biological Chemistry, 289(13), 9195–9207.

Kurokawa, S., Hill, K. E., McDonald, W. H., & Burk, R. F. (2012). Long isoform mouse selenoprotein P (Sepp1) supplies rat myoblast L8 cells with selenium via endocytosis mediated by heparin binding properties and apolipoprotein E receptor-2 (ApoER2). Journal of Biological Chemistry, 287(34), 28717–28726.

Kwak, G. H., & Kim, H. Y. (2017). MsrB3 deficiency induces cancer cell apoptosis through p53-independent and ER stress-dependent pathways. Archives of Biochemistry and Biophysics, 621, 1–5.

Lee, B. C., Lee, S. G., Choo, M. K., Kim, J. H., Lee, H. M., Kim, S., Fomenko, D. E., Kim, H. Y., Park, J. M., & Gladyshev, V. N. (2017). Selenoprotein MsrB1 promotes anti-inflammatory cytokine gene expression in macrophages and controls immune response in vivo. Scientific Reports, 7(1), 5119.

Lee, J. H., Kwon, J. H., Jeon, Y. H., Ko, K. Y., Lee, S. R., & Kim, I. Y. (2014). Pro178 and Pro183 of selenoprotein S are essential residues for interaction with p97(VCP) during endoplasmic reticulum-associated degradation. The Journal of Biological Chemistry, 289(20), 13758–13768.

Lee, J. H., Park, K. J., Jang, J. K., Jeon, Y. H., Ko, K. Y., Kwon, J. H., Lee, S. R., & Kim, I. Y. (2015). Selenoprotein S-dependent selenoprotein K binding to p97(VCP) protein is essential for endoplasmic reticulum-associated degradation. The Journal of Biological Chemistry, 290(50), 29941–29952.

Li, H., He, Q., Meng, F., Feng, X., Chen, J., Li, L., & Liu, J. (2018). Methionine sulfoxide reductase B1 regulates proliferation and invasion by affecting mitogen-activated protein kinase pathway and epithelial-mesenchymal transition in cells. Biochemical and Biophysical Research Communications, 496(3), 806–813.

Li, M., Cheng, W., Luo, J., Hu, X., Nie, T., Lai, H., Zheng, X., Li, F., & Li, H. (2017). Loss of selenocysteine insertion sequence binding protein 2 suppresses the proliferation, migration/invasion and hormone secretion of human trophoblast cells via the PI3K/Akt and ERK signaling pathway. Placenta, 55, 81–89.

Li, M., Cheng, W., Nie, T., Lai, H., Hu, X., Luo, J., Li, F., & Li, H. (2018). Selenoprotein K mediates the proliferation, migration, and invasion of human choriocarcinoma cells by negatively regulating human chorionic gonadotropin expression via ERK, p38 MAPK, and akt signaling pathway. Biological Trace Element Research, 184(1), 47–59.

Li, X., Chen, M., Yang, Z., Wang, W., Lin, H., & Xu, S. (2018). Selenoprotein S silencing triggers mouse hepatoma cells apoptosis and necrosis involving in intracellular calcium imbalance and ROS-mPTP-ATP. Biochimica et Biophysica Acta. General Subjects, 1862(10), 2113–2123.

Liu, J. S., & Cui, Z. J. (2019). Pancreatic stellate cells serve as a brake mechanism on pancreatic acinar cell calcium signaling modulated by methionine sulfoxide Reductase Expression. Cells, 8(2), e109.

Liu, L., Geng, X., Cai, Y., Copple, B., Yoshinaga, M., Shen, J., Nebert, D. W., Wang, H., & Liu, Z. (2018). Hepatic ZIP8 deficiency is associated with disrupted selenium homeostasis, liver pathology, and tumor formation. American Journal of Physiology Gastrointestinal and Liver Physiology, 315(4), G569–G579.

Luchman, H. A., Villemaire, M. L., Bismar, T. A., Carlson, B. A., & Jirik, F. R. (2014). Prostate epithelium-specific deletion of the selenocysteine tRNA gene Trsp leads to early onset intraepithelial neoplasia. American Journal of Pathology, 184(3), 871–877.

Marciel, M. P., & Hoffmann, P. R. (2019). Molecular mechanisms by which selenoprotein K regulates immunity and cancer. Biological Trace Element Research, 192(1), 60–68.

Marciel, M. P., Khadka, V. S., Deng, Y., Kilicaslan, P., Pham, A., Bertino, P., Lee, K., Chen, S., Glibetic, N., Hoffmann, F. W., Matter, M. L., & Hoffmann, P. R. (2018). Selenoprotein K deficiency inhibits melanoma by reducing calcium flux required for tumor growth and metastasis. Oncotarget, 9(17), 13407–13422.

Mariclair, A. R., Frederick, P. B., & Marla, J. B. (2010). The neuroprotective functions of selenoprotein M and its role in cytosolic calcium regulation. Antioxidants and Redox Signaling, 12(7), 809–818.

Marino, M., Stoilova, T., Giorgi, C., Bachi, A., Cattaneo, A., Auricchio, A., Pinton, P., & Zito, E. (2015). SEPN1, endoplasmic reticulum-localized selenoprotein linked to skeletal muscle pathology, counteracts hyperoxidation by means of redox-regulating SERCA2 pump activity. Human Molecular Genetics, 24(7), 1843–1855.

Men, L., Yu, S., Yao, J., Li, Y., Ren, D., & Du, J. (2018). Selenoprotein S protects against adipocyte death through mediation of the IRE1α-sXBP1 pathway. Biochemical and Biophysical Research Communications, 503(4), 2866–2871.

Méplan, C., Dragsted, L. O., Ravn-Haren, G., Tjonneland, A., Vogel, U., & Hesketh, J. (2013). Association between polymorphisms in glutathione peroxidase and selenoprotein P genes, glutathione peroxidase activity, HRT use and breast cancer risk. PLoS One, 8(9), e73316.

Min, Z., Guo, Y., Sun, M., Hussain, S., Zhao, Y., Guo, D., Huang, H., Heng, L., Zhang, F., Ning, Q., Han, Y., Xu, P., Zhong, N., Sun, J., & Lu, S. (2018). Selenium-sensitive miRNA-181a-5p targeting SBP2 regulates selenoproteins expression in cartilage. Journal of Cellular and Molecular Medicine, 22(12), 5888–5898.

Misu, H., Takamura, T., Takayama, H., Hayashi, H., Matsuzawa-Nagata, N., Kurita, S., Ishikura, K., Ando, H., Takeshita, Y., Ota, T., Sakurai, M., Yamashita, T., Mizukoshi, E., Yamashita, T., Honda, M., Miyamoto, K., Kubota, T., Kubota, N., Kadowaki, T., Kim, H. J., Lee, I. K., Minokoshi, Y., Saito, Y., Takahashi, K., Yamada, Y., Takakura, N., & Kaneko, S. (2010). A liver-derived secretory protein, selenoprotein P, causes insulin resistance. Cell Metabolism, 12(5), 483–495.

Misu, H., Takayama, H., Saito, Y., Mita, Y., Kikuchi, A., Ishii, K. A., Chikamoto, K., Kanamori, T., Tajima, N., Lan, F., Takeshita, Y., Honda, M., Tanaka, M., Kato, S., Matsuyama, N., Yoshioka, Y., Iwayama, K., Tokuyama, K., Akazawa, N., Maeda, S., Takekoshi, K., Matsugo, S., Noguchi, N., Kaneko, S., & Takamura, T. (2017). Deficiency of the hepatokine selenoprotein P increases responsiveness to exercise in mice through upregulation of reactive oxygen species and AMP-activated protein kinase in muscle. Nature Medicine, 23(4), 508–516.

Müller, M. F., Florian, S., Pommer, S., Osterhoff, M., Esworthy, R. S., Chu, F. F., Brigelius-Flohé, R., & Kipp, A. P. (2013). Deletion of glutathione peroxidase-2 inhibits azoxymethane-induced colon cancer development. PLoS One, 8(8), e72055.

Naiki, T., Naiki-Ito, A., Asamoto, M., Kawai, N., Tozawa, K., Etani, T., Sato, S., Suzuki, S., Shirai, T., Kohri, K., & Takahashi, S. (2014). GPX2 overexpression is involved in cell proliferation and prognosis of castration-resistant prostate cancer. Carcinogenesis, 35(9), 1962–1967.

Naiki, T., Naiki-Ito, A., Iida, K., Etani, T., Kato, H., Suzuki, S., Yamashita, Y., Kawai, N., Yasui, T., & Takahashi, S. (2018). GPX2 promotes development of bladder cancer with squamous cell differentiation through the control of apoptosis. Oncotarget, 9(22), 15847–15859.

Ojeda, M. L., Carreras, O., Sobrino, P., Murillo, M. L., & Nogales, F. (2017). Biological implications of selenium in adolescent rats exposed to binge drinking: Oxidative, immunologic and apoptotic balance. Toxicology and Applied Pharmacology, 329, 165–172.

Pelosof, L., Yerram, S., Armstrong, T., Chu, N., Danilova, L., Yanagisawa, B., Hidalgo, M., Azad, N., & Herman, J. G. (2017). GPX3 promoter methylation predicts platinum sensitivity in colorectal cancer. Epigenetics, 12(7), 540–550.

Pérez, S., Taléns-Visconti, R., Rius-Pérez, S., Finamor, I., & Sastre, J. (2017). Redox signaling in the gastrointestinal tract. Free Radical Biology and Medicine, 104, 75–103.

Pozzer, D., Varone, E., Chernorudskiy, A., Schiarea, S., Missiroli, S., Giorgi, C., Pinton, P., Canato, M., Germinario, E., Nogara, L., Blaauw, B., & Zito, E. (2019). A maladaptive ER stress response triggers dysfunction in highly active muscles of mice with selenon loss. Redox Biology, 20, 354–366.

Raninga, P. V., Di Trapani, G., Vuckovic, S., & Tonissen, K. F. (2016). TrxR1 inhibition overcomes both hypoxia-induced and acquired bortezomib resistance in multiple myeloma through NF-кβ inhibition. Cell Cycle, 15(4), 559–572.

Raninga, P. V., Di Trapani, G., Vuckovic, S., Bhatia, M., & Tonissen, K. F. (2015). Inhibition of thioredoxin 1 leads to apoptosis in drug-resistant multiple myeloma. Oncotarget, 6(17), 15410–15424.

Reeves, M. A., Bellinger, F. P., & Berry, M. J. (2010). The neuroprotective functions of selenoprotein M and its role in cytosolic calcium regulation. Antioxidants and Redox Signaling, 12(7), 809–818.

Ren, B., Huang, Y., Zou, C., Wu, Y., Huang, Y., Ni, J., & Tian, J. (2019). Transcriptional regulation of selenoprotein F by heat shock factor 1 during selenium supplementation and stress response. Cells, 8(5), e479.

Rocca, C., Boukhzar, L., Granieri, M. C., Alsharif, I., Mazza, R., Lefranc, B., Tota, B., Leprince, J., Cerra, M. C., Anouar, Y., & Angelone, T. (2018). A selenoprotein T-derived peptide protects the heart against ischaemia/reperfusion injury through inhibition of apoptosis and oxidative stress. Acta Physiologica (Oxford), 223(4), e13067.

Romitti, M., Wajner, S. M., Ceolin, L., Ferreira, C. V., Ribeiro, R. V., Rohenkohl, H. C., Weber, Sde, S., Lopez, P. L., Fuziwara, C. S., Kimura, E. T., & Maia, A. L. (2016). MAPK and SHH pathways modulate type 3 deiodinase expression in papillary thyroid carcinoma. Endocrine Related Cancer, 23(3), 135–146.

Santesmasses, D., Mariotti, M., & Gladyshev, V. N. (2019). Tolerance to selenoprotein loss differs between human and mouse. Molecular Biology and Evolution, 27, msz218.

Sattar, H., Yang, J., Zhao, X., Cai, J., Liu, Q., Ishfaq, M., Yang, Z., Chen, M., Zhang, Z., & Xu, S. (2018). Selenoprotein-U (SelU) knockdown triggers autophagy through PI3K-Akt-mTOR pathway inhibition in rooster sertoli cells. Metallmics, 10(7), 929–940.

Schweizer, U., Schlicker, C., Braun, D., Köhrle, J., & Steegborn, C. (2014). Crystal structure of mammalian selenocysteine-dependent iodothyronine deiodinase suggests a peroxiredoxin-like catalytic mechanism". Proceedings of the National Academy of Sciences of the United States of America, 111(29), 10526–10531.

Sengupta, A., Lichti, U. F., Carlson, B. A., Cataisson, C., Ryscavage, A. O., Mikulec, C., Conrad, M., Fischer, S. M., Hatfield, D. L., & Yuspa, S. H. (2013). Targeted disruption of glutathione peroxidase 4 in mouse skin epithelial cells impairs postnatal hair follicle morphogenesis that is partially rescued through inhibition of COX-2. Journal of Investigative Dermatology, 133(7), 1731–1741.

Short, S. P., & Williams, C. S. (2017). Selenoproteins in tumorigenesis and cancer progression. Advances in Cancer Research, 136, 49–83.

Singh, M. P., Kim, K. Y., Kwak, G. H., Baek, S. H., & Kim, H. Y. (2017). Methionine sulfoxide reductase A protects against lipopolysaccharide-induced septic shock via negative regulation of the proinflammatory responses. Archives of Biochemistry and Biophysics, 631, 42–48.

Singh, M., Venugopal, C., Tokar, T., McFarlane, N., Subapanditha, M. K., Qazi, M., Bakhshinyan, D., Vora, P., Murty, N. K., Jurisica, I., & Singh, S. K. (2018). Therapeutic targeting of the premetastatic stage in human lung-to-brain metastasis. Cancer Research, 78(17), 5124–5134.

Strauss, E., Tomczak, J., Staniszewski, R., & Oszkinis, G. (2018). Associations and interactions between variants in selenoprotein genes, selenoprotein levels and the development of abdominal aortic aneurysm, peripheral arterial disease, and heart failure. PLoS One, 13(9), e0203350.

Sun, X., Yuan, Y., Xiao, Y., Lu, Q., Yang, L., Chen, C., & Guo, Q. (2018). Long non-coding RNA, Bmcob, regulates osteoblastic differentiation of bone marrow mesenchymal stem cells. Biochemical and Biophysical Research Communications, 506(3), 536–542.

Tang, J., He, A., Yan, H., Jia, G., Liu, G., Chen, X., Cai, J., Tian, G., Shang, H., & Zhao, H. (2018). Damage to the myogenic differentiation of C2C12 cells by heat stress is associated with up-regulation of several selenoproteins. Scientific Reports, 8(1), 10601.

Topkas, E., Cai, N., Cumming, A., Hazar-Rethinam, M., Gannon, O. M., Burgess, M., Saunders, N. A., & Endo-Munoz, L. (2016). Auranofin is a potent suppressor of osteosarcoma metastasis. Oncotarget, 7(1), 831–844.

Tsuji, P. A., Carlson, B. A., Yoo, M. H., Naranjo-Suarez, S., Xu, X. M., He, Y., Asaki, E., Seifried, H. E., Reinhold, W. C., Davis, C. D., Gladyshev, V. N., & Hatfield, D. L. (2015). The 15kDa selenoprotein and thioredoxin reductase 1 promote colon cancer by different pathways. PLoS One, 10(4), e0124487.

Varone, E., Pozzer, D., Di Modica, S., Chernorudskiy, A., Nogara, L., Baraldo, M., Cinquanta, M., Fumagalli, S.,Villar-Quiles, R. N., De Simoni, M. G., Blaauw, B., Ferreiro, A., & Zito, E. (2019). Selenon (SEPN1) protects skeletal muscle from saturated fatty acid-induced ER stress and insulin resistance. Redox Biology, 24, 101176.

Vindry, C., Guillin, O., Mangeot, P. E., Ohlmann, T., & Chavatte, L. (2019). A versatile strategy to reduce UGA-selenocysteine recoding efficiency of the ribosome using CRISPR-Cas9-viral-like-particles targeting selenocysteine-tRNA[Ser]Sec gene. Cells, 8(6), e574.

Wang, C., Li, R., Huang, Y., Wang, M., Yang, F., Huang, D., Wu, C., Li, Y., Tang, Y., Zhang, R., & Cheng, J. (2017). Selenoprotein K modulate intracellular free Ca2+ by regulating expression of calcium homoeostasis endoplasmic reticulum protein. Biochemical and Biophysical Research Communications, 484(4), 734–739.

Wang, H., Luo, K., Tan, L. Z., Ren, B. G., Gu, L. Q., Michalopoulos, G., Luo, J. H., & Yu, Y. P. (2012). p53-induced gene 3 mediates cell death induced by glutathione peroxidase 3. Journal of Biological Chemistry, 287(20), 16890–16902.

Watrowski, R., Dan, C. T., Fabjani, G., Schuster, E., Fischer, M., & Zeillinger, R. (2016). The 811 C/T polymorphism in the 3' untranslated region of the selenoprotein 15kDa (Sep15) gene and breast cancer in Caucasian women. Tumour Biology, 37(1), 1009–1015.

Xue, J., Min, Z., Xia, Z., Cheng, B., Lan, B., Zhang, F., Han, Y., Wang, K., & Sun, J. (2018). The hsa-miR-181a-5p reduces oxidation resistance by controlling SECISBP2 in osteoarthritis. BMC Musculoskeletal Disorders, 19(1), 355.

Yan, J., Fei, Y., Han, Y., & Lu, S. (2016). Selenoprotein O deficiencies suppress chondrogenic differentiation of ATDC5 cells. Cell Biology International, 40(10), 1033–1040.

Yang, W. S., SriRamaratnam, R., Welsch, M. E., Shimada, K., Skouta, R., Viswanathan, V. S., Cheah, J. H., Clemons, P. A., Shamji, A. F., Clish, C. B., Brown, L. M., Girotti, A. W., Cornish, V. W., Schreiber, S. L., & Stockwell, B. R. (2014). Regulation of ferroptotic cancer cell death by GPX4. Cell, 156(1–2), 317–331.

Yang, W., Shen, Y., Wei, J., & Liu, F. (2015). MicroRNA-153/Nrf-2/GPx1 pathway regulates radiosensitivity and stemness of glioma stem cells via reactive oxygen species. Oncotarget, 6(26), 22006–22027.

Ye, Y., Bian, W., Fu, F., Hu, J., & Liu, H. (2018). Selenoprotein S inhibits inflammation-induced vascular smooth muscle cell calcification. Journal of Biological Inorganic Chemistry, 23(5), 739–751.

Yu, D., Zhang, Z. W., Yao, H. D., Li, S., & Xu, S. W. (2014). Antioxidative role of selenoprotein W in oxidant-induced chicken splenic lymphocyte death. Biometals, 27(2), 277–291.

Yu, D., Zhang, Z., Yao, H., Li, S., & Xu, S. W. (2015). The role of selenoprotein W in inflammatory injury in chicken immune tissues and cultured splenic lymphocyte. Biometals, 28(1), 75–87.

Zhao, H., Li, J., Li, X., Han, C., Zhang, Y., Zheng, L., & Guo, M. (2015). Silencing GPX3 expression promotes tumor metastasis in human thyroid cancer. Current Protein and Peptide Science, 16(4), 316–321.

Zhao, L., Zheng, Y. Y., Chen, Y., Ma, Y. T., Yang, Y. N., Li, X. M., Ma, X., & Xie, X. (2018). Association of genetic polymorphisms of SelS with Type 2 diabetes in a Chinese population. Bioscience Reports, 38(6), BSR20181696.

Zhou, J., Li, C., Gu, G., Wang, Q., & Guo, M. (2018). Selenoprotein N was required for the regulation of selenium on the uterine smooth muscle contraction in mice. Biological Trace Element Research, 183(1), 138–146.

Zhu, S., Zhang, Q., Sun, X., Zeh, H. J., Lotze, M. T., Kang, R., & Tang, D. (2017). HSPA5 regulates ferroptotic cell death in cancer cells. Cancer Research, 77(8), 2064–2077.

Published
2020-04-23
How to Cite
Stanishevska, N. V. (2020). Selenoproteins and their emerging roles in signaling pathways . Regulatory Mechanisms in Biosystems, 11(2), 186-199. https://doi.org/10.15421/022028