Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Selenium Donors at the Junction of Inflammatory Diseases

Author(s): Rama Alhasan, Ammar Kharma, Pierre Leroy, Claus Jacob and Caroline Gaucher*

Volume 25, Issue 15, 2019

Page: [1707 - 1716] Pages: 10

DOI: 10.2174/1381612825666190701153903

Price: $65

Abstract

Selenium is an essential non-metal trace element, and the imbalance in the bioavailability of selenium is associated with many diseases ranking from acute respiratory distress syndrome, myocardial infarction and renal failure (Se overloading) to diseases associated with chronic inflammation like inflammatory bowel diseases, rheumatoid arthritis, and atherosclerosis (Se unload). The only source of selenium is the diet (animal and cereal sources) and its intestinal absorption is limiting for selenocysteine and selenomethionine synthesis and incorporation in selenoproteins. In this review, after establishing the link between selenium and inflammatory diseases, we envisaged the potential of selenium nanoparticles and organic selenocompounds to compensate the deficit of selenium intake from the diet. With high selenium loading, nanoparticles offer a low dosage to restore selenium bioavailability whereas organic selenocompounds can play a role in the modulation of their antioxidant or antiinflammatory activities.

Keywords: Selenium, redox, inflammation, organoselenium, selenium nanoparticles, selenoproteins.

[1]
Ursini F, Bindoli A. The role of selenium peroxidases in the protection against oxidative damage of membranes. Chem Phys Lipids 1987; 44(2-4): 255-76. [http://dx.doi.org/10.1016/0009-3084(87)90053-3]. [PMID: 3311419].
[2]
Gaucher C, Boudier A, Dahboul F, Parent M, Leroy P. S-nitrosation/denitrosation in cardiovascular pathologies: Facts and concepts for the rational design of S-nitrosothiols. Curr Pharm Des 2013; 19(3): 458-72. [http://dx.doi.org/10.2174/138161213804143635]. [PMID: 22920903].
[3]
Asahi M, Fujii J, Takao T, et al. The oxidation of selenocysteine is involved in the inactivation of glutathione peroxidase by nitric oxide donor. J Biol Chem 1997; 272(31): 19152-7. [http://dx.doi.org/10.1074/jbc.272.31.19152]. [PMID: 9235904].
[4]
Fujii J, Taniguchi N. Down regulation of superoxide dismutases and glutathione peroxidase by reactive oxygen and nitrogen species. Free Radic Res 1999; 31(4): 301-8. [http://dx.doi.org/10.1080/10715769900300861]. [PMID: 10517534].
[5]
Dobashi K, Asayama K, Nakane T, Kodera K, Hayashibe H, Nakazawa S. Induction of glutathione peroxidase in response to inactivation by nitric oxide. Free Radic Res 2001; 35(3): 319-27. [http://dx.doi.org/10.1080/10715760100300851]. [PMID: 11697130].
[6]
Cha W, Meyerhoff ME. Catalytic generation of nitric oxide from S-nitrosothiols using immobilized organoselenium species. Biomaterials 2007; 28(1): 19-27. [http://dx.doi.org/10.1016/j.biomaterials.2006.08.019]. [PMID: 16959311].
[7]
Cai W, Wu J, Xi C, Ashe AJ III, Meyerhoff ME. Carboxyl-ebselen-based layer-by-layer films as potential antithrombotic and antimicrobial coatings. Biomaterials 2011; 32(31): 7774-84. [http://dx.doi.org/10.1016/j.biomaterials.2011.06.075]. [PMID: 21794909].
[8]
Weng Y, Song Q, Zhou Y, et al. Immobilization of selenocystamine on TiO2 surfaces for in situ catalytic generation of nitric oxide and potential application in intravascular stents. Biomaterials 2011; 32(5): 1253-63. [http://dx.doi.org/10.1016/j.biomaterials.2010.10.039]. [PMID: 21093045].
[9]
Yang Z, Yang Y, Xiong K, et al. Nitric oxide producing coating mimicking endothelium function for multifunctional vascular stents. Biomaterials 2015; 63: 80-92. [http://dx.doi.org/10.1016/j.biomaterials.2015.06.016]. [PMID: 26093790].
[10]
Mugesh G, du Mont W-W, Sies H. Chemistry of biologically important synthetic organoselenium compounds. Chem Rev 2001; 101(7): 2125-79. [http://dx.doi.org/10.1021/cr000426w]. [PMID: 11710243].
[11]
Ávila DS, Gubert P, Palma A, et al. An organotellurium compound with antioxidant activity against excitotoxic agents without neurotoxic effects in brain of rats. Brain Res Bull 2008; 76(1-2): 114-23. [http://dx.doi.org/10.1016/j.brainresbull.2007.12.008]. [PMID: 18395620].
[12]
Ramoutar RR, Brumaghim JL. Antioxidant and anticancer properties and mechanisms of inorganic selenium, oxo-sulfur, and oxo-selenium compounds. Cell Biochem Biophys 2010; 58(1): 1-23. [http://dx.doi.org/10.1007/s12013-010-9088-x]. [PMID: 20632128].
[13]
Combs GF Jr. Selenium in global food systems. Br J Nutr 2001; 85(5): 517-47. [http://dx.doi.org/10.1079/BJN2000280]. [PMID: 11348568].
[14]
Rayman MP, Rayman MP. The argument for increasing selenium intake. Proc Nutr Soc 2002; 61(2): 203-15. [http://dx.doi.org/10.1079/PNS2002153]. [PMID: 12133202].
[15]
Surai PF. elenium in food and feed: Selenomethionine and beyond. urai PF, ed^eds, Selenium in nutrition and health Nottingham University Press: Nottingham, UK 2006; pp. 151-212.
[16]
Rayman MP, Thompson AJ, Bekaert B, et al. Randomized controlled trial of the effect of selenium supplementation on thyroid function in the elderly in the United Kingdom. Am J Clin Nutr 2008; 87(2): 370-8. [http://dx.doi.org/10.1093/ajcn/87.2.370]. [PMID: 18258627].
[17]
Selenium RAS. In: Bowman B RR, ed eds, In Present Knowldege in Nutrition ILSI Press: Washington, DC, USA. 2001.361.
[18]
Stoffaneller R, Morse NL. A review of dietary selenium intake and selenium status in Europe and the Middle East. Nutrients 2015; 7(3): 1494-537. [http://dx.doi.org/10.3390/nu7031494]. [PMID: 25734564].
[19]
Medicine Io. Dietary Reference Intakes for Vitamin C. Vitamin E, Selenium, and Carotenoids 2000.
[20]
Whanger PD. Selenium and its relationship to cancer: An update. Br J Nutr 2004; 91(1): 11-28. [http://dx.doi.org/10.1079/BJN20031015]. [PMID: 14748935].
[21]
Yamashita Y, Yamashita M. Identification of a novel selenium-containing compound, selenoneine, as the predominant chemical form of organic selenium in the blood of bluefin tuna. J Biol Chem 2010; 285(24): 18134-8. [http://dx.doi.org/10.1074/jbc.C110.106377]. [PMID: 20388714].
[22]
Hartmanis MG, Sliwkowski MX. Selenomethionine-containing thiolase and 3-hydroxybutyryl-CoA dehydrogenase from Clostridium kluyveri. Curr Top Cell Regul 1985; 27: 479-86. [http://dx.doi.org/10.1016/B978-0-12-152827-0.50048-7]. [PMID: 4092496].
[23]
Whanger PD. Selenocompounds in plants and animals and their biological significance. J Am Coll Nutr 2002; 21(3): 223-32. [http://dx.doi.org/10.1080/07315724.2002.10719214]. [PMID: 12074249].
[24]
Quijano MA, Moreno P, Gutiérrez AM, Pérez-Conde MC, Cámara C. Selenium speciation in animal tissues after enzymatic digestion by high-performance liquid chromatography coupled to inductively coupled plasma mass spectrometry. J Mass Spectrom 2000; 35(7): 878-84. [http://dx.doi.org/10.1002/1096-9888(200007)35:7<878:AID-JMS12>3.0.CO;2-2]. [PMID: 10934441].
[25]
Bryszewska MA, Måge A. Determination of selenium and its compounds in marine organisms. J Trace Elem Med Biol 2015; 29: 91-8. [http://dx.doi.org/10.1016/j.jtemb.2014.10.004]. [PMID: 25468190].
[26]
Drake EN. Cancer chemoprevention: Selenium as a prooxidant, not an antioxidant. Med Hypotheses 2006; 67(2): 318-22. [http://dx.doi.org/10.1016/j.mehy.2006.01.058]. [PMID: 16574336].
[27]
Okuno T, Motobayashi S, Ueno H, Nakamuro K. Identification of mouse selenomethionine α,γ-elimination enzyme: Cystathionine γ-lyase catalyzes its reaction to generate methylselenol. Biol Trace Elem Res 2005; 108(1-3): 245-57. [http://dx.doi.org/10.1385/BTER:108:1-3:245]. [PMID: 16327076].
[28]
Suzuki KT, Kurasaki K, Suzuki N. Selenocysteine β-lyase and methylselenol demethylase in the metabolism of Se-methylated selenocompounds into selenide. Biochim Biophys Acta 2007; 1770(7): 1053-61. [http://dx.doi.org/10.1016/j.bbagen.2007.03.007]. [PMID: 17451884].
[29]
Lu J, Berndt C, Holmgren A. Metabolism of selenium compounds catalyzed by the mammalian selenoprotein thioredoxin reductase. Biochim Biophys Acta 2009; 1790(11): 1513-9. [http://dx.doi.org/10.1016/j.bbagen.2009.04.013]. [PMID: 19406206].
[30]
Labunskyy VM, Hatfield DL, Gladyshev VN. Selenoproteins: Molecular pathways and physiological roles. Physiol Rev 2014; 94(3): 739-77. [http://dx.doi.org/10.1152/physrev.00039.2013]. [PMID: 24987004].
[31]
Rose AH, Hoffmann PR. Selenoproteins and cardiovascular stress. Thromb Haemost 2015; 113(3): 494-504. [http://dx.doi.org/10.1160/TH14-07-0603]. [PMID: 25354851].
[32]
Canolty NL, Nasset ES. Intestinal absorption of free and protein-bound dietary methionine in the rat. J Nutr 1975; 105(7): 867-77. [http://dx.doi.org/10.1093/jn/105.7.867]. [PMID: 1138031].
[33]
Nickel A, Kottra G, Schmidt G, Danier J, Hofmann T, Daniel H. Characteristics of transport of selenoamino acids by epithelial amino acid transporters. Chem Biol Interact 2009; 177(3): 234-41. [http://dx.doi.org/10.1016/j.cbi.2008.09.008]. [PMID: 18845132].
[34]
Bergeron MJ, Clémençon B, Hediger MA, Markovich D. SLC13 family of Na+-coupled di- and tri-carboxylate/sulfate transporters. Mol Aspects Med 2013; 34(2-3): 299-312. [http://dx.doi.org/10.1016/j.mam.2012.12.001]. [PMID: 23506872].
[35]
Speckmann B, Bidmon H-J, Borchardt A, Sies H, Steinbrenner H. Intestinal selenoprotein P in epithelial cells and in plasma cells. Arch Biochem Biophys 2014; 541: 30-6. [http://dx.doi.org/10.1016/j.abb.2013.10.011]. [PMID: 24157689].
[36]
Burk RF, Hill KE. Selenoprotein P-expression, functions, and roles in mammals. Biochim Biophys Acta 2009; 1790(11): 1441-7. [http://dx.doi.org/10.1016/j.bbagen.2009.03.026]. [PMID: 19345254].
[37]
Burk RF, Hill KE, Selenoprotein P. An extracellular protein with unique physical characteristics and a role in selenium homeostasis. Annu Rev Nutr 2005; 25: 215-35. [http://dx.doi.org/10.1146/annurev.nutr.24.012003.132120]. [PMID: 16011466].
[38]
Saito Y, Hayashi T, Tanaka A, et al. Selenoprotein P in human plasma as an extracellular phospholipid hydroperoxide glutathione peroxidase. Isolation and enzymatic characterization of human selenoprotein p. J Biol Chem 1999; 274(5): 2866-71. [http://dx.doi.org/10.1074/jbc.274.5.2866]. [PMID: 9915822].
[39]
Takebe G, Yarimizu J, Saito Y, et al. A comparative study on the hydroperoxide and thiol specificity of the glutathione peroxidase family and selenoprotein P. J Biol Chem 2002; 277(43): 41254-8. [http://dx.doi.org/10.1074/jbc.M202773200]. [PMID: 12185074].
[40]
Hill KE, Xia Y, Åkesson B, Boeglin ME, Burk RF. Selenoprotein P concentration in plasma is an index of selenium status in selenium-deficient and selenium-supplemented Chinese subjects. J Nutr 1996; 126(1): 138-45. [http://dx.doi.org/10.1093/jn/126.1.138]. [PMID: 8558294].
[41]
Duffield AJ, Thomson CD, Hill KE, Williams S. An estimation of selenium requirements for New Zealanders. Am J Clin Nutr 1999; 70(5): 896-903. [http://dx.doi.org/10.1093/ajcn/70.5.896]. [PMID: 10539752].
[42]
Hurst R, Armah CN, Dainty JR, et al. Establishing optimal selenium status: Results of a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 2010; 91(4): 923-31. [http://dx.doi.org/10.3945/ajcn.2009.28169]. [PMID: 20181815].
[43]
Xia Y, Hill KE, Byrne DW, Xu J, Burk RF. Effectiveness of selenium supplements in a low-selenium area of China. Am J Clin Nutr 2005; 81(4): 829-34. [http://dx.doi.org/10.1093/ajcn/81.4.829]. [PMID: 15817859].
[44]
Olson GE, Winfrey VP, Hill KE, Burk RF. Megalin mediates selenoprotein P uptake by kidney proximal tubule epithelial cells. J Biol Chem 2008; 283(11): 6854-60. [http://dx.doi.org/10.1074/jbc.M709945200]. [PMID: 18174160].
[45]
Olson GE, Winfrey VP, Nagdas SK, Hill KE, Burk RF. Apolipoprotein E receptor-2 (ApoER2) mediates selenium uptake from selenoprotein P by the mouse testis. J Biol Chem 2007; 282(16): 12290-7. [http://dx.doi.org/10.1074/jbc.M611403200]. [PMID: 17314095].
[46]
Saito Y, Sato N, Hirashima M, Takebe G, Nagasawa S, Takahashi K. Domain structure of bi-functional selenoprotein P. Biochem J 2004; 381(Pt 3): 841-6. [http://dx.doi.org/10.1042/BJ20040328]. [PMID: 15117283].
[47]
Kurokawa S, Hill KE, McDonald WH, Burk RF. 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). J Biol Chem 2012; 287(34): 28717-26. [http://dx.doi.org/10.1074/jbc.M112.383521]. [PMID: 22761431].
[48]
Arteel GE, Mostert V, Oubrahim H, Briviba K, Abel J, Sies H. Protection by selenoprotein P in human plasma against peroxynitrite-mediated oxidation and nitration. Biol Chem 1998; 379(8-9): 1201-5. [PMID: 9792455].
[49]
Traulsen H, Steinbrenner H, Buchczyk DP, Klotz LO, Sies H. Selenoprotein P protects low-density lipoprotein against oxidation. Free Radic Res 2004; 38(2): 123-8. [http://dx.doi.org/10.1080/10715760320001634852]. [PMID: 15104205].
[50]
Steinbrenner H, Bilgic E, Alili L, Sies H, Brenneisen P. Selenoprotein P protects endothelial cells from oxidative damage by stimulation of glutathione peroxidase expression and activity. Free Radic Res 2006; 40(9): 936-43. [http://dx.doi.org/10.1080/10715760600806248]. [PMID: 17015273].
[51]
Rea HM, Thomson CD, Campbell DR, Robinson MF. Relation between erythrocyte selenium concentrations and glutathione peroxidase (EC 1.11.1.9) activities of New Zealand residents and visitors to New Zealand. Br J Nutr 1979; 42(2): 201-8. [http://dx.doi.org/10.1079/BJN19790107]. [PMID: 113028].
[52]
Rotruck JT, Pope AL, Ganther HE, Hoekstra WG. Prevention of oxidative damage to rat erythrocytes by dietary selenium. J Nutr 1972; 102(5): 689-96. [http://dx.doi.org/10.1093/jn/102.5.689]. [PMID: 5022203].
[53]
Beckett GJ, Arthur JR. Selenium and endocrine systems. J Endocrinol 2005; 184(3): 455-65. [http://dx.doi.org/10.1677/joe.1.05971]. [PMID: 15749805].
[54]
Flohe L, Günzler WA, Schock HH. Glutathione peroxidase: A selenoenzyme. FEBS Lett 1973; 32(1): 132-4. [http://dx.doi.org/10.1016/0014-5793(73)80755-0]. [PMID: 4736708].
[55]
Grossmann A, Wendel A. Non-reactivity of the selenoenzyme glutathione peroxidase with enzymatically hydroperoxidized phospholipids. Eur J Biochem 1983; 135(3): 549-52. [http://dx.doi.org/10.1111/j.1432-1033.1983.tb07687.x]. [PMID: 6413205].
[56]
Takahashi K, Cohen HJ. Selenium-dependent glutathione peroxidase protein and activity: Immunological investigations on cellular and plasma enzymes. Blood 1986; 68(3): 640-5. [PMID: 3742048].
[57]
Tham DM, Whitin JC, Kim KK, Zhu SX, Cohen HJ. Expression of extracellular glutathione peroxidase in human and mouse gastrointestinal tract. Am J Physiol 1998; 275(6): G1463-71. [PMID: 9843785].
[58]
Speckmann B, Bidmon H-J, Pinto A, Anlauf M, Sies H, Steinbrenner H. Induction of glutathione peroxidase 4 expression during enterocytic cell differentiation. J Biol Chem 2011; 286(12): 10764-72. [http://dx.doi.org/10.1074/jbc.M110.216028]. [PMID: 21252226].
[59]
Kipp AP, Müller MF, Göken EM, Deubel S, Brigelius-Flohé R. The selenoproteins GPx2, TrxR2 and TrxR3 are regulated by Wnt signalling in the intestinal epithelium. Biochim Biophys Acta 2012; 1820(10): 1588-96. [http://dx.doi.org/10.1016/j.bbagen.2012.05.016]. [PMID: 22683372].
[60]
Florian S, Krehl S, Loewinger M, et al. Loss of GPx2 increases apoptosis, mitosis, and GPx1 expression in the intestine of mice. Free Radic Biol Med 2010; 49(11): 1694-702. [http://dx.doi.org/10.1016/j.freeradbiomed.2010.08.029]. [PMID: 20828612].
[61]
Weitzel F, Wendel A. Selenoenzymes regulate the activity of leukocyte 5-lipoxygenase via the peroxide tone. J Biol Chem 1993; 268(9): 6288-92. [PMID: 8454601].
[62]
Schnurr K, Belkner J, Ursini F, Schewe T, Kühn H. The selenoenzyme phospholipid hydroperoxide glutathione peroxidase controls the activity of the 15-lipoxygenase with complex substrates and preserves the specificity of the oxygenation products. J Biol Chem 1996; 271(9): 4653-8. [http://dx.doi.org/10.1074/jbc.271.9.4653]. [PMID: 8617728].
[63]
Imai H, Narashima K, Arai M, Sakamoto H, Chiba N, Nakagawa Y. Suppression of leukotriene formation in RBL-2H3 cells that overexpressed phospholipid hydroperoxide glutathione peroxidase. J Biol Chem 1998; 273(4): 1990-7. [http://dx.doi.org/10.1074/jbc.273.4.1990]. [PMID: 9442035].
[64]
Brigelius-Flohé R. Glutathione peroxidases and redox-regulated transcription factors. Biol Chem 2006; 387(10-11): 1329-35. [http://dx.doi.org/10.1515/BC.2006.166].
[65]
Yang K, Ma W, Liang H, Ouyang Q, Tang C, Lai L. Dynamic simulations on the arachidonic acid metabolic network. PLOS Comput Biol 2007; 3(3): e55-. [http://dx.doi.org/10.1371/journal.pcbi.0030055]. [PMID: 17381237].
[66]
Yang K, Bai H, Ouyang Q, Lai L, Tang C. Finding multiple target optimal intervention in disease-related molecular network. Mol Syst Biol 2008; 4: 228-8. [http://dx.doi.org/10.1038/msb.2008.60]. [PMID: 18985027].
[67]
Pei J, Yin N, Ma X, Lai L. Systems biology brings new dimensions for structure-based drug design. J Am Chem Soc 2014; 136(33): 11556-65. [http://dx.doi.org/10.1021/ja504810z]. [PMID: 25061983].
[68]
Yang WS. SriRamaratnam R, Welsch ME, et al Regulation of ferroptotic cancer cell death by GPX4. Cell 2014; 156(1-2): 317-31. [http://dx.doi.org/10.1016/j.cell.2013.12.010]. [PMID: 24439385].
[69]
Yang WS, Stockwell BR. Ferroptosis: Death by lipid peroxidation. Trends Cell Biol 2016; 26(3): 165-76. [http://dx.doi.org/10.1016/j.tcb.2015.10.014]. [PMID: 26653790].
[70]
Li C, Deng X, Xie X, Liu Y, Friedmann Angeli JP, Lai L. Activation of Glutathione Peroxidase 4 as a Novel Anti-inflammatory Strategy. Front Pharmacol 2018; 9: 1120-0. [http://dx.doi.org/10.3389/fphar.2018.01120]. [PMID: 30337875].
[71]
Meng H, Liu Y, Lai L. Diverse ways of perturbing the human arachidonic acid metabolic network to control inflammation. Acc Chem Res 2015; 48(8): 2242-50. [http://dx.doi.org/10.1021/acs.accounts.5b00226]. [PMID: 26237215].
[72]
Brigelius-Flohé R, Maiorino M. Glutathione peroxidases. Biochim Biophys Acta 2013; 1830: 3289-303.
[73]
Utomo A, Jiang X, Furuta S, et al. Identification of a novel putative non-selenocysteine containing phospholipid hydroperoxide glutathione peroxidase (NPGPx) essential for alleviating oxidative stress generated from polyunsaturated fatty acids in breast cancer cells. J Biol Chem 2004; 279(42): 43522-9. [http://dx.doi.org/10.1074/jbc.M407141200]. [PMID: 15294905].
[74]
Kryukov GV, Castellano S, Novoselov SV, et al. Characterization of mammalian selenoproteomes. Science 2003; 300(5624): 1439-43. [http://dx.doi.org/10.1126/science.1083516]. [PMID: 12775843].
[75]
Arnér ESJ. Focus on mammalian thioredoxin reductases--important selenoproteins with versatile functions. Biochim Biophys Acta 2009; 1790(6): 495-526. [http://dx.doi.org/10.1016/j.bbagen.2009.01.014]. [PMID: 19364476].
[76]
Damdimopoulos AE, Miranda-Vizuete A, Treuter E, Gustafsson JA, Spyrou G. An alternative splicing variant of the selenoprotein thioredoxin reductase is a modulator of estrogen signaling. J Biol Chem 2004; 279(37): 38721-9. [http://dx.doi.org/10.1074/jbc.M402753200]. [PMID: 15199063].
[77]
Damdimopoulou PEM-VA, Miranda-Vizuete A, Arnér ES, Gustafsson JA, Damdimopoulos AE. The human thioredoxin reductase-1 splice variant TXNRD1_v3 is an atypical inducer of cytoplasmic filaments and cell membrane filopodia. Biochim Biophys Acta 2009; 1793(10): 1588-96. [http://dx.doi.org/10.1016/j.bbamcr.2009.07.007]. [PMID: 19654027].
[78]
Cebula M, Moolla N, Capovilla A, Arnér ES. The rare TXNRD1_v3 (“v3”) splice variant of human thioredoxin reductase 1 protein is targeted to membrane rafts by N-acylation and induces filopodia independently of its redox active site integrity. J Biol Chem 2013; 288(14): 10002-11. [http://dx.doi.org/10.1074/jbc.M112.445932]. [PMID: 23413027].
[79]
Arnér ESJ, Holmgren A. Physiological functions of thioredoxin and thioredoxin reductase. Eur J Biochem 2000; 267(20): 6102-9. [http://dx.doi.org/10.1046/j.1432-1327.2000.01701.x]. [PMID: 11012661].
[80]
Sengupta R, Ryter SW, Zuckerbraun BS, Tzeng E, Billiar TR, Stoyanovsky DA. Thioredoxin catalyzes the denitrosation of low-molecular mass and protein S-nitrosothiols. Biochemistry 2007; 46(28): 8472-83. [http://dx.doi.org/10.1021/bi700449x]. [PMID: 17580965].
[81]
Benhar M, Forrester MT, Hess DT, Stamler JS. Regulated protein denitrosylation by cytosolic and mitochondrial thioredoxins. Science 2008; 320(5879): 1050-4. [http://dx.doi.org/10.1126/science.1158265]. [PMID: 18497292].
[82]
Holmgren A. Biochemistry. SNO removal. Science 2008; 320(5879): 1019-20. [http://dx.doi.org/10.1126/science.1159246]. [PMID: 18497281].
[83]
Sengupta R, Holmgren A. Thioredoxin and thioredoxin reductase in relation to reversible S-nitrosylation. Antioxid Redox Signal 2013; 18(3): 259-69. [http://dx.doi.org/10.1089/ars.2012.4716]. [PMID: 22702224].
[84]
Nikitovic D, Holmgren A. S-nitrosoglutathione is cleaved by the thioredoxin system with liberation of glutathione and redox regulating nitric oxide. J Biol Chem 1996; 271(32): 19180-5. [http://dx.doi.org/10.1074/jbc.271.32.19180]. [PMID: 8702596].
[85]
Belcastro E, Gaucher C, Corti A, Leroy P, Lartaud I, Pompella A. Regulation of protein function by S-nitrosation and S-glutathionylation: Processes and targets in cardiovascular pathophysiology. Biol Chem 2017; 398(12): 1267-93. [http://dx.doi.org/10.1515/hsz-2017-0150]. [PMID: 28822219].
[86]
Belcastro E, Wu W, Fries-Raeth I, et al. Oxidative stress enhances and modulates protein S-nitrosation in smooth muscle cells exposed to S-nitrosoglutathione. Nitric Oxide 2017; 69: 10-21. [http://dx.doi.org/10.1016/j.niox.2017.07.004]. [PMID: 28743484].
[87]
Choi H, Allahdadi KJ, Tostes RC, Webb RC. Augmented S-nitrosylation contributes to impaired relaxation in angiotensin II hypertensive mouse aorta: Role of thioredoxin reductase. J Hypertens 2011; 29(12): 2359-68. [http://dx.doi.org/10.1097/HJH.0b013e32834d2554]. [PMID: 22025239].
[88]
Wu W, Gaucher C, Diab R, et al. Time lasting S-nitrosoglutathione polymeric nanoparticles delay cellular protein S-nitrosation. Eur J Pharm Biopharm 2015; 89: 1-8. [http://dx.doi.org/10.1016/j.ejpb.2014.11.005]. [PMID: 25448077].
[89]
Takagi Y, Gon Y, Todaka T, et al. Expression of thioredoxin is enhanced in atherosclerotic plaques and during neointima formation in rat arteries. Lab Invest 1998; 78(8): 957-66.
[90]
Michalke B. Selenium speciation in human serum of cystic fibrosis patients compared to serum from healthy persons. J Chromatogr A 2004; 1058(1-2): 203-8. [http://dx.doi.org/10.1016/S0021-9673(04)01416-5]. [PMID: 15595669].
[91]
Benstoem C, Goetzenich A, Kraemer S, et al. Selenium and its supplementation in cardiovascular disease--what do we know? Nutrients 2015; 7(5): 3094-118. [http://dx.doi.org/10.3390/nu7053094]. [PMID: 25923656].
[92]
Zhang X, Liu C, Guo J, Song Y. Selenium status and cardiovascular diseases: Meta-analysis of prospective observational studies and randomized controlled trials. Eur J Clin Nutr 2016; 70(2): 162-9. [http://dx.doi.org/10.1038/ejcn.2015.78]. [PMID: 25990689].
[93]
Schomburg L, Schweizer U. Hierarchical regulation of selenoprotein expression and sex-specific effects of selenium. Biochim Biophys Acta 2009; 1790(11): 1453-62. [http://dx.doi.org/10.1016/j.bbagen.2009.03.015]. [PMID: 19328222].
[94]
Geerling BJ, Badart-Smook A, Stockbrügger RW, Brummer RJ. Comprehensive nutritional status in patients with long-standing Crohn disease currently in remission. Am J Clin Nutr 1998; 67(5): 919-26. [http://dx.doi.org/10.1093/ajcn/67.5.919]. [PMID: 9583850].
[95]
Geerling BJ, Badart-Smook A, Stockbrügger RW, Brummer RJ. Comprehensive nutritional status in recently diagnosed patients with inflammatory bowel disease compared with population controls. Eur J Clin Nutr 2000; 54(6): 514-21. [http://dx.doi.org/10.1038/sj.ejcn.1601049]. [PMID: 10878655].
[96]
Weisshof R, Chermesh I. Micronutrient deficiencies in inflammatory bowel disease. Curr Opin Clin Nutr Metab Care 2015; 18(6): 576-81. [http://dx.doi.org/10.1097/MCO.0000000000000226]. [PMID: 26418823].
[97]
Castro Aguilar-Tablada T, Navarro-Alarcón M, Quesada Granados J, Samaniego Sánchez C, Rufián-Henares JÁ, Nogueras-Lopez F. Ulcerative colitis and Crohn’s disease are associated with decreased serum selenium concentrations and increased cardiovascular Risk. Nutrients 2016; 8(12): 780. [http://dx.doi.org/10.3390/nu8120780]. [PMID: 27916926].
[98]
Ojuawo A, Keith L. The serum concentrations of zinc, copper and selenium in children with inflammatory bowel disease. Cent Afr J Med 2002; 48(9-10): 116-9. [PMID: 14562534].
[99]
Andoh A, Hirashima M, Maeda H, et al. Serum selenoprotein-P levels in patients with inflammatory bowel disease. Nutrition 2005; 21(5): 574-9. [http://dx.doi.org/10.1016/j.nut.2004.08.025]. [PMID: 15850963].
[100]
Hiller F, Besselt K, Deubel S, Brigelius-Flohé R, Kipp AP. GPx2 induction is mediated through STAT transcription factors during acute colitis. Inflamm Bowel Dis 2015; 21(9): 2078-89. [http://dx.doi.org/10.1097/MIB.0000000000000464]. [PMID: 26115075].
[101]
Barrett CW, Singh K, Motley AK, et al. Dietary selenium deficiency exacerbates DSS-induced epithelial injury and AOM/DSS-induced tumorigenesis. PLoS One 2013; 8(7): e67845-5. [http://dx.doi.org/10.1371/journal.pone.0067845]. [PMID: 23861820].
[102]
Tirosh O, Levy E, Reifen R. High selenium diet protects against TNBS-induced acute inflammation, mitochondrial dysfunction, and secondary necrosis in rat colon. Nutrition 2007; 23(11-12): 878-86. [http://dx.doi.org/10.1016/j.nut.2007.08.019]. [PMID: 17936198].
[103]
Kaur R, Thakur S, Rastogi P, Kaushal N. Resolution of Cox mediated inflammation by Se supplementation in mouse experimental model of colitis. PLoS One 2018; 13(7)e0201356 [http://dx.doi.org/10.1371/journal.pone.0201356]. [PMID: 30063735].
[104]
Hiller F, Oldorff L, Besselt K, Kipp AP. Differential acute effects of selenomethionine and sodium selenite on the severity of colitis. Nutrients 2015; 7(4): 2687-706. [http://dx.doi.org/10.3390/nu7042687]. [PMID: 25867950].
[105]
Zhu C, Zhang S, Song C, et al. Selenium nanoparticles decorated with Ulva lactuca polysaccharide potentially attenuate colitis by inhibiting NF-κB mediated hyper inflammation. J Nanobiotechnology 2017; 15(1): 20-0. [http://dx.doi.org/10.1186/s12951-017-0252-y]. [PMID: 28270147].
[106]
Firestein GS. Evolving concepts of rheumatoid arthritis. Nature 2003; 423(6937): 356-61. [http://dx.doi.org/10.1038/nature01661]. [PMID: 12748655].
[107]
Yu N, Han F, Lin X, Tang C, Ye J, Cai X. The association between serum selenium levels with rheumatoid arthritis. Biol Trace Elem Res 2016; 172(1): 46-52. [http://dx.doi.org/10.1007/s12011-015-0558-2]. [PMID: 26581918].
[108]
Maryam S, Zahra R, Mandana K. Selenium and autoimmune diseases: A review article. Curr Rheumatol Rev 2018; 14: 1-12.
[109]
Parnham MJ, Winkelmann J, Leyck S. Macrophage, lymphocyte and chronic inflammatory responses in selenium deficient rodents. Association with decreased glutathione peroxidase activity. Int J Immunopharmacol 1983; 5(5): 455-61. [http://dx.doi.org/10.1016/0192-0561(83)90022-X]. [PMID: 6654541].
[110]
Vieira AT, Silveira KD, Arruda MCC, et al. Treatment with Selemax®, a selenium-enriched yeast, ameliorates experimental arthritis in rats and mice. Br J Nutr 2012; 108(10): 1829-38. [http://dx.doi.org/10.1017/S0007114512000013]. [PMID: 22273003].
[111]
Peretz A, Siderova V, Nève J. Selenium supplementation in rheumatoid arthritis investigated in a double blind, placebo-controlled trial. Scand J Rheumatol 2001; 30(4): 208-12. [http://dx.doi.org/10.1080/030097401316909549]. [PMID: 11578015].
[112]
Deyab G, Hokstad I, Aaseth J, et al. Effect of anti-rheumatic treatment on selenium levels in inflammatory arthritis. J Trace Elem Med Biol 2018; 49: 91-7. [http://dx.doi.org/10.1016/j.jtemb.2018.05.001]. [PMID: 29895378].
[113]
Hercberg S, Galan P, Preziosi P, et al. The SU.VI.MAX Study: A randomized, placebo-controlled trial of the health effects of antioxidant vitamins and minerals. Arch Intern Med 2004; 164(21): 2335-42. [http://dx.doi.org/10.1001/archinte.164.21.2335]. [PMID: 15557412].
[114]
Assmann KE, Andreeva VA, Jeandel C, Hercberg S, Galan P, Kesse-Guyot E. Healthy aging 5 years after a period of daily supplementation with antioxidant nutrients: A post hoc analysis of the french randomized trial SU.VI.MAX. Am J Epidemiol 2015; 182(8): 694-704. [http://dx.doi.org/10.1093/aje/kwv105]. [PMID: 26374140].
[115]
Rayman MP, Stranges S, Griffin BA, Pastor-Barriuso R, Guallar E. Effect of supplementation with high-selenium yeast on plasma lipids: A randomized trial. Ann Intern Med 2011; 154(10): 656-65. [http://dx.doi.org/10.7326/0003-4819-154-10-201105170-00005]. [PMID: 21576533].
[116]
Sneddon AA, Wu H-C, Farquharson A, et al. Regulation of selenoprotein GPx4 expression and activity in human endothelial cells by fatty acids, cytokines and antioxidants. Atherosclerosis 2003; 171(1): 57-65. [http://dx.doi.org/10.1016/j.atherosclerosis.2003.08.008]. [PMID: 14642406].
[117]
Guo Z, Ran Q, Roberts LJ II, et al. Suppression of atherogenesis by overexpression of glutathione peroxidase-4 in apolipoprotein E-deficient mice. Free Radic Biol Med 2008; 44(3): 343-52. [http://dx.doi.org/10.1016/j.freeradbiomed.2007.09.009]. [PMID: 18215741].
[118]
Dabkowski ER, Williamson CL, Hollander JM. Mitochondria-specific transgenic overexpression of phospholipid hydroperoxide glutathione peroxidase (GPx4) attenuates ischemia/reperfusion-associated cardiac dysfunction. Free Radic Biol Med 2008; 45(6): 855-65. [http://dx.doi.org/10.1016/j.freeradbiomed.2008.06.021]. [PMID: 18638546].
[119]
Wingler K, Müller C, Schmehl K, Florian S, Brigelius-Flohé R. Gastrointestinal glutathione peroxidase prevents transport of lipid hydroperoxides in CaCo-2 cells. Gastroenterology 2000; 119(2): 420-30. [http://dx.doi.org/10.1053/gast.2000.9521]. [PMID: 10930377].
[120]
Poirier J, Cockell KA, Scoggan KA, et al. High-dose supplemental selenite to male Syrian hamsters fed hypercholesterolaemic diets alters Ldlr, Abcg8 and Npc1l1 mRNA expression and lowers plasma cholesterol concentrations. Br J Nutr 2012; 108(2): 257-66. [http://dx.doi.org/10.1017/S0007114511005587]. [PMID: 22152646].
[121]
Bleys J, Navas-Acien A, Guallar E. Serum selenium and diabetes in U.S. adults. Diabetes Care 2007; 30(4): 829-34. [http://dx.doi.org/10.2337/dc06-1726]. [PMID: 17392543].
[122]
Agrahari V, Agrahari V. Facilitating the translation of nanomedicines to a clinical product: Challenges and opportunities. Drug Discov Today 2018; 23(5): 974-91. [http://dx.doi.org/10.1016/j.drudis.2018.01.047]. [PMID: 29406263].
[123]
Lin Z, Li Y, Gong G, et al. Restriction of H1N1 influenza virus infection by selenium nanoparticles loaded with ribavirin via resisting caspase-3 apoptotic pathway. Int J Nanomedicine 2018; 13: 5787-97. [http://dx.doi.org/10.2147/IJN.S177658]. [PMID: 30310281].
[124]
Zhao S, Yu Q, Pan J, et al. Redox-responsive mesoporous selenium delivery of doxorubicin targets MCF-7 cells and synergistically enhances its anti-tumor activity. Acta Biomater 2017; 54: 294-306. [http://dx.doi.org/10.1016/j.actbio.2017.02.042]. [PMID: 28267598].
[125]
Zhang J, Zhou X, Yu Q, et al. Epigallocatechin-3-gallate (EGCG)-stabilized selenium nanoparticles coated with Tet-1 peptide to reduce amyloid-β aggregation and cytotoxicity. ACS Appl Mater Interfaces 2014; 6(11): 8475-87. [http://dx.doi.org/10.1021/am501341u]. [PMID: 24758520].
[126]
Huang X, Chen X, Chen Q, Yu Q, Sun D, Liu J. Investigation of functional selenium nanoparticles as potent antimicrobial agents against superbugs. Acta Biomater 2016; 30: 397-407. [http://dx.doi.org/10.1016/j.actbio.2015.10.041]. [PMID: 26518106].
[127]
Malhotra S, Welling MN, Mantri SB, Desai K. In vitro and in vivo antioxidant, cytotoxic, and anti-chronic inflammatory arthritic effect of selenium nanoparticles. J Biomed Mater Res B Appl Biomater 2016; 104(5): 993-1003. [http://dx.doi.org/10.1002/jbm.b.33448]. [PMID: 25994972].
[128]
Tan VlC, Hinchman A, Williams R. Nanostructured biomedical selenium at the biological interface (Review). Biointerphases 2018; 13(6): 06D301.
[129]
Comasseto JV. Selenium and tellurium chemistry: Historical background. J Braz Chem Soc 2010; 21: 2027-31. [http://dx.doi.org/10.1590/S0103-50532010001100003].
[130]
Cantor AH, Scott ML, Noguchi T. Biological Availability of Selenium in Feedstuffs and Selenium Compounds for Prevention of Exudative Diathesis in Chicks. J Nutr 1975; 105: 96-105. [http://dx.doi.org/10.1093/jn/105.1.96].
[131]
Seebach D, Peleties N. Mono-, Bis-, and Tris(phenylseleno) methyllithium (Selenium-Stabilized Carbanions). Angew Chem Int Ed Engl 1969; 8: 450-1. [http://dx.doi.org/10.1002/anie.196904502].
[132]
Sharpless KB, Lauer RF. Selenium dioxide oxidation of olefins. Evidence for the intermediacy of allylseleninic acids. J Am Chem Soc 1972; 94: 7154-5. [http://dx.doi.org/10.1021/ja00775a050].
[133]
Klayman DLG W H. H Organic selenium compounds: Their chemistry and biology 1973.
[134]
Z R The Chemistry of Organic Selenium and Tellurium Compounds 2014.
[135]
Schulz S. Covalently Bonded Compounds of Heavy Group 15/16 Elements – Synthesis, structure and potential application in material sciences. Coord Chem Rev 2015; 297-298: 49-76.
[136]
Malik MA, Afzaal M, O’Brien P. Precursor chemistry for main group elements in semiconducting materials. Chem Rev 2010; 110(7): 4417-46. [http://dx.doi.org/10.1021/cr900406f]. [PMID: 20481563].
[137]
Bochmann M. Metal Chalcogenide Materials: Chalcogenolato complexes as “single-source” precursors. Chem Vap Depos 1996; 2: 85-96. [http://dx.doi.org/10.1002/cvde.19960020302].
[138]
Tamba BI A-ST. Trace Elements Alleviate Pain in Mice and Humans 2017.
[http://dx.doi.org/10.1016/B978-0-12-805186-3.00017-5]
[139]
Reilly C. Selenium in Food and Health 1997.
[140]
Ihnat M. Occurrence and distribution of selenium 1989; 33431.
[141]
Soriano-Garcia M. Organoselenium compounds as potential therapeutic and chemopreventive agents: a review. Curr Med Chem 2004; 11(12): 1657-69. [http://dx.doi.org/10.2174/0929867043365053]. [PMID: 15180570].
[142]
Yu SC, Borchert A, Kuhn H, Ivanov I. Synthesis of a new seleninic acid anhydride and mechanistic studies into its glutathione peroxidase activity. Chemistry 2008; 14(23): 7066-71. [http://dx.doi.org/10.1002/chem.200800694]. [PMID: 18604859].
[143]
Back TG, Moussa Z. Remarkable activity of a novel cyclic seleninate ester as a glutathione peroxidase mimetic and its facile in situ generation from allyl 3-hydroxypropyl selenide. J Am Chem Soc 2002; 124(41): 12104-5. [http://dx.doi.org/10.1021/ja028030k]. [PMID: 12371844].
[144]
Manna D, Mugesh G. A chemical model for the inner-ring deiodination of thyroxine by iodothyronine deiodinase. Angew Chem Int Ed Engl 2010; 49(48): 9246-9. [http://dx.doi.org/10.1002/anie.201005235]. [PMID: 20941724].
[145]
Radhakrishna PM, Sharada KC, Vagdevi HM, et al. Synthesis and antibacterial activity of novel organoselenium compounds. Int J Chem 2010; 2(2)
[146]
Chagas PM, Rosa SG, Sari MHM, et al. Evaluation of the pharmacological properties of salicylic acid-derivative organoselenium: 2-hydroxy-5-selenocyanatobenzoic acid as an anti-inflammatory and antinociceptive compound. Pharmacol Biochem Behav 2014; 118: 87-95. [http://dx.doi.org/10.1016/j.pbb.2013.12.022]. [PMID: 24398148].
[147]
Sarma BK, Manna D, Minoura M, Mugesh G. Synthesis, structure, spirocyclization mechanism, and glutathione peroxidase-like antioxidant activity of stable spirodiazaselenurane and spirodiazatellurane. J Am Chem Soc 2010; 132(15): 5364-74. [http://dx.doi.org/10.1021/ja908080u]. [PMID: 20345146].
[148]
Lamani DS, Bhowmick D, Mugesh G. Spirodiazaselenuranes: synthesis, structure and antioxidant activity. Org Biomol Chem 2012; 10(39): 7933-43. [http://dx.doi.org/10.1039/c2ob26156a]. [PMID: 22932965].
[149]
Parashiva Prabhu C, Phadnis PP, Wadawale AP, et al. Synthesis, characterization, structures and antioxidant activity of nicotinoyl based organoselenium compounds. J Organomet Chem 2012; 713: 42-50. [http://dx.doi.org/10.1016/j.jorganchem.2012.04.014].
[150]
Shiva K. Effects of a Garlic Active Based Growth Promoter on Growth Performance and Specific Pathogenic Intestinal Microbial Counts of Broiler Chicks. Int J Poult Sci 2010.
[151]
Kumar S, Sharma N, Maurya IK, et al. Facile synthesis, structural evaluation, antimicrobial activity and synergistic effects of novel imidazo[1,2-a]pyridine based organoselenium compounds. Eur J Med Chem 2016; 123: 916-24. [http://dx.doi.org/10.1016/j.ejmech.2016.07.076]. [PMID: 27565415].
[152]
Park Y-J, Koketsu M, Kim JM, et al. 1,3-selenazol-4-one derivatives inhibit inducible nitric oxide-mediated nitric oxide production in lipopolysaccharide-induced BV-2 cells. Biol Pharm Bull 2003; 26(12): 1657-60. [http://dx.doi.org/10.1248/bpb.26.1657]. [PMID: 14646166].
[153]
Oliveira J, Straliotto MR, Mancini G, et al. Atheroprotective action of a modified organoselenium compound: In vitro evidence. An Acad Bras Cienc 2016; 88(3)(Suppl.): 1953-65. [http://dx.doi.org/10.1590/0001-3765201620150760]. [PMID: 27737337].

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy